Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116.764
Filter
Add more filters








Publication year range
1.
Cell Commun Signal ; 22(1): 386, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090602

ABSTRACT

BACKGROUND: T-LAK cell-oriented protein kinase (TOPK) strongly promotes the malignant proliferation of cancer cells and is recognized as a promising biomarker of tumor progression. Psoriasis is a common inflammatory skin disease featured by excessive proliferation of keratinocytes. Although we have previously reported that topically inhibiting TOPK suppressed psoriatic manifestations in psoriasis-like model mice, the exact role of TOPK in psoriatic inflammation and the underlying mechanism remains elusive. METHODS: GEO datasets were analyzed to investigate the association of TOPK with psoriasis. Skin immunohistochemical (IHC) staining was performed to clarify the major cells expressing TOPK. TOPK conditional knockout (cko) mice were used to investigate the role of TOPK-specific deletion in IMQ-induced psoriasis-like dermatitis in mice. Flow cytometry was used to analyze the alteration of psoriasis-related immune cells in the lesional skin. Next, the M5-induced psoriasis cell model was used to identify the potential mechanism by RNA-seq, RT-RCR, and western blotting. Finally, the neutrophil-neutralizing antibody was used to confirm the relationship between TOPK and neutrophils in psoriasis-like dermatitis in mice. RESULTS: We found that TOPK levels were strongly associated with the progression of psoriasis. TOPK was predominantly increased in the epidermal keratinocytes of psoriatic lesions, and conditional knockout of TOPK in keratinocytes suppressed neutrophils infiltration and attenuated psoriatic inflammation. Neutrophils deletion by neutralizing antibody greatly diminished the suppressive effect of TOPK cko in psoriasis-like dermatitis in mice. In addition, topical application of TOPK inhibitor OTS514 effectively attenuated already-established psoriasis-like dermatitis in mice. Mechanismly, RNA-seq revealed that TOPK regulated the expression of some genes in the IL-17 signaling pathway, such as neutrophils chemokines CXCL1, CXCL2, and CXCL8. TOPK modulated the expression of neutrophils chemokines via activating transcription factors STAT3 and NF-κB p65 in keratinocytes, thereby promoting neutrophils infiltration and psoriasis progression. CONCLUSIONS: This study identified a crucial role of TOPK in psoriasis by regulating neutrophils infiltration, providing new insights into the pathogenesis of psoriasis.


Subject(s)
Keratinocytes , Mitogen-Activated Protein Kinase Kinases , Neutrophil Infiltration , Psoriasis , Animals , Humans , Mice , Disease Models, Animal , Disease Progression , Imiquimod , Keratinocytes/pathology , Keratinocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Neutrophils/pathology , Psoriasis/pathology , Psoriasis/genetics , Signal Transduction , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Up-Regulation , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism
2.
Sci Rep ; 14(1): 17926, 2024 08 02.
Article in English | MEDLINE | ID: mdl-39095643

ABSTRACT

Colorectal cancer (CRC) is a complex malignancy with poorly understood molecular mechanisms, necessitating the identification of genetic markers. Although Ubiquitin domain-containing protein 1 (UBTD1) has received significant attention in the study of human cancers, its specific role in CRC is yet to be fully clarified. This study sought to examine how UBTD1 expression was associated with various clinical and pathological characteristics of CRC, and to determine its prognostic significance and biological function, utilizing data from clinical samples and large-scale databases. Notably, UBTD1 expression was found to be upregulated in CRC, resulting in decreased survival rates and unfavorable clinical characteristics such as advanced T, N, and pathological stages. The findings of the multivariate Cox regression analysis illustrated that UBTD1 expression upregulation is a significant independent marker of unfavorable outcomes in CRC patients. An examination of the functional enrichment of UBTD1 and the genes it co-expresses indicated that it could serve as an oncogene by modulating the expression of genes implicated in crucial tumorigenesis pathways and functions. Additionally, immune cell infiltration analysis suggested a link between UBTD1 levels and various immune cells, particularly macrophages. In conclusion, the use of UBTD1 as a biomarker for both the prognosis and diagnosis of CRC has promising prospects for further investigation and therapeutic approaches.


Subject(s)
Biomarkers, Tumor , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Prognosis , Male , Female , Middle Aged , Up-Regulation
3.
J Immunol Res ; 2024: 9512251, 2024.
Article in English | MEDLINE | ID: mdl-39108258

ABSTRACT

Macrophage alternative activation is involved in kidney fibrosis. Previous researches have documented that the transcriptional regulators Yes-associated protein (Yap)/transcriptional coactivator with PDZ-binding motif (Taz) are linked to organ fibrosis. However, limited knowledge exists regarding the function and mechanisms of their downstream molecules in regulating macrophage activation and kidney fibrosis. In this paper, we observed that the Hippo pathway was suppressed in macrophages derived from fibrotic kidneys in mice. Knockout of Taz or Tead1 in macrophages inhibited the alternative activation of macrophages and reduced kidney fibrosis. Additionally, by using bone marrow-derived macrophages (BMDMs), we investigated that knockout of Taz or Tead1 in macrophages impeded both cell proliferation and migration. Moreover, deletion of Tead1 reduces p-Smad3 and Smad3 abundance in macrophages. And chromatin immunoprecipitation (ChIP) assays showed that Tead1 could directly bind to the promoter region of Smad3. Collectively, these results indicate that Tead1 knockout in macrophages could reduce TGFß1-induced phosphorylation Smad3 via transcriptional downregulation of Smad3, thus suppressing macrophage alternative activation and IRI-induced kidney fibrosis.


Subject(s)
DNA-Binding Proteins , Fibrosis , Macrophage Activation , Macrophages , Mice, Knockout , Smad3 Protein , TEA Domain Transcription Factors , Transcription Factors , Animals , Smad3 Protein/metabolism , Smad3 Protein/genetics , Mice , Macrophages/immunology , Macrophages/metabolism , Macrophage Activation/genetics , Macrophage Activation/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Kidney/pathology , Kidney/metabolism , Signal Transduction , Up-Regulation , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/metabolism , Kidney Diseases/immunology , Hippo Signaling Pathway , Disease Models, Animal , Transforming Growth Factor beta1/metabolism , Mice, Inbred C57BL , Male , Phosphorylation , Cell Proliferation , Acyltransferases
4.
J Cardiovasc Pharmacol ; 84(2): 239-249, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39115722

ABSTRACT

ABSTRACT: The study aimed to investigate the pathogenesis of sepsis-induced cardiomyopathy, a leading cause of mortality in septic patients. Transcriptome data from cecal ligation and puncture-induced septic mice were analyzed at different time points (24, 48, and 72 hours) using GSE171546 data. Through weighted gene co-expression network analysis, time series, and differential expression analyses, key time-series differentially expressed genes were identified. In addition, single-cell sequencing data (GSE207363) were used for both differential and pseudotime analyses to pinpoint differentially expressed genes specific to endothelial cells. The study highlighted Spock2, S100a9, S100a8, and Xdh as differential genes specific to endothelial cells in a time-dependent manner. Immunofluorescence validation confirmed the increased expression of SPOCK2 in the endothelial cells of cecal ligation and puncture-induced septic mice. Furthermore, in vitrostudies showed that deletion of Spock2 significantly increased LPS-induced apoptosis and necrosis in human umbilical vein endothelial cells. In conclusion, SPOCK2 expression was increased in septic cardiac endothelial cells and LPS-induced human umbilical vein endothelial cells and may play a protective role.


Subject(s)
Apoptosis , Cardiomyopathies , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Mice, Inbred C57BL , Sepsis , Animals , Sepsis/metabolism , Sepsis/genetics , Sepsis/complications , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Male , Time Factors , Transcriptome , Cells, Cultured , Mice, Knockout , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Regulatory Networks , Necrosis , Databases, Genetic , Signal Transduction , Gene Expression Profiling , Gene Expression Regulation , Lipopolysaccharides/pharmacology , Up-Regulation , Single-Cell Analysis , Mice , Calgranulin B
5.
Nat Commun ; 15(1): 6525, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117630

ABSTRACT

Reactive astrocytes play a pivotal role in the pathogenesis of neurological diseases; however, their functional phenotype and the downstream molecules by which they modify disease pathogenesis remain unclear. Here, we genetically increase P2Y1 receptor (P2Y1R) expression, which is upregulated in reactive astrocytes in several neurological diseases, in astrocytes of male mice to explore its function and the downstream molecule. This astrocyte-specific P2Y1R overexpression causes neuronal hyperexcitability by increasing both astrocytic and neuronal Ca2+ signals. We identify insulin-like growth factor-binding protein 2 (IGFBP2) as a downstream molecule of P2Y1R in astrocytes; IGFBP2 acts as an excitatory signal to cause neuronal excitation. In neurological disease models of epilepsy and stroke, reactive astrocytes upregulate P2Y1R and increase IGFBP2. The present findings identify a mechanism underlying astrocyte-driven neuronal hyperexcitability, which is likely to be shared by several neurological disorders, providing insights that might be relevant for intervention in diverse neurological disorders.


Subject(s)
Astrocytes , Insulin-Like Growth Factor Binding Protein 2 , Neurons , Receptors, Purinergic P2Y1 , Up-Regulation , Animals , Astrocytes/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , Neurons/metabolism , Male , Mice , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y1/genetics , Mice, Transgenic , Epilepsy/metabolism , Epilepsy/genetics , Epilepsy/physiopathology , Mice, Inbred C57BL , Humans , Calcium Signaling , Disease Models, Animal
6.
Cell Death Dis ; 15(8): 574, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117638

ABSTRACT

Necrosome activation following TLR- or cytokine receptor-signaling results in cell death by necroptosis which is characterized by the rupture of cell membranes and the consequent release of intracellular contents to the extracellular milieu. While necroptosis exacerbates various inflammatory diseases, the mechanisms through which the inflammatory responses are regulated are not clear. We show that the necrosome activation of macrophages results in an upregulation of various pathways, including the mitogen-activated protein kinase (MAPK) cascade, which results in an elevation of the inflammatory response and consequent expression of several cytokines and chemokines. Programming for this upregulation of inflammatory response occurs during the early phase of necrosome activation and proceeds independently of cell death but depends on the activation of the receptor-interacting protein kinase-1 (RipK1). Interestingly, necrosome activation also results in an upregulation of IFNß, which in turn exerts an inhibitory effect on the maintenance of inflammatory response through the repression of MAPK-signaling and an upregulation of Zfp36. Activation of the interferon-induced gene factor-3 (ISGF3) results in the expression of ZFP36 (TTP), which induces the post-transcriptional degradation of mRNAs of various inflammatory cytokines and chemokines through the recognition of AU-rich elements in their 3'UTR. Furthermore, ZFP-36 inhibits IFNß-, but not TNFα- induced necroptosis. Overall, these results reveal the molecular mechanism through which IFNß, a pro-inflammatory cytokine, induces the expression of ZFP-36, which in turn inhibits necroptosis and halts the maintenance of the inflammatory response.


Subject(s)
Cytokines , Intracellular Signaling Peptides and Proteins , Necroptosis , Protein Serine-Threonine Kinases , Tristetraprolin , Tristetraprolin/metabolism , Tristetraprolin/genetics , Animals , Cytokines/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Macrophages/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , RAW 264.7 Cells , Up-Regulation/drug effects , Interferon-beta/metabolism , Mice, Inbred C57BL , Gene Expression Regulation
7.
Cell Death Dis ; 15(8): 578, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117671

ABSTRACT

Approximately 70% of treatment failures in nasopharyngeal carcinoma (NPC) patients are attributed to distant metastasis, yet the underlying mechanisms remain unclear. RNA 5-methylcytosine (m5C) is an emerging regulatory modification that controls gene expression and plays a critical role in tumor progression. However, there is little information on the potential roles of RNA m5C modification in NPC metastasis. In this study, we found that the m5C reader Aly/REF export factor (ALYREF) is significantly upregulated in NPC, whereby its high expression is associated with metastasis and poor prognosis. ALYREF overexpression was found to promote tumor metastasis of NPC cells in vitro and in vivo. Mechanistically, m5C-modified NOTCH1 mRNA was identified as a target of ALYREF. Moreover, ALYREF was found to upregulate NOTCH1 expression by enhancing its RNA stability in an m5C modification-dependent manner, thereby promoting the activation of the NOTCH signaling pathway and facilitating NPC metastasis. Overall, our data reveal the crucial role of ALYREF in NPC metastasis and provide a potential therapeutic target for NPC.


Subject(s)
Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Neoplasm Metastasis , RNA Stability , RNA, Messenger , Receptor, Notch1 , Humans , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/metabolism , Cell Line, Tumor , RNA, Messenger/metabolism , RNA, Messenger/genetics , Animals , Gene Expression Regulation, Neoplastic , Mice, Nude , Male , Female , Mice , Signal Transduction , Mice, Inbred BALB C , Up-Regulation/genetics , Carcinoma/metabolism , Carcinoma/genetics , Carcinoma/pathology , Middle Aged
8.
BMC Cardiovasc Disord ; 24(1): 411, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118024

ABSTRACT

AIMS: To assess the correlation between high-sensitivity C-reactive protein (Hs-CRP) and the prevalence of cardiovascular disease (CVD) among individuals with diabetes. METHODS: A total of 1,555 participants from the National Health and Nutrition Examination Survey were enrolled in this cross-sectional study after excluding individuals without diabetes and those who lacked data on Hs-CRP, diabetes and CVD. All participants were divided into four groups based on quartiles of Hs-CRP: Q1 (≤ 1.20 mg/L), Q2 (1.20-2.86 mg/L), Q3 (2.86-6.40 mg/L), and Q4 (> 6.40 mg/L). Logistic regression analysis, subgroup analysis and restricted cubic spline (RCS) analysis were used to evaluate the correlation between Hs-CRP and the prevalence of CVD in individuals with diabetes. RESULTS: In univariate logistic regression analysis, a higher level of Hs-CRP was associated with a higher prevalence of CVD (P < 0.05). In the multivariate logistic regression analysis adjusting for confounders, the correlation between Hs-CRP and the prevalence of CVD remained significant (Q3 vs. Q1, OR: 1.505, 95% CI: 1.056-2.147, P = 0.024; Q4 vs. Q1, OR: 1.711, 95% CI: 1.171-2.499, P = 0.006; log10(Hs-CRP), OR: 1.504, 95% CI: 1.168-1.935, P = 0.002). Further subgroup analysis showed that a higher Hs-CRP was independently associated with a higher prevalence of CVD in the < 60 years, male, non-hypertension and non-hypercholesterolemia subgroups (P < 0.05). Additionally, RCS analysis revealed a linear positive correlation between Hs-CRP and CVD prevalence (P for nonlinearity = 0.244). CONCLUSION: A higher level of Hs-CRP was closely related to a higher prevalence of CVD in people with diabetes.


Subject(s)
Biomarkers , C-Reactive Protein , Cardiovascular Diseases , Diabetes Mellitus , Nutrition Surveys , Humans , Male , C-Reactive Protein/analysis , C-Reactive Protein/metabolism , Middle Aged , Female , Cross-Sectional Studies , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Prevalence , Diabetes Mellitus/epidemiology , Diabetes Mellitus/blood , Diabetes Mellitus/diagnosis , Biomarkers/blood , Adult , Aged , Risk Factors , Linear Models , Multivariate Analysis , Logistic Models , Up-Regulation , Odds Ratio , Risk Assessment
9.
Cells ; 13(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39120269

ABSTRACT

Mast cells are the major effector cells that mediate IgE-dependent allergic reactions. We sought to use integrated network analysis to identify genomic biomarkers associated with high response in IgE-mediated activation of primary human mast cells. Primary human mast cell cultures derived from 262 normal donors were categorized into High, Average and Low responder groups according to their activation response profiles. Transcriptome analysis was used to identify genes that were differentially expressed in different responder cultures in their baseline conditions, and the data were analyzed by constructing a personalized perturbed profile (PEEP). For upregulated genes, the construction of PEEP for each individual sample of all three responder groups revealed that High responders exhibited a higher percentage of "perturbed" samples whose PEEP values lay outside the normal range of expression. Moreover, the integration of PEEP of four selected upregulated genes into distinct sets of combinatorial profiles demonstrated that the specific pattern of upregulated expression of these four genes, in a tandem combination, was observed exclusively among the High responders. In conclusion, this combinatorial approach was useful in identifying a set of genomic biomarkers that are associated with high degranulation response in human mast cell cultures derived from the blood of a cohort of normal donors.


Subject(s)
Biomarkers , Cell Degranulation , Immunoglobulin E , Mast Cells , Humans , Mast Cells/metabolism , Immunoglobulin E/metabolism , Biomarkers/metabolism , Gene Expression Profiling , Genomics , Transcriptome/genetics , Up-Regulation/genetics , Cells, Cultured
10.
Turk J Gastroenterol ; 35(6): 453-464, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-39114901

ABSTRACT

The pathogenesis mechanism of acute gastric mucosal lesions (AGML) is still unclear; further exploration is urgently needed to find a new therapeutic target. This study aimed to investigate whether morphine might regulate the expression and function of transient receptor potential ankyrin 1 (TRPA1) through a cyclic adenosine monophosphate/protein kinase A (cAMP/PKA)-dependent pathway, thereby alleviating gastric mucosal lesions caused by water-immersion restraint stress (WIRS). Rats were administered with intrathecal morphine, TRPA1 antagonist (HC-030031), µ-opioid receptor antagonist, or protein kinase A inhibitor (H-89), respectively, before WIRS. After 6 hours of WIRS, microscopic lesions, hematoxylin and eosin staining, and transmission electron microscopy were applied to assess the damage of the gastric mucosa. Real-time polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay were conducted to detect the levels of TRPA1 and substance P (SP) in the dorsal root ganglia (DRG) and gastric tissues. In addition, immunofluorescence was used to explore the possible co-expression of TRPA1 and µ-opioid receptors in the DRG. The results indicated that WIRS upregulated TRPA1 and SP in gastric mucosa, and HC-030031 or H-89 could alleviate gastric mucosal lesions caused by WIRS (P < .0001). Morphine was found to suppress both WIRS-induced gastric mucosal lesions (P < .0001) and the upregulation of TRPA1 (P = .0086) and SP (P = .0013). Both TRPA1 and SP play important roles in the pathogenesis of WIRS-induced AGML. Exogenous gastroprotective strategies reduce elevated levels of TRPA1 via the cAMP/PKA-dependent pathway. Inhibition of TRPA1 upregulation in the DRG is critical for intrathecal morphine preconditioning-induced gastric protection.


Subject(s)
Ganglia, Spinal , Gastric Mucosa , Isoquinolines , Morphine , Rats, Sprague-Dawley , Restraint, Physical , TRPA1 Cation Channel , Up-Regulation , Animals , Morphine/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Up-Regulation/drug effects , TRPA1 Cation Channel/metabolism , Male , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Restraint, Physical/adverse effects , Rats , Isoquinolines/pharmacology , Acetanilides/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Purines/pharmacology , Stress, Psychological/complications , Immersion , Receptors, Opioid, mu/metabolism , Cyclic AMP/metabolism , Sulfonamides
11.
Virol J ; 21(1): 197, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182136

ABSTRACT

Serine/threonine kinase receptor-associated protein (STRAP) serves as a scaffold protein and is engaged in a variety of cellular activities, although its importance in antiviral innate immunity is unknown. We discovered that STRAP works as an interferon (IFN)-inducible positive regulator, facilitating type I IFN signaling during pseudorabies virus infection. Mechanistically, STRAP interacts with TBK1 to activate type I IFN signaling. Both the CT and WD40 7 - 6 domains contribute to the function of STRAP. Furthermore, TBK1 competes with PRV-UL50 for binding to STRAP, and STRAP impedes the degradation of TBK1 mediated by PRV-UL50, thereby increasing the interaction between STRAP and TBK1. Overall, these findings reveal a previously unrecognized role for STRAP in innate antiviral immune responses during PRV infection. STRAP could be a potential therapeutic target for viral infectious diseases.


Subject(s)
Herpesvirus 1, Suid , Immunity, Innate , Interferon Type I , Protein Serine-Threonine Kinases , Animals , Cell Line , Herpesvirus 1, Suid/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Pseudorabies/immunology , Pseudorabies/virology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Signal Transduction , Up-Regulation
12.
J Orthop Surg Res ; 19(1): 511, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39192334

ABSTRACT

OBJECTIVE: Delayed fracture healing is a common complication of fractures that significantly impacts human health. This study aimed to explore the role of LINC00339 (lncRNA) in delayed fracture healing to provide new directions for its treatment. METHODS: This study included 82 patients with fractures healing in a normal manner and 90 patients experiencing delayed fracture healing. Levels of LINC00339, miR-16-5p, and osteogenic marker-related mRNAs were measured using RT-qPCR. The predictive potential of LINC00339 for delayed fracture healing was validated using ROC curve analysis. The interaction between LINC00339 and miR-16-5p was validated using dual-luciferase reporter assays and RIP experiments. CCK-8 was used to assess cell proliferation, and apoptosis rates were measured by flow cytometry. RESULTS: LINC00339 was significantly upregulated in delayed fracture healing patients and exhibited strong predictive ability for this condition. Overexpression of LINC00339 inhibited osteoblast proliferation, promoted apoptosis, and reduced mRNA levels of osteogenic markers (P < 0.05). miR-16-5p was recognized as a target mRNA of LINC00339, with LINC00339 exerting negative regulation on miR-16-5p, while overexpression of miR-16-5p mitigated the inhibitory effects of LINC00339 on fracture healing (P < 0.05). CONCLUSION: This research indicated that LINC00339 may serve as a diagnostic marker for delayed fracture healing and revealed the function of the LINC00339/miR-16-5p axis on fracture healing by regulating osteoblasts.


Subject(s)
Apoptosis , Cell Proliferation , Fracture Healing , MicroRNAs , RNA, Long Noncoding , Fracture Healing/genetics , Fracture Healing/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , MicroRNAs/genetics , Cell Proliferation/genetics , Apoptosis/genetics , Male , Female , Osteoblasts/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Middle Aged , Adult , Up-Regulation , Cells, Cultured
13.
Nat Commun ; 15(1): 6549, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095385

ABSTRACT

The placenta is crucial for fetal development, yet the impact of environmental stressors such as arsenic exposure remains poorly understood. We apply single-cell RNA sequencing to analyze the response of the mouse placenta to arsenic, revealing cell-type-specific gene expression, function, and pathological changes. Notably, the Prap1 gene, which encodes proline-rich acidic protein 1 (PRAP1), is significantly upregulated in 26 placental cell types including various trophoblast cells. Our study shows a female-biased increase in PRAP1 in response to arsenic and localizes it in the placenta. In vitro and ex vivo experiments confirm PRAP1 upregulation following arsenic treatment and demonstrate that recombinant PRAP1 protein reduces arsenic-induced cytotoxicity and downregulates cell cycle pathways in human trophoblast cells. Moreover, PRAP1 knockdown differentially affects cell cycle processes, proliferation, and cell death depending on the presence of arsenic. Our findings provide insights into the placental response to environmental stress, offering potential preventative and therapeutic approaches for environment-related adverse outcomes in mothers and children.


Subject(s)
Arsenic , Placenta , Single-Cell Analysis , Trophoblasts , Female , Pregnancy , Placenta/metabolism , Placenta/drug effects , Animals , Humans , Mice , Trophoblasts/metabolism , Trophoblasts/drug effects , Trophoblasts/cytology , Arsenic/toxicity , Sequence Analysis, RNA , Stress, Physiological/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Up-Regulation/drug effects , Mice, Inbred C57BL
14.
Cells ; 13(16)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39195242

ABSTRACT

CKLF-like MARVEL transmembrane domain-containing 3 (CMTM3), a member of the CMTM family that is closely related to tumor occurrence and progression, plays crucial roles in the immune system, cardiovascular system, and male reproductive system. Recently, CMTM3 has emerged as a potential target for treating diseases related to bone formation. However, additional studies are needed to understand the mechanisms by which CMTM3 regulates the process of osteogenic differentiation. In this study, we observed a significant downregulation of Cmtm3 expression during the transdifferentiation of C2C12 myoblasts into osteoblasts induced by BMP4. Cmtm3 overexpression suppressed proliferation and osteogenic differentiation in BMP4-induced C2C12 cells, whereas its knockdown conversely facilitated the process. Mechanistically, Cmtm3 overexpression upregulated both the protein and mRNA levels of p53 and p21. Conversely, Cmtm3 knockdown exerted the opposite effects. Additionally, we found that Cmtm3 interacts with p53 and increases protein stability by inhibiting proteasome-mediated ubiquitination and degradation. Notably, Trp53 downregulation abrogated the inhibitory effect of Cmtm3 on BMP4-induced proliferation and osteogenic differentiation of C2C12 myoblasts. Collectively, our findings provide key insights into the role of CMTM3 in regulating myoblast proliferation and transdifferentiation into osteoblasts, highlighting its significance in osteogenesis research.


Subject(s)
Cell Proliferation , Cell Transdifferentiation , Chemokines , MARVEL Domain-Containing Proteins , Myoblasts , Osteogenesis , Tumor Suppressor Protein p53 , Animals , Humans , Mice , Bone Morphogenetic Protein 4/metabolism , Cell Line , Chemokines/metabolism , MARVEL Domain-Containing Proteins/metabolism , MARVEL Domain-Containing Proteins/genetics , Myoblasts/metabolism , Myoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/cytology , Osteogenesis/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/genetics
15.
Sci Rep ; 14(1): 19635, 2024 08 23.
Article in English | MEDLINE | ID: mdl-39179631

ABSTRACT

L-type amino acid transporter 1 (LAT1) is upregulated in various cancer types and contributes to disease progression. Previous studies have demonstrated or suggested that hypoxia-inducible factors (HIFs), the key transcription factors in hypoxic responses, control the expression of LAT1 gene in several types of cancer cells. However, this regulatory relationship has not been investigated yet in colorectal cancer (CRC), one of the cancer types in which the increased LAT1 expression holds prognostic significance. In this study, we found that neither LAT1 mRNA nor protein is induced under hypoxic condition (1% O2) in CRC HT-29 cells in vitro, regardless of the prominent HIF-1/2α accumulation and HIFs-dependent upregulation of glucose transporter 1. The hypoxic treatment generally did not increase either the mRNA or protein expression of LAT1 in eight CRC cell lines tested, in contrast to the pronounced upregulation by amino acid restriction. Interestingly, knockdown of von Hippel-Lindau ubiquitin ligase to inhibit the proteasomal degradation of HIFs caused an accumulation of HIF-2α and increased the LAT1 expression in certain CRC cell lines. This study contributes to delineating the molecular mechanisms responsible for the pathological expression of LAT1 in CRC cells, emphasizing the ambiguity of HIFs-dependent transcriptional upregulation of LAT1 across cancer cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit , Large Neutral Amino Acid-Transporter 1 , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , HT29 Cells , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cell Line, Tumor , Cell Hypoxia , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
17.
Anal Cell Pathol (Amst) ; 2024: 6217134, 2024.
Article in English | MEDLINE | ID: mdl-39184399

ABSTRACT

Background: Gastric cancer (GC) is the most common malignant tumor and ranks third in the world. LncRNA H19 (H19), one of the members of lncRNA, is overexpressed in various tumors. However, many undetermined molecular mechanisms by which H19 promotes GC progression still need to be further investigated. Methodology. A series of experiments was used to confirm the undetermined molecular mechanism including wound healing and transwell assays. Key Results. In this study, a significant upregulation of H19 expression was detected in GC cells and tissues. The poor overall survival was observed in GC patient with high H19 expression. Overexpression of H19 promoted the migration of GC cells, while knockdown of H19 significantly inhibited cell migration. Moreover, miR-148a-3p had a certain negative correlation with H19. Luciferase reporter assay confirmed that H19 could directly bind to miR-148a-3p. As expected, miR-148a mimics inhibited cell migration and invasion induced by H19 overexpression. The above findings proved that H19 functions as a miRNA sponge and verified that miR-148a-3p is the H19-associated miRNA in GC. We also confirmed that SOX-12 expression was upregulated in GC patient's samples. SOX-12 expression was positively correlated with expression of H19 and was able to directly bind to miR-148a-3p. Importantly, in vitro wound healing assay showed that knockout of SOX-12 could reverse the promoting effect of H19 overexpression on cell migration. Conclusion: In conclusion, H19 has certain application value in the diagnosis and prognosis of GC. Specifically, H19 accelerates GCs to migration and metastasis by miR-138a-3p/SOX-12 axis.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , MicroRNAs , Neoplasm Metastasis , RNA, Long Noncoding , SOXC Transcription Factors , Stomach Neoplasms , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Movement/genetics , Cell Line, Tumor , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Up-Regulation/genetics , Male , Middle Aged , Female , Neoplasm Invasiveness , Base Sequence
19.
Cancer Rep (Hoboken) ; 7(8): e2159, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39118262

ABSTRACT

BACKGROUND: Ring finger protein 135 (RNF135) is an E3 ubiquitin ligase that has been implicated in the tumorigenesis of multiple human malignancies. However, whether RNF135 plays a role in the development of human osteosarcoma (OS) remains unknown. METHODS: RNF135 expression in 20 human OS and 20 human osteochondroma specimens were evaluated by means of immunohistochemistry staining. The effects of shRNA-mediated RNF135 knockdown on human OS cell growth and apoptosis were evaluated through a panel of in vitro studies on cell proliferation, colony formation, exposure of phosphatidylserine on the cell surface, and caspase 3/7 activation. The protein levels of PI3K, AKT, and p-AKT were determined by western blot analysis. RESULTS: We detected significantly higher RNF135 levels in human OS tissues than human osteochondroma tissues. In in vitro studies, shRNA-mediated RNF135 knockdown in human OS cells inhibited proliferation and induced apoptosis. In addition, RNF135 knockdown reduced PI3K and p-AKT protein levels and activated caspase 3 and 7. CONCLUSIONS: These results supported that RNF135 contributes to human OS development through PI3K/AKT-dependent mechanisms. Targeting RNF135 may provide a new therapeutic approach for treating this human malignancy.


Subject(s)
Apoptosis , Bone Neoplasms , Cell Proliferation , Osteosarcoma , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Ubiquitin-Protein Ligases , Female , Humans , Male , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Osteochondroma/pathology , Osteochondroma/genetics , Osteochondroma/metabolism , Osteosarcoma/pathology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
20.
J Orthop Surg Res ; 19(1): 487, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154011

ABSTRACT

BACKGROUND: Osteoporosis (OP) is a high-incidence bone disease that is prone to osteoporotic fractures (OF), so it has attracted widespread attention. AIM: This study investigated the specific expression and role of miR-331 in patients with OP and OF. The findings have profound implications for the clinical prevention and treatment of these conditions. METHODS: The study included 60 OP patients, 46 OF patients, and 40 healthy controls. The expression level of miR-331-3p was detected using RT-qPCR. BMP2 was used to stimulate differentiation in MC3T3-E1 cells. After induction, the expression activity of osteogenic differentiation-related gene markers was detected using RT-qPCR. The target gene analysis was conducted using a luciferase reporter assay. RESULTS: The levels of miR-331-3p were significantly elevated, while NRP2 levels were significantly reduced in OF patients. Post-surgery, miR-331-3p levels decreased over time. MiR-331-3p was found to negatively regulate the luciferase activity of NPR2 in MC3T3-E1 cells. Furthermore, overexpression of miR-331-3p inhibited cell proliferation and decreased the levels of osteoblast differentiation markers. CONCLUSION: The up-regulation of miR-331-3p can promote OP and might also encourage the occurrence of OF by regulating NRP2. However, this needs further verification.


Subject(s)
MicroRNAs , Neuropilin-2 , Osteoporosis , Osteoporotic Fractures , MicroRNAs/genetics , Humans , Osteoporotic Fractures/genetics , Osteoporotic Fractures/metabolism , Female , Osteoporosis/genetics , Osteoporosis/metabolism , Mice , Animals , Middle Aged , Aged , Neuropilin-2/genetics , Neuropilin-2/metabolism , Male , Cell Differentiation/genetics , Osteoblasts/metabolism , Cell Proliferation/genetics , Osteogenesis/genetics , Osteogenesis/physiology , Gene Expression/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL