Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Int J Mol Sci ; 25(12)2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38928092

ABSTRACT

Lung adenocarcinoma (LUAD) is the most widespread cancer in the world, and its development is associated with complex biological mechanisms that are poorly understood. Here, we revealed a marked upregulation in the mRNA level of C1orf131 in LUAD samples compared to non-tumor tissue samples in The Cancer Genome Atlas (TCGA). Depletion of C1orf131 suppressed cell proliferation and growth, whereas it stimulated apoptosis in LUAD cells. Mechanistic investigations revealed that C1orf131 knockdown induced cell cycle dysregulation via the AKT and p53/p21 signalling pathways. Additionally, C1orf131 knockdown blocked cell migration through the modulation of epithelial-mesenchymal transition (EMT) in lung adenocarcinoma. Notably, we identified the C1orf131 protein nucleolar localization sequence, which included amino acid residues 137-142 (KKRKLT) and 240-245 (KKKRKG). Collectively, C1orf131 has potential as a novel therapeutic marker for patients in the future, as it plays a vital role in the progression of lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Proto-Oncogene Proteins c-akt , Signal Transduction , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Cell Proliferation/genetics , Cell Movement/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Apoptosis/genetics , Disease Progression , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , A549 Cells
2.
Protein Expr Purif ; 221: 106501, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38782081

ABSTRACT

Nanobodies are single-variable domain antibodies with excellent properties, which are evolving as versatile tools to guide cognate antigens in vitro and in vivo for biological research, diagnosis, and treatment. Given their simple structure, nanobodies are readily produced in multiple systems. However, selecting an appropriate expression system is crucial because different conditions might cause proteins to produce different folds or post-translational modifications (PTMs), and these differences often result in different functions. At present, the strategies of PTMs are rarely reported. The GFP nanobody can specifically target the GFP protein. Here, we engineered a GFP nanobody fused with 6 × His tag and Fc tag, respectively, and expressed in bacteria and mammalian cells. The 6 × His-GFP-nanobody was produced from Escherichia coli at high yields and the pull-down assay indicated that it can precipitate the GFP protein. Meanwhile, the Fc-GFP-nanobody can be expressed in HEK293T cells, and the co-immunoprecipitation experiment can trace and target the GFP-tagged protein in vivo. Furthermore, some different PTMs in antigen-binding regions have been identified after using mass spectrometry (MS) to analyze the GFP nanobodies, which are expressed in prokaryotes and eukaryotes. In this study, a GFP nanobody was designed, and its binding ability was verified by using the eukaryotic and prokaryotic protein expression systems. In addition, this GFP nanobody was transformed into a useful instrument for more in-depth functional investigations of GFP fusion proteins. MS was further used to explore the reason for the difference in binding ability, providing a novel perspective for the study of GFP nanobodies and protein expression purification.


Subject(s)
Escherichia coli , Green Fluorescent Proteins , Protein Processing, Post-Translational , Recombinant Fusion Proteins , Single-Domain Antibodies , Humans , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/isolation & purification , Single-Domain Antibodies/immunology , HEK293 Cells , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/biosynthesis , Protein Engineering/methods , Gene Expression
3.
Front Nutr ; 11: 1364841, 2024.
Article in English | MEDLINE | ID: mdl-38765814

ABSTRACT

Background: Nephritis is a pivotal catalyst in chronic kidney disease (CKD) progression. Although epidemiological studies have explored the impact of plasma circulating metabolites and drugs on nephritis, few have harnessed genetic methodologies to establish causal relationships. Methods: Through Mendelian randomization (MR) in two substantial cohorts, spanning large sample sizes, we evaluated over 100 plasma circulating metabolites and 263 drugs to discern their causal effects on nephritis risk. The primary analytical tool was the inverse variance weighted (IVW) analysis. Our bioinformatic scrutiny of GSE115857 (IgA nephropathy, 86 samples) and GSE72326 (lupus nephritis, 238 samples) unveiled anomalies in lipid metabolism and immunological characteristics in nephritis. Thorough sensitivity analyses (MR-Egger, MR-PRESSO, leave-one-out analysis) were undertaken to verify the instrumental variables' (IVs) assumptions. Results: Unique lipoprotein-related molecules established causal links with diverse nephritis subtypes. Notably, docosahexaenoic acid (DHA) emerged as a protective factor for acute tubulointerstitial nephritis (ATIN) (OR1 = 0.84, [95% CI 0.78-0.90], p1 = 0.013; OR2 = 0.89, [95% CI 0.82-0.97], p2 = 0.007). Conversely, multivitamin supplementation minus minerals notably increased the risk of ATIN (OR = 31.25, [95% CI 9.23-105.85], p = 0.004). Reduced α-linolenic acid (ALA) levels due to lipid-lowering drugs were linked to both ATIN (OR = 4.88, [95% CI 3.52-6.77], p < 0.001) and tubulointerstitial nephritis (TIN) (OR = 7.52, [95% CI 2.78-20.30], p = 0.042). While the non-renal drug indivina showed promise for TIN treatment, the use of digoxin, hydroxocobalamin, and liothyronine elevated the risk of chronic tubulointerstitial nephritis (CTIN). Transcriptome analysis affirmed that anomalous lipid metabolism and immune infiltration are characteristic of IgA nephropathy and lupus nephritis. The robustness of these causal links was reinforced by sensitivity analyses and leave-one-out tests, indicating no signs of pleiotropy. Conclusion: Dyslipidemia significantly contributes to nephritis development. Strategies aimed at reducing plasma low-density lipoprotein levels or ALA supplementation may enhance the efficacy of existing lipid-lowering drug regimens for nephritis treatment. Renal functional status should also be judiciously considered with regard to the use of nonrenal medications.

4.
Int J Mol Sci ; 25(7)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38612387

ABSTRACT

Apobec-1 complementation factor (A1CF) functions as an RNA-binding cofactor for APO-BEC1-mediated C-to-U conversion during RNA editing and as a hepatocyte-specific regulator in the alternative pre-mRNA splicing of metabolic enzymes. Its role in RNA editing has not been clearly established. Western blot, co-immunoprecipitation (Co-IP), immunofluorescence (IF), methyl thiazolyl tetrazolium (MTT), and 5-ethynyl-2'-deoxyuridine (EdU) assays were used to examine the role of A1CF beyond RNA editing in renal carcinoma cells. We demonstrated that A1CF interacts with NKRF, independent of RNA and DNA, without affecting its expression or nuclear translocation; however, it modulates p65(Ser536) phosphorylation and IFN-ß levels. Truncation of A1CF or deletion on NKRF revealed that the RRM1 domain of A1CF and the p65 binding motif of NKRF are required for their interaction. Deletion of RRM1 on A1CF abrogates NKRF binding, and the decrease in IFN-ß expression and p65(Ser536) phosphorylation was induced by A1CF. Moreover, full-length A1CF, but not an RRM1 deletion mutant, promoted cell proliferation in renal carcinoma cells. Perturbation of A1CF levels in renal carcinoma cells altered anchorage-independent growth and tumor progression in nude mice. Moreover, p65(Ser536) phosphorylation and IFN-ß expression were lower, but ki67 was higher in A1CF-overexpressing tumor tissues of a xenograft mouse model. Notably, primary and metastatic samples from renal cancer patients exhibited high A1CF expression, low p65(Ser536) phosphorylation, and decreased IFN-ß levels in renal carcinoma tissues compared with the corresponding paracancerous tissues. Our results indicate that A1CF-decreased p65(Ser536) phosphorylation and IFN-ß levels may be caused by A1CF competitive binding to the p65-combined site on NKRF and demonstrate the direct binding of A1CF independent of RNA or DNA in signal pathway regulation and tumor promotion in renal carcinoma cells.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Humans , Mice , APOBEC-1 Deaminase , Carcinoma, Renal Cell/genetics , Disease Models, Animal , DNA , Kidney Neoplasms/genetics , Mice, Nude , Phosphorylation , RNA , RNA-Binding Proteins , Interferon-beta
5.
bioRxiv ; 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37873089

ABSTRACT

Astrocyte specification during development is influenced by both intrinsic and extrinsic factors, but the precise contribution of each remains poorly understood. Here we show that septal astrocytes from Nkx2.1 and Zic4 expressing progenitor zones are allocated into non-overlapping domains of the medial (MS) and lateral septal nuclei (LS) respectively. Astrocytes in these areas exhibit distinctive molecular and morphological features tailored to the unique cellular and synaptic circuit environment of each nucleus. Using single-nucleus (sn) RNA sequencing, we trace the developmental trajectories of cells in the septum and find that neurons and astrocytes undergo region and developmental stage-specific local cell-cell interactions. We show that expression of the classic morphogens Sonic hedgehog (Shh) and Fibroblast growth factors (Fgfs) by MS and LS neurons respectively, functions to promote the molecular specification of local astrocytes in each region. Finally, using heterotopic cell transplantation, we show that both morphological and molecular specifications of septal astrocytes are highly dependent on the local microenvironment, regardless of developmental origins. Our data highlights the complex interplay between intrinsic and extrinsic factors shaping astrocyte identities and illustrates the importance of the local environment in determining astrocyte functional specialization.

6.
bioRxiv ; 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37873286

ABSTRACT

When interacting with their environment, animals must balance exploratory and defensive behavior to evaluate and respond to potential threats. The lateral septum (LS) is a structure in the ventral forebrain that calibrates the magnitude of behavioral responses to stress-related external stimuli, including the regulation of threat avoidance. The complex connectivity between the LS and other parts of the brain, together with its largely unexplored neuronal diversity, makes it difficult to understand how defined LS circuits control specific behaviors. Here, we describe a mouse model in which a population of neurons with a common developmental origin (Nkx2.1-lineage neurons) are absent from the LS. Using a combination of circuit tracing and behavioral analyses, we found that these neurons receive inputs from the perifornical area of the anterior hypothalamus (PeFAH) and are specifically activated in stressful contexts. Mice lacking Nkx2.1-lineage LS neurons display increased exploratory behavior even under stressful conditions. Our study extends the current knowledge about how defined neuronal populations within the LS can evaluate contextual information to select appropriate behavioral responses. This is a necessary step towards understanding the crucial role that the LS plays in neuropsychiatric conditions where defensive behavior is dysregulated, such as anxiety and aggression disorders.

7.
Front Oncol ; 12: 894485, 2022.
Article in English | MEDLINE | ID: mdl-36324576

ABSTRACT

Background: Lung adenocarcinoma (LUAD) is the most common respiratory globallywith a poor prognosis. Lipid metabolism is extremely important for the occurrence and development of cancer. However, the role of genes involved in lipid metabolism in LUAD development is unclear. We aimed to identify the abnormal lipid metabolism pathway of LUAD, construct a novel prognostic model of LUAD, and discover novel biomarkers involved in lipid metabolism in LUAD. Methods: Based on differentially expressed genes involved in lipid metabolism in LUAD samples from The Cancer Genome Atlas (TCGA), abnormal lipid metabolism pathways in LUAD were analyzed. The lasso penalized regression analysis was performed on the TCGA cohort (training set) to construct a risk score formula. The predictive ability of the risk score was validated in the Gene Expression Omnibus (GEO) dataset (validation set) using Kaplan-Meier analysis and ROC curves. Finally, based on CRISPR gene editing technology, hematopoietic prostaglandin D synthase (HPGDS) was knocked out in A549 cell lines, the changes in lipid metabolism-related markers were detected by western blotting, and the changes in cell migration were detected by transwell assay. Results: Based on the differential genes between lung cancer tissue and normal tissue, we found that the arachidonic acid metabolism pathway is an abnormal lipid metabolism pathway in both lung adenocarcinoma and lung squamous cell carcinoma. Based on the sample information of TCGA and abnormally expressed lipid metabolism-related genes, a 9-gene prognostic risk score was successfully constructed and validated in the GEO dataset. Finally, we found that knockdown of HPGDS in A549 cell lines promoted lipid synthesis and is more invasive than in control cells. Rescue assays showed that ACSL1 knockdown reversed the pro-migration effects of HPGDS knockdown. The knockdown of HPGDS promoted migration response by upregulating the expression of the lipid metabolism key enzymes ACSL1 and ACC. Conclusion: The genes involved in lipid metabolism are associated with the occurrence and development of LUAD. HPGDS can be a therapeutic target of a potential lipid metabolism pathway in LUAD, and the therapeutic target of lipid metabolism genes in LUAD should be studied further.

8.
Bioeng Transl Med ; 7(3): e10344, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36176601

ABSTRACT

Effective skin wound healing is a complex process involving anti-inflammation, fibrosis, matrix reconstruction, and angiogenesis. This work aimed to integrate the macrophage-mediated anti-inflammation and fibroblast-assisted matrix reconstruction for enhanced skin wound healing. Herein, we utilized the cytomembranes derived from repolarized M2 macrophages and fibroblasts to prepare the natural biologics. Results showed that the inflammatory M1 macrophages were repolarized to M2 phenotype by the M2 macrophage cytomembranes. As a consequence, the cytomembranes of M2 macrophage could facilitate the wound closure in mice. Furthermore, the addition of fibroblast membranes to the macrophage cytomembranes contributed to a better matrix reconstruction, neovascularization and angiogenesis. Next, we used a transforming growth factor-ß (TGF-ß) inhibitor to attenuate cutaneous scar formation. Therefore, our modality could promote skin wound healing and effectively suppress scar formation in the preclinical murine skin wounds. The cytomembrane biologics might provide a biocompatible and versatile tool for wound healing.

9.
Gene ; 832: 146559, 2022 Jul 20.
Article in English | MEDLINE | ID: mdl-35569765

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is one of the most lethal genitourinary tumors with rapid progression and metastasis. Selenoprotein S (SELS), which is broadly expressed in human tissues, has been reported to be involved in ER homeostasis and inflammation. However, the biological roles of SELS in ccRCC remain unclear. In this study, we found that SELS expression was significantly higher in ccRCC and correlated with multiple clinicopathological features. Overexpression of SELS could promote cell proliferation and inhibit apoptosis in 786-O cells, whereas silence of SELS elicited opposite effect. Further mechanistic studies revealed that SELS enhanced cell proliferation and inhibited apoptosis through activating AKT/GSK3ß/NF-κB signaling pathway. Besides, SELS could stabilize c-Myc by preventing ubiquitin-proteasome-mediated degradation. Interestingly, we found that SELS could also inhibit migration of ccRCC cell likely through repressing epithelial-mesenchymal transition (EMT). Collectively, our findings suggested that SELS promoted tumor progression, and inhibited apoptosis and migration through AKT/GSK3ß/NF-κB signaling pathway and EMT in ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Apoptosis , Carcinogenesis/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Kidney Neoplasms/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Selenoproteins , Signal Transduction
10.
Cell Rep ; 38(8): 110416, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35196485

ABSTRACT

Neuron-glia interactions play a critical role in the regulation of synapse formation and circuit assembly. Here we demonstrate that canonical Sonic hedgehog (Shh) pathway signaling in cortical astrocytes acts to coordinate layer-specific synaptic connectivity. We show that the Shh receptor Ptch1 is expressed by cortical astrocytes during development and that Shh signaling is necessary and sufficient to promote the expression of genes involved in regulating synaptic development and layer-enriched astrocyte molecular identity. Loss of Shh in layer V neurons reduces astrocyte complexity and coverage by astrocytic processes in tripartite synapses; conversely, cell-autonomous activation of Shh signaling in astrocytes promotes cortical excitatory synapse formation. Furthermore, Shh-dependent genes Lrig1 and Sparc distinctively contribute to astrocyte morphology and synapse formation. Together, these results suggest that Shh secreted from deep-layer cortical neurons acts to specialize the molecular and functional features of astrocytes during development to shape circuit assembly and function.


Subject(s)
Astrocytes , Hedgehog Proteins , Astrocytes/metabolism , Hedgehog Proteins/metabolism , Neurogenesis/physiology , Neurons/metabolism , Synapses/metabolism
11.
Biosci Rep ; 41(11)2021 11 26.
Article in English | MEDLINE | ID: mdl-34735568

ABSTRACT

Glucocorticoids (GCs) have been widely used in clinical treatment as anti-inflammatory, anti-shock and immunosuppressive medicines. However, the effect of excessive GCs on immune response and metabolism of kidney remains unclear. Here, we profiled the gene expression of kidney from mice with high-dose dexamethasone (DEX) treatment. A total of 1193 differentially expressed genes (DEGs) were screened in DEX treatment group compared with the saline group, including 715 down- regulated and 478 up-regulated. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of these DEGs showed extracellular matrix (ECM)-receptor interaction, cell adhesion molecules signaling pathway were significantly enriched, and that the vast majority of DEGs were involved in monocarboxylic acid metabolism, leukocyte cell-cell adhesion and fatty acid metabolism. Gene set enrichment analysis (GSEA) revealed that DEGs were strongly associated with immune-response and cell adhesion gene sets, such as Fc γ R-mediated phagocytosis, leukocyte transendothelial migration, T-cell receptor signaling pathway, cell adhesion, ECM-receptor interaction and focal adhesion-associated pathways. KEGG pathway analysis of differentially expressed kinases (DEKs) showed T-cell receptor and forkhead box class O signaling pathway were enriched. Furthermore, we found multiple protein kinases expression were dysregulated greatly after dexamethasone treatment, including classical effector of GCs stimulation-serum and GC-regulated kinase. These protein kinases are involved in multiple signaling pathways in mice kidney, such as mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. We profiled the gene expression of the kidney from high-dose dexamethasone-treated mice and provided important information for further study the mechanism of side effects of GCs in clinical therapy.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Dexamethasone/adverse effects , Kidney/metabolism , Metabolism/drug effects , Protein Kinases/biosynthesis , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/immunology , Cell Movement/drug effects , Computational Biology , Dexamethasone/administration & dosage , Dexamethasone/immunology , Gene Expression Regulation/drug effects , Immunity/drug effects , Inflammation/metabolism , Injections, Intraperitoneal , Kidney/drug effects , Lipid Metabolism/drug effects , Male , Mice, Inbred C57BL , Protein Kinases/drug effects , Protein Kinases/genetics , Signal Transduction/drug effects
12.
Biomaterials ; 278: 121161, 2021 11.
Article in English | MEDLINE | ID: mdl-34601198

ABSTRACT

Immunotherapy emerges as a potential therapeutic strategy against tumor relapse. However, immunosuppressive tumor microenvironment poses an obstacle to immunotherapy. Of particular note is that macrophages are abundant in solid tumors and tumor-associated macrophages (TAMs) are mainly anti-inflammatory and protumoral. Therefore, re-educating TAMs will be critical for improving the antitumor efficacy of immunotherapy. Herein we engineered a macrophage-derived implantable vaccine for suppressing postsurgical tumor relapse. The vaccine comprised hybrid cytomembranes from macrophages/tumor cells and an immunoadjuvant, cytosinephosphate-guanosine oligodeoxynucleotides (CpG ODNs). The vaccine was further embedded into a calcium alginate hydrogel for tissue-localized delivery. Results show that the vaccine could induce the shift from anti-inflammatory M2-like TAMs to proinflammatory M1-like macrophage. Moreover, the vaccine stimulated systemic immunity by facilitating dendritic cells (DCs) maturation and memory T (T EM) cell activation, forming a self-replenishing circulation in tumor microenvironment. Consequently, the vaccine could prevent the postsurgical tumor relapse at both the primary and distant tumor sites. In addition, the lung metastasis was also reduced by the vaccine implantation in mice. The multifunctional vaccine prepared from biomacromolecule and nature-derived material provides a biocompatible and versatile tool for re-educating TAMs and preventing postsurgical tumor recurrence.


Subject(s)
Neoplasm Recurrence, Local , Vaccines , Animals , Immunotherapy , Macrophages , Mice , Neoplasm Recurrence, Local/prevention & control , Tumor Microenvironment
13.
Nature ; 594(7863): 341-343, 2021 06.
Article in English | MEDLINE | ID: mdl-34089037
14.
In Vitro Cell Dev Biol Anim ; 57(5): 501-509, 2021 May.
Article in English | MEDLINE | ID: mdl-34002283

ABSTRACT

Metastasis is inevitable in about 30% of patients with primary renal cell carcinoma after nephrectomy treatment. APOBEC1 complementation factor (A1CF), an RNA binding protein, participates in tumor progressions such as growth, apoptosis, differentiation, and invasion. Here, we explored biological functions of A1CF and provided a new insight into renal cell carcinoma metastasis. Wound healing assay was conducted to detect migration in A1CF overexpression and knockdown stable cell lines. Quantitative PCR and western blot assays were utilized to test transcriptional and translation levels of A1CF and SMAD3 in A1CF overexpression and knockdown renal carcinoma cells. Nuclear and cytoplasmic protein separation assays were conducted to evaluate the subcellular distribution of A1CF and SMAD3. Immunoprecipitation assay was conducted to detect the interaction between A1CF and SMAD3. Our study demonstrated A1CF overexpression facilitated cell migration in renal carcinoma cells. A1CF deficiency downregulated expression of SMAD3, Snail1, and N-cadherin. In addition, A1CF promoted nucleus translocation of SMAD3 and interacted with SMAD3. SMAD3 knockdown attenuated cell migration induced by A1CF overexpression. Our study suggested A1CF facilitated cell migration by promoting nucleus translocation of SMAD3 in renal cell carcinoma cells.


Subject(s)
APOBEC-1 Deaminase/metabolism , Active Transport, Cell Nucleus , Carcinoma, Renal Cell/pathology , Cell Movement , Kidney Neoplasms/pathology , Smad3 Protein/metabolism , Blotting, Western , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Immunoprecipitation , Kidney Neoplasms/metabolism , Polymerase Chain Reaction
15.
J Microbiol Biotechnol ; 31(2): 207-216, 2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33144548

ABSTRACT

Supplement of high-protein food plays an important role in improving the symptoms of malnutrition and the immune capacity of the body, but the association of high-protein diet and gut microbiota remained unaddressed. Here, we systematically analyzed the internal organs and gut microbiota in C57(WT) or PD-1H-depleted (KO) mice (T cells were activated) fed with pupae or feed for six weeks. We observed that the body weight gain in the mice fed with pupae increased less significantly than that of the feed group, while the villi and small intestine lengths in the pupa group were reduced compared with that of mice given feed. However, the average body weight of the KO mice increased compared with that of the WT mice fed with pupae or feed. Pupae increased the concentration of blood glucose in WT, but not in KO mice. Moreover, in the feed group, there was no difference in the weight of the internal organs between the WT and KO mice, but in the pupae-fed group, liver weight was decreased and spleen weight was increased compared with that of KO mice. The amounts/plural/amounts of Melainabacteria, Chloroflexi, and Armatimonadetes were specifically upregulated by pupae, and this upregulation was weakened or eliminated by PD-1H depletion. Some bacteria with high abundance in the feed-fed KO mice, such as Deferribacteres, Melainabacteria, Acidobacteria, Bacteroidetes, Spirochaetes and Verrucomicrobia, were decreased in pupae-fed KO mice, and Proteobacteria and Deinococcus were specifically enriched in pupae-fed KO mice. Bacteroidetes, Firmicutes and Akkermansia were associated with weight loss in the pupaefed group while Lachnospiraceae and Anaerobiospirillum were related glucose metabolism and energy consumption. Based on high-throughput sequencing, we discovered that some gut bacteria specifically regulated the metabolism of a high-protein diet, and PD-1H deficiency improved life quality and sustained blood glucose. Moreover, PD-1H responses to high-protein diet through modulating the type and quantity of gut bacteria. These findings provide evidence about the association among gut microbiota, T cell activation (for PD-1H depletion) and high-protein diet metabolism, have important theoretical significance for nutrition and health research.


Subject(s)
Dietary Proteins/metabolism , Gastrointestinal Microbiome , Programmed Cell Death 1 Receptor/deficiency , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Biodiversity , Blood Glucose/metabolism , Diet, High-Protein , Feces/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics
16.
Development ; 147(22)2020 11 16.
Article in English | MEDLINE | ID: mdl-33060132

ABSTRACT

The mammalian cortex is populated by neurons derived from neural progenitors located throughout the embryonic telencephalon. Excitatory neurons are derived from the dorsal telencephalon, whereas inhibitory interneurons are generated in its ventral portion. The transcriptional regulator PRDM16 is expressed by radial glia, neural progenitors present in both regions; however, its mechanisms of action are still not fully understood. It is unclear whether PRDM16 plays a similar role in neurogenesis in both dorsal and ventral progenitor lineages and, if so, whether it regulates common or unique networks of genes. Here, we show that Prdm16 expression in mouse medial ganglionic eminence (MGE) progenitors is required for maintaining their proliferative capacity and for the production of proper numbers of forebrain GABAergic interneurons. PRDM16 binds to cis-regulatory elements and represses the expression of region-specific neuronal differentiation genes, thereby controlling the timing of neuronal maturation. PRDM16 regulates convergent developmental gene expression programs in the cortex and MGE, which utilize both common and region-specific sets of genes to control the proliferative capacity of neural progenitors, ensuring the generation of correct numbers of cortical neurons.


Subject(s)
Cerebral Cortex/metabolism , DNA-Binding Proteins/metabolism , GABAergic Neurons/metabolism , Interneurons/metabolism , Neural Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cerebral Cortex/cytology , DNA-Binding Proteins/genetics , GABAergic Neurons/cytology , Interneurons/cytology , Mice , Neural Stem Cells/cytology , Transcription Factors/genetics
17.
J Cancer ; 11(23): 6874-6882, 2020.
Article in English | MEDLINE | ID: mdl-33123278

ABSTRACT

Accumulating evidence indicates that hotspot p53 mutants have gain-of-function in promoting cell migration and tumor metastasis. However, the molecular mechanisms are not completely understood. Here, we show that a hotspot mutation, p53-R273H, promotes non-small cell lung cancer (NSCLC) cell migration and upregulates the mRNA and protein expression of neuraminidase-1 (NEU1), a sialidase involved in cell proliferation, cell migration and tumorigenesis. Silencing of NEU1 leads to upregulation of integrin ß4 which significantly inhibits NSCLC cell migration induced by p53-R273H. Mechanistically, p53-R273H promotes NEU1 transcription via activation of AKT signaling. Importantly, NEU1 expression is upregulated in human NSCLC samples harboring mutant p53 and is associated with poor clinical outcome. Overall, this study highlights an important role of NEU1 in p53-R273H-induced NSCLC cell migration and provides a potential target for NSCLC diagnosis and treatment.

18.
Environ Pollut ; 266(Pt 3): 115278, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32853872

ABSTRACT

Atmospheric ambient gaseous ammonia (NH3), the most abundant alkaline gas, affects public health and climate change through its key role in the formation of secondary aerosols via reactions with acidic gases. Estimation of the contributions of ammonia sources is very challenging in the urban atmosphere. Stable nitrogen isotope ratio (δ15N) measurements have shown that urban aerosol NH4+ and gaseous NH3 are derived from fossil fuel combustion-related (FF) sources, such as coal combustion, NH3 slip, and vehicle exhaust, and volatilization-related sources, such as agriculture and urban water volatilization. Biomass burning (BB) sources, especially residential biofuel, can produce vast quantities of NH3 and other pollutants and may greatly influence air quality and contribute to increased urban NH3 emissions. In the present study, we continually collected PM2.5 samples at three urban sites in Central China during autumn and analyzed the major water-soluble ions and δ15N values of aerosol NH4+. The concentrations of NH4+ increased as the temperature decreased close to winter, whereas the δ15N values did not show this pattern. According to the Bayesian model after isotope fractionation correction, FF sources contributed to 56.4 ± 17.1%, 46.4 ± 18.2%, and 51.8 ± 14.9% of aerosol NH4+ in Nanchang, Wuhan, and Changsha, respectively, throughout autumn. The contributions from BB sources were 34.5 ± 20.4%, 46.4 ± 21.4%, and 40.4 ± 17.4% for Nanchang, Wuhan, and Changsha, respectively. We also found the fraction of aerosol NH4+ from BB increased in all three cities from September to November 2017, which was likely caused by increased heating demands with the decrease in temperature during the season. Furthermore, BB was responsible for a severe haze event (maximum PM2.5 of 205.69 µg/m3) in Nanchang. These findings suggest government controls to improve air quality should include BB sources in addition to FF sources.


Subject(s)
Air Pollutants/analysis , Ammonium Compounds/analysis , Aerosols/analysis , Bayes Theorem , Biomass , China , Cities , Environmental Monitoring , Particulate Matter/analysis , Seasons
19.
Environ Sci Pollut Res Int ; 27(28): 35031-35039, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32583119

ABSTRACT

Aerosol acidity (pH), one of key properties of fine-mode particulate (PM2.5), depends largely on nitrate and sulfate in particle. The mass contribution of nitrate relative to sulfate in PM2.5 has tended to increase in many regions globally, but how this change affects aerosol pH remains in debate. In this way, we measured PM2.5 ionic species and oxygen isotopic composition of nitrate in the eastern China, and predicted aerosol pH using the ISORROPIA-II model. When nitrate to sulfate molar ratio increases and thus PM2.5 is gradually enriched in ammonium nitrate (NH4NO3), aerosol pH tends to increase. The oxidation of nitrogen dioxide (NO2) by hydroxyl radical is responsible for most of nitrate formation (generally above 60%). These indicate that nitrate formation through gas-to-particle conversion involving ammonia and nitric acid results in increasing aerosol pH with increasing molar ratio of nitrate to sulfate. Conversely, aerosol pH is expected to decrease with increasing relative abundance of nitrate as ammonia emissions are lowered. Our research concludes that it should be considered to reduce aerosol NH4NO3 by reducing the precursors of nitric oxide and ammonia emissions, to substantially improve the air quality (i.e., reduce PM2.5 levels and potential nitrate deposition) in China.


Subject(s)
Air Pollutants/analysis , Particulate Matter/analysis , Aerosols/analysis , China , Environmental Monitoring , Hydrogen-Ion Concentration , Sulfates
20.
Int J Mol Sci ; 21(5)2020 Mar 07.
Article in English | MEDLINE | ID: mdl-32156004

ABSTRACT

High-dose dexamethasone (DEX) is used to treat chemotherapy-induced nausea and vomiting or to control immunotherapy-related autoimmune diseases in clinical practice. However, the underlying mechanisms of high-dose DEX in tumor progression remain unaddressed. Therefore, we explored the effects of high-dose DEX on tumor progression and the potential mechanisms of its anti-tumor function using immunohistochemistry, histological examination, real-time quantitative PCR (qPCR), and Western blotting. Tumor volume, blood vessel invasion, and levels of the cell proliferation markers Ki67 and c-Myc and the anti-apoptotic marker Bcl2 decreased in response to high-dose DEX. However, the cell apoptosis marker cleaved caspase 3 increased significantly in mice treated with 50 mg/kg DEX compared with controls. Some genes associated with immune responses were significantly downregulated following treatment with 50 mg/kg DEX e.g., Cxcl9, Cxcl10, Cd3e, Gzmb, Ifng, Foxp3, S100a9, Arg1, and Mrc1. In contrast, the M1-like tumor-associated macrophages (TAMs) activation marker Nos2 was shown to be increased. Moreover, the expression of peroxisome proliferator-activated receptors α and γ (Pparα and Pparg, respectively) was shown to be significantly upregulated in livers or tumors treated with DEX. However, high-dose DEX treatment decreased the expression of glucose and lipid metabolic pathway-related genes such as glycolysis-associated genes (Glut1, Hk2, Pgk1, Idh3a), triglyceride (TG) synthesis genes (Gpam, Agpat2, Dgat1), exogenous free fatty acid (FFA) uptake-related genes (Fabp1, Slc27a4, and CD36), and fatty acid oxidation (FAO) genes (Acadm, Acaa1, Cpt1a, Pnpla2). In addition, increased serum glucose and decreased serum TG and non-esterified fatty acid (NEFA) were observed in DEX treated-xenografted tumor mice. These findings indicate that high-dose DEX-inhibited tumor progression is a complicated process, not only activated by M1-like TAMs, but also decreased by the uptake and consumption of glucose and lipids that block the raw material and energy supply of cancer cells. Activated M1-like TAMs and inefficient glucose and lipid metabolism delayed tumor cell growth and promoted apoptosis. These findings have important implications for the application of DEX combined with drugs that target key metabolism pathways for tumor therapy in clinical practice.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Lewis Lung/drug therapy , Dexamethasone/pharmacology , Glycolysis/drug effects , Lipid Metabolism/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Glucose/metabolism , Mice , Mice, Inbred C57BL , PPAR alpha/metabolism , PPAR gamma/metabolism , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL