Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 14: 1153915, 2023.
Article in English | MEDLINE | ID: mdl-37153549

ABSTRACT

Macrophage infiltration into adipose tissue is a key pathological factor inducing adipose tissue dysfunction and contributing to obesity-induced inflammation and metabolic disorders. In this review, we aim to present the most recent research on macrophage heterogeneity in adipose tissue, with a focus on the molecular targets applied to macrophages as potential therapeutics for metabolic diseases. We begin by discussing the recruitment of macrophages and their roles in adipose tissue. While resident adipose tissue macrophages display an anti-inflammatory phenotype and promote the development of metabolically favorable beige adipose tissue, an increase in pro-inflammatory macrophages in adipose tissue has negative effects on adipose tissue function, including inhibition of adipogenesis, promotion of inflammation, insulin resistance, and fibrosis. Then, we presented the identities of the newly discovered adipose tissue macrophage subtypes (e.g. metabolically activated macrophages, CD9+ macrophages, lipid-associated macrophages, DARC+ macrophages, and MFehi macrophages), the majority of which are located in crown-like structures within adipose tissue during obesity. Finally, we discussed macrophage-targeting strategies to ameliorate obesity-related inflammation and metabolic abnormalities, with a focus on transcriptional factors such as PPARγ, KLF4, NFATc3, and HoxA5, which promote macrophage anti-inflammatory M2 polarization, as well as TLR4/NF-κB-mediated inflammatory pathways that activate pro-inflammatory M1 macrophages. In addition, a number of intracellular metabolic pathways closely associated with glucose metabolism, oxidative stress, nutrient sensing, and circadian clock regulation were examined. Understanding the complexities of macrophage plasticity and functionality may open up new avenues for the development of macrophage-based treatments for obesity and other metabolic diseases.


Subject(s)
Adipose Tissue , Macrophages , Metabolic Diseases , Obesity , Adipose Tissue/immunology , Macrophages/classification , Macrophages/immunology , Obesity/immunology , Obesity/therapy , Metabolic Diseases/immunology , Metabolic Diseases/therapy , Humans , Inflammation/immunology , Inflammation/therapy , Adipogenesis/immunology , Cell Polarity
2.
Front Endocrinol (Lausanne) ; 13: 873699, 2022.
Article in English | MEDLINE | ID: mdl-35909571

ABSTRACT

Chronic low-grade inflammation in adipose tissue (AT) is a hallmark of obesity and contributes to various metabolic disorders, such as type 2 diabetes and cardiovascular diseases. Inflammation in ATs is characterized by macrophage infiltration and the activation of inflammatory pathways mediated by NF-κB, JNK, and NLRP3 inflammasomes. Adipokines, hepatokines and myokines - proteins secreted from AT, the liver and skeletal muscle play regulatory roles in AT inflammation via endocrine, paracrine, and autocrine pathways. For example, obesity is associated with elevated levels of pro-inflammatory adipokines (e.g., leptin, resistin, chemerin, progranulin, RBP4, WISP1, FABP4, PAI-1, Follistatin-like1, MCP-1, SPARC, SPARCL1, and SAA) and reduced levels of anti-inflammatory adipokines such as adiponectin, omentin, ZAG, SFRP5, CTRP3, vaspin, and IL-10. Moreover, some hepatokines (Fetuin A, DPP4, FGF21, GDF15, and MANF) and myokines (irisin, IL-6, and DEL-1) also play pro- or anti-inflammatory roles in AT inflammation. This review aims to provide an updated understanding of these organokines and their role in AT inflammation and related metabolic abnormalities. It serves to highlight the molecular mechanisms underlying the effects of these organokines and their clinical significance. Insights into the roles and mechanisms of these organokines could provide novel and potential therapeutic targets for obesity-induced inflammation.


Subject(s)
Adipokines , Adipose Tissue , Inflammation , Obesity , Adipokines/metabolism , Adipose Tissue/metabolism , Cytokines/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Obesity/complications , Obesity/metabolism , Retinol-Binding Proteins, Plasma/metabolism
3.
World J Stem Cells ; 14(1): 104-116, 2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35126831

ABSTRACT

BACKGROUND: Type 1 diabetes (T1D), a chronic metabolic and autoimmune disease, seriously endangers human health. In recent years, mesenchymal stem cell (MSC) transplantation has become an effective treatment for diabetes. Menstrual blood-derived endometrial stem cells (MenSC), a novel MSC type derived from the decidual endometrium during menstruation, are expected to become promising seeding cells for diabetes treatment because of their noninvasive collection procedure, high proliferation rate and high immunomodulation capacity. AIM: To comprehensively compare the effects of MenSC and umbilical cord-derived MSC (UcMSC) transplantation on T1D treatment, to further explore the potential mechanism of MSC-based therapies in T1D, and to provide support for the clinical application of MSC in diabetes treatment. METHODS: A conventional streptozotocin-induced T1D mouse model was established, and the effects of MenSC and UcMSC transplantation on their blood glucose and serum insulin levels were detected. The morphological and functional changes in the pancreas, liver, kidney, and spleen were analyzed by routine histological and immunohistochemical examinations. Changes in the serum cytokine levels in the model mice were assessed by protein arrays. The expression of target proteins related to pancreatic regeneration and apoptosis was examined by western blot. RESULTS: MenSC and UcMSC transplantation significantly improved the blood glucose and serum insulin levels in T1D model mice. Immunofluorescence analysis revealed that the numbers of insulin+ and CD31+ cells in the pancreas were significantly increased in MSC-treated mice compared with control mice. Subsequent western blot analysis also showed that vascular endothelial growth factor (VEGF), Bcl2, Bcl-xL and Proliferating cell nuclear antigen in pancreatic tissue was significantly upregulated in MSC-treated mice compared with control mice. Additionally, protein arrays indicated that MenSC and UcMSC transplantation significantly downregulated the serum levels of interferon γ and tumor necrosis factor α and upregulated the serum levels of interleukin-6 and VEGF in the model mice. Additionally, histological and immunohistochemical analyses revealed that MSC transplantation systematically improved the morphologies and functions of the liver, kidney, and spleen in T1D model mice. CONCLUSION: MenSC transplantation significantly improves the symptoms in T1D model mice and exerts protective effects on their main organs. Moreover, MSC-mediated angiogenesis, antiapoptotic effects and immunomodulation likely contribute to the above improvements. Thus, MenSC are expected to become promising seeding cells for clinical diabetes treatment due to their advantages mentioned above.

4.
Plant Mol Biol ; 104(4-5): 429-450, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32808190

ABSTRACT

KEY MESSAGE: OsWRKY36 represses plant height and grain size by inhibiting gibberellin signaling. Plant height and grain size are important agronomic traits affecting yield in cereals, including rice. Gibberellins (GAs) are plant hormones that promote plant growth and developmental processions such as stem elongation and grain size. WRKYs are transcription factors that regulate stress tolerance and plant development including height and grain size. However, the relationship between GA signaling and WRKY genes is still poorly understood. Here, we characterized a small grain and semi-dwarf 3 (sgsd3) mutant in rice cv. Hwayoung (WT). A T-DNA insertion in the 5'-UTR of OsWRKY36 induced overexpression of OsWRKY36 in the sgsd3 mutant, likely leading to the mutant phenotype. This was confirmed by the finding that overexpression of OsWRKY36 caused a similar small grain and semi-dwarf phenotype to the sgsd3 mutant whereas knock down and knock out caused larger grain phenotypes. The sgsd3 mutant was also hyposensitive to GA and accumulated higher mRNA and protein levels of SLR1 (a GA signaling DELLA-like inhibitor) compared with the WT. Further assays showed that OsWRKY36 enhanced SLR1 transcription by directly binding to its promoter. In addition, we found that OsWRKY36 can protect SLR1 from GA-mediated degradation. We thus identified a new GA signaling repressor OsWRKY36 that represses GA signaling through stabilizing the expression of SLR1.


Subject(s)
Oryza/growth & development , Oryza/genetics , Plant Proteins/genetics , Transcription Factors/genetics , 5' Untranslated Regions , DNA, Bacterial , Gene Expression Regulation, Plant , Gibberellins/metabolism , Mutation , Oryza/cytology , Phenotype , Plant Cells , Plant Proteins/chemistry , Plant Proteins/metabolism , Plant Stems/cytology , Plant Stems/genetics , Promoter Regions, Genetic , Protein Stability , RNA Interference , Seeds/genetics , Seeds/growth & development , Signal Transduction , Transcription Factors/chemistry , Transcription Factors/metabolism
5.
Stem Cell Res Ther ; 11(1): 346, 2020 08 08.
Article in English | MEDLINE | ID: mdl-32771033

ABSTRACT

BACKGROUND: Research into the pathogenesis of endometriosis (EMs) would substantially promote its effective treatment and early diagnosis. However, the aetiology of EMs is poorly understood and controversial despite the progress in EMs research in the last several decades. Currently, accumulating evidence has shed light on the importance of endometrial stem cells (EnSCs) residing in the basal layer of endometrium in the establishment and progression of endometriotic lesions. Therefore, we aimed to identify the differences between EnSCs isolated from the ectopic lesions of EMs patients (EnSC-EM-EC) and EnSCs isolated from eutopic endometrium of control group (EnSC-Control). We further performed preliminary exploration of the potential signalling pathways involved in the above abnormalities. METHODS: EnSC-EM-EC (n = 12) and EnSC-Control (n = 13) were successfully isolated. Then, the proliferative capacity, migratory capacity and angiogenic potential of EnSCs were evaluated by conventional MTT assay, flow cytometry, wound healing assay, transwell assay, tube formation assay and chick embryo chorioallantoic membrane assay respectively. The expression of 11 angiogenesis-associated biological factors and 11 cytokines secreted by EnSCs and 17 adhesion molecules expressed on EnSCs were determined by protein array assays respectively. Differentially expressed genes (DEGs) between EnSC-EM-EC and EnSC-Control were analysed by RNA-sequence. RESULTS: EnSC-EM-EC exhibited unique biological characteristics, including prolonged mitosis, enhanced migratory capacity and enhanced angiogenic potential. Greater amounts of angiogenic factors (especially VEGF and PDGF) were secreted by EnSC-EM-EC than by EnSC-Control; however, the distinct profiles of cytokines secreted by EnSC-EM-EC and adhesion molecules expressed by EnSC-EM-EC require further investigation. A total of 523 DEGs between EnSC-EM-EC and EnSC-Control were identified and analysed using the KEGG and Gene Ontology databases. CONCLUSIONS: Our results not only improve the understanding of EMs but also contribute to the development of EnSC-EM-EC as a tool for EMs drug discovery. These cells could be of great help in exploiting promising therapeutic targets and new biomarkers for EMs treatment and prognosis.


Subject(s)
Endometriosis , Mesenchymal Stem Cells , Transcriptome , Adult , Animals , Chick Embryo , Endometriosis/genetics , Endometriosis/pathology , Endometrium , Female , Humans , Mesenchymal Stem Cells/metabolism , Middle Aged , Neovascularization, Pathologic , Stem Cells
6.
Med Microbiol Immunol ; 208(6): 781-792, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31187242

ABSTRACT

Staphylococcal enterotoxins (SEs), as typical superantigens, exhibit promising antitumour activity in the clinic, but their unavoidable side effects related to fever and emesis seriously limit their application for the treatment of malignant tumours. Fortunately, the identification of Staphylococcal enterotoxin-like toxins (SEls), which possess amino acid sequences similar to those of classical SEs but exhibit no or low emetic activity, has provided a set of potential immunomodulatory candidates for cancer therapy. The aim of this study was to examine the effect of SElQ on lymphocyte activation and to further demonstrate its antitumour activity both in vitro and in vivo. High-purity SElQ was successfully harvested, and in vitro results confirmed that SElQ can significantly activate mouse- and human-derived lymphocytes in a dose-dependent manner, particularly CD4+ and CD8+ T cells, which showed significant increases in both percentage and absolute number. Further examination revealed that in addition to the originally recognized TCR Vß5 and 21, TCR Vß14, 17 and 18 were activated in SElQ-induced human PBMCs. Moreover, the expression of IL-2 and IFN-γ was significantly upregulated in vitro and in vivo after SElQ treatment. Based on the findings that SElQ induces lymphocyte activation and cytokine release, we then confirmed its antitumour activity both in vitro and in vivo. The data showed that treatment with a low concentration of SElQ (30 µg/mouse) could inhibit the growth of tumours by approximately 30% and no significant toxicity was observed. Taken together, our results demonstrated that SElQ can significantly induce T cell activation and cytokine release and further elicit substantial antitumour activity and thus provide support for the potential application of SElQ in cancer immunotherapy.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Enterotoxins/metabolism , Lymphocyte Activation/drug effects , Superantigens/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Immunologic , Female , Healthy Volunteers , Humans , Mice, Inbred BALB C , Receptors, Antigen, T-Cell/metabolism
7.
Biol Open ; 8(5)2019 May 02.
Article in English | MEDLINE | ID: mdl-31036750

ABSTRACT

Recently, menstrual blood-derived endometrial stem cells (MenSCs) have become attractive for stem cell based therapy due to their abundance, easy and non-invasive extraction and isolation process, high proliferative capacity, and multi-lineage differentiation potential. MenSC-based therapies for various diseases are being extensively researched. However, the high death rate and poor engraftment in sites of damaged tissues reduce the therapeutic value of these stem cells for transplantation. In theory, periodic stem cell transplantation is an alternative strategy to overcome the challenge of the loss of beneficial stem cell-derived effects due to the rapid disappearance of the stem cells in vivo However, periodic stem cell transplantation requires sufficient amounts of the desired stem cells with a low number of subculture passages. Our previous results have demonstrated that primary MenSCs mainly reside in the deciduous endometrium, and considerable amounts of deciduous endometrium intertwined with menstrual blood clots were discarded after conventional density gradient centrifugation (DGC). Therefore, the aim of this study was to determine whether primary MenSCs exist in the sedimentation of the deciduous endometrium after DGC and further to evaluate the isolation of MenSCs by direct red blood cell lysis treatment. As expected, our results confirmed that substantial amounts of primary MenSCs still remain in the sedimentation after DGC and indicated that MenSC isolation by directly lysing the red blood cells not only guaranteed substantial amounts of superior MenSCs with a low number of subculture passages, but also was time efficient and economical, providing a solid support for extensive clinical application.

8.
Plant Cell Rep ; 38(2): 183-194, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30499032

ABSTRACT

KEY MESSAGE: GARS encodes an enzyme catalyzing the second step of purine nucleotide biosynthesis and plays an important role to maintain the development of chloroplasts in juvenile plants by affecting the expression of plastid-encoded genes. A series of rice white striped mutants were previously described. In this research, we characterized a novel gars mutant with white striped leaves at the seedling stage. By positional cloning, we identified the mutated gene, which encodes a glycinamide ribonucleotide synthetase (GARS) that catalyzes the second step of purine nucleotide biosynthesis. Thylakoid membranes were less abundant in the albinic sectors of mutant seedling leaves compared to the wild type. The expression levels of genes involved in chlorophyll synthesis and photosynthesis were changed. Contents of ATP, ADP, AMP, GTP and GDP, which are crucial for plant growth and development, were decreased in the mutant seedlings. Complementation and CrispR tests confirmed the role of the GARS allele, which was expressed in all rice tissues, especially in the leaves. GARS protein displayed a typical chloroplast location pattern in rice protoplasts. Our results indicated that GARS was involved in chloroplast development at early leaf development by affecting the expression of plastid-encoded genes.


Subject(s)
Carbon-Nitrogen Ligases/genetics , Chloroplasts/metabolism , Genes, Plant , Oryza/enzymology , Oryza/genetics , Purine Nucleotides/biosynthesis , Biosynthetic Pathways/genetics , Carbon-Nitrogen Ligases/metabolism , Chlorophyll/biosynthesis , Chloroplasts/ultrastructure , Gene Expression Regulation, Plant , Mutation/genetics , Phenotype , Photosynthesis/genetics , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Breed Sci ; 65(5): 411-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26719744

ABSTRACT

Seed storability in rice (Oryza sativa L.) is an important agronomic trait. Two segregating populations with N22 (indica) as a common parent, viz. a set of 122 backcross-inbred lines (BILs) derived from the backcross Nanjing35 (japonica)/N22//Nanjing35 and another population comprising 189 recombinant inbred lines (RILs) from the cross of USSR5 (japonica) and N22, were studied to detect quantitative trait loci (QTL) controlling seed storability. Germination percentage (GP) was used to evaluate seed storability after aging treated under three different conditions, viz. natural, artificial and combined aging treatments. A total of seven QTLs were identified on chromosomes 1, 2, 5, 6 and 9. Among them, a major QTL, qSSn-9, was common in the two populations. In contrast, four QTLs (qSSnj-2-1, qSSn-2-2, qSSn-5 and qSSn-6) were detected in BILs and the QTL qSSn-1 was identified in RILs, which was a new QTL for seed storability. The N22-derived alleles increased the seed storability at all the loci except qSSnj-2-1. We also investigated the effect of QTLs using five selected lines with high storability from BILs and verified qSSn-5 with a near-isogenic line (NIL). These results provide an opportunity for pyramiding or map-based cloning major QTLs for seed storability in rice.

SELECTION OF CITATIONS
SEARCH DETAIL
...