Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 279
Filter
1.
BMC Genomics ; 25(1): 526, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807051

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a complicated neurodegenerative disease. Neuron-glial cell interactions are an important but not fully understood process in the progression of AD. We used bioinformatic methods to analyze single-nucleus RNA sequencing (snRNA-seq) data to investigate the cellular and molecular biological processes of AD. METHOD: snRNA-seq data were downloaded from Gene Expression Omnibus (GEO) datasets and reprocessed to identify 240,804 single nuclei from healthy controls and patients with AD. The cellular composition of AD was further explored using Uniform Manifold Approximation and Projection (UMAP). Enrichment analysis for the functions of the DEGs was conducted and cell development trajectory analyses were used to reveal underlying cell fate decisions. iTALK was performed to identify ligand-receptor pairs among various cell types in the pathological ecological microenvironment of AD. RESULTS: Six cell types and multiple subclusters were identified based on the snRNA-seq data. A subcluster of neuron and glial cells co-expressing lncRNA-SNHG14, myocardin-related transcription factor A (MRTFA), and MRTFB was found to be more abundant in the AD group. This subcluster was enriched in mitogen-activated protein kinase (MAPK)-, immune-, and apoptosis-related pathways. Through molecular docking, we found that lncRNA-SNHG14 may bind MRTFA and MRTFB, resulting in an interaction between neurons and glial cells. CONCLUSIONS: The findings of this study describe a regulatory relationship between lncRNA-SNHG14, MRTFA, and MRTFB in the six main cell types of AD. This relationship may contribute to microenvironment remodeling in AD and provide a theoretical basis for a more in-depth analysis of AD.


Subject(s)
Alzheimer Disease , Neuroglia , Neurons , Single-Cell Analysis , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Humans , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Cellular Microenvironment/genetics , Computational Biology/methods
2.
Mamm Genome ; 35(2): 256-279, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38538990

ABSTRACT

Unexplained recurrent miscarriage (URM) is a common pregnancy complication with few effective therapies. Moreover, little is known regarding the role of pyroptosis in the regulation of the URM immune microenvironment. To address this issue, gene expression profiles of publicly available placental datasets GSE22490 and GSE76862 were downloaded from the Gene Expression Omnibus database. Pyroptosis-related differentially expressed genes were identified and a total of 16 differentially expressed genes associated with pyroptosis were detected, among which 1 was upregulated and 15 were downregulated. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses indicated that the functionally enriched modules and pathways of these genes are closely related to immune and inflammatory responses. Four hub genes were identified: BTK, TLR8, NLRC4, and TNFSF13B. BTK, TLR8, and TNFSF13B were highly connected with immune cells, according to the correlation analysis of four hub genes and 20 different types of immune cells (p < 0.05). The four hub genes were used as research objects to construct the interaction networks. Chorionic villus tissue was used for quantitative real-time polymerase chain reaction and western blot to confirm the expression levels of hub genes, and the results showed that the expression of the four hub genes was significantly decreased in the chorionic villus tissue in the URM group. Collectively, the present study indicates that perhaps pyroptosis is essential to the diversity and complexity of the URM immune microenvironment, and provides a theoretical basis and research ideas for subsequent target gene verification and mechanism research.


Subject(s)
Abortion, Habitual , Pyroptosis , Humans , Female , Pyroptosis/genetics , Abortion, Habitual/genetics , Abortion, Habitual/immunology , Pregnancy , Gene Expression Profiling , Gene Regulatory Networks , Gene Ontology , Placenta/metabolism , Placenta/immunology , Transcriptome , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Gene Expression Regulation
3.
Reprod Biol Endocrinol ; 20(1): 54, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35313911

ABSTRACT

BACKGROUND: Both mild and conventional controlled ovarian stimulation are the frequently used protocols for poor ovarian responders. However, there are some debates about which treatment is better. Moreover, little is known about the follicular physiology after the two ovarian stimulation protocols. This study was intended to investigate the features in granulosa cells and follicular fluid micro-environment after the two different ovarian stimulation protocols in poor responders. METHODS: Granulosa cells RNA were sequenced using Illumina Hiseq technology. Specific differently expressed genes and proteins were verified by real-time quantitative PCR and Western blot analysis. Moreover, hormone and cytokine concentrations in the follicular fluid were measured by electrochemiluminescence immunoassay and enzyme-linked immunoabsorbent assay. The correlation between the results of molecular experiments and the laboratory outcomes were analyzed by Spearman correlation analysis. RESULTS: The differentially expressed genes between the two groups were involved in 4 signaling pathways related to the follicular development; three proteins pertinent to the TGF-ß signaling pathway were expressed differently in granulosa cells between the two, and the constituents in the follicular fluid were also different. Further, a correlation between the TGF-ß signaling pathway and the good-quality embryo was observed. CONCLUSIONS: The present study made a comparison for the first time in the transcriptome of human granulosa cells and the follicular fluid micro-environment between poor responders with the conventional controlled ovarian stimulation or the mild ovarian stimulation, showing that the TGF-ß signaling pathway may correlate with the good-quality of embryos in the mild group, which may be instrumental to the choice of optimal management for IVF patients.


Subject(s)
Follicular Fluid/metabolism , Granulosa Cells/metabolism , Infertility, Female/genetics , Ovulation Induction/methods , Transcriptome , Adult , Case-Control Studies , Cellular Microenvironment/genetics , Female , Follicular Fluid/chemistry , Gene Expression Profiling , Granulosa Cells/chemistry , High-Throughput Nucleotide Sequencing , Humans , Infertility, Female/metabolism , Infertility, Female/physiopathology , Oocyte Retrieval , Ovarian Reserve/genetics , Ovulation/genetics , Sequence Analysis, DNA , Treatment Failure
4.
J Exp Med ; 219(3)2022 03 07.
Article in English | MEDLINE | ID: mdl-35089323

ABSTRACT

Inflammation is associated with bone marrow failure syndromes, but how specific molecules impact the bone marrow microenvironment is not well elucidated. We report a novel role for the miR-145 target, Toll/interleukin-1 receptor domain containing adaptor protein (TIRAP), in driving bone marrow failure. We show that TIRAP is overexpressed in various types of myelodysplastic syndromes (MDS) and suppresses all three major hematopoietic lineages. TIRAP expression promotes up-regulation of Ifnγ, leading to myelosuppression through Ifnγ-Ifnγr-mediated release of the alarmin, Hmgb1, which disrupts the bone marrow endothelial niche. Deletion of Ifnγ blocks Hmgb1 release and is sufficient to reverse the endothelial defect and restore myelopoiesis. Contrary to current dogma, TIRAP-activated Ifnγ-driven bone marrow suppression is independent of T cell function or pyroptosis. In the absence of Ifnγ, TIRAP drives myeloproliferation, implicating Ifnγ in suppressing the transformation of MDS to acute leukemia. These findings reveal novel, noncanonical roles of TIRAP, Hmgb1, and Ifnγ in the bone marrow microenvironment and provide insight into the pathophysiology of preleukemic syndromes.


Subject(s)
Bone Marrow Failure Disorders/etiology , Bone Marrow Failure Disorders/metabolism , Endothelium/metabolism , HMGB1 Protein/metabolism , Interferon-gamma/metabolism , Membrane Glycoproteins/genetics , Myelopoiesis/genetics , Receptors, Interleukin-1/genetics , Animals , Biomarkers , Bone Marrow Failure Disorders/pathology , Cellular Microenvironment/genetics , Disease Susceptibility , Gene Expression , Hematopoiesis/genetics , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Receptors, Interleukin-1/metabolism
5.
Life Sci Alliance ; 5(3)2022 03.
Article in English | MEDLINE | ID: mdl-34853163

ABSTRACT

Acute-on-chronic liver failure (ACLF) is clinical syndrome with high mortality rate. This study aimed to perform detailed transcriptomic analysis in liver cirrhosis-based ACLF rats to elucidate ACLF pathogenesis. ACLF was induced by combined porcine serum with D-galactosamine and lipopolysaccharide. Gene expression profile of liver tissues from ACLF rats was generated by transcriptome sequencing to reveal the molecular mechanism. ACLF rats successfully developed with typical characteristics. Total of 2,354/3,576 differentially expressed genes were identified when ACLF was compared to liver cirrhosis and normal control, separately. The functional synergy analysis revealed prominent immune dysregulation at ACLF stage, whereas metabolic disruption was significantly down-regulated. Relative proportions of innate immune-related cells showed significant elevation of monocytes and macrophages, whereas adaptive immune-related cells were reduced. The seven differentially expressed genes underlying the ACLF molecular mechanisms were externally validated, among them THBS1, IL-10, and NR4A3 expressions were confirmed in rats, patient transcriptomics, and liver biopsies, verifying their potential value in the ACLF pathogenesis. This study indicates immune-metabolism disorder in ACLF rats, which may provide clinicians new targets for improving intervention strategies.


Subject(s)
Acute-On-Chronic Liver Failure/etiology , Acute-On-Chronic Liver Failure/metabolism , Disease Susceptibility , Energy Metabolism , Immunity , Acute-On-Chronic Liver Failure/pathology , Animals , Biomarkers , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Disease Models, Animal , Disease Susceptibility/immunology , Gene Expression Profiling , Gene Expression Regulation , Liver/immunology , Liver/metabolism , Liver/pathology , Rats , Transcriptome
6.
Immunology ; 165(2): 206-218, 2022 02.
Article in English | MEDLINE | ID: mdl-34775606

ABSTRACT

The immune system plays fundamental roles in the mammary gland, shaping developmental processes and controlling inflammation during infection and cancer.Here, we reveal unanticipated heterogeneity in the myeloid cell compartment duringdevelopment of virgin, pregnant, lactating and involuting mouse mammary glands,and in milk. We investigate the functional consequences of individual and compoundchemokine receptor deficiency on cell recruitment. Diverse myeloid cell recruitmentwas also shown in models of sterile inflammation and bacterial infection.Strikingly, we have shown that inflammation and infection can alter the abundanceof terminal end buds, a key developmental structure, within the pubertal mammarygland. This previously unknown effect of inflammatory burden during puberty couldhave important implications for understanding pubertal development.


Subject(s)
Disease Susceptibility , Mastitis/etiology , Mastitis/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Animals , Biomarkers , Biopsy , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry , Granulocytes/immunology , Granulocytes/metabolism , Immunohistochemistry , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mastitis/pathology , Mice , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/pathology
7.
Front Immunol ; 12: 770852, 2021.
Article in English | MEDLINE | ID: mdl-34868029

ABSTRACT

Autoimmune thyroiditis (AIT) is the most prevalent autoimmune endocrine disease, with a higher incidence in women than in men. Immunological abnormalities may lead to the impairment of ovarian folliculogenesis; however, whether the presence of AIT affects immunological microenvironment in follicles remains controversial. We performed a cross-sectional study including 122 patients, aged 20-40 years, who underwent IVF/ICSI treatment owing to isolated male or tube factor infertility. Patients were divided into AIT and control groups according to clinical presentation, thyroid function, and thyroid autoantibody measurements. Follicular fluid was collected and the distribution of cytokines/chemokines in follicular fluid was measured by flow cytometry using multiplex bead assays between the two groups. Based on differences in levels of intrafollicular chemokines and cytokines between the AIT and control groups, the relevant inflammatory cascade was further demonstrated. Among the 12 chemokines analyzed, three (CXCL9, CXCL10, and CXCL11) showed significantly elevated levels in the follicular fluid of patients with AIT. Among the 11 cytokines detected, compared with those in the control group, significantly higher levels of IFNγ were observed in patients with AIT. IFNγ dose-dependently stimulated the expression and secretion of CXCL9/10/11 in cultured primary granulosa cells. The percentage of CXCR3+ T lymphocytes was significantly elevated in the follicular microenvironment of patients with AIT. We concluded that the IFNγ-CXCL9/10/11-CXCR3+ T lymphocyte inflammatory cascade is activated in the follicular microenvironment of patients with AIT. These findings indicate that a considerable immune imbalance occurred in the follicular microenvironment of patients with AIT.


Subject(s)
Cellular Microenvironment/immunology , Cytokines/immunology , Follicular Fluid/immunology , Thyroiditis, Autoimmune/immunology , Adult , Cells, Cultured , Cellular Microenvironment/genetics , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Chemokine CXCL11/genetics , Chemokine CXCL11/immunology , Chemokine CXCL11/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Chemokine CXCL9/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Fertilization in Vitro , Flow Cytometry , Follicular Fluid/metabolism , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction , Sperm Injections, Intracytoplasmic , Thyroiditis, Autoimmune/genetics , Thyroiditis, Autoimmune/metabolism
8.
Cells ; 10(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34943805

ABSTRACT

Direct reprogramming of fibroblasts into CM-like cells has emerged as an attractive strategy to generate induced CMs (iCMs) in heart regeneration. However, low conversion rate, poor purity, and the lack of precise conversion of iCMs are still present as significant challenges. In this review, we summarize the recent development in understanding the molecular mechanisms of cardiac reprogramming with various strategies to achieve more efficient iCMs. reprogramming. Specifically, we focus on the identified critical roles of transcriptional regulation, epigenetic modification, signaling pathways from the cellular microenvironment, and cell cycling regulation in cardiac reprogramming. We also discuss the progress in delivery system optimization and cardiac reprogramming in human cells related to preclinical applications. We anticipate that this will translate cardiac reprogramming-based heart therapy into clinical applications. In addition to optimizing the cardiogenesis related transcriptional regulation and signaling pathways, an important strategy is to modulate the pathological microenvironment associated with heart injury, including inflammation, pro-fibrotic signaling pathways, and the mechanical properties of the damaged myocardium. We are optimistic that cardiac reprogramming will provide a powerful therapy in heart regenerative medicine.


Subject(s)
Cellular Reprogramming , Heart/physiology , Regeneration , Translational Science, Biomedical , Animals , Cellular Microenvironment/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Humans , Regeneration/genetics
9.
RNA Biol ; 18(sup2): 866-880, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34843419

ABSTRACT

Subcellular localization of transcripts is highly associated with regulation of gene expression, synthesis of protein, and also the development of the human brain cortex. Although many mechanisms are prevalent in the occurrence of neuroinflammation, the mechanisms based on differences in subcellular localization of transcripts have not been explored. To characterize the dynamic profile of nuclear and cytoplasmic transcripts during the progress of haemorrhage-induced neuroinflammation, we isolated nucleo-cytoplasmic RNA fractions of oxyhaemoglobin (oxy-Hb) treated microglia cells and sequenced both fractions. We discovered that cytoplasmic retained genes were the major forces to maintain the neuroinflammatory microenvironment with 10 hub genes and 40 conserved genes were identified. Moreover, antisense RNA Gm44096 and lincRNA Gm47270, which co-expressed with a crowd of inflammatory genes in the cytoplasm, were discovered as regulatory strategies for sustaining the neuroinflammatory microenvironment. Thus, our study provides a new perspective on understanding haemorrhage-induced neuroinflammation and also reveals a mechanism of lncRNA responsible for maintaining the neuroinflammatory microenvironment.


Subject(s)
Cell Nucleus/metabolism , Cellular Microenvironment/genetics , Cytoplasm/metabolism , Neuroinflammatory Diseases/etiology , RNA Transport , Animals , Cell Line , Cell Nucleus/genetics , Computational Biology/methods , Cytoplasm/genetics , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Gene Ontology , Hemorrhage/complications , Mice , Microglia/metabolism , Neuroinflammatory Diseases/metabolism , RNA, Long Noncoding/genetics
10.
J Cell Mol Med ; 25(21): 10236-10247, 2021 11.
Article in English | MEDLINE | ID: mdl-34647423

ABSTRACT

N6-methyladenosine (m6A) modification is one of the most prevalent RNA modification forms of eukaryotic mRNA and is an important post-transcriptional mechanism for regulating genes. However, the role of m6A modification in the regulation of severe asthma has never been reported. Thus, we aimed to investigate the m6A regulator-mediated RNA methylation modification patterns and immune microenvironment infiltration characterization in severe asthma. In this study, 87 healthy controls and 344 severe asthma cases from the U-BIOPRED (Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes) programme were used to systematically evaluate the m6A modification patterns mediated by 27 m6A regulators and to investigate the effects of m6A modification on immune microenvironment characteristics. We found that 16 m6A regulators were abnormal and identified two key m6A regulators (YTHDF3 and YTHDC1) and three m6A modification patterns. The study of infiltration characteristics of immune microenvironment found that pattern 2 had more infiltrating immune cells and more active immune response. Besides, it was found that the eosinophils which are very important for severe asthma were affected by YTHDF3 and EIF3B. We also verified key m6A regulators with merip-seq and found that they were mainly distributed in exons and enriched in 3'UTR. In conclusion, our findings suggested that m6A modification plays a key role in severe asthma, and may be able to guide the future strategy of immunotherapy.


Subject(s)
Adenosine/analogs & derivatives , Asthma/etiology , Asthma/metabolism , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , RNA, Messenger/genetics , Adenosine/metabolism , Animals , Asthma/diagnosis , Biomarkers , Computational Biology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Methylation , Mice , Models, Biological , RNA, Messenger/metabolism , ROC Curve , Severity of Illness Index , Transcriptome
11.
Biomolecules ; 11(10)2021 09 25.
Article in English | MEDLINE | ID: mdl-34680038

ABSTRACT

Cellular metabolism alterations have been recognized as one of the most predominant hallmarks of colorectal cancers (CRCs). It is precisely regulated by many oncogenic signaling pathways in all kinds of regulatory levels, including transcriptional, post-transcriptional, translational and post-translational levels. Among these regulatory factors, epigenetics play an essential role in the modulation of cellular metabolism. On the one hand, epigenetics can regulate cellular metabolism via directly controlling the transcription of genes encoding metabolic enzymes of transporters. On the other hand, epigenetics can regulate major transcriptional factors and signaling pathways that control the transcription of genes encoding metabolic enzymes or transporters, or affecting the translation, activation, stabilization, or translocation of metabolic enzymes or transporters. Interestingly, epigenetics can also be controlled by cellular metabolism. Metabolites not only directly influence epigenetic processes, but also affect the activity of epigenetic enzymes. Actually, both cellular metabolism pathways and epigenetic processes are controlled by enzymes. They are highly intertwined and are essential for oncogenesis and tumor development of CRCs. Therefore, they are potential therapeutic targets for the treatment of CRCs. In recent years, both epigenetic and metabolism inhibitors are studied for clinical use to treat CRCs. In this review, we depict the interplay between epigenetics and cellular metabolism in CRCs and summarize the underlying molecular mechanisms and their potential applications for clinical therapy.


Subject(s)
Cellular Microenvironment/genetics , Colorectal Neoplasms/genetics , Epigenesis, Genetic/genetics , Transcription Factors/metabolism , Carcinogenesis/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic/genetics , Histones/metabolism , Humans , Signal Transduction/genetics
12.
Sci Rep ; 11(1): 21110, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702961

ABSTRACT

Cryptococcus deneoformans is an opportunistic fungal pathogen that infects the lungs via airborne transmission and frequently causes fatal meningoencephalitis. Claudins (Cldns), a family of proteins with 27 members found in mammals, form the tight junctions within epithelial cell sheets. Cldn-4 and 18 are highly expressed in airway tissues, yet the roles of these claudins in respiratory infections have not been clarified. In the present study, we analyzed the roles of Cldn-4 and lung-specific Cldn-18 (luCldn-18) in host defense against C. deneoformans infection. luCldn-18-deficient mice exhibited increased susceptibility to pulmonary infection, while Cldn-4-deficient mice had normal fungal clearance. In luCldn-18-deficient mice, production of cytokines including IFN-γ was significantly decreased compared to wild-type mice, although infiltration of inflammatory cells including CD4+ T cells into the alveolar space was significantly increased. In addition, luCldn-18 deficiency led to high K+ ion concentrations in bronchoalveolar lavage fluids and also to alveolus acidification. The fungal replication was significantly enhanced both in acidic culture conditions and in the alveolar spaces of luCldn-18-deficient mice, compared with physiological pH conditions and those of wild-type mice, respectively. These results suggest that luCldn-18 may affect the clinical course of cryptococcal infection indirectly through dysregulation of the alveolar space microenvironment.


Subject(s)
Cellular Microenvironment/immunology , Claudins/deficiency , Cryptococcosis/immunology , Cryptococcus/immunology , Lung/immunology , Pneumonia/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cellular Microenvironment/genetics , Claudins/immunology , Cryptococcosis/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Lung/microbiology , Mice , Mice, Knockout , Organ Specificity/genetics , Organ Specificity/immunology , Pneumonia/genetics , Pneumonia/microbiology
13.
Cell Stress Chaperones ; 26(6): 937-944, 2021 11.
Article in English | MEDLINE | ID: mdl-34495492

ABSTRACT

Human periodontal ligament fibroblast (HPDLF) is a major component of the resident cells in the periodontal microenvironment, and plays important roles in periodontitis through multiple mechanisms. Although lipopolysaccharide (LPS) has been shown to cause endoplasmic reticulum (ER) stress and activate the unfolded protein response (UPR) in HPDLF, the mechanisms governing HPDLF function in periodontitis are unclear. In this study, we tested the ability of unfolded protein response (UPR) to regulate HPDLF in vitro and examined the underlying mechanisms. We found LPS-pretreated HPDLF induced macrophage polarization toward M1 phenotype. UPR activation reduced the inflammatory cytokine production and downregulated the expression of TLR4 in HPDLF. The phosphorylation of NF-κB p65 and I-κB was also inhibited by UPR activation. Our findings demonstrate that the connection of LPS, UPR, and HPDLF may represent a new concrete theory of innate immunity regulation in periodontal diseases, and suggest that targeting of UPR in HPDLF may be developed as a potent therapy for periodontitis.


Subject(s)
Inflammation/genetics , Periodontal Ligament/metabolism , Periodontitis/genetics , Unfolded Protein Response/genetics , Cell Polarity/genetics , Cellular Microenvironment/genetics , Endoplasmic Reticulum Stress/drug effects , Fibroblasts/metabolism , Humans , Immunity, Innate/genetics , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/pathology , NF-kappa B/genetics , Periodontal Ligament/pathology , Periodontitis/therapy , Phosphorylation , Transcription Factor RelA/genetics
14.
Endocrinology ; 162(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34467975

ABSTRACT

Islet function is critical for normal glucose homeostasis. Unlike adult ß cells, fetal and neonatal islets are more proliferative and have decreased insulin secretion in response to stimuli. However, the underlying mechanisms governing functional maturity of islets have not been completely elucidated. Pancreatic islets comprise different cell types. The microenvironment of islets and interactions between these cell types are critical for ß-cell development and maturation. Thus, the study of intact islets is optimal to identify novel molecular mechanisms controlling islet functional development. Transcriptomes and genome-wide histone landscapes of H3K4me3, H3K27me3, and H3K27Ac from intact islets isolated from 2- and 10-week-old Sprague-Dawley rats were integrated to elucidate genes and pathways modulating islet development, as well as the contribution of epigenetic regulation. A total of 4489 differentially expressed genes were identified; 2289 and 2200 of them were up- and down-regulated in 10-week islets, respectively. Ingenuity Pathway Analysis revealed critical pathways regulating functional maturation of islets, including nutrient sensing, neuronal function, immune function, cell replication, and extracellular matrix. Furthermore, we identified significant changes in enrichment of H3K4me3, H3K27me3, and H3K27Ac marks, which correlated with expression changes of genes critical for islet function. These histone marks were enriched at critical transcription factor-binding motifs, such as Hoxa9, C/EBP-ß, Gata1, Foxo1, E2f1, E2f3, and Mafb. In addition, our chromatin immunoprecipitation sequencing data revealed multiple potential bivalent genes whose poised states changed with maturation. Collectively, our current study identified critical novel pathways for mature islet function and suggested a role for histone modifications in regulating islet development and maturation.


Subject(s)
Cell Differentiation/genetics , Insulin-Secreting Cells/physiology , Islets of Langerhans/growth & development , Animals , Cellular Microenvironment/genetics , Energy Metabolism/genetics , Epigenesis, Genetic/physiology , Epigenome/physiology , Gene Expression Regulation , Islets of Langerhans/immunology , Islets of Langerhans/innervation , Islets of Langerhans/physiology , Rats , Rats, Sprague-Dawley , Transcriptome/physiology
15.
Arq. bras. neurocir ; 40(3): 253-256, 15/09/2021.
Article in English | LILACS | ID: biblio-1362147

ABSTRACT

Tumor heterogeneity is the concept that different tumor cells provide distinct biomorphological lesions, gene expressions, proliferation, microenvironment and graduated capacity of metastatic lesions. Brain tumor heterogeneity has been recently discussed about the interesting interaction of chronic inflammation, microenvironment, epigenetics and glioma steam cells. Brain tumors remain a challenge with regards to medication and disease, due to the lack of treatment options and unsatisfactory results. These results might be the result of the brain tumor heterogeneity and its multiple resistance mechanisms to chemo and radiotherapy.


Subject(s)
Neoplastic Stem Cells/cytology , Brain Neoplasms/genetics , Genetic Heterogeneity , Gene Expression Profiling , Glioma/genetics , Receptor Protein-Tyrosine Kinases/genetics , Drug Resistance, Neoplasm/genetics , Stem Cell Niche/genetics , Tumor Microenvironment , Clonal Evolution/genetics , Cellular Microenvironment/genetics , RNA-Seq
16.
Genome Med ; 13(1): 133, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34412659

ABSTRACT

BACKGROUND: The enrichment of Gram-negative bacteria of oral origin in the esophageal microbiome has been associated with the development of metaplasia. However, to date, no study has comprehensively assessed the relationships between the esophageal microbiome and the host. METHODS: Here, we examine the esophageal microenvironment in gastro-esophageal reflux disease and metaplasia using multi-omics strategies targeting the microbiome and host transcriptome, followed by targeted culture, comparative genomics, and host-microbial interaction studies of bacterial signatures of interest. RESULTS: Profiling of the host transcriptome from esophageal mucosal biopsies revealed profound changes during metaplasia. Importantly, five biomarkers showed consistent longitudinal changes with disease progression from reflux disease to metaplasia. We showed for the first time that the esophageal microbiome is distinct from the salivary microbiome and the enrichment of Campylobacter species as a consistent signature in disease across two independent cohorts. Shape fitting and matrix correlation identified associations between the microbiome and host transcriptome profiles, with a novel co-exclusion relationship found between Campylobacter and napsin B aspartic peptidase. Targeted culture of Campylobacter species from the same cohort revealed a subset of isolates to have a higher capacity to survive within primary human macrophages. Comparative genomic analyses showed these isolates could be differentiated by specific genomic features, one of which was validated to be associated with intracellular fitness. Screening for these Campylobacter strain-specific signatures in shotgun metagenomics data from another cohort showed an increase in prevalence with disease progression. Comparative transcriptomic analyses of primary esophageal epithelial cells exposed to the Campylobacter isolates revealed expression changes within those infected with strains with high intracellular fitness that could explain the increased likelihood of disease progression. CONCLUSIONS: We provide a comprehensive assessment of the esophageal microenvironment, identifying bacterial strain-specific signatures with high relevance to progression of metaplasia.


Subject(s)
Barrett Esophagus/etiology , Barrett Esophagus/metabolism , Biomarkers , Cellular Microenvironment , Disease Susceptibility , Esophagus/metabolism , Adult , Barrett Esophagus/pathology , Cellular Microenvironment/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Esophagus/microbiology , Esophagus/pathology , Female , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/etiology , Gene Expression Profiling , Gram-Negative Bacterial Infections/complications , Gram-Negative Bacterial Infections/microbiology , Host-Pathogen Interactions/genetics , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Mast Cells/immunology , Mast Cells/metabolism , Metaplasia , Microbiota , Middle Aged , Models, Biological , RNA, Ribosomal, 16S
17.
Dev Cell ; 56(13): 1875-1883, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34256927

ABSTRACT

Cell fate decisions are critical for adequate tissue development, maintenance and regeneration. In the mammary gland, epithelial cell fates are tightly controlled by the microenvironment. Here, we review how cell fate decisions are regulated by components of the microenvironment during mammary gland development and how pathological changes in the microenvironment can alter cell fates, leading to malignancy. Specifically, we describe the current understanding of how mammary cell fate is controlled and directed by three elements: the extracellular matrix, the immune microenvironment, and hormones-and how these elements can converge to create microenvironments that promote a fourth element: DNA damage.


Subject(s)
Cellular Microenvironment/genetics , Extracellular Matrix/genetics , Mammary Glands, Animal/growth & development , Mammary Glands, Human/growth & development , Animals , Breast/growth & development , Breast/pathology , Cell Differentiation/genetics , Cell Lineage/genetics , Female , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment
18.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34205072

ABSTRACT

Two-photon microscopy enables monitoring cellular dynamics and communication in complex systems, within a genuine environment, such as living tissues and, even, living organisms. Particularly, its application to understand cellular interactions in the immune system has brought unique insights into pathophysiologic processes in vivo. Simultaneous multiplexed imaging is required to understand the dynamic orchestration of the multiple cellular and non-cellular tissue compartments defining immune responses. Here, we present an improvement of our previously developed method, which allowed us to achieve multiplexed dynamic intravital two-photon imaging, by using a synergistic strategy. This strategy combines a spectrally broad range of fluorophore emissions, a wave-mixing concept for simultaneous excitation of all targeted fluorophores, and an unmixing algorithm based on the calculation of spectral similarities with previously measured fluorophore fingerprints. The improvement of the similarity spectral unmixing algorithm here described is based on dimensionality reduction of the mixing matrix. We demonstrate its superior performance in the correct pixel-based assignment of probes to tissue compartments labeled by single fluorophores with similar spectral fingerprints, as compared to the full-dimensional similarity spectral unmixing approach.


Subject(s)
Cell Communication/genetics , Cellular Microenvironment/genetics , Microscopy, Fluorescence/methods , Molecular Imaging/methods , Algorithms , Cell Line , Fluorescent Dyes/chemistry , Photons
19.
PLoS One ; 16(7): e0254802, 2021.
Article in English | MEDLINE | ID: mdl-34310634

ABSTRACT

Spermatozoa released from testes undergo a maturation process and acquire the capacity to fertilize ova through epididymal transit. The epididymis is divided into four regions, each with unique morphology, gene profile, luminal microenvironment and distinct function. To study the functions of relevant genes in the epididymal initial segment (IS), a novel IS-specific mouse model, Lcn9-Cre knock-in (KI) mouse line was generated via CRISPR/Cas9 technology. The TAG stop codon was replaced by a 2A-NLS-Cre cassette, resulting in the co-expression of Lcn9 and Cre recombinase. IS-specific Cre expression was first observed from postnatal day 17. Using the Rosa26tdTomato reporter mice, the Cre-mediated DNA recombination was detected exclusively in principal cells. The epididymal IS-specific Cre activity in vivo was further confirmed using Lcn9-Cre mice crossed with a mouse strain carrying Tsc1 floxed alleles (Tsc1flox/+). Cre expression did not affect either normal development or male fecundity. Different from any epididymis-specific Cre mice reported previously, the novel Lcn9-Cre mouse line can be used to introduce entire IS-specific conditional gene editing for gene functional study.


Subject(s)
Cellular Microenvironment/genetics , Epididymis/metabolism , Integrases/genetics , Lipocalins/genetics , Alleles , Animals , Epididymis/anatomy & histology , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Recombination, Genetic/genetics , Spermatozoa/growth & development , Spermatozoa/metabolism , Testis/growth & development , Testis/metabolism
20.
Dev Cell ; 56(13): 1821-1832, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34192527

ABSTRACT

Developmental myelination is a protracted process that extends well into postnatal life. Cell-intrinsic mechanisms operate in myelin-forming oligodendrocytes, as well as microenvironmental interactions that guide and modulate every aspect of myelination, from oligodendrocyte precursor cell migration to oligodendrocyte differentiation and the formation of stable myelin internodes. During development and throughout adult life, neuron-oligodendroglial interactions shape activity and experience-dependent myelin adaptations to fine-tune neural circuit dynamics and promote healthy neurological function.


Subject(s)
Cellular Microenvironment/genetics , Myelin Sheath/genetics , Neurons/metabolism , Oligodendroglia/metabolism , Cell Differentiation/genetics , Cell Movement/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...