RESUMO
Sugarcane smut caused by Sporisorium scitamineum represents the most destructive disease in the sugarcane industry, causing host hormone disruption and producing a black whip-like sorus in the apex of the stalk. In this study, the gibberellin metabolic pathway was found to respond to S. scitamineum infection, and the contents of bioactive gibberellins were significantly reduced in the leaves of diseased plants. The gibberellin receptor gene ScGID1 was identified and significantly downregulated. ScGID1 localized in both the nucleus and cytoplasm and had the highest expression level in the leaves. Eight proteins that interact with ScGID1 were screened out using a yeast two-hybrid assay. Novel DELLA proteins named ScGAI1a and ScGA20ox2, key enzymes in GA biosynthesis, were both found to interact with ScGID1 in a gibberellin-independent manner. Transcription factor trapping with a yeast one-hybrid system identified 50 proteins that interacted with the promoter of ScGID1, among which ScS1FA and ScPLATZ inhibited ScGID1 transcription, while ScGDSL promoted transcription. Overexpression of ScGID1 in transgenic Nicotiana benthamiana plants could increase plant height and promote flowering. These results not only contribute to improving our understanding of the metabolic regulatory network of sugarcane gibberellin but also expand our knowledge of the interaction between sugarcane and pathogens.
Assuntos
Regulação da Expressão Gênica de Plantas , Giberelinas , Proteínas de Plantas , Saccharum , Saccharum/genética , Saccharum/metabolismo , Giberelinas/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Nicotiana/genética , Nicotiana/metabolismo , Doenças das Plantas/genética , Doenças das Plantas/microbiologia , Folhas de Planta/metabolismo , Folhas de Planta/genética , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genéticaRESUMO
Background: Non-alcoholic fatty liver disease (NAFLD) and heart failure (HF) are related conditions with an increasing incidence. However, the mechanism underlying their association remains unclear. This study aimed to explore the shared pathogenic mechanisms and common biomarkers of NAFLD and HF through bioinformatics analyses and experimental validation. Methods: NAFLD and HF-related transcriptome data were extracted from the Gene Expression Omnibus (GEO) database (GSE126848 and GSE26887). Differential analysis was performed to identify common differentially expressed genes (co-DEGs) between NAFLD and HF. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) were conducted to explore the functions and regulatory pathways of co-DEGs. Protein-protein interaction (PPI) network and support vector machine-recursive feature elimination (SVM-RFE) methods were used to screen common key DEGs. The diagnostic value of common key DEGs was assessed by receiver operating characteristic (ROC) curve and validated with external datasets (GSE89632 and GSE57345). Finally, the expression of biomarkers was validated in mouse models. Results: A total of 161 co-DEGs were screened out in NAFLD and HF patients. GO, KEGG, and GSEA analyses indicated that these co-DEGs were mainly enriched in immune-related pathways. PPI network revealed 14 key DEGs, and SVM-RFE model eventually identified two genes (CD163 and CCR1) as common key DEGs for NAFLD and HF. Expression analysis revealed that the expression levels of CD163 and CCR1 were significantly down-regulated in HF and NAFLD patients. ROC curve analysis showed that CD163 and CCR1 had good diagnostic values for HF and NAFLD. Single-gene GSEA suggested that CD163 and CCR1 were mainly engaged in immune responses and inflammation. Experimental validation indicated unbalanced macrophage polarization in HF and NAFLD mouse models, and the expression of CD163 and CCR1 were significantly down-regulated. Conclusion: This study identified M2 polarization impairment characterized by decreased expression of CD163 and CCR1 as a common pathogenic pathway in NAFLD and HF. The downregulation of CD163 and CCR1 may reflect key pathological changes in the development and progression of NAFLD and HF, suggesting their potential as diagnostic and therapeutic targets.
Assuntos
Biomarcadores , Biologia Computacional , Perfilação da Expressão Gênica , Insuficiência Cardíaca , Hepatopatia Gordurosa não Alcoólica , Mapas de Interação de Proteínas , Hepatopatia Gordurosa não Alcoólica/genética , Animais , Humanos , Camundongos , Insuficiência Cardíaca/genética , Biologia Computacional/métodos , Transcriptoma , Redes Reguladoras de Genes , Modelos Animais de Doenças , Ontologia Genética , Bases de Dados Genéticas , Antígenos CD/genética , Receptores de Superfície Celular/genética , Transdução de Sinais/genética , Antígenos de Diferenciação Mielomonocítica/genética , Camundongos Endogâmicos C57BL , MasculinoRESUMO
The axon guidance cue netrin-1 signals through its receptor DCC (deleted in colorectal cancer) to attract commissural axons to the midline. Variants in DCC are frequently associated with congenital mirror movements (CMMs). A CMM-associated variant in the cytoplasmic tail of DCC is located in a conserved motif predicted to bind to a regulator of actin dynamics called the WAVE (Wiskott-Aldrich syndrome protein-family verprolin homologous protein) regulatory complex (WRC). Here, we explored how this variant affects DCC function and may contribute to CMM. We found that a conserved WRC-interacting receptor sequence (WIRS) motif in the cytoplasmic tail of DCC mediated the interaction between DCC and the WRC. This interaction was required for netrin-1-mediated axon guidance in cultured rodent commissural neurons. Furthermore, the WIRS motif of Fra, the Drosophila DCC ortholog, was required for attractive signaling in vivo at the Drosophila midline. The CMM-associated R1343H variant of DCC, which altered the WIRS motif, prevented the DCC-WRC interaction and impaired axon guidance in cultured commissural neurons and in Drosophila. The findings reveal the WRC as a pivotal component of netrin-1-DCC signaling and uncover a molecular mechanism explaining how a human genetic variant in the cytoplasmic tail of DCC may lead to CMM.
Assuntos
Orientação de Axônios , Receptor DCC , Proteínas de Drosophila , Netrina-1 , Netrina-1/metabolismo , Netrina-1/genética , Receptor DCC/metabolismo , Receptor DCC/genética , Animais , Humanos , Orientação de Axônios/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Ratos , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Axônios/metabolismo , Axônios/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genética , Transdução de Sinais , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Camundongos , Neurônios/metabolismo , Células HEK293 , Receptores de NetrinaRESUMO
KEY MESSAGE: The Arabidopsis RNA helicase LOS4 plays a key role in regulating pre-mRNA splicing of the genes EIN2, CTR1, and ERS2 in ethylene signaling pathway. The plant hormone ethylene plays diverse roles in plant growth, development, and responses to stress. Ethylene is perceived by the membrane-bound ethylene receptors complex, and then triggers downstream components, such as EIN2, to initiate signal transduction into the nucleus, leading to the activation of ethylene-responsive genes. Over the past decades, substantial information has been accumulated regarding gene cloning, protein-protein interactions, and downstream gene expressions in the ethylene pathway. However, our understanding of mRNA post-transcriptional processing and modification of key genes in the ethylene signaling pathway remains limited. This study aims to provide evidence demonstrating the involvement of the Arabidopsis RNA helicase LOS4 in pre-mRNA splicing of the genes EIN2, CTR1, and ERS2 in ethylene signaling pathway. Various genetic approaches including RNAi gene silencing, CRISPR-Cas9 gene editing, and amino acid mutations were employed in this study. When LOS4 was silenced or knocked down, the ethylene sensitivity of etiolated seedlings was significantly enhanced. Further investigation revealed errors in the EIN2 pre-mRNA splicing when LOS4 was knocked down. In addition, aberrant pre-mRNA splicing was observed in the ERS2 and CTR1 genes in the pathway. Biochemical assays indicated that the los4-2 (E94K) mutant protein exhibited increased ATP binding and enhanced ATP hydrolytic activity. Conversely, the los4-1 (G364R) mutant had reduced substrate RNA binding and lower ATP binding activities. These findings significantly advanced our comprehension of the regulatory functions and molecular mechanisms of RNA helicase in ethylene signaling.
Assuntos
Proteínas de Arabidopsis , Arabidopsis , Etilenos , Regulação da Expressão Gênica de Plantas , Splicing de RNA , Transdução de Sinais , Etilenos/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Transdução de Sinais/genética , Splicing de RNA/genética , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genética , Precursores de RNA/genética , Precursores de RNA/metabolismo , RNA Helicases/metabolismo , RNA Helicases/genética , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Proteínas QuinasesRESUMO
Solute carrier family 37 member 4 (SLC37A4) is known to regulate glucose-6-phosphate transport from cytoplasm to the lumen of the endoplasmic reticulum, which serves to maintain glucose homeostasis. Glycogen storage disease type 1b (GSD1b) is caused by a mutation of SLC37A4, leading to a glycogenolysis defect. Although GSD1b cases are known to be complicated by periodontitis, the etiological molecular basis remains unclear. The present study investigated the effects of SLC37A4 on gingival barrier function. Examinations of immortalized human gingival epithelial (IHGE) cells showed SLC37A4 localized in the endoplasmic reticulum. SLC37A4 knockout decreased expression of JAM1, a tight junction-related protein, in IHGE cells. Using in silico analysis to investigate potential transcription factor binding sites, H6 family homeobox 3 (HMX3) was shown to be related to JAM1 expression. In HMX3-knockdown IHGE cells, JAM1 expression was markedly suppressed. Furthermore, HMX3 was scarcely detected in SLC37A4-knockout cells, while HMX3 overexpression restored JAM1 expression in those cells. Finally, using a three-dimensional multilayered gingival epithelial tissue model, knockout of SLC37A4 was also found to increase permeability to lipopolysaccharide and peptidoglycan, which was dependent on JAM1 expression. Specific downregulation of HMX3 by SLC37A4 and the consequent decrease in JAM1 expression provides findings indicating a molecular basis for the reduction in barrier function of gingival epithelial tissues in GSD1b cases.
Assuntos
Células Epiteliais , Gengiva , Receptores de Superfície Celular , Humanos , Gengiva/metabolismo , Células Epiteliais/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Regulação da Expressão Gênica , Linhagem Celular , Lipopolissacarídeos , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Doença de Depósito de Glicogênio Tipo I , AntiportersRESUMO
Atherosclerosis results from lipid-driven inflammation of the arterial wall that fails to resolve. Imbalances in macrophage accumulation and function, including diminished migratory capacity and defective efferocytosis, fuel maladaptive inflammation and plaque progression. The neuroimmune guidance cue netrin-1 has dichotomous roles in inflammation partly due to its multiple receptors; in atherosclerosis, netrin-1 promotes macrophage survival and retention via its receptor Unc5b. To minimize the pleiotropic effects of targeting netrin-1, we tested the therapeutic potential of deleting Unc5b in mice with advanced atherosclerosis. We generated Unc5bfl/flCx3cr1creERT2/WT mice, which allowed conditional deletion of Un5b (∆Unc5bMØ) in monocytes and macrophages by tamoxifen injection. After inducing advanced atherosclerosis by hepatic PCSK9 overexpression and western diet feeding for 20 wk, Unc5b was deleted and hypercholesterolemia was normalized to simulate clinical lipid management. Deletion of myeloid Unc5b led to a 40% decrease in atherosclerotic plaque burden and reduced plaque complexity compared to Unc5bfl/flCx3cr1WT/WT littermate controls (CtrlMØ). Consistently, plaque macrophage content was reduced by 50% in ∆Unc5bMØ mice due to reduced plaque Ly6Chi monocyte recruitment and macrophage retention. Compared to CtrlMØ mice, plaques in ∆Unc5bMØ mice had reduced necrotic area and fewer apoptotic cells, which correlated with improved efferocytotic capacity by Unc5b-deficient macrophages in vivo and in vitro. Beneficial changes in macrophage dynamics in the plaque upon Unc5b deletion were accompanied by an increase in atheroprotective T cell populations, including T-regulatory and Th2 cells. Our data identify Unc5b in advanced atherosclerosis as a therapeutic target to induce pro-resolving restructuring of the plaque immune cells and to promote atherosclerosis regression.
Assuntos
Aterosclerose , Macrófagos , Receptores de Netrina , Placa Aterosclerótica , Animais , Masculino , Camundongos , Aterosclerose/imunologia , Aterosclerose/patologia , Aterosclerose/metabolismo , Inflamação/patologia , Inflamação/metabolismo , Inflamação/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Receptores de Netrina/metabolismo , Netrina-1/metabolismo , Netrina-1/genética , Placa Aterosclerótica/patologia , Placa Aterosclerótica/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genéticaRESUMO
BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disease that causes cognitive dysfunction in older adults. One of the AD pathological factors, ß-Amyloid (Aß), triggers inflammatory responses and phagocytosis of microglia. C-type lectin domain family 5 member A (CLEC5A) induces over-reactive inflammatory responses in several virus infections. Yet, the role of CLEC5A in AD progression remains unknown. This study aimed to elucidate the contribution of CLEC5A to Aß-induced microglial activation and behavioral deficits. METHODS: The AD mouse model was crossed with Clec5a knockout mice for subsequent behavioral and pathological tests. The memory deficit was revealed by the Morris water maze, while the nociception abnormalities were examined by the von Frey filament and hotplate test. The Aß deposition and microglia recruitment were identified by ELISA and immunohistochemistry. The inflammatory signals were identified by ELISA and western blotting. In the Clec5a knockdown microglial cell model and Clec5a knockout primary microglia, the microglial phagocytosis was revealed using the fluorescent-labeled Aß. RESULTS: The AD mice with Clec5a knockout improved Aß-induced memory deficit and abnormal nociception. These mice have reduced Aß deposition and increased microglia coverage surrounding the amyloid plaque, suggesting the involvement of CLEC5A in AD progression and Aß clearance. Moreover, the phagocytosis was also increased in the Aß-stressed Clec5a knockdown microglial cell lines and Clec5a knockout primary microglia. CONCLUSION: The Clec5a knockout ameliorates AD-like deficits by modulating microglial Aß clearance. This study implies that targeting microglial Clec5a could offer a promising approach to mitigate AD progression.
Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Modelos Animais de Doenças , Lectinas Tipo C , Camundongos Knockout , Microglia , Animais , Lectinas Tipo C/metabolismo , Lectinas Tipo C/deficiência , Lectinas Tipo C/genética , Microglia/metabolismo , Microglia/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/genética , Camundongos , Peptídeos beta-Amiloides/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Camundongos Transgênicos , Aprendizagem em Labirinto/fisiologia , Fagocitose , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genéticaRESUMO
Distal metastases result from metastatic microenvironment and tumour epithelial cell interactions, the cellular heterogeneity of primary colorectal cancer (CRC) and liver metastases (LM) was evaluated by integrating single-cell sequencing data, and the collected gene expression data from metastatic epithelial cell subsets was used to construct a prognostic model and to identify intercellular receptor-ligand interactions between epithelial and immune cells in CRC and LM. Multiplex immunofluorescence staining, and in vitro wound healing, cell migration and cell apoptosis assays were performed to further explore the biological relevance of identified potential regulatory molecules. In this study, approximately 17 epithelial cell subtypes were detected, with Epi-11 cells being highly expressed in LM tissues compared with CRC samples. Furthermore, patients with high expression of the metastasis-related genetic profile of Epi-11 had a poorer prognosis. By predicting receptor-ligand interactions, Epi-11 cells were found to interact more with myeloid and T/natural killer cells in LM tissues when compared to primary CRC samples, which was mediated by the PLXNB1/SEMA4D axis. In addition, high SEMA4D expression was correlated with decreased overall survival of patients with CRC, whereas PLXNB1 was not. SEMA4D knockdown prevented the migration and promoted the apoptosis of HCT116 cells in vitro. In summary, Epi-11 cells, an important subset of epithelial cells, may drive the LM of CRC and act by crosstalk with immune cells through the PLXNB1/SEMA4D signalling axis.
Assuntos
Movimento Celular , Neoplasias Colorretais , Neoplasias Hepáticas , Proteínas do Tecido Nervoso , Receptores de Superfície Celular , Semaforinas , Humanos , Masculino , Antígenos CD/metabolismo , Antígenos CD/genética , Apoptose , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/imunologia , Prognóstico , Semaforinas/metabolismo , Semaforinas/genética , Transdução de Sinais , Microambiente Tumoral , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismoRESUMO
Background/aim: Autosomal recessive polycystic kidney disease is an inherited kidney disease. This study aims to detect rare and common DNA variants of the PKHD1 gene using next-generation sequencing (NGS) and to classify them in terms of being pathogenic according to The American College of Medical Genetics and Genomics. Materials and methods: NGS analysis was performed on the DNA of 304 patients who were referred to Ege University Molecular Medicine Laboratory with suspected polycystic kidney disease. Results: As a result, a total of 82 different DNA variants, 16 of which were novel, were detected. The breakdown of the variants found is as follows: 73 (89.02%) were missense variants, six (7.32%) nonsense variants, two (2.44%) frameshift deletions, and one (1.22%) nonframeshift deletion. According to The American College of Medical Genetics and Genomics classification of these variants, 26 were benign (Class 5), two were likely benign (Class 4), 36 were of uncertain significance (Class 3), and nine were likely pathogenic (Class 2), nine of which are pathogenic variants (Class 1). Heterozygosity was found in 39 (63.9%) patients, homozygosity in six (9.8%) patients, compound heterozygosity in 12 (19.7%) patients, and complex genotype in four (6.6%) patients in which variants in Class 1, Class 2 and Class 3 were determined according to ACMG classification. When the exon distributions of the DNA variants detected in the PKHD1 gene were analyzed, the most common exons of the DNA variant are exon 32 (n = 9), exon 58 (n = 8), exon 67 (n = 6), exon 61 (n = 5), 30 exons (n = 4). Conclusion: This fast and economical molecular diagnostic approach will provide a reliable prenatal diagnostic option, enabling definitive disease diagnosis and the identification of carriers.
Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Receptores de Superfície Celular , Humanos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Turquia , Feminino , Masculino , Receptores de Superfície Celular/genética , Adulto , Rim Policístico Autossômico Recessivo/genética , Adolescente , Criança , Adulto Jovem , Variação Genética/genética , Pré-EscolarRESUMO
Background/Objectives: The phytosulfokine receptor (PSKR) gene family plays a crucial role in regulating plant growth, development, and stress response. Here, the PSKR gene family was characterized in Triticum aestivum L. The study aimed to bridge knowledge gaps and clarify the functional roles of TaPSKRs to create a solid foundation for examining the structure, functions, and regulatory aspects. Methods: The investigation involved genome-wide identification of PSKRs through collection and chromosomal assignment, followed by phylogenetic analysis and gene expression profiling. Additionally, interactions with their interactors were stimulated and analyzed to elucidate their function. Results: The wide-genome inspection of all TaPSKRs led to 25 genes with various homeologs, resulting in 57 TaPSKR members distributed among the A, B, and D subgenomes. Investigating the expression of 61 TaPSKR cDNAs in RNA-seq datasets generated from different growth stages at 14, 21, and 60 days old and diverse tissues such as leaves, shoots, and roots provided further insight into their functional purposes. The expression profile of the TaPSKRs resulted in three key clusters. Gene cluster 1 (GC 1) is partially associated with root growth, suggesting that specific TaPSKRs control root development. The GC 2 cluster targeted genes that show high levels of expression in all tested leaf growth stages and the early developmental stage of the shoots and roots. Furthermore, the GC 3 cluster was composed of genes that are constantly expressed, highlighting their crucial role in regulating various processes during the entire life cycle of wheat. Molecular docking simulations showed that phytosulfokine type α (PSK-α) interacted with all TaPSKRs and had a strong binding affinity with certain TaPSKR proteins, encompassing TaPSKR1A, TaPSKR3B, and TaPSKR13A, that support their involvement in PSK signaling pathways. The crucial arbitration of the affinity may depend on interactions between wheat PSK-α and PSKRs, especially in the LRR domain region. Conclusions: These discoveries deepened our knowledge of the role of the TaPSKR gene family in wheat growth and development, opening up possibilities for further studies to enhance wheat durability and yield via focused innovation approaches.
Assuntos
Regulação da Expressão Gênica de Plantas , Simulação de Acoplamento Molecular , Proteínas de Plantas , Triticum , Triticum/genética , Triticum/crescimento & desenvolvimento , Triticum/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Proteínas de Plantas/química , Filogenia , Biologia Computacional/métodos , Genoma de Planta/genética , Perfilação da Expressão Gênica/métodos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/químicaRESUMO
Macrophages play a pivotal role in tissue homeostasis, pathogen defense, and inflammation resolution. M1 and M2 macrophage phenotypes represent two faces in a spectrum of responses to microenvironmental changes, crucial in both physiological and pathological conditions. Neuraminidase 1 (Neu1), a lysosomal and cell surface sialidase responsible for removing terminal sialic acid residues from glycoconjugates, modulates several macrophage functions, including phagocytosis and Toll-like receptor (TLR) signaling. Current evidence suggests that Neu1 expression influences M1/M2 macrophage phenotype alterations in the context of cardiovascular diseases, indicating a potential role for Neu1 in macrophage polarization. For this reason, we investigated the impact of Neu1 deficiency on macrophage polarization in vitro and in vivo. Using bone marrow-derived macrophages (BMDMs) and peritoneal macrophages from Neu1 knockout (Neu1-/- ) mice and wild-type (WT) littermate controls, we demonstrated that Neu1-deficient macrophages exhibit an aberrant M2-like phenotype, characterized by elevated macrophage mannose receptor 1 (MMR/CD206) expression and reduced responsiveness to M1 stimuli. This M2-like phenotype was also observed in vivo in peritoneal and splenic macrophages. However, lymph node (LN) macrophages from Neu1-/- mice exhibited phenotypic alterations with reduced CD206 expression. Further analysis revealed that peripheral LNs from Neu1-/- mice were highly fibrotic, with overexpression of transforming growth factor-beta 1 (TGF-ß1) and hyperactivated TGF-ß signaling in LN macrophages. Consistently, TGF-ß1 was found to alter M1/M2 macrophage polarization in vitro. Our findings showed that Neu1 deficiency prompts macrophages towards an M2 phenotype and that microenvironmental changes, particularly increased TGF-ß1 in fibrotic tissues such as peripheral LNs in Neu1-/- mice, further influence M1/M2 macrophage polarization, highlighting its sensitivity to the local microenvironment. Therapeutic interventions targeting Neu1 or TGF-ß signaling pathways may offer the potential to regulate macrophage behavior across different diseases.
Assuntos
Microambiente Celular , Fibrose , Linfonodos , Macrófagos , Camundongos Knockout , Neuraminidase , Animais , Camundongos , Macrófagos/imunologia , Macrófagos/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Neuraminidase/deficiência , Neuraminidase/genética , Neuraminidase/metabolismo , Camundongos Endogâmicos C57BL , Ativação de Macrófagos , Lectinas Tipo C/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/deficiência , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Células Cultivadas , Transdução de Sinais , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/deficiência , Receptor de Manose , Fenótipo , Fator de Crescimento Transformador beta1/metabolismoRESUMO
Complete elucidation of members of the gustatory receptor (Gr) family in lepidopteran insects began in the silkworm Bombyx mori. Grs of lepidopteran insects were initially classified into four subfamilies based on the results of phylogenetic studies and analyses of a few ligands. However, with further ligand analysis, it has become clear that plant secondary metabolites are important targets not only for Grs in the bitter subfamily but also for the Drosophila melanogaster Gr43a orthologue subfamily and Grs in the sugar subfamily. Gene knockout experiments showed that B. mori Gr6 (BmGr6) and BmGr9 are involved in the recognition of the feeding-promoting compounds chlorogenic acid and isoquercetin in mulberry leaves by the maxillary palps, suggesting that these Grs are responsible for palpation-dependent host recognition without biting. On the other hand, BmGr expression was also confirmed in nonsensory organs. Midgut enteroendocrine cells that produce specific neuropeptides were shown to express specific BmGrs, suggesting that BmGrs are involved in the induction of endocrine secretion in response to changes in the midgut contents. Furthermore, gene knockout experiments indicated that BmGr6 is indeed involved in the secretion of myosuppressin. On the other hand, BmGr9 was shown to induce signal transduction that is not derived from the intracellular signaling cascade mediated by G proteins but from the fructose-regulated cation channel of BmGr9 itself. Cryogenic electron microscopy revealed the mechanism by which the ion channel of the BmGr9 homotetramer opens upon binding of fructose to the ligand-binding pocket. Research on BmGrs has contributed greatly to our understanding of the functions and roles of Grs in insects.
Assuntos
Bombyx , Proteínas de Insetos , Receptores de Superfície Celular , Animais , Bombyx/genética , Bombyx/metabolismo , Bombyx/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/química , Proteínas de Insetos/metabolismo , Proteínas de Insetos/genética , Proteínas de Insetos/química , Transdução de Sinais , FilogeniaRESUMO
The two-kidney, one-clip (2K1C) Goldblatt rodent model elicits a reduction in renal blood flow (RBF) in the clipped kidney (CK). The reduced RBF and oxygen bio-ability causes the accumulation of the tricarboxylic cycle intermediary, α-ketoglutarate, which activates the oxoglutarate receptor-1 (OXGR1). In the kidney, OXGR1 is abundantly expressed in intercalated cells (ICs) of the collecting duct (CD), thus contributing to sodium transport and electrolyte balance. The (pro)renin receptor (PRR), a member of the renin-angiotensin system (RAS), is a key regulator of sodium reabsorption and blood pressure (BP) that is expressed in ICs. The PRR is upregulated in 2K1C rats. Here, we tested the hypothesis that chronic reduction in RBF in the CK leads to OXGR1-dependent PRR upregulation in the CD and alters sodium balance and BP in 2K1C mice. To determine the role of OXGR1 in regulating the PRR in the CDs during renovascular hypertension, we performed 2K1C Goldblatt surgery (clip = 0.13 mm internal gap, 14 days) in two groups of male mice: (1) mice treated with Montelukast (OXGR1 antagonist; 5 mg/Kg/day); (2) OXGR1-/- knockout mice. Wild-type and sham-operated mice were used as controls. After 14 days, 2K1C mice showed increased systolic BP (SBP) (108 ± 11 vs. control 82 ± 5 mmHg, p < 0.01) and a lower natriuretic response after the saline challenge test. The CK group showed upregulation of erythropoietin, augmented α-ketoglutarate, and increased PRR expression in the renal medulla. The CK of OXGR1 knockout mice and mice subjected to the OXGR1 antagonist elicited impaired PRR upregulation, attenuated SBP, and better natriuretic responses. In 2K1C mice, the effect of reduced RBF on the OXGR1-dependent PRR upregulation in the CK may contribute to the anti-natriuretic and increased SBP responses.
Assuntos
Túbulos Renais Coletores , Receptores de Superfície Celular , Sódio , Regulação para Cima , Animais , Camundongos , Túbulos Renais Coletores/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genética , Masculino , Sódio/metabolismo , Hipertensão Renovascular/metabolismo , Hipertensão Renovascular/genética , Pressão Sanguínea , Camundongos Knockout , Receptor de Pró-Renina , Rim/metabolismo , Modelos Animais de Doenças , Sistema Renina-Angiotensina , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Purinérgicos P2RESUMO
A 5-year-old girl was admitted due to one episode of melena and one episode of hematemesis. Upon admission, gastroscopy revealed esophageal and gastric varices. Abdominal CT scan, MRI, and color Doppler ultrasound suggested cirrhosis, intrahepatic bile duct dilation, and bilateral kidney enlargement. Genetic testing identified compound heterozygous mutations in the PKHD1 gene: c.2264C>T (p.Pro755Leu) and c.1886T>C (p.Val629Ala). The c.2264C>T (p.Pro755Leu) mutation is a known pathogenic variant with previous reports, while c.1886T>C (p.Val629Ala) is a novel mutation predicted to have pathogenic potential according to Mutation Taster and PolyPhen2. The child was diagnosed with autosomal recessive polycystic kidney disease. In children presenting with gastrointestinal bleeding without obvious causes, particularly those with liver or kidney disease, consideration should be given to the possibility of autosomal recessive polycystic kidney disease, and genetic testing should be conducted for definitive diagnosis when necessary.
Assuntos
Rim Policístico Autossômico Recessivo , Humanos , Feminino , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/complicações , Pré-Escolar , Mutação , Receptores de Superfície Celular/genéticaRESUMO
CD163, a scavenger receptor with anti-inflammatory function expressed exclusively on monocytes/macrophages, is dysregulated in cases of diabetes complications. This study aimed to characterize circulating CD163+ monocytes in the presence (D+Comps) or absence (D-Comps) of diabetes-related complications. RNA-sequencing and mass cytometry were conducted on CD163+ monocytes in adults with long-duration diabetes and D+Comps or D-Comps. Out of 10,868 differentially expressed genes identified between D+Comps and D-Comps, 885 were up-regulated and 190 were down-regulated with a ≥ 1.5-fold change. In D+Comps, 'regulation of centrosome cycle' genes were enriched 6.7-fold compared to the reference genome. MIR27A, MIR3648-1, and MIR23A, the most up-regulated and CD200R1, the most down-regulated gene, were detected in D+Comps from the list of 75 'genes of interest'. CD163+ monocytes in D+Comps had a low proportion of recruitment markers CCR5, CD11b, CD11c, CD31, and immune regulation markers CD39 and CD86. A gene-protein network identified down-regulated TLR4 and CD11b as 'hub-nodes'. In conclusion, this study reports novel insights into CD163+ monocyte dysregulation in diabetes-related complications. Enriched centrosome cycle genes and up-regulated miRNAs linked to apoptosis, coupled with down-regulated monocyte activation, recruitment, and immune regulation, suggest functionally distinct CD163+ monocytes in cases of diabetes complications. Further investigation is needed to confirm their role in diabetes-related tissue damage.
Assuntos
Antígenos CD , Antígenos de Diferenciação Mielomonocítica , MicroRNAs , Monócitos , Receptores de Superfície Celular , Humanos , Antígenos CD/genética , Antígenos CD/metabolismo , Monócitos/metabolismo , Monócitos/imunologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , MicroRNAs/genética , Feminino , Masculino , Pessoa de Meia-Idade , Complicações do Diabetes/genética , Complicações do Diabetes/imunologia , Complicações do Diabetes/sangue , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Perfilação da Expressão Gênica , Idoso , Regulação da Expressão Gênica , Adulto , Redes Reguladoras de Genes , BiomarcadoresRESUMO
Echinochloa crus-galli (L.) P. Beauv is a monocotyledonous weed that seriously infests rice fields. Florpyrauxifen-benzyl, a novel synthetic auxin herbicide commercialized in China in 2018, is an herbicide for controlling E. crus-galli. However, a suspected resistant population (R) collected in 2012 showed resistance to the previously unused florpyrauxifen-benzyl. Whole-plant dose-response bioassay indicated that the R population evolved high resistance to quinclorac and florpyrauxifen-benzyl. Pretreatment with P450 inhibitors did not influence the GR50 of E. crus-galli to florpyrauxifen-benzyl. The expression of target receptor EcAFB4 was down-regulated in the R population, leading to the reduced response to florpyrauxifen-benzyl (suppresses over-production of ethylene and ABA). We verified this resistance mechanism in the knockout OsAFB4 in Oryza sativa L. The Osafb4 mutants exhibited high resistance to florpyrauxifen-benzyl and moderate resistance to quinclorac. Furthermore, DNA methylation in the EcAFB4 promoter regulated its low expression in the R population after florpyrauxifen-benzyl treatment. In summary, the low expression of the auxin receptor EcAFB4 confers target resistance to the synthetic auxin herbicide florpyrauxifen-benzyl in the R- E. crus-galli.
Assuntos
Echinochloa , Resistência a Herbicidas , Herbicidas , Proteínas de Plantas , Echinochloa/efeitos dos fármacos , Echinochloa/genética , Echinochloa/metabolismo , Herbicidas/farmacologia , Resistência a Herbicidas/genética , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genética , Oryza/genética , Oryza/metabolismo , Oryza/efeitos dos fármacos , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Ácidos Indolacéticos/metabolismo , Ácidos Indolacéticos/farmacologia , Quinolinas/farmacologia , Plantas Daninhas/efeitos dos fármacos , Plantas Daninhas/genética , Plantas Daninhas/metabolismoRESUMO
BACKGROUND: Leber congenital amaurosis 1 (LCA1), caused by mutations in GUCY2D, is a rare inherited retinal disease that typically causes blindness in early childhood. The aim of this study was to evaluate the safety and preliminary efficacy of ascending doses of ATSN-101, a subretinal AAV5 gene therapy for LCA1. METHODS: 15 patients with genetically confirmed biallelic mutations in GUCY2D were included in this phase 1/2 study. All patients received unilateral subretinal injections of ATSN-101. In the dose-escalation phase, three adult cohorts (n=3 each) were treated with three ascending doses: 1·0â×â1010 vg/eye (low dose), 3·0â×â1010 vg/eye (middle dose), and 1·0â×â1011 vg/eye (high dose). In the dose-expansion phase, one adult cohort (n=3) and one paediatric cohort (n=3) were treated at the high dose. The primary endpoint was the incidence of treatment-emergent adverse events (TEAEs), and secondary endpoints included full-field stimulus test (FST) and best-corrected visual acuity (BCVA). A multi-luminance mobility test (MLMT) was also done. Data through the 12-month main study period are reported. FINDINGS: Patients were enrolled between Sept 12, 2019, and May 5, 2022. A total of 68 TEAEs were observed, 56 of which were related to the surgical procedure. No serious TEAE was related to the study drug. Ocular inflammation was mild and reversible with steroid treatment. For patients who received the high dose, mean change in dark-adapted FST was 20·3 decibels (dB; 95% CI 6·6 to 34·0) for treated eyes and 1·1 dB (-3·7 to 5·9) for untreated eyes at month 12 (white stimulus); improvements were first observed at day 28 and persisted over 12 months (p=0·012). Modest improvements in BCVA were also observed (p=0·10). Three of six patients who received the high dose and did the MLMT achieved the maximum score in the treated eye. INTERPRETATION: ATSN-101 is well tolerated 12 months after treatment, with no drug-related serious adverse events. Clinically significant improvements in retinal sensitivity were sustained in patients receiving the high dose. FUNDING: Atsena Therapeutics.
Assuntos
Terapia Genética , Guanilato Ciclase , Amaurose Congênita de Leber , Receptores de Superfície Celular , Adolescente , Adulto , Criança , Humanos , Terapia Genética/métodos , Guanilato Ciclase/genética , Injeções Intraoculares , Amaurose Congênita de Leber/genética , Mutação , Receptores de Superfície Celular/genética , Resultado do Tratamento , Acuidade VisualRESUMO
The malignant behavior and immune escape ability of cancer cells lead to therapeutic failure and poor prognosis for patients with various cancers, including colon cancer. Plexin domain containing 1 (PLXDC1) was initially identified to exert key roles in tumor by regulating angiogenesis and has recently proved to be involved in cell proliferation and migration of glioblastoma and gastric cancer cells. However, its roles in colon cancer remain unclear. In this study, the online bioinformatics databases confirmed high expression of PLXDC1 in colon cancer specimens, which was associated with cancer stages and nodal metastasis. Similarly, the increased expression of PLXDC1 was also validated in our collected samples and colon cancer cells. Moreover, patients with high expression of PLXDC1 had shorter survival, indicating that PLXDC1 might be a potential prognostic predictor for colon cancer patients. Notably, targeting PLXDC1 inhibited cancer cell viability and invasion, and enhanced cell apoptosis. Intriguingly, Tumor Immune Estimation Resource database confirmed that PLXDC1 expression was related to various tumor-infiltrating immune cells in colon adenocarcinoma including macrophages, and its expression was also correlated with M2-like macrophage markers. In vitro, colon cancer cells with PLXDC1 downregulation had a reduced ability to recruit and polarize macrophage towards M2 phenotype by decreasing the percentage of CD206+ cells and M2-like markers (CD206, CD163, arginase1, and interleukin 10 [IL-10]). Moreover, PLXDC1 knockdown attenuated M2 macrophage-mediated promotion in cancer cell viability and invasion. Mechanically, inhibition of PLXDC1 suppressed activation of the IL-6/Signal transducer and activator of transcription 3 (STAT3) signaling. Reactivating the above pathway by transfection with IL-6 plasmids reversed the suppressive effects of PLXDC1 knockdown on cancer cell malignant behaviors, macrophage recruitment and M2-like polarization. Thus, PLXDC1 downregulation may inhibit the malignancy of colon cancer cells and their ability to recruit and polarize macrophages towards M2 phenotype by blocking the IL-6/STAT3 pathway. Together, targeting PLXDC1 may attenuate the progression of colon cancer by direct roles in cancer cells and indirect roles in macrophage polarization, representing a promising therapeutic target for colon cancer patients.
Assuntos
Biomarcadores Tumorais , Neoplasias do Colo , Macrófagos , Humanos , Neoplasias do Colo/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/genética , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/genética , Prognóstico , Macrófagos/metabolismo , Macrófagos/patologia , Linhagem Celular Tumoral , Progressão da Doença , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genética , Masculino , Feminino , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Regulação Neoplásica da Expressão GênicaRESUMO
Purpose: Cognitive dysfunction caused by chronic cerebral hypoperfusion (CCH) is the leading cause of vascular dementia. Therefore, it is necessary to explore the mechanism that causes cerebral injury and find an effective therapy. Methods: Bone marrow mononuclear cells (BMMNCs) were extracted to detect the activity by CCK-8 kit and verify the transfection efficiency using reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR). A CCH rat model was established. Superparamagnetic iron oxide nanoparticles (BMPs)-PEI-Slit2/BMMNCs were injected into the tail vein and intervened with an external magnetic field. Hematoxylin and eosin staining was used to observe the pathological changes in brain tissue. The Slit/Robo pathway-related proteins Slit2 and Robo4 were detected by RT-qPCR and Western blotting. Results: The neurological score of the CCH group significantly increased compared with that of the sham group (P<0.05). The levels of brain injury markers S-100ß and NSE were significantly higher in the CCH group than in the sham group (P<0.05). Neuronal apoptosis in the frontal cortex and hippocampus of CCH rats significantly increased compared with that of the sham group (P<0.05). The expression levels of Slit2 and Robo4 mRNAs and proteins in brain tissue of CCH rats significantly increased (P<0.05). The neurological function scores of CCH rats treated with BMP-PEI-Slit2/BMMNC significantly increased after Robo4 siRNA administration (P<0.05). Conclusion: BMP combination with the CCH-related gene Slit2 can effectively improve the efficiency of BMMNC transplantation in treatment.
Assuntos
Isquemia Encefálica , Disfunção Cognitiva , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas do Tecido Nervoso , Animais , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ratos , Disfunção Cognitiva/terapia , Disfunção Cognitiva/etiologia , Isquemia Encefálica/terapia , Isquemia Encefálica/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Humanos , Masculino , Nanopartículas de Magnetita/administração & dosagem , Nanopartículas de Magnetita/química , Nanopartículas Magnéticas de Óxido de Ferro/administração & dosagem , Células da Medula Óssea , Apoptose/genética , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Ratos Sprague-Dawley , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Terapia Genética/métodos , Proteínas RoundaboutRESUMO
Chickens are the most thoroughly domesticated vertebrate species, and after long-continued natural and artificial selection, they now show rich phenotypic diversity. In particular, feathered legs present in domestic chickens are a characteristic that is carefully selected by advanced breeders. Previous studies have identified the key mutations responsible for feathered legs on chromosomes 13 and 15; however, not all chickens can be easily distinguished based on these two markers. In this study, whole-genome resequencing of 29 Bamaxiaogu chickens (BXCs) yielded 12,201,978 valid single-nucleotide polymorphisms (SNPs) and 2,792,426 valid insertions and deletions (InDels). Population structure analysis based on SNPs revealed that the test samples came from the same natural population. Based on these findings, we used an SNP- and InDel-based genome-wide association study (GWAS) to investigate the genetic basis of feathered legs in chickens. GWAS results revealed that 2 SNPs located in the introns of cubilin (CUBN; SNP1, chr2:19885382T>A) and recombinant Ras suppressor protein 1 (RSU1) genes (SNP2, chr2:20002551G>A), as well as an InDel (InDel1, chr2:19884383TG>T) on CUBN, were all significantly associated with the presence of feathered legs. Diagnostic testing demonstrated that SNP1 effectively differentiated between chickens with feathered legs and those with clean legs (leg without feathers) within the BXC population and may thus be considered an effective marker of feathered legs in BXC. In contrast, other loci did not show the same discriminatory power. This study not only presents a new variant of feathered legs but also provides valuable novel insights into the underlying mechanisms of variation in the feathered-legs trait among chickens.
Birds display remarkable diversity in the distribution and morphology of scales and feathers on their legs. However, the genetic and developmental mechanisms controlling this diversity are complex and remain largely unknown. Feathered legs are a phenotypic trait of domestic chickens, which have undergone intense selection. Previous studies have shown that the 2 major loci controlling feathered legs are located on chromosomes 13 and 15. In this study, we used a single-nucleotide polymorphism- and insertion and deletion-based genome-wide association study to elucidate the genetic basis of feathered legs in chickens. Additionally, we report a novel mutation in the cubilin gene associated with feathered legs in Bamaxiaogu chickens.