Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Blood ; 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38635773

ABSTRACT

Pseudouridine is the most prevalent RNA modification, and its aberrant function is implicated in various human diseases. However, the specific impact of pseudouridylation on hematopoiesis remains poorly understood. In this study, we investigated the role of tRNA pseudouridylation in erythropoiesis and its association with mitochondrial myopathy, lactic acidosis, and sideroblastic anemia syndrome (MLASA) pathogenesis. By utilizing patient-specific induced pluripotent stem cells (iPSCs) carrying a genetic PUS1 mutation and a corresponding mutant mouse model, we demonstrated impaired erythropoiesis in MLASA iPSCs and anemia in the MLASA mouse model. Both MLASA iPSCs and mouse erythroblasts exhibited compromised mitochondrial function and impaired protein synthesis. Mechanistically, we revealed that PUS1 deficiency resulted in reduced mitochondrial tRNA levels due to pseudouridylation loss, leading to aberrant mitochondrial translation. Screening of mitochondrial supplements aimed at enhancing respiration or heme synthesis showed limited effect in promoting erythroid differentiation. Interestingly, the mTOR inhibitor rapamycin facilitated erythroid differentiation in MLASA-iPSCs by suppressing mTOR signaling and protein synthesis, and consistent results were observed in the MLASA mouse model. Importantly, rapamycin treatment effectively ameliorated anemia phenotypes in the MLASA patient. Our findings provide novel insights into the crucial role of mitochondrial tRNA pseudouridylation in governing erythropoiesis and present potential therapeutic strategies for anemia patients facing challenges related to protein translation.

2.
Oncogene ; 43(19): 1476-1487, 2024 May.
Article in English | MEDLINE | ID: mdl-38514854

ABSTRACT

RNA-binding proteins (RBPs) are critical regulators for RNA transcription and translation. As a key member of RBPs, ELAV-like family protein 2 (CELF2) has been shown to regulate RNA splicing and embryonic hematopoietic development and was frequently seen dysregulated in acute myeloid leukemia (AML). However, the functional role(s) of CELF2 in hematopoiesis and leukemogenesis has not been fully elucidated. In the current study, we showed that Celf2 deficiency in hematopoietic system led to enhanced HSCs self-renewal and differentiation toward myeloid cells in mice. Loss of Celf2 accelerated myeloid cell transformation and AML development in MLL-AF9-induced AML murine models. Gene expression profiling integrated with RNA immunoprecipitation sequencing (RIP-Seq), together with biochemical experiments revealed that CELF2 deficiency stabilizes FAT10 mRNA, promotes FAT10 translation, thereby increases AKT phosphorylation and mTORC1 signaling pathway activation. Notably, combination therapy with a mTORC1 inhibitor (Rapamycin) and a MA9/DOTL1 inhibitor (EPZ-5676) reduced the leukemia burden in MLL-AF9 mice lacking Celf2 in vivo. Our study elucidated a novel mechanism by which the CELF2/FAT10-AKT/mTORC1 axis regulates the proliferation of normal blood cells and the development of AML, thus providing potential therapeutic targets for myeloid leukemia suppression.


Subject(s)
CELF Proteins , Leukemia, Myeloid, Acute , Mechanistic Target of Rapamycin Complex 1 , Nerve Tissue Proteins , RNA-Binding Proteins , Animals , Humans , Mice , CELF Proteins/genetics , CELF Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics
3.
iScience ; 27(3): 109265, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38450158

ABSTRACT

Pseudouridylation plays a regulatory role in various physiological and pathological processes. A prime example is the mitochondrial myopathy, lactic acidosis, and sideroblastic anemia syndrome (MLASA), characterized by defective pseudouridylation resulting from genetic mutations in pseudouridine synthase 1 (PUS1). However, the roles and mechanisms of pseudouridylation in normal erythropoiesis and MLASA-related anemia remain elusive. We established a mouse model carrying a point mutation (R110W) in the enzymatic domain of PUS1, mimicking the common mutation in human MLASA. Pus1-mutant mice exhibited anemia at 4 weeks old. Impaired mitochondrial oxidative phosphorylation was also observed in mutant erythroblasts. Mechanistically, mutant erythroblasts showed defective pseudouridylation of targeted tRNAs, altered tRNA profiles, decreased translation efficiency of ribosomal protein genes, and reduced globin synthesis, culminating in ineffective erythropoiesis. Our study thus provided direct evidence that pseudouridylation participates in erythropoiesis in vivo. We demonstrated the critical role of pseudouridylation in regulating tRNA homeostasis, cytoplasmic translation, and erythropoiesis.

4.
Dev Dyn ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38193537

ABSTRACT

BACKGROUND: Tripartite motif (TRIM46) is a relatively novel protein that belongs to tripartite motif family. TRIM46 organizes parallel microtubule arrays on the axons, which are important for neuronal polarity and axonal function. TRIM46 is highly expressed in the brain, but its biological function in adults has not yet been determined. RESULTS: Trim46 knockout (KO) rat line was established using CRISPR/cas9. Trim46 KO rats had smaller hippocampus sizes, fewer neuronal dendritic arbors and dendritic spines, and shorter and more distant axon initial segment. Furthermore, the protein interaction between endogenous TRIM46 and FK506 binding protein 5 (FKBP5) in brain tissues was determined; Trim46 KO increased hippocampal FKBP5 protein levels and decreased hippocampal protein kinase B (Akt) phosphorylation, gamma-aminobutyric acid type A receptor subunit alpha1 (GABRA1) and glutamate ionotropic receptor NMDA type subunit 1 (NMDAR1) protein levels. Trim46 KO rats exhibited hypoactive behavioral changes such as reduced spontaneous activity, social interaction, sucrose preference, impaired prepulse inhibition (PPI), and short-term reference memory. CONCLUSIONS: These results demonstrate the significant impact of Trim46 KO on brain structure and behavioral function. This study revealed a novel potential association of TRIM46 with dendritic development and neuropsychiatric behavior, providing new insights into the role of TRIM46 in the brain.

5.
Sci Adv ; 9(15): eadf2695, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37058569

ABSTRACT

Hundreds of pathogenic variants of mitochondrial DNA (mtDNA) have been reported to cause mitochondrial diseases, which still lack effective treatments. It is a huge challenge to install these mutations one by one. We repurposed the DddA-derived cytosine base editor to incorporate a premature stop codon in the mtProtein-coding genes to ablate mitochondrial proteins encoded in the mtDNA (mtProteins) instead of installing pathogenic variants and generated a library of both cell and rat resources with mtProtein depletion. In vitro, we depleted 12 of 13 mtProtein-coding genes with high efficiency and specificity, resulting in decreased mtProtein levels and impaired oxidative phosphorylation. Moreover, we generated six conditional knockout rat strains to ablate mtProteins using Cre/loxP system. Mitochondrially encoded ATP synthase membrane subunit 8 and NADH:ubiquinone oxidoreductase core subunit 1 were specifically depleted in heart cells or neurons, resulting in heart failure or abnormal brain development. Our work provides cell and rat resources for studying the function of mtProtein-coding genes and therapeutic strategies.


Subject(s)
Codon, Nonsense , Mitochondria , Rats , Animals , Base Sequence , Mitochondria/genetics , Mitochondria/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mutation
6.
Mol Ther Nucleic Acids ; 32: 1-12, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-36942261

ABSTRACT

An animal model harboring pathogenic mitochondrial DNA (mtDNA) mutations is important to understand the biological links between mtDNA variation and mitochondrial diseases. DdCBE, a DddA-derived cytosine base editor, has been utilized in zebrafish, mice, and rats for tC sequence-context targeting and human mitochondrial disease modeling. However, human pathogenic mtDNA mutations other than the tC context cannot be manipulated. Here, we screened the combination of different DdCBE pairs at pathogenic mtDNA mutation sites with nC (n for a, g, or c) context and identified that the left-G1333C (L1333C) + right G1333N (R1333N) pair could mediate C⋅G-to-T⋅A conversion effectively at aC sites in rat C6 cells. The editing efficiency at disease-associated mtDNA mutation sites within aC context was further confirmed to be up to 67.89% in vivo. Also, the installed disease-associated mtDNA mutations were germline transmittable. Moreover, the edited rats showed impaired cardiac function and mitochondrial function, resembling human mitochondrial disease symptoms. In summary, for the first time, we expanded the DdCBE targeting scope to an aC motif and installed the pathogenic mutation in rats to model human mitochondrial diseases.

7.
Dis Model Mech ; 16(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-35855640

ABSTRACT

When pathological hypertrophy progresses to heart failure (HF), the prognosis is often very poor. Therefore, it is crucial to find new and effective intervention targets. Here, myocardium-specific Trim44 knockout rats were generated using CRISPR-Cas9 technology. Cardiac phenotypic observations revealed that Trim44 knockout affected cardiac morphology at baseline. Rats with Trim44 deficiency exhibited resistance to cardiac pathological changes in response to stimulation via isoproterenol (ISO) treatment, including improvement of cardiac remodeling and dysfunction by morphological and functional observations, reduced myocardial fibrosis and reduced expression of molecular markers of cardiac stress. Furthermore, signal transduction validation associated with growth and hypertrophy development in vivo and in vitro demonstrated that Trim44 deficiency inhibited the activation of signaling pathways involved in myocardial hypertrophy, especially response to pathological stress. In conclusion, the present study indicates that Trim44 knockout attenuates ISO-induced pathological cardiac remodeling through blocking the AKT/mTOR/GSK3ß/P70S6K signaling pathway. This is the first study to demonstrate the function and importance of Trim44 in the heart at baseline and under pathological stress. Trim44 could be a novel therapeutic target for prevention of cardiac hypertrophy and HF.


Subject(s)
Proto-Oncogene Proteins c-akt , Ventricular Remodeling , Animals , Cardiomegaly/genetics , Isoproterenol/metabolism , Isoproterenol/pharmacology , Isoproterenol/therapeutic use , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , TOR Serine-Threonine Kinases/metabolism , Ventricular Remodeling/physiology
8.
Int J Cardiol ; 363: 149-158, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35714719

ABSTRACT

Dilated cardiomyopathy (DCM) is a major cause of heart failure. LMNA variants contribute to 6-10% DCM cases, but the underlying mechanisms remain incompletely understood. Here, we reported two patients carrying the LMNA c.1621C > T/ p.R541C variant and generated a knock-in mouse model (LmnaRC) to study the role of this variant in DCM pathogenesis. We found LmnaRC/RC mice exhibited ventricular dilation and reduced systolic functions at 6 months after birth. The LmnaRC/RC cardiomyocytes increased in size but no nuclear morphology defects were detected. Transcriptomic and microscopic analyses revealed suppressed gene expression and perturbed ultrastructure in LmnaRC/RC mitochondria. These defects were associated with increased heterochromatin structures and epigenetic markers including H3K9me2/3. Together, these data implied that the LMNA c.1621C > T/ p.R541C variant enhanced heterochromatic gene suppression and disrupted mitochondria functions as a cause of DCM.


Subject(s)
Cardiomyopathy, Dilated , Lamin Type A/metabolism , Animals , Cardiomyopathy, Dilated/complications , Disease Models, Animal , Humans , Lamin Type A/genetics , Mice , Mutation/genetics , Myocytes, Cardiac/metabolism
9.
Mol Neurobiol ; 59(7): 4612-4629, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35589918

ABSTRACT

Paraoxonase 1 (PON1) plays an anti-inflammatory role in the cardiovascular system. Levels of serum PON1 and polymorphisms in this gene were linked to Alzheimer's disease (AD) and Parkinson disease (PD), but its function in the neuroimmune system and AD is not clear. To address this issue, we used Pon1 knockout rats previously generated by our lab to investigate the role of Pon1 in microglia. Knockout of Pon1 in rat brain tissues protected against LPS-induced microglia activation. Pon1 deficiency in rat primary microglia increased Trem2 (triggering receptor expressed in myeloid cells 2) expression, phagocytosis, and IL-10 (M2-phenotype marker) release, but decreased production of pro-inflammatory cytokines such as IL-1ß, IL-6, and IL-18 especially TNF-α (M1-phenotype markers) induced by LPS. Pon1 deficiency in rat primary microglia activated Trem2 pathway but decreased LPS-induced ERK activation. The phagocytosis-promoting effect of Pon1 knockout could be reversed by administration of recombinant PON1 protein. The interaction between PON1 and TREM2 was verified by co-immunoprecipitation (co-IP) using rat brain tissues or over-expressed BV2 cell lysates, which might be involved in lysosomal localization of TREM2. Furthermore, Pon1 knockout also enhanced microglial phagocytosis and clearance of exogenous Aß by an intrahippocampal injection and decrease the transcription of cytokines such as IL-1ß, IL-6, and TNF-α in vivo. These results suggest that Pon1 knockout facilitates microglial phagocytosis and inhibits the production of proinflammatory cytokines both in vivo and in vitro, in which the interaction between Pon1 and Trem2 may be involved. These findings provide novel insights into the role of PON1 in neuroinflammation and highlight TREM2 as a potential target for Alzheimer's disease therapy.


Subject(s)
Alzheimer Disease , Aryldialkylphosphatase , Membrane Glycoproteins , Microglia , Receptors, Immunologic , Alzheimer Disease/metabolism , Animals , Aryldialkylphosphatase/genetics , Biomarkers/metabolism , Cytokines/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/metabolism , Microglia/metabolism , Phagocytosis/genetics , Rats , Receptors, Immunologic/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Animal Model Exp Med ; 5(2): 172-182, 2022 04.
Article in English | MEDLINE | ID: mdl-35474613

ABSTRACT

BACKGROUND: Inflammation is a complex physiological and pathological process. Although many types of inflammation are well characterized, their physiological functions are largely unknown. tRNA aspartic acid methyltransferase 1 (TRDMT1) has been implicated as a stress-related protein, but its intrinsic biological role is unclear. METHODS: We constructed a Trdmt1 knockout rat and adopted the LPS-induced sepsis model. Survival curve, histopathological examination, expression of inflammatory factors, and protein level of TLR4 pathway were analyzed. RESULTS: Trdmt1 deletion had no obvious impact on development and growth. Trdmt1 deletion slightly increased the mortality during aging. Our data showed that Trdmt1 strongly responded in LPS-treated rats, and Trdmt1 knockout rats were vulnerable to LPS treatment with declined survival rate. We also observed more aggravated tissue damage and more cumulative functional cell degeneration in LPS-treated knockout rats compared with control rats. Further studies showed upregulated TNF-α level in liver, spleen, lung, and serum tissues, which may be explained by enhanced p65 and p38 phosphorylation. CONCLUSIONS: Our data demonstrated that Trdmt1 plays a protective role in inflammation by regulating the TLR4-NF-κB/MAPK-TNF-α pathway. This work provides useful information to understand the TRDMT1 function in inflammation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , MAP Kinase Signaling System , NF-kappa B , Toll-Like Receptor 4 , Tumor Necrosis Factor-alpha , Animals , DNA (Cytosine-5-)-Methyltransferases/metabolism , Inflammation , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Rats , Signal Transduction , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Life Sci ; 297: 120485, 2022 May 15.
Article in English | MEDLINE | ID: mdl-35304126

ABSTRACT

AIMS: Multiple mitochondrial dysfunction (MMD) can lead to complex damage of mitochondrial structure and function, which then lead to the serious damage of various metabolic pathways including cerebral abnormalities. However, the effects of MMD on heart, a highly mitochondria-dependent tissue, are still unclear. In this study, we use iron-sulfur cluster assembly 1 (Isca1), which has been shown to cause MMD syndromes type 5 (MMDS5), to verify the above scientific question. MAIN METHODS: We generated myocardium-specific Isca1 knockout rat (Isca1flox/flox/α-MHC-Cre) using CRISPR-Cas9 technology. Echocardiography, magnetic resonance imaging (MRI), histopathological examinations and molecular markers detection demonstrated phenotypic characteristics of our model. Immunoprecipitation, immunofluorescence co-location, mitochondrial activity, ATP generation and iron ions detection were used to verify the molecular mechanism. KEY FINDINGS: This study was the first to verify the effects of Isca1 deficiency on cardiac development in vivo, that is cardiomyocytes suffer from mitochondria damage and iron metabolism disorder, which leads to myocardial oncosis and eventually heart failure and body death in rat. Furthermore, forward and reverse validation experiments demonstrated that six-transmembrane epithelial antigen of prostate 3 (STEAP3), a new interacting molecule for ISCA1, plays an important role in iron metabolism and energy generation impairment induced by ISCA1 deficiency. SIGNIFICANCE: This result provides theoretical basis for understanding of MMDS pathogenesis, especially on heart development and the pathological process of heart diseases, and finally provides new clues for searching clinical therapeutic targets of MMDS.


Subject(s)
Cardiomyopathies , Iron Metabolism Disorders , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Iron Metabolism Disorders/metabolism , Male , Mitochondria/genetics , Mitochondria/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Rats
12.
Leukemia ; 36(4): 1111-1122, 2022 04.
Article in English | MEDLINE | ID: mdl-34853439

ABSTRACT

SETD5 mutations were identified as the genetic causes of neurodevelopmental disorders. While the whole-body knockout of Setd5 in mice leads to embryonic lethality, the role of SETD5 in adult stem cell remains unexplored. Here, a critical role of Setd5 in hematopoietic stem cells (HSCs) is identified. Specific deletion of Setd5 in hematopoietic system significantly increased the number of immunophenotypic HSCs by promoting HSC proliferation. Setd5-deficient HSCs exhibited impaired long-term self-renewal capacity and multiple-lineage differentiation potentials under transplantation pressure. Transcriptome analysis of Setd5-deficient HSCs revealed a disruption of quiescence state of long-term HSCs, a cause of the exhaustion of functional HSCs. Mechanistically, SETD5 was shown to regulate HSC quiescence by mediating the release of promoter-proximal paused RNA polymerase II (Pol II) on E2F targets in cooperation with HCF-1 and PAF1 complex. Taken together, these findings reveal an essential role of SETD5 in regulating Pol II pausing-mediated maintenance of adult stem cells.


Subject(s)
Hematopoietic Stem Cells , RNA Polymerase II , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Homeostasis , Humans , Methyltransferases , Mice , Mice, Knockout , RNA Polymerase II/genetics , Transcription Factors
13.
Transgenic Res ; 31(1): 107-118, 2022 02.
Article in English | MEDLINE | ID: mdl-34709566

ABSTRACT

Uncoupling protein 1 (UCP1) was found exclusively in the inner membranes of the mitochondria of brown adipose tissue (BAT). We found that UCP1 was also expressed in heart tissue and significantly upregulated in isoproterenol (ISO)-induced acute myocardial ischemia (AMI) rat model. The present study is to determine the underlying mechanism involved in the UCP1 upregulation in ISO-induced AMI rat model. The Ucp1-/- rats were generated by CRISPR-Cas9 system and presented decreased BAT volume. 2-months old Sprague Dawley (SD) wild-type (WT) and Ucp1-/- rats were treated with ISO intraperitoneally 30 mg/kg once a day for 3 consecutive days to establish AMI model. In saline group, the echocardiographic parameters, serum markers of myocardial injury cardiac troponin I (cTnI), creatine kinase isoenzyme MB (CK-MB), oxidant malondialdehyde (MDA), antioxidant superoxide dismutase (SOD) or fibrosis were comparable between WT and Ucp1-/- rats. ISO treatment induced worse left ventricle (LV) hypertrophy, myocardial fibrosis, increased higher cTnI, CK-MB and MDA and decreased lower SOD level in Ucp1-/- rats compared with that of WT rats. Ucp1-/- rats also presented lower myocardial phosphocreatine (PCr)/ATP-ratio, which demonstrated worse cardiac energy regulation defect. ISO treatment induced the phosphorylation of AMP-activated protein kinase (AMPK) activation, subsequently the phosphorylation of mammalian target of rapamycin (mTOR) inhibition and peroxisome proliferators-activated receptor α (PPARα) activation in WT rats, whereas activation of AMPK/mTOR/PPARα pathways significantly inhibited in Ucp1-/- rats. To sum up, UCP1 knockout aggravated ISO-induced AMI by inhibiting AMPK/mTOR/PPARα pathways in rats. Increasing UCP1 expression in heart tissue may be a cytoprotective therapeutic strategy for AMI.


Subject(s)
AMP-Activated Protein Kinases , Myocardial Ischemia , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Isoproterenol/metabolism , Isoproterenol/toxicity , Mammals/metabolism , Myocardial Ischemia/metabolism , Myocardium/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Peroxisome Proliferators/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Uncoupling Protein 1/metabolism
14.
Neuron ; 110(5): 874-890.e7, 2022 03 02.
Article in English | MEDLINE | ID: mdl-34932943

ABSTRACT

Self-grooming is a complex behavior with important biological functions and pathological relevance. How the brain coordinates with the spinal cord to generate the repetitive movements of self-grooming remains largely unknown. Here, we report that in the caudal part of the spinal trigeminal nucleus (Sp5C), neurons that express Cerebellin-2 (Cbln2+) form a neural circuit to the cervical spinal cord to maintain repetitive orofacial self-grooming. Inactivation of Cbln2+ Sp5C neurons blocked both sensory-evoked and stress-induced repetitive orofacial self-grooming. Activation of these neurons triggered short-latency repetitive forelimb movements that resembled orofacial self-grooming. The Cbln2+ Sp5C neurons were monosynaptically innervated by both somatosensory neurons in the trigeminal ganglion and paraventricular hypothalamic neurons. Among the divergent projections of Cbln2+ Sp5C neurons, a descending pathway that innervated motor neurons and interneurons in the cervical spinal cord was necessary and sufficient for repetitive orofacial self-grooming. These data reveal a brain-to-spinal sensorimotor loop for repetitive self-grooming in mice.


Subject(s)
Brain , Neurons , Animals , Grooming , Hypothalamus , Mice , Neurons/physiology , Spinal Cord
16.
Cell Rep ; 37(5): 109926, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731629

ABSTRACT

Interferon regulatory factor 3 (IRF3) is an essential transductor for initiation of many immune responses. Here, we show that lncRNA-ISIR directly binds IRF3 to promote its phosphorylation, dimerization, and nuclear translocation, along with enhanced target gene productions. In vivo lncRNA-ISIR deficiency results in reduced IFN production, uncontrolled viral replication, and increased mortality. The human homolog, AK131315, also binds IRF3 and promotes its activation. More important, AK131315 expression is positively correlated with type I interferon (IFN-I) level and severity in patients with lupus. Mechanistically, in resting cells, IRF3 is bound to suppressor protein Flightless-1 (Fli-1), which keeps its inactive state. Upon infection, IFN-I-induced lncRNA-ISIR binds IRF3 at DNA-binding domain in cytoplasm and removes Fli-1's association from IRF3, consequently facilitating IRF3 activation. Our results demonstrate that IFN-I-inducible lncRNA-ISIR feedback strengthens IRF3 activation by removing suppressive Fli-1 in immune responses, revealing a method of lncRNA-mediated modulation of transcription factor (TF) activation.


Subject(s)
Interferon Regulatory Factor-3/metabolism , Lupus Erythematosus, Systemic/metabolism , Macrophages, Peritoneal/metabolism , RNA, Long Noncoding/metabolism , Vesicular Stomatitis/metabolism , Animals , Case-Control Studies , Chlorocebus aethiops , Disease Models, Animal , Gene Silencing , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , RAW 264.7 Cells , RNA, Long Noncoding/genetics , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Vero Cells , Vesicular Stomatitis/genetics , Vesicular Stomatitis/immunology , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/pathogenicity
18.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34607953

ABSTRACT

Chemokine production by epithelial cells is important for neutrophil recruitment during viral infection, the appropriate regulation of which is critical for restraining inflammation and attenuating subsequent tissue damage. Epithelial cell expression of long noncoding RNAs (lncRNAs), RNA-binding proteins, and their functional interactions during viral infection and inflammation remain to be fully understood. Here, we identified an inducible lncRNA in the Cxcl2 gene locus, lnc-Cxcl2, which could selectively inhibit Cxcl2 expression in mouse lung epithelial cells but not in macrophages. lnc-Cxcl2-deficient mice exhibited increased Cxcl2 expression, enhanced neutrophils recruitment, and more severe inflammation in the lung after influenza virus infection. Mechanistically, nucleus-localized lnc-Cxcl2 bound to Cxcl2 promoter, recruited a ribonucleoprotein La, which inhibited the chromatin accessibility of chemokine promoters, and consequently inhibited Cxcl2 transcription in cis However, unlike mouse lnc-Cxcl2, human lnc-CXCL2-4-1 inhibited multiple immune cytokine expressions including chemokines in human lung epithelial cells. Together, our results demonstrate a self-protecting mechanism within epithelial cells to restrain chemokine and neutrophil-mediated inflammation, providing clues for better understanding chemokine regulation and epithelial cell function in lung viral infection.


Subject(s)
Chemokine CXCL2/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , RNA, Long Noncoding/genetics , A549 Cells , Animals , Cell Line, Tumor , Chemokine CXCL2/metabolism , Chromatin/metabolism , Epithelial Cells/metabolism , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Humans , Inflammation/prevention & control , Inflammation Mediators , Influenza A virus/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/virology , Promoter Regions, Genetic/genetics , RAW 264.7 Cells , Vesicular Stomatitis/immunology , Vesicular Stomatitis/pathology , Vesicular stomatitis Indiana virus/immunology
19.
Front Genet ; 12: 642079, 2021.
Article in English | MEDLINE | ID: mdl-33790947

ABSTRACT

Objective: By exploring the effects of miR-29a-5p knockout on neurological damage after acute ischemic stroke, we aim to deepen understanding of the molecular mechanisms of post-ischemic injury and thus provide new ideas for the treatment of ischemic brain injury. Methods: miR-29a-5p knockout rats and wild-type SD rats were subjected to transient middle cerebral artery occlusion (MCAO). miR-29a levels in plasma, cortex, and basal ganglia of ischemic rats, and in plasma and neutrophils of ischemic stroke patients, as well as hypoxic glial cells were detected by real-time PCR. The infarct volume was detected by TTC staining and the activation of astrocytes and microglia was detected by western blotting. Results: The expression of miR-29a-5p was decreased in parallel in blood and brain tissue of rat MCAO models. Besides, miR-29a-5p levels were reduced in the peripheral blood of acute stroke patients. Knockout of miR-29a enhanced infarct volume of the MCAO rat model, and miR-29a knockout showed M1 polarization of microglia in the MCAO rat brain. miR-29a knockout in rats after MCAO promoted astrocyte proliferation and increased glutamate release. Conclusion: Knockout of miR-29a in rats promoted M1 microglial polarization and increased glutamate release, thereby aggravating neurological damage in experimental stroke rat models.

SELECTION OF CITATIONS
SEARCH DETAIL
...