Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.469
Filter
1.
Sci Rep ; 14(1): 11020, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38745067

ABSTRACT

The absence of stimulator of interferon genes (STING) in 129.B6.Fcgr2b-deficient mice rescue lupus phenotypes. The administration of a STING inhibitor (ISD017) into the young 129.B6.Fcgr2b-deficient mice prevents lupus nephritis development. This study mainly aimed to evaluate the effects of STING inhibition (ISD107) on established SLE in mice to prove that ISD017 could be a good therapeutic drug to reverse the already set-up autoimmunity and kidney impairment. Twenty-four-week-old Fcgr2b-deficient mice were treated with cyclophosphamide (25 mg/kg, intraperitoneal, once per week), ISD017 (10 mg/kg, intraperitoneal, three times per week), or control vehicle for 8 weeks, and were analyzed for phenotypes. Both ISD017 and cyclophosphamide treatment increased long-term survival and reduced the severity of glomerulonephritis in Fcgr2b-deficient mice. While cyclophosphamide reduced activated B cells (B220+GL-7+), ISD017 decreased activated T cells (CD4+CD69+) and neutrophils (Ly6c+Ly6g+) in Fcgr2b-deficient mice. In addition, ISD017 reduced IL-1ß and interferon-inducible genes. In summary, ISD017 treatment in symptomatic 129.B6.Fcgr2b-deficient mice reduced the severity of glomerulonephritis and increased long-term survival. ISD017 worked comparably to cyclophosphamide for treating lupus nephritis in 129.B6.Fcgr2b-deficient mice. ISD017 reduced activated T cells and neutrophils, while cyclophosphamide targeted activated B cells. These results suggested that STING inhibitors can potentially be a new therapeutic drug for treating lupus.


Subject(s)
Cyclophosphamide , Membrane Proteins , Receptors, IgG , Animals , Mice , Membrane Proteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Cyclophosphamide/pharmacology , Receptors, IgG/genetics , Receptors, IgG/metabolism , Lupus Nephritis/drug therapy , Lupus Nephritis/pathology , Glomerulonephritis/drug therapy , Mice, Knockout , Female , Disease Models, Animal , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics , Mice, Inbred C57BL
2.
Biochem Biophys Res Commun ; 714: 149973, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38657444

ABSTRACT

Acute respiratory distress syndrome (ARDS) is characterized by acute diffuse inflammatory lung injury with a high mortality rate. Mesenchymal stromal cells (MSC) are pluripotent adult cells that can be extracted from a variety of tissues, including the lung. Lung-resident MSC (LR-MSC) located around vascular vessels and act as important regulators of lung homeostasis, regulating the balance between lung injury and repair processes. LR-MSC support the integrity of lung tissue by modulating immune responses and releasing trophic factors. Studies have reported that the STING pathway is involved in the progression of lung injury inflammation, but the specific mechanism is unclear. In this study, we found that STING deficiency could ameliorate lipopolysaccharides (LPS)-induced acute lung injury, STING knockout (STING KO) LR-MSC had an enhanced treatment effect on acute lung injury. STING depletion protected LR-MSC from LPS-induced apoptosis. RNA-sequencing and Western blot results showed that STING KO LR-MSC expressed higher levels of MSC immunoregulatory molecules, such as Igfbp4, Icam1, Hgf and Cox2, than WT LR-MSC. This study highlights that LR-MSC have a therapeutic role in acute lung injury, and we demonstrate that STING deficiency can enhance the immunomodulatory function of LR-MSC in controlling lung inflammation. Thus, STING can be used as an intervention target to enhance the therapeutic effect of MSC.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Lung , Membrane Proteins , Mesenchymal Stem Cells , Mice, Inbred C57BL , Animals , Lipopolysaccharides/toxicity , Mesenchymal Stem Cells/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/deficiency , Lung/pathology , Lung/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Acute Lung Injury/metabolism , Mice , Mice, Knockout , Apoptosis , Male
3.
J Biol Chem ; 299(12): 105455, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37949232

ABSTRACT

The Akt-Rheb-mTORC1 pathway plays a crucial role in regulating cell growth, but the mechanisms underlying the activation of Rheb-mTORC1 by Akt remain unclear. In our previous study, we found that CBAP was highly expressed in human T-ALL cells and primary tumors, and its deficiency led to reduced phosphorylation of TSC2/S6K1 signaling proteins as well as impaired cell proliferation and leukemogenicity. We also demonstrated that CBAP was required for Akt-mediated TSC2 phosphorylation in vitro. In response to insulin, CBAP was also necessary for the phosphorylation of TSC2/S6K1 and the dissociation of TSC2 from the lysosomal membrane. Here we report that CBAP interacts with AKT and TSC2, and knockout of CBAP or serum starvation leads to an increase in TSC1 in the Akt/TSC2 immunoprecipitation complexes. Lysosomal-anchored CBAP was found to override serum starvation and promote S6K1 and 4EBP1 phosphorylation and c-Myc expression in a TSC2-dependent manner. Additionally, recombinant CBAP inhibited the GAP activity of TSC2 complexes in vitro, leading to increased Rheb-GTP loading, likely due to the competition between TSC1 and CBAP for binding to the HBD domain of TSC2. Overexpression of the N26 region of CBAP, which is crucial for binding to TSC2, resulted in a decrease in mTORC1 signaling and an increase in TSC1 association with the TSC2/AKT complex, ultimately leading to increased GAP activity toward Rheb and impaired cell proliferation. Thus, we propose that CBAP can modulate the stability of TSC1-TSC2 as well as promote the translocation of TSC1/TSC2 complexes away from lysosomes to regulate Rheb-mTORC1 signaling.


Subject(s)
Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins , Proto-Oncogene Proteins c-akt , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Humans , Cell Proliferation , Guanosine Triphosphate/metabolism , Immunoprecipitation , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism
4.
J Virol ; 97(12): e0143823, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37991364

ABSTRACT

IMPORTANCE: Herpes simplex virus 1 (HSV-1) establishes lifelong latency in neuronal cells. Following a stressor, the virus reactivates from latency, virus is shed at the periphery and recurrent disease can occur. During latency, the viral lncRNA termed the latency-associated transcript (LAT) is known to accumulate to high abundance. The LAT is known to impact many aspects of latency though the molecular events involved are not well understood. Here, we utilized a human neuronal cell line model of HSV latency and reactivation (LUHMES) to identify the molecular-binding partners of the LAT during latency. We found that the LAT binds to both the cellular protein, TMEM43, and HSV-1 genomes in LUHMES cells. Additionally, we find that knockdown of TMEM43 prior to infection results in a decreased ability of HSV-1 to establish latency. This work highlights a potential mechanism for how the LAT facilitates the establishment of HSV-1 latency in human neurons.


Subject(s)
Cell Nucleus , Genome, Viral , Herpes Simplex , Herpesvirus 1, Human , RNA, Long Noncoding , Virus Latency , Humans , Cell Line , Herpes Simplex/genetics , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , RNA, Long Noncoding/genetics , Virus Activation/genetics , Virus Latency/genetics , Cell Nucleus/metabolism , Cell Nucleus/virology , Neurons/metabolism , Neurons/virology , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Genome, Viral/genetics
5.
Cells ; 12(12)2023 06 13.
Article in English | MEDLINE | ID: mdl-37371084

ABSTRACT

The axoneme and accessory structures of flagella are critical for sperm motility and male fertilization. Sperm production needs precise and highly ordered gene expression to initiate and sustain the many cellular processes that result in mature spermatozoa. Here, we identified a testis enriched gene transmembrane protein 232 (Tmem232), which is essential for the structural integrity of the spermatozoa flagella axoneme. Tmem232 knockout mice were generated for in vivo analyses of its functions in spermatogenesis. Phenotypic analysis showed that deletion of Tmem232 in mice causes male-specific infertility. Transmission electron microscopy together with scanning electron microscopy were applied to analyze the spermatozoa flagella and it was observed that the lack of TMEM232 caused failure of the cytoplasm removal and the absence of the 7th outer microtubule doublet with its corresponding outer dense fiber (ODF). Co-IP assays further identified that TMEM232 interacts with ODF family protein ODF1, which is essential to maintain sperm motility. In conclusion, our findings indicate that TMEM232 is a critical protein for male fertility and sperm motility by regulating sperm cytoplasm removal and maintaining axoneme integrity.


Subject(s)
Infertility, Male , Membrane Proteins , Sperm Motility , Sperm Tail , Animals , Male , Mice , Infertility, Male/genetics , Infertility, Male/metabolism , Mice, Knockout , Semen , Sperm Motility/genetics , Sperm Tail/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism
6.
Nature ; 618(7964): 402-410, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225994

ABSTRACT

Membrane-shaping proteins characterized by reticulon homology domains play an important part in the dynamic remodelling of the endoplasmic reticulum (ER). An example of such a protein is FAM134B, which can bind LC3 proteins and mediate the degradation of ER sheets through selective autophagy (ER-phagy)1. Mutations in FAM134B result in a neurodegenerative disorder in humans that mainly affects sensory and autonomic neurons2. Here we report that ARL6IP1, another ER-shaping protein that contains a reticulon homology domain and is associated with sensory loss3, interacts with FAM134B and participates in the formation of heteromeric multi-protein clusters required for ER-phagy. Moreover, ubiquitination of ARL6IP1 promotes this process. Accordingly, disruption of Arl6ip1 in mice causes an expansion of ER sheets in sensory neurons that degenerate over time. Primary cells obtained from Arl6ip1-deficient mice or from patients display incomplete budding of ER membranes and severe impairment of ER-phagy flux. Therefore, we propose that the clustering of ubiquitinated ER-shaping proteins facilitates the dynamic remodelling of the ER during ER-phagy and is important for neuronal maintenance.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Ubiquitinated Proteins , Ubiquitination , Animals , Humans , Mice , Autophagy/genetics , Endoplasmic Reticulum/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ubiquitinated Proteins/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Intracellular Membranes/metabolism
7.
Nature ; 613(7942): 160-168, 2023 01.
Article in English | MEDLINE | ID: mdl-36477540

ABSTRACT

Multilocular adipocytes are a hallmark of thermogenic adipose tissue1,2, but the factors that enforce this cellular phenotype are largely unknown. Here, we show that an adipocyte-selective product of the Clstn3 locus (CLSTN3ß) present in only placental mammals facilitates the efficient use of stored triglyceride by limiting lipid droplet (LD) expansion. CLSTN3ß is an integral endoplasmic reticulum (ER) membrane protein that localizes to ER-LD contact sites through a conserved hairpin-like domain. Mice lacking CLSTN3ß have abnormal LD morphology and altered substrate use in brown adipose tissue, and are more susceptible to cold-induced hypothermia despite having no defect in adrenergic signalling. Conversely, forced expression of CLSTN3ß is sufficient to enforce a multilocular LD phenotype in cultured cells and adipose tissue. CLSTN3ß associates with cell death-inducing DFFA-like effector proteins and impairs their ability to transfer lipid between LDs, thereby restricting LD fusion and expansion. Functionally, increased LD surface area in CLSTN3ß-expressing adipocytes promotes engagement of the lipolytic machinery and facilitates fatty acid oxidation. In human fat, CLSTN3B is a selective marker of multilocular adipocytes. These findings define a molecular mechanism that regulates LD form and function to facilitate lipid utilization in thermogenic adipocytes.


Subject(s)
Adipocytes , Calcium-Binding Proteins , Lipid Metabolism , Membrane Proteins , Animals , Female , Humans , Mice , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Placenta , Triglycerides/metabolism , Endoplasmic Reticulum/metabolism , Lipid Droplets/metabolism , Fatty Acids/metabolism , Hypothermia/metabolism , Thermogenesis
8.
Cell Mol Life Sci ; 79(7): 389, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35773608

ABSTRACT

EWI2 is a transmembrane immunoglobulin superfamily (IgSF) protein that physically associates with tetraspanins and integrins. It inhibits cancer cells by influencing the interactions among membrane molecules including the tetraspanins and integrins. The present study revealed that, upon EWI2 silencing or ablation, the elevated movement and proliferation of cancer cells in vitro and increased cancer metastatic potential and malignancy in vivo are associated with (i) increases in clustering, endocytosis, and then activation of EGFR and (ii) enhancement of Erk MAP kinase signaling. These changes in signaling make cancer cells (i) undergo partial epithelial-to-mesenchymal (EMT) for more tumor progression and (ii) proliferate faster for better tumor formation. Inhibition of EGFR or Erk kinase can abrogate the cancer cell phenotypes resulting from EWI2 removal. Thus, to inhibit cancer cells, EWI2 prevents EGFR from clustering and endocytosis to restrain its activation and signaling.


Subject(s)
Antigens, CD , Endocytosis , ErbB Receptors , Membrane Proteins , Neoplasms , Antigens, CD/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Integrins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
9.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35022233

ABSTRACT

Synaptic cell-adhesion molecules (CAMs) organize the architecture and properties of neural circuits. However, whether synaptic CAMs are involved in activity-dependent remodeling of specific neural circuits is incompletely understood. Leucine-rich repeat transmembrane protein 3 (LRRTM3) is required for the excitatory synapse development of hippocampal dentate gyrus (DG) granule neurons. Here, we report that Lrrtm3-deficient mice exhibit selective reductions in excitatory synapse density and synaptic strength in projections involving the medial entorhinal cortex (MEC) and DG granule neurons, accompanied by increased neurotransmitter release and decreased excitability of granule neurons. LRRTM3 deletion significantly reduced excitatory synaptic innervation of hippocampal mossy fibers (Mf) of DG granule neurons onto thorny excrescences in hippocampal CA3 neurons. Moreover, LRRTM3 loss in DG neurons significantly decreased mossy fiber long-term potentiation (Mf-LTP). Remarkably, silencing MEC-DG circuits protected against the decrease in the excitatory synaptic inputs onto DG and CA3 neurons, excitability of DG granule neurons, and Mf-LTP in Lrrtm3-deficient mice. These results suggest that LRRTM3 may be a critical factor in activity-dependent synchronization of the topography of MEC-DG-CA3 excitatory synaptic connections. Collectively, our data propose that LRRTM3 shapes the target-specific structural and functional properties of specific hippocampal circuits.


Subject(s)
Cortical Synchronization/physiology , Hippocampus/physiology , Membrane Proteins/metabolism , Nerve Net/physiology , Nerve Tissue Proteins/metabolism , Synapses/physiology , Animals , CA3 Region, Hippocampal/metabolism , Dentate Gyrus/metabolism , Entorhinal Cortex/metabolism , Long-Term Potentiation , Membrane Proteins/deficiency , Mice, Knockout , Mossy Fibers, Hippocampal/metabolism , Nerve Tissue Proteins/deficiency , Neurons/metabolism , Pseudopodia/metabolism , Synaptic Transmission/physiology
10.
Sci China Life Sci ; 65(1): 193-205, 2022 01.
Article in English | MEDLINE | ID: mdl-33740186

ABSTRACT

Pulmonary surfactant is a lipid-protein complex secreted by alveolar type II epithelial cells and is essential for the maintenance of the delicate structure of mammalian alveoli to promote efficient gas exchange across the air-liquid barrier. The Golgi apparatus plays an important role in pulmonary surfactant modification and secretory trafficking. However, the physiological function of the Golgi apparatus in the transport of pulmonary surfactants is unclear. In the present study, deletion of GM130, which encodes for a matrix protein of the cis-Golgi cisternae, was shown to induce the disruption of the Golgi structure leading to impaired secretion of lung surfactant proteins and lipids. Specifically, the results of in vitro and in vivo analysis indicated that the loss of GM130 resulted in trapping of Sftpa in the endoplasmic reticulum, Sftpb and Sftpc accumulation in the Golgi apparatus, and an increase in the compensatory secretion of Sftpd. Moreover, global and epithelial-specific GM130 knockout in mice resulted in an enlargement of alveolar airspace and an increase in alveolar epithelial autophagy; however, surfactant repletion partially rescued the enlarged airspace defects in GM130-deficient mice. Therefore, our results demonstrate that GM130 and the mammalian Golgi apparatus play a critical role in the control of surfactant protein secretion in pulmonary epithelial cells.


Subject(s)
Autoantigens/physiology , Golgi Apparatus/metabolism , Membrane Proteins/physiology , Pulmonary Alveoli/metabolism , Pulmonary Surfactants/metabolism , Animals , Autoantigens/genetics , Golgi Apparatus/pathology , Golgi Apparatus/ultrastructure , Lung/pathology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout
11.
J Pediatr Endocrinol Metab ; 35(1): 11-18, 2022 Jan 27.
Article in English | MEDLINE | ID: mdl-34668366

ABSTRACT

Peroxisomal disorders are a heterogeneous group of diseases caused by mutations in a large number of genes. One of the genetic disorders known to cause this situation is ACBD5 (Acyl-CoA binding-domain-containing-5) gene mutations that have been described in recent years. Here, we report two siblings with a novel homozygous nonsense variation (c.1297C>T, p.Arg433*) in ACBD5 (NM_145698.4) gene using Clinical Exome Sequencing (Sophia Genetics).


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Membrane Proteins/genetics , Mutation , Peroxisomal Disorders/genetics , Adaptor Proteins, Signal Transducing/deficiency , Audiometry , Child , Child, Preschool , Female , Humans , Membrane Proteins/deficiency , Peroxisomal Disorders/diagnosis , Peroxisomal Disorders/physiopathology , Retinal Dystrophies/genetics
12.
Sci China Life Sci ; 65(1): 107-118, 2022 01.
Article in English | MEDLINE | ID: mdl-34279819

ABSTRACT

As the leading cause of worldwide hospital-acquired infection, Clostridioides difficile (C. difficile) infection has caused heavy economic and hospitalized burden, while its pathogenesis is not fully understood. Toxin B (TcdB) is one of the major virulent factors of C. difficile. Recently, CSPG4 and FZD2 were reported to be the receptors that mediate TcdB cellular entry. However, genetic ablation of genes encoding these receptors failed to completely block TcdB entry, implicating the existence of alternative receptor(s) for this toxin. Here, by employing the CRISPR-Cas9 screen in CSPG4-deficient HeLa cells, we identified LDL receptor-related protein-1 (LRP1) as a novel receptor for TcdB. Knockout of LRP1 in both CSPG4-deficient HeLa cells and colonic epithelium Caco2 cells conferred cells with increased TcdB resistance, while LRP1 overexpression sensitized cells to TcdB at a low concentration. Co-immunoprecipitation assay showed that LRP1 interacts with full-length TcdB. Moreover, CROPs domain, which is dispensable for TcdB's interaction with CSPG4 and FZD2, is sufficient for binding to LRP1. As such, our study provided evidence for a novel mechanism of TcdB entry and suggested potential therapeutic targets for treating C. difficile infection.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , CRISPR-Cas Systems , Chondroitin Sulfate Proteoglycans/deficiency , Chondroitin Sulfate Proteoglycans/metabolism , Clostridioides difficile/pathogenicity , Clostridium Infections , Frizzled Receptors , HeLa Cells , Humans , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Oligopeptides/metabolism
13.
FEBS Lett ; 596(4): 427-436, 2022 02.
Article in English | MEDLINE | ID: mdl-34939667

ABSTRACT

T helper cells, especially Th1 and Th17 cells, were reported to play a pivotal role in the pathogenesis of inflammatory bowel disease (IBD). However, the underlying factors regulating T cell functions in IBD progression remain to be fully elucidated. Here, we revealed that IL-17RD/Sef exacerbates DSS-induced colitis by regulating the balance of T cell subsets and their secretion of associated cytokines. We also observed that IL-17RD/Sef promotes colitis-associated tumorigenesis and negatively correlates with survival in both mouse and colorectal cancer patients. Our results suggested that IL-17RD/Sef functions as a regulator of T cell subsets to promote the inflammatory responses in the pathogenesis of IBD and colitis-associated colon cancer.


Subject(s)
Carcinogenesis/immunology , Colitis/immunology , Membrane Proteins/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Colitis/chemically induced , Colitis/genetics , Colitis/mortality , Colon/immunology , Colon/pathology , Dextran Sulfate/administration & dosage , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lymphocyte Count , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Survival Analysis , Th1 Cells/pathology , Th17 Cells/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
14.
Nature ; 600(7888): 324-328, 2021 12.
Article in English | MEDLINE | ID: mdl-34819670

ABSTRACT

Activation-induced cytidine deaminase (AID) catalyses the deamination of deoxycytidines to deoxyuracils within immunoglobulin genes to induce somatic hypermutation and class-switch recombination1,2. AID-generated deoxyuracils are recognized and processed by subverted base-excision and mismatch repair pathways that ensure a mutagenic outcome in B cells3-6. However, why these DNA repair pathways do not accurately repair AID-induced lesions remains unknown. Here, using a genome-wide CRISPR screen, we show that FAM72A is a major determinant for the error-prone processing of deoxyuracils. Fam72a-deficient CH12F3-2 B cells and primary B cells from Fam72a-/- mice exhibit reduced class-switch recombination and somatic hypermutation frequencies at immunoglobulin and Bcl6 genes, and reduced genome-wide deoxyuracils. The somatic hypermutation spectrum in B cells from Fam72a-/- mice is opposite to that observed in mice deficient in uracil DNA glycosylase 2 (UNG2)7, which suggests that UNG2 is hyperactive in FAM72A-deficient cells. Indeed, FAM72A binds to UNG2, resulting in reduced levels of UNG2 protein in the G1 phase of the cell cycle, coinciding with peak AID activity. FAM72A therefore causes U·G mispairs to persist into S phase, leading to error-prone processing by mismatch repair. By disabling the DNA repair pathways that normally efficiently remove deoxyuracils from DNA, FAM72A enables AID to exert its full effects on antibody maturation. This work has implications in cancer, as the overexpression of FAM72A that is observed in many cancers8 could promote mutagenesis.


Subject(s)
B-Lymphocytes , DNA Glycosylases , DNA Mismatch Repair , Immunoglobulin Class Switching , Membrane Proteins , Mutation , Neoplasm Proteins , Somatic Hypermutation, Immunoglobulin , Animals , Female , Humans , Mice , B-Lymphocytes/metabolism , CRISPR-Cas Systems , DNA Glycosylases/antagonists & inhibitors , DNA Glycosylases/metabolism , Epistasis, Genetic , HEK293 Cells , Immunoglobulin Class Switching/genetics , Immunoglobulin Switch Region/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Somatic Hypermutation, Immunoglobulin/genetics
15.
PLoS Pathog ; 17(11): e1010114, 2021 11.
Article in English | MEDLINE | ID: mdl-34843584

ABSTRACT

Malaria is a hazardous disease caused by Plasmodium parasites and often results in lethal complications, including malaria-associated acute respiratory distress syndrome (MA-ARDS). Parasite sequestration in the microvasculature is often observed, but its role in malaria pathogenesis and complications is still incompletely understood. We used skeleton binding protein-1 (SBP-1) KO parasites to study the role of sequestration in experimental MA-ARDS. The sequestration-deficiency of these SBP-1 KO parasites was confirmed with bioluminescence imaging and by measuring parasite accumulation in the lungs with RT-qPCR. The SBP-1 KO parasites induced similar lung pathology in the early stage of experimental MA-ARDS compared to wildtype (WT) parasites. Strikingly, the lung pathology resolved subsequently in more than 60% of the SBP-1 KO infected mice, resulting in prolonged survival despite the continuous presence of the parasite. This spontaneous disease resolution was associated with decreased inflammatory cytokine expression measured by RT-qPCR and lower expression of cytotoxic markers in pathogenic CD8+ T cells in the lungs of SBP-1 KO infected mice. These data suggest that SBP-1-mediated parasite sequestration and subsequent high parasite load are not essential for the development of experimental MA-ARDS but inhibit the resolution of the disease.


Subject(s)
Lung/parasitology , Malaria/complications , Membrane Proteins/deficiency , Plasmodium berghei/pathogenicity , Protozoan Proteins/metabolism , Respiratory Distress Syndrome/prevention & control , Animals , Disease Progression , Female , Lung/metabolism , Lung/pathology , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Protozoan Proteins/genetics , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/parasitology , Respiratory Distress Syndrome/pathology
16.
Sci Rep ; 11(1): 21817, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34751190

ABSTRACT

Proliferating cell nuclear antigen (PCNA) plays a critical role as a processivity clamp for eukaryotic DNA polymerases and a binding platform for many DNA replication and repair proteins. The enzymatic activities of PCNA loading and unloading have been studied extensively in vitro. However, the subcellular locations of PCNA loaders, replication complex C (RFC) and CTF18-RFC-like-complex (RLC), and PCNA unloader ATAD5-RLC remain elusive, and the role of their subunits RFC2-5 is unknown. Here we used protein fractionation to determine the subcellular localization of RFC and RLCs and affinity purification to find molecular requirements for the newly defined location. All RFC/RLC proteins were detected in the nuclease-resistant pellet fraction. RFC1 and ATAD5 were not detected in the non-ionic detergent-soluble and nuclease-susceptible chromatin fractions, independent of cell cycle or exogenous DNA damage. We found that small RFC proteins contribute to maintaining protein levels of the RFC/RLCs. RFC1, ATAD5, and RFC4 co-immunoprecipitated with lamina-associated polypeptide 2 (LAP2) α which regulates intranuclear lamin A/C. LAP2α knockout consistently reduced detection of RFC/RLCs in the pellet fraction, while marginally affecting total protein levels. Our findings strongly suggest that PCNA-mediated DNA transaction occurs through regulatory machinery associated with nuclear structures, such as the nuclear matrix.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , DNA-Binding Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Replication Protein C/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , Animals , Cell Cycle , Cell Fractionation , Cells, Cultured , Chromatin/metabolism , DNA Damage , DNA Replication , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Stability , Protein Subunits , Replication Protein C/chemistry , Subcellular Fractions/metabolism
17.
Cell Death Dis ; 12(11): 1049, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34741033

ABSTRACT

Heterotopic ossification (HO) represents a common problem after tendon injury with no effective treatment yet being developed. Tenomodulin (Tnmd), the best-known mature marker for tendon lineage cells, has important effects in tendon tissue aging and function. We have reported that loss of Tnmd leads to inferior early tendon repair characterized by fibrovascular scaring and therefore hypothesized that its lack will persistently cause deficient repair during later stages. Tnmd knockout (Tnmd-/-) and wild-type (WT) animals were subjected to complete Achilles tendon surgical transection followed by end-to-end suture. Lineage tracing revealed a reduction in tendon-lineage cells marked by ScleraxisGFP, but an increase in alpha smooth muscle actin myofibroblasts in Tnmd-/- tendon scars. At the proliferative stage, more pro-inflammatory M1 macrophages and larger collagen II cartilaginous template were detected in this group. At the remodeling stage, histological scoring revealed lower repair quality in the injured Tnmd-/- tendons, which was coupled with higher HO quantified by micro-CT. Tendon biomechanical properties were compromised in both groups upon injury, however we identified an abnormal stiffening of non-injured Tnmd-/- tendons, which possessed higher static and dynamic E-moduli. Pathologically thicker and abnormally shaped collagen fibrils were observed by TEM in Tnmd-/- tendons and this, together with augmented HO, resulted in diminished running capacity of Tnmd-/- mice. These novel findings demonstrate that Tnmd plays a protecting role against trauma-induced endochondral HO and can inspire the generation of novel therapeutics to accelerate repair.


Subject(s)
Achilles Tendon/pathology , Membrane Proteins/deficiency , Ossification, Heterotopic/etiology , Ossification, Heterotopic/pathology , Wound Healing , Wounds and Injuries/complications , Achilles Tendon/ultrastructure , Actins/metabolism , Animals , Bromodeoxyuridine/metabolism , Cell Count , Chondrogenesis , Cicatrix/pathology , Elastic Modulus , Elasticity , Extracellular Matrix/metabolism , Fibrillar Collagens/metabolism , Fibrillar Collagens/ultrastructure , Genotype , Green Fluorescent Proteins/metabolism , Inflammation/pathology , Macrophages/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Viscosity
18.
Cell Death Dis ; 12(11): 966, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667161

ABSTRACT

Numb, a stem cell fate determinant, acts as a tumor suppressor and is closely related to a wide variety of malignancies. Intrahepatic cholangiocarcinoma (iCCA) originates from hepatic progenitors (HPCs); however, the role of Numb in HPC malignant transformation and iCCA development is still unclear. A retrospective cohort study indicated that Numb was frequently decreased in tumor tissues and suggests poor prognosis in iCCA patients. Consistently, in a chemically induced iCCA mouse model, Numb was downregulated in tumor cells compared to normal cholangiocytes. In diet-induced chronic liver injury mouse models, Numb ablation significantly promoted histological impairment, HPC expansion, and tumorigenesis. Similarly, Numb silencing in cultured iCCA cells enhanced cell spheroid growth, invasion, metastasis, and the expression of stem cell markers. Mechanistically, Numb was found to bind to the Notch intracellular domain (NICD), and Numb ablation promoted Notch signaling; this effect was reversed when Notch signaling was blocked by γ-secretase inhibitor treatment. Our results suggested that loss of Numb plays an important role in promoting HPC expansion, HPC malignant transformation, and, ultimately, iCCA development in chronically injured livers. Therapies targeting suppressed Numb are promising for the treatment of iCCA.


Subject(s)
Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Liver/pathology , Membrane Proteins/deficiency , Nerve Tissue Proteins/deficiency , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Bile Duct Neoplasms/genetics , Body Weight , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Proliferation , Cholangiocarcinoma/genetics , Down-Regulation/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Ki-67 Antigen/metabolism , Liver Cirrhosis/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ Size , Prognosis , Protein Domains , Receptors, Notch/chemistry , Transcription Factor HES-1/metabolism
19.
Biochem Biophys Res Commun ; 582: 144-149, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34715405

ABSTRACT

The chemical synapse is one type of cell-adhesion system that transmits information from a neuron to another neuron in the complex neuronal network in the brain. Synaptic transmission is the rate-limiting step during the information processing in the neuronal network and its plasticity is involved in cognitive functions. Thus, morphological and electrophysiological analyses of synapses are of particular importance in neuroscience research. In the current study, we applied super-resolved three-dimensional stimulated emission depletion (3D-STED) microscopy for the morphological analyses of synapses. This approach allowed us to estimate the precise number of excitatory and inhibitory synapses in the mouse hippocampal tissue. We discovered a region-specific increase in excitatory synapses in a model mouse of autism spectrum disorder, Neuroligin-3 KO, with this method. This type of analysis will open a new field in developmental neuroscience in the future.


Subject(s)
Autism Spectrum Disorder/genetics , CA1 Region, Hippocampal/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Membrane Proteins/genetics , Microscopy/methods , Nerve Tissue Proteins/genetics , Neurons/metabolism , Synapses/genetics , Animals , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , CA1 Region, Hippocampal/diagnostic imaging , CA1 Region, Hippocampal/pathology , Cell Adhesion Molecules, Neuronal/deficiency , Cognition/physiology , Disease Models, Animal , Gene Knockout Techniques , Homer Scaffolding Proteins/genetics , Homer Scaffolding Proteins/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy/instrumentation , Nerve Tissue Proteins/deficiency , Neuroimaging/instrumentation , Neuroimaging/methods , Neurons/pathology , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission/physiology
20.
J Immunol ; 207(11): 2688-2698, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34697226

ABSTRACT

Regulation of BCR signaling has important consequences for generating effective Ab responses to pathogens and preventing production of autoreactive B cells during development. Currently defined functions of Fc receptor-like (FCRL) 1 include positive regulation of BCR-induced calcium flux, proliferation, and Ab production; however, the mechanistic basis of FCRL1 signaling and its contributions to B cell development remain undefined. Molecular characterization of FCRL1 signaling shows phosphotyrosine-dependent associations with GRB2, GRAP, SHIP-1, and SOS1, all of which can profoundly influence MAPK signaling. In contrast with previous characterizations of FCRL1 as a strictly activating receptor, we discover a role for FCRL1 in suppressing ERK activation under homeostatic and BCR-stimulated conditions in a GRB2-dependent manner. Our analysis of B cells in Fcrl1 -/- mice shows that ERK suppression by FCRL1 is associated with a restriction in the number of cells surviving splenic maturation in vivo. The capacity of FCRL1 to modulate ERK activation presents a potential for FCRL1 to be a regulator of peripheral B cell tolerance, homeostasis, and activation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/immunology , GRB2 Adaptor Protein/immunology , Membrane Proteins/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HEK293 Cells , Humans , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...