Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Cell ; 2024 May 09.
Article En | MEDLINE | ID: mdl-38754421

Clinical trials have identified ARID1A mutations as enriched among patients who respond favorably to immune checkpoint blockade (ICB) in several solid tumor types independent of microsatellite instability. We show that ARID1A loss in murine models is sufficient to induce anti-tumor immune phenotypes observed in ARID1A mutant human cancers, including increased CD8+ T cell infiltration and cytolytic activity. ARID1A-deficient cancers upregulated an interferon (IFN) gene expression signature, the ARID1A-IFN signature, associated with increased R-loops and cytosolic single-stranded DNA (ssDNA). Overexpression of the R-loop resolving enzyme, RNASEH2B, or cytosolic DNase, TREX1, in ARID1A-deficient cells prevented cytosolic ssDNA accumulation and ARID1A-IFN gene upregulation. Further, the ARID1A-IFN signature and anti-tumor immunity were driven by STING-dependent type I IFN signaling, which was required for improved responsiveness of ARID1A mutant tumors to ICB treatment. These findings define a molecular mechanism underlying anti-tumor immunity in ARID1A mutant cancers.

2.
Cell Death Dis ; 15(1): 56, 2024 01 16.
Article En | MEDLINE | ID: mdl-38225225

Although KRASG12C inhibitors have shown promising activity in lung adenocarcinomas harbouring KRASG12C, acquired resistance to these therapies eventually occurs in most patients. Re-expression of KRAS is thought to be one of the main causes of acquired resistance. However, the mechanism through which cancer cells re-express KRAS is not fully understood. Here, we report that the Hedgehog signal is induced by KRASG12C inhibitors and mediates KRAS re-expression in cancer cells treated with a KRASG12C inhibitor. Further, KRASG12C inhibitors induced the formation of primary cilia and activated the Hedgehog-GLI-1 pathway. GLI-1 binds to the KRAS promoter region, enhancing KRAS promoter activity and KRAS expression. Inhibition of GLI using siRNA or the smoothened (Smo) inhibitor suppressed re-expression of KRAS in cells treated with a KRASG12C inhibitor. In addition, we demonstrate that KRASG12C inhibitors decreased Aurora kinase A (AURKA) levels in cancer cells, and inhibition of AURKA using siRNA or inhibitors led to increased expression levels of GLI-1 and KRAS even in the absence of KRAS inhibitor. Ectopic expression of AURKA attenuated the effect of KRASG12C inhibitors on the expression of GLI-1 and re-expression of KRAS. Together, these findings demonstrate the important role of AURKA, primary cilia, and Hedgehog signals in the re-expression of KRAS and therefore the induction of acquired resistance to KRASG12C inhibitors, and provide a rationale for targeting Hedgehog signalling to overcome acquired resistance to KRASG12C inhibitors.


Hedgehog Proteins , Lung Neoplasms , Humans , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Aurora Kinase A/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mutation/genetics , RNA, Small Interfering/metabolism
3.
Clin Immunol ; 257: 109819, 2023 12.
Article En | MEDLINE | ID: mdl-37918467

Innate immune response is critical for the control of Listeria monocytogenes infection. Here, we identified developmentally regulated GTP-binding protein 2 (DRG2) in macrophages as a major regulator of the innate immune response against L. monocytogenes infection. Both whole-body DRG2 knockout (KO) mice and macrophage-specific DRG2 KO mice had low levels of IL-6 during early infection and increased susceptibility to L. monocytogenes infection. Following an initial impaired inflammatory response of macrophages upon i.p. L. monocytogenes infection, DRG2-/- mice showed delayed recruitment of neutrophils and monocytes into the peritoneal cavity, which led to elevated bacterial burden, inflammatory cytokine production at a late infection time point, and liver micro-abscesses. DRG2 deficiency decreased the transcriptional activity of NF-κB and impaired the inflammatory response of both bone marrow-derived and peritoneal macrophages upon L. monocytogenes stimulation. Our findings reveal that DRG2 in macrophages is critical for the initial inflammatory response and protection against L. monocytogenes infection.


GTP-Binding Proteins , Listeria monocytogenes , Listeriosis , Macrophages , Animals , Mice , Immunity, Innate , Listeriosis/immunology , Macrophages/immunology , Mice, Knockout , Monocytes , GTP-Binding Proteins/metabolism
4.
Elife ; 122023 10 23.
Article En | MEDLINE | ID: mdl-37869988

The fidelity of motor control requires the precise positional arrangement of motor pools and the establishment of synaptic connections between them. During neural development in the spinal cord, motor nerves project to specific target muscles and receive proprioceptive input from these muscles via the sensorimotor circuit. LIM-homeodomain transcription factors are known to play a crucial role in successively restricting specific motor neuronal fates. However, their exact contribution to limb-based motor pools and locomotor circuits has not been fully understood. To address this, we conducted an investigation into the role of Isl2, a LIM-homeodomain transcription factor, in motor pool organization. We found that deletion of Isl2 led to the dispersion of motor pools, primarily affecting the median motor column (MMC) and lateral motor column (LMC) populations. Additionally, hindlimb motor pools lacked Etv4 expression, and we observed reduced terminal axon branching and disorganized neuromuscular junctions in Isl2-deficient mice. Furthermore, we performed transcriptomic analysis on the spinal cords of Isl2-deficient mice and identified a variety of downregulated genes associated with motor neuron (MN) differentiation, axon development, and synapse organization in hindlimb motor pools. As a consequence of these disruptions, sensorimotor connectivity and hindlimb locomotion were impaired in Isl2-deficient mice. Taken together, our findings highlight the critical role of Isl2 in organizing motor pool position and sensorimotor circuits in hindlimb motor pools. This research provides valuable insights into the molecular mechanisms governing motor control and its potential implications for understanding motor-related disorders in humans.


DNA-Binding Proteins , Transcription Factors , Animals , Humans , Mice , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Motor Neurons/physiology , Spinal Cord/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Cell Mol Gastroenterol Hepatol ; 15(1): 213-236, 2023.
Article En | MEDLINE | ID: mdl-36167263

BACKGROUND & AIMS: Histamine in the stomach traditionally is considered to regulate acid secretion but also has been reported to participate in macrophage differentiation, which plays an important role in tissue homeostasis. Therefore, this study aimed to uncover the precise role of histamine in mediating macrophage differentiation and in maintaining stomach homeostasis. METHODS: Here, we expand on this role using histidine decarboxylase knockout (Hdc-/-) mice with hypertrophic gastropathy. In-depth in vivo studies were performed in Hdc-/- mice, germ-free Hdc-/- mice, and bone-marrow-transplanted Hdc-/- mice. The stomach macrophage populations and function were characterized by flow cytometry. To identify stomach macrophages and find the new macrophage population, we performed single-cell RNA sequencing analysis on Hdc+/+ and Hdc-/- stomach tissues. RESULTS: Single-cell RNA sequencing and flow cytometry of the stomach cells of Hdc-/- mice showed alterations in the ratios of 3 distinct tissue macrophage populations (F4/80+Il1bhigh, F4/80+CD93+, and F4/80-MHC class IIhighCD74high). Tissue macrophages of the stomachs of Hdc-/- mice showed impaired phagocytic activity, increasing the bacterial burden of the stomach and attenuating hypertrophic gastropathy in germ-free Hdc-/- mice. The transplantation of bone marrow cells of Hdc+/+ mice to Hdc-/- mice recovered the normal differentiation of stomach macrophages and relieved the hypertrophic gastropathy of Hdc-/- mice. CONCLUSIONS: This study showed the importance of histamine signaling in tissue macrophage differentiation and maintenance of gastric homeostasis through the suppression of bacterial overgrowth in the stomach.


Cell Differentiation , Histamine , Macrophages , Stomach , Animals , Mice , Histamine/physiology , Histidine Decarboxylase/genetics , Stomach/microbiology , Blind Loop Syndrome , Mice, Knockout
6.
Adv Sci (Weinh) ; 9(15): e2103526, 2022 05.
Article En | MEDLINE | ID: mdl-35322595

Kidney organoids derived from human pluripotent stem cells (hPSCs) have extensive potential for disease modelling and regenerative medicine. However, the limited vascularization and immaturity of kidney organoids have been still remained to overcome. Extracellular matrix (ECM) can provide mechanical support and a biochemical microenvironment for cell growth and differentiation. Here in vitro methods using a kidney decellularized extracellular matrix (dECM) hydrogel to culture hPSC-derived kidney organoids, which have extensive vascular network and their own endothelial cells, are reported. Single-cell transcriptomics reveal that the vascularized kidney organoids cultured using the kidney dECM have more mature patterns of glomerular development and higher similarity to human kidney than those cultured without the kidney dECM. Differentiation of α-galactosidase A (GLA)-knock-out hPSCs generated using CRISPR/Cas9 into kidney organoids by the culture method using kidney dECM efficiently recapitulate Fabry nephropathy with vasculopathy. Transplantation of kidney organoids with kidney dECM into kidney of mouse accelerates the recruitment of endothelial cells from the host mouse kidney and maintains vascular integrity with the more organized slit diaphragm-like structures than those without kidney dECM. The kidney dECM methodology for inducing extensive vascularization and maturation of kidney organoids can be applied to studies for kidney development, disease modeling, and regenerative medicine.


Organoids , Pluripotent Stem Cells , Animals , Decellularized Extracellular Matrix , Endothelial Cells , Humans , Kidney , Mice
7.
Brief Bioinform ; 23(2)2022 03 10.
Article En | MEDLINE | ID: mdl-35226074

The development of autoimmune diseases following SARS-CoV-2 infection, including multisystem inflammatory syndrome, has been reported, and several mechanisms have been suggested, including molecular mimicry. We developed a scalable, comparative immunoinformatics pipeline called cross-reactive-epitope-search-using-structural-properties-of-proteins (CRESSP) to identify cross-reactive epitopes between a collection of SARS-CoV-2 proteomes and the human proteome using the structural properties of the proteins. Overall, by searching 4 911 245 proteins from 196 352 SARS-CoV-2 genomes, we identified 133 and 648 human proteins harboring potential cross-reactive B-cell and CD8+ T-cell epitopes, respectively. To demonstrate the robustness of our pipeline, we predicted the cross-reactive epitopes of coronavirus spike proteins, which were recognized by known cross-neutralizing antibodies. Using single-cell expression data, we identified PARP14 as a potential target of intermolecular epitope spreading between the virus and human proteins. Finally, we developed a web application (https://ahs2202.github.io/3M/) to interactively visualize our results. We also made our pipeline available as an open-source CRESSP package (https://pypi.org/project/cressp/), which can analyze any two proteomes of interest to identify potentially cross-reactive epitopes between the proteomes. Overall, our immunoinformatic resources provide a foundation for the investigation of molecular mimicry in the pathogenesis of autoimmune and chronic inflammatory diseases following COVID-19.


Computational Biology/methods , Epitopes/chemistry , Epitopes/immunology , SARS-CoV-2/immunology , Software , Viral Proteins/chemistry , Viral Proteins/immunology , Algorithms , Cross Reactions/immunology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Models, Molecular , Molecular Mimicry , Neural Networks, Computer , Proteome , Proteomics/methods , Structure-Activity Relationship , Web Browser
8.
BMC Immunol ; 22(1): 30, 2021 05 12.
Article En | MEDLINE | ID: mdl-33980160

BACKGROUND: Triggering receptor expressed on myeloid cells 2 (TREM2) signaling is considered to regulate anti-inflammatory responses in macrophages, dendritic cell maturation, osteoclast development, induction of obesity, and Alzheimer's disease pathogenesis. However, little is known regarding the effect of TREM2 on natural killer (NK) cells. RESULTS: Here, we demonstrated for the first time that CD3-CD122+NK1.1+ precursor NK (pNK) cells expressed TREM2 and their population increased in TREM2-overexpressing transgenic (TREM2-TG) mice compared with that in female C57BL/6 J wild type (WT) mice. Both NK cell-activating receptors and NK cell-associated genes were expressed at higher levels in various tissues of TREM2-TG mice than in WT mice. In addition, bone marrow-derived hematopoietic stem cells (HSCs) of TREM2-TG mice (TG-HSCs) successfully differentiated into NK cells in vitro, with a higher yield from TG-HSCs than from WT-HSCs. In contrast, TREM2 signaling inhibition by TREM2-Ig or a phosphatidylinositol 3-kinase (PI3K) inhibitor affected the expression of the NK cell receptor repertoire and decreased the expression levels of NK cell-associated genes, resulting in significant impairment of NK cell differentiation. Moreover, in melanoma-bearing WT mice, injection of bone marrow cells from TREM2-TG mice exerted greater antitumor effects than that with cells from WT control mice. CONCLUSIONS: Collectively, our data clearly showed that TREM2 promoted NK cell development and tumor regression, suggesting TREM2 as a new candidate for cancer immunotherapy.


Bone Marrow Cells/immunology , Killer Cells, Natural/immunology , Melanoma/immunology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Animals , Bone Marrow Transplantation , CD3 Complex/metabolism , Cell Differentiation , Female , Humans , Immunotherapy, Adoptive , Interleukin-2 Receptor beta Subunit/metabolism , Melanoma/therapy , Melanoma, Experimental , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental , Receptors, Immunologic/genetics
9.
Cancers (Basel) ; 11(9)2019 Sep 06.
Article En | MEDLINE | ID: mdl-31489935

TREM2 (triggering receptor expressed on myeloid cells) is involved in the development of malignancies. However, the function of TREM2 in colorectal cancer has not been clearly elucidated. Here, we investigated TREM2 function for the first time in colorectal epithelial cancer cells and demonstrated that TREM2 is a novel tumor suppressor in colorectal carcinoma. Blockade of TREM2 significantly promoted the proliferation of HT29 colorectal carcinoma cells by regulating cell cycle-related factors, such as p53 phosphorylation and p21 and cyclin D1 protein levels. HT29 cell migration was also increased by TREM2 inhibition via MMP9 (matrix metalloproteinase 9) expression upregulation. Furthermore, we found that the tumor suppressor effects of TREM2 were associated with Wnt/ß-catenin and extracellular signal-regulated kinase (ERK) signaling. Importantly, the effect of TREM2 in the suppression of tumor development was demonstrated by in vivo and in vitro assays, as well as in human colon cancer patient tissue arrays. Overall, our results identify TREM2 as a potential prognostic biomarker and therapeutic target for colorectal cancer.

10.
Diabetes ; 64(1): 117-27, 2015 Jan.
Article En | MEDLINE | ID: mdl-25114293

Triggering receptor expressed on myeloid cells 2 (TREM2) is known to be involved in the anti-inflammatory response and osteoclast development. However, the role of TREM2 in adipogenesis or obesity has not yet been defined. The effect of TREM2 on adipogenesis and obesity was investigated in TREM2 transgenic (TG) mice on a high-fat diet (HFD). To block TREM2 signaling, a neutralizing fusion protein specific for TREM2 (TREM2-Ig) was used. TG mice were much more obese than wild-type mice after feeding with an HFD, independent of the quantity of food intake. These HFD-fed TG mice manifested adipocyte hypertrophy, glucose and insulin resistance, and hepatic steatosis. The expression of adipogenic regulator genes, such as peroxisome proliferator-activated receptor γ and CCAAT/enhancer-binding protein α, was markedly increased in HFD-fed TG mice. Additionally, HFD-fed TG mice exhibited decreased Wnt10b expression and increased GSK-3ß (glycogen synthase kinase-3ß)-mediated ß-catenin phosphorylation. In contrast, the blockade of TREM2 signaling using TREM2-Ig resulted in the inhibition of adipocyte differentiation in vitro and a reduction in body weight in vivo by downregulating the expression of adipogenic regulators. Our data demonstrate that TREM2 promotes adipogenesis and diet-induced obesity by upregulating adipogenic regulators in conjunction with inhibiting the Wnt10b/ß-catenin signaling pathway.


Adipocytes/metabolism , Adipogenesis/physiology , Membrane Glycoproteins/metabolism , Myeloid Cells/metabolism , Obesity/metabolism , Receptors, Immunologic/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/immunology , Animals , Calorimetry, Indirect , Cell Differentiation/physiology , Diet, High-Fat , Energy Metabolism/physiology , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Insulin Resistance/physiology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Obesity/genetics , Obesity/immunology , Primary Cell Culture , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Signal Transduction/physiology , Wnt Signaling Pathway/physiology
11.
Biochem Biophys Res Commun ; 409(4): 792-8, 2011 Jun 17.
Article En | MEDLINE | ID: mdl-21621523

Major histocompatibility complex (MHC) class I is a major host defense mechanism against viral infections such as type 16 and type 18 of the human papillomavirus (HPV). Here, we found that the E6 oncogene from HPV16, but not HPV18, suppressed MHC I expression. Ectopic expression of HPV16E6 in HeLa cells, which are infected with HPV18, suppressed MHC I expression, and that knockdown by antisense or siRNA of the HPV16E6 strongly enhanced MHC I expression in Caski cells, which are infected with HPV18, but not HPV16. The expression of HPV16E6 strongly enhanced cellular resistance to cytotoxic T lymphocytes (CTLs)-mediated lytic activity, and knockdown of HPV16E6 by antisense had the opposite effect. The regulation of HPV16E6-mediated MHC I suppression might be through the regulation of lymphotoxin (LT) and its receptor, LTßR. In addition, cells from the spleen and liver of LTα- or LTßR-deficient mice showed increased MHC I expression. Overall, these results demonstrated that the E6 oncogene of HPV16 might play an important role in cell transformation and cancer development through LT-mediated MHC I downregulation in humans.


Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Human papillomavirus 16/genetics , Lymphotoxin-alpha/genetics , Lymphotoxin-beta/genetics , Oncogene Proteins, Viral/genetics , Repressor Proteins/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology , Animals , Female , Gene Knockdown Techniques , HeLa Cells , Histocompatibility Antigens Class I/immunology , Human papillomavirus 18/genetics , Humans , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Up-Regulation , Uterine Cervical Neoplasms/immunology
...