Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26.845
Filtrer
1.
Cell Death Dis ; 15(7): 515, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39025844

RÉSUMÉ

Although multiple myeloma (MM) responds well to immunotherapeutic treatment, certain portions of MM are still unresponsive or relapse after immunotherapy. Other immune molecules are needed for the immunotherapy of MM. Here, we revealed that leukocyte immunoglobulin-like receptor B4 (LILRB4) was highly expressed in multiple myeloma cell lines and patient samples and that the expression of LILRB4 was adversely correlated with the overall survival of MM patients. Knockdown of LILRB4 efficiently delayed the growth of MM cells both in vitro and in vivo. Mechanistically, IKZF1 transactivated LILRB4 expression to trigger the downstream of STAT3-PFKFB1 pathways to support MM cell proliferation. Blockade of LILRB4 signaling by blocking antibodies can effectively inhibit MM progression. Our data show that targeting LILRB4 is potentially an additional therapeutic strategy for the immunotherapeutic treatment of MM.


Sujet(s)
Myélome multiple , Récepteurs immunologiques , Facteur de transcription STAT-3 , Transduction du signal , Myélome multiple/anatomopathologie , Myélome multiple/métabolisme , Myélome multiple/génétique , Humains , Facteur de transcription STAT-3/métabolisme , Animaux , Lignée cellulaire tumorale , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Souris , Prolifération cellulaire , Facteur de transcription Ikaros/métabolisme , Facteur de transcription Ikaros/génétique , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Femelle , Régulation de l'expression des gènes tumoraux , Mâle
2.
Nat Commun ; 15(1): 5694, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38972873

RÉSUMÉ

Tumor-associated myeloid-derived cells (MDCs) significantly impact cancer prognosis and treatment responses due to their remarkable plasticity and tumorigenic behaviors. Here, we integrate single-cell RNA-sequencing data from different cancer types, identifying 29 MDC subpopulations within the tumor microenvironment. Our analysis reveals abnormally expanded MDC subpopulations across various tumors and distinguishes cell states that have often been grouped together, such as TREM2+ and FOLR2+ subpopulations. Using deconvolution approaches, we identify five subpopulations as independent prognostic markers, including states co-expressing TREM2 and PD-1, and FOLR2 and PDL-2. Additionally, TREM2 alone does not reliably predict cancer prognosis, as other TREM2+ macrophages show varied associations with prognosis depending on local cues. Validation in independent cohorts confirms that FOLR2-expressing macrophages correlate with poor clinical outcomes in ovarian and triple-negative breast cancers. This comprehensive MDC atlas offers valuable insights and a foundation for futher analyses, advancing strategies for treating solid cancers.


Sujet(s)
Glycoprotéines membranaires , Cellules myéloïdes , Tumeurs , Récepteurs immunologiques , Analyse sur cellule unique , Microenvironnement tumoral , Humains , Analyse sur cellule unique/méthodes , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Cellules myéloïdes/métabolisme , Cellules myéloïdes/anatomopathologie , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Pronostic , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Femelle , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/génétique , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Antigène CD274/métabolisme , Antigène CD274/génétique
3.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39000444

RÉSUMÉ

The taurine transporter (TauT, SLC6A6) is a member of the solute carrier 6 (SLC6) family, which plays multiple physiological roles. The SLC6 family is divided into four subfamilies: GABA (γ-aminobutyric acid), monoamine, glycine and neutral amino acid transporters. Proteins from the GABA group, including the taurine transporter, are primarily considered therapeutic targets for treating central nervous system disorders. However, recent studies have suggested that inhibitors of SLC6A6 could also serve as anticancer agents. Overexpression of TauT has been associated with the progression of colon and gastric cancer. The pool of known ligands of this transporter is limited and the exact spatial structure of taurine transporter remains unsolved. Understanding its structure could aid in the development of novel inhibitors. Therefore, we utilized homology modelling techniques to create models of TauT. Docking studies and molecular dynamics simulations were conducted to describe protein-ligand interactions. We compared the obtained information for TauT with literature data on other members of the GABA transporter group. Our in silico analysis allowed us to characterize the transporter structure and point out amino acids crucial for ligand binding: Glu406, Gly62 and Tyr138. The significance of selected residues was confirmed through structural studies of mutants. These results will aid in the development of novel taurine transporter inhibitors, which can be explored as anticancer agents.


Sujet(s)
Transporteurs de GABA , Protéines de transport membranaire , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Humains , Transporteurs de GABA/métabolisme , Transporteurs de GABA/composition chimique , Transporteurs de GABA/génétique , Protéines de transport membranaire/composition chimique , Protéines de transport membranaire/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/génétique , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/composition chimique , Glycoprotéines membranaires/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Ligands , Séquence d'acides aminés , Liaison aux protéines
4.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-39000569

RÉSUMÉ

Regulation of neuroinflammation is critical for maintaining central nervous system (CNS) homeostasis and holds therapeutic promise in autoimmune diseases such as multiple sclerosis (MS). Previous studies have highlighted the significance of selective innate signaling in triggering anti-inflammatory mechanisms, which play a protective role in an MS-like disease, experimental autoimmune encephalomyelitis (EAE). However, the individual intra-CNS administration of specific innate receptor ligands or agonists, such as for toll-like receptor 7 (TLR7) and nucleotide-binding oligomerization-domain-containing protein 2 (NOD2), failed to elicit the desired anti-inflammatory response in EAE. In this study, we investigated the potential synergistic effect of targeting both TLR7 and NOD2 simultaneously to prevent EAE progression. Our findings demonstrate that simultaneous intrathecal administration of NOD2- and TLR7-agonists led to synergistic induction of Type I IFN (IFN I) and effectively suppressed EAE in an IFN I-dependent manner. Suppression of EAE was correlated with a significant decrease in the infiltration of monocytes, granulocytes, and natural killer cells, reduced demyelination, and downregulation of IL-1ß, CCL2, and IFNγ gene expression in the spinal cord. These results underscore the therapeutic promise of concurrently targeting the TLR7 and NOD2 pathways in alleviating neuroinflammation associated with MS, paving the way for novel and more efficacious treatment strategies.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Sclérose en plaques , Protéine adaptatrice de signalisation NOD2 , Récepteur de type Toll-7 , Animaux , Récepteur de type Toll-7/métabolisme , Récepteur de type Toll-7/agonistes , Protéine adaptatrice de signalisation NOD2/métabolisme , Protéine adaptatrice de signalisation NOD2/génétique , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/métabolisme , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Sclérose en plaques/traitement médicamenteux , Sclérose en plaques/métabolisme , Souris , Souris de lignée C57BL , Immunité innée/effets des médicaments et des substances chimiques , Femelle , Moelle spinale/métabolisme , Moelle spinale/anatomopathologie , Moelle spinale/effets des médicaments et des substances chimiques , Glycoprotéines membranaires/métabolisme , Interféron de type I/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
5.
Cell Mol Life Sci ; 81(1): 302, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39008111

RÉSUMÉ

DNAX-activating protein of 12 kDa (DAP12) is a transmembrane adapter protein expressed in lymphoid and myeloid lineage cells. It interacts with several immunoreceptors forming functional complexes that trigger intracellular signaling pathways. One of the DAP12 associated receptors is the triggering receptor expressed on myeloid cells 2 (TREM2). Mutations in both DAP12 and TREM2 have been linked to neurodegenerative diseases. However, mechanisms involved in the regulation of subcellular trafficking and turnover of these proteins are not well understood. Here, we demonstrate that proteasomal degradation of DAP12 is increased in the absence of TREM2. Interestingly, unassembled DAP12 is also retained in early secretory compartments, including the endoplasmic reticulum (ER) and the ER-Golgi intermediate compartment (ERGIC), thereby preventing its transport to the plasma membrane. We also show that unassembled DAP12 interacts with the retention in ER sorting receptor 1 (RER1). The deletion of endogenous RER1 decreases expression of functional TREM2-DAP12 complexes and membrane proximal signaling, and resulted in almost complete inhibition of phagocytic activity in THP-1 differentiated macrophage-like cells. These results indicate that RER1 acts as an important regulator of DAP12 containing immunoreceptor complexes and immune cell function.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Réticulum endoplasmique , Glycoprotéines membranaires , Récepteurs immunologiques , Voie de sécrétion , Humains , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Réticulum endoplasmique/métabolisme , Voie de sécrétion/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Cellules HEK293 , Transduction du signal , Phagocytose/génétique , Macrophages/métabolisme , Transport des protéines , Liaison aux protéines , Animaux , Appareil de Golgi/métabolisme , Protéines du transport vésiculaire/métabolisme , Protéines du transport vésiculaire/génétique , Membrane cellulaire/métabolisme
6.
Commun Biol ; 7(1): 845, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987622

RÉSUMÉ

Adult Neural Stem Cells (aNSCs) in the ventricular-subventricular zone (V-SVZ) are largely quiescent. Here, we characterize the mechanism underlying the functional role of a cell-signalling inhibitory protein, LRIG1, in the control of aNSCs proliferation. Using Lrig1 knockout models, we show that Lrig1 ablation results in increased aNSCs proliferation with no change in neuronal progeny and that this hyperproliferation likely does not result solely from activation of the epidermal growth factor receptor (EGFR). Loss of LRIG1, however, also leads to impaired activation of transforming growth factor beta (TGFß) and bone morphogenic protein (BMP) signalling. Biochemically, we show that LRIG1 binds TGFß/BMP receptors and the TGFß1 ligand. Finally, we show that the consequences of these interactions are to facilitate SMAD phosphorylation. Collectively, these data suggest that unlike in embryonic NSCs where EGFR may be the primary mechanism of action, in aNSCs, LRIG1 and TGFß pathways function together to fulfill their inhibitory roles.


Sujet(s)
Protéines morphogénétiques osseuses , Prolifération cellulaire , Glycoprotéines membranaires , Cellules souches neurales , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Facteur de croissance transformant bêta/métabolisme , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Souris , Protéines morphogénétiques osseuses/métabolisme , Souris knockout , Cellules souches adultes/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Protéines de tissu nerveux
7.
Hematology ; 29(1): 2372482, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38994874

RÉSUMÉ

BACKGROUND: CD83 are closely related to the pathogenesis of immune thrombocytopenia (ITP), but the exact mechanism remains unclear. AIM: To explore the relationship between CD83 and CD4+ T cell subsets and clarify the role of CD83 in the pathogenesis of ITP. METHODS: RT-qPCR and Flow cytometry were used to illustrate CD83 expression. The downregulation and overexpression of DC-CD83 were co-cultured with CD4+ T cells to detect cell proliferation, co-cultured supernatant cytokines and Tregs expression. RESULTS: The results indicate that the ITP patients showed higher expression of CD83 than the healthy controls. The proliferation of CD4+ T cells was inhibited by downregulation of DCs-CD83 but promoted by overexpression of DCs-CD83. siRNA-CD83 inhibited proinflammatory IFN-γ and IL-17 secretion while raising TGF-ß, IL-10 concentrations. Overexpression of DCs-CD83 promoted Tregs expression. CONCLUSION: The Th1/Th2 and Th17/Tregs polarization were reversed via interfering DCs with siRNA-CD83. CD83 plays an important role in ITP pathogenesis, suggesting novel treatment for ITP patients.


Sujet(s)
Antigènes CD , , Immunoglobulines , Glycoprotéines membranaires , Purpura thrombopénique idiopathique , Humains , Purpura thrombopénique idiopathique/immunologie , Purpura thrombopénique idiopathique/anatomopathologie , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Antigènes CD/métabolisme , Immunoglobulines/génétique , Immunoglobulines/métabolisme , Femelle , Mâle , Adulte , Adulte d'âge moyen , Cytokines/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme
8.
Front Immunol ; 15: 1415565, 2024.
Article de Anglais | MEDLINE | ID: mdl-38989285

RÉSUMÉ

How the microbiome regulates responses of systemic innate immune cells is unclear. In the present study, our purpose was to document a novel mechanism by which the microbiome mediates crosstalk with the systemic innate immune system. We have identified a family of microbiome Bacteroidota-derived lipopeptides-the serine-glycine (S/G) lipids, which are TLR2 ligands, access the systemic circulation, and regulate proinflammatory responses of splenic monocytes. To document the role of these lipids in regulating systemic immunity, we used oral gavage with an antibiotic to decrease the production of these lipids and administered exogenously purified lipids to increase the systemic level of these lipids. We found that decreasing systemic S/G lipids by decreasing microbiome Bacteroidota significantly enhanced splenic monocyte proinflammatory responses. Replenishing systemic levels of S/G lipids via exogenous administration returned splenic monocyte responses to control levels. Transcriptomic analysis demonstrated that S/G lipids regulate monocyte proinflammatory responses at the level of gene expression of a small set of upstream inhibitors of TLR and NF-κB pathways that include Trem2 and Irf4. Consistent with enhancement in proinflammatory cytokine responses, decreasing S/G lipids lowered gene expression of specific pathway inhibitors. Replenishing S/G lipids normalized gene expression of these inhibitors. In conclusion, our results suggest that microbiome-derived S/G lipids normally establish a level of buffered signaling activation necessary for well-regulated innate immune responses in systemic monocytes. By regulating gene expression of inflammatory pathway inhibitors such as Trem2, S/G lipids merit broader investigation into the potential dysfunction of other innate immune cells, such as microglia, in diseases such as Alzheimer's disease.


Sujet(s)
Monocytes , Transduction du signal , Monocytes/immunologie , Monocytes/métabolisme , Monocytes/effets des médicaments et des substances chimiques , Animaux , Souris , Microbiote/immunologie , Souris de lignée C57BL , Immunité innée , Récepteur de type Toll-2/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Lipopeptides/pharmacologie , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Facteur de transcription NF-kappa B/métabolisme , Inflammation/immunologie , Facteurs de régulation d'interféron/métabolisme , Facteurs de régulation d'interféron/génétique , Mâle , Lipides , Rate/immunologie , Rate/métabolisme , Cytokines/métabolisme , Femelle
9.
J Neuroinflammation ; 21(1): 166, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956653

RÉSUMÉ

BACKGROUND: Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM. METHODS: A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR. RESULTS: Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation. CONCLUSIONS: HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.


Sujet(s)
Anxiété , Diabète de type 2 , Alimentation riche en graisse , Hypoxie , Glycoprotéines membranaires , Souris de lignée C57BL , Récepteur à l'interféron alpha-bêta , Récepteurs immunologiques , Transduction du signal , Animaux , Souris , Alimentation riche en graisse/effets indésirables , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Anxiété/étiologie , Anxiété/métabolisme , Transduction du signal/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Hypoxie/métabolisme , Hypoxie/complications , Mâle , Diabète de type 2/complications , Diabète de type 2/métabolisme , Diabète de type 2/psychologie , Récepteur à l'interféron alpha-bêta/métabolisme , Récepteur à l'interféron alpha-bêta/génétique , Diabète expérimental/complications , Diabète expérimental/métabolisme , Microglie/métabolisme , Facteur de transcription STAT-1/métabolisme , Syndrome d'apnées obstructives du sommeil/complications , Syndrome d'apnées obstructives du sommeil/métabolisme , Syndrome d'apnées obstructives du sommeil/psychologie
10.
Invest Ophthalmol Vis Sci ; 65(8): 10, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38958972

RÉSUMÉ

Purpose: Retinopathy of prematurity (ROP) results from postnatal hyperoxia exposure in premature infants and is characterized by aberrant neovascularization of retinal blood vessels. Epithelial membrane protein-2 (EMP2) regulates hypoxia-inducible factor (HIF)-induced vascular endothelial growth factor (VEGF) production in the ARPE-19 cell line and genetic knock-out of Emp2 in a murine oxygen-induced retinopathy (OIR) model attenuates neovascularization. We hypothesize that EMP2 blockade via intravitreal injection protects against neovascularization. Methods: Ex vivo choroid sprouting assay was performed, comparing media and human IgG controls versus anti-EMP2 antibody (Ab) treatment. In vivo, eyes from wild-type (WT) mice exposed to hyperoxia from postnatal (P) days 7 to 12 were treated with P12 intravitreal injections of control IgG or anti-EMP2 Abs. Neovascularization was assessed at P17 by flat mount imaging. Local and systemic effects of anti-EMP2 Ab treatment were assessed. Results: Choroid sprouts treated with 30 µg/mL of anti-EMP2 Ab demonstrated a 48% reduction in vessel growth compared to control IgG-treated sprouts. Compared to IgG-treated controls, WT OIR mice treated with 4 µg/g of intravitreal anti-EMP2 Ab demonstrated a 42% reduction in neovascularization. They demonstrated down-regulation of retinal gene expression in pathways related to vasculature development and up-regulation in genes related to fatty acid oxidation and tricarboxylic acid cycle respiratory electron transport, compared to controls. Anti-EMP2 Ab-treated OIR mice did not exhibit gross retinal histologic abnormalities, vision transduction abnormalities, or weight loss. Conclusions: Our results suggest that EMP2 blockade could be a local and specific treatment modality for retinal neovascularization in oxygen-induced retinopathies, without systemic adverse effects.


Sujet(s)
Animaux nouveau-nés , Modèles animaux de maladie humaine , Injections intravitréennes , Souris de lignée C57BL , Oxygène , Néovascularisation rétinienne , Rétinopathie du prématuré , Animaux , Souris , Oxygène/toxicité , Néovascularisation rétinienne/métabolisme , Néovascularisation rétinienne/prévention et contrôle , Néovascularisation rétinienne/anatomopathologie , Rétinopathie du prématuré/traitement médicamenteux , Rétinopathie du prématuré/métabolisme , Glycoprotéines membranaires/antagonistes et inhibiteurs , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Hyperoxie/complications , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Humains
11.
Nat Commun ; 15(1): 6067, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39025856

RÉSUMÉ

After recognizing its ligand lipopolysaccharide, Toll-like receptor 4 (TLR4) recruits adaptor proteins to the cell membrane, thereby initiating downstream signaling and triggering inflammation. Whether this recruitment of adaptor proteins is dependent solely on protein-protein interactions is unknown. Here, we report that the sphingolipid sphinganine physically interacts with the adaptor proteins MyD88 and TIRAP and promotes MyD88 recruitment in macrophages. Myeloid cell-specific deficiency in serine palmitoyltransferase long chain base subunit 2, which encodes the key enzyme catalyzing sphingolipid biosynthesis, decreases the membrane recruitment of MyD88 and inhibits inflammatory responses in in vitro bone marrow-derived macrophage and in vivo sepsis models. In a melanoma mouse model, serine palmitoyltransferase long chain base subunit 2 deficiency decreases anti-tumor myeloid cell responses and increases tumor growth. Therefore, sphinganine biosynthesis is required for the initiation of TLR4 signal transduction and serves as a checkpoint for macrophage pattern recognition in sepsis and melanoma mouse models.


Sujet(s)
Macrophages , Mélanome , Facteur de différenciation myéloïde-88 , Sepsie , Serine C-palmitoyltransferase , Sphingosine , Récepteur de type Toll-4 , Animaux , Récepteur de type Toll-4/métabolisme , Sepsie/métabolisme , Macrophages/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Souris , Sphingosine/analogues et dérivés , Sphingosine/métabolisme , Mélanome/métabolisme , Mélanome/anatomopathologie , Mélanome/génétique , Serine C-palmitoyltransferase/métabolisme , Serine C-palmitoyltransferase/génétique , Humains , Transduction du signal , Modèles animaux de maladie humaine , Inflammation/métabolisme , Récepteurs à l'interleukine-1/métabolisme , Récepteurs à l'interleukine-1/génétique , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Souris de lignée C57BL , Souris knockout , Cellules HEK293 , Lipopolysaccharides
12.
J Exp Clin Cancer Res ; 43(1): 183, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951916

RÉSUMÉ

BACKGROUND: Leukocyte Ig-like receptor B family 4 (LILRB4) as an immune checkpoint on myeloid cells is a potential target for tumor therapy. Extensive osteolytic bone lesion is the most characteristic feature of multiple myeloma. It is unclear whether ectopic LILRB4 on multiple myeloma regulates bone lesion. METHODS: The conditioned medium (CM) from LILRB4-WT and -KO cells was used to analyze the effects of LILRB4 on osteoclasts and osteoblasts. Xenograft, syngeneic and patient derived xenograft models were constructed, and micro-CT, H&E staining were used to observe the bone lesion. RNA-seq, cytokine array, qPCR, the activity of luciferase, Co-IP and western blotting were used to clarify the mechanism by which LILRB4 mediated bone damage in multiple myeloma. RESULTS: We comprehensively analyzed the expression of LILRB4 in various tumor tissue arrays, and found that LILRB4 was highly expressed in multiple myeloma samples. The patient's imaging data showed that the higher the expression level of LILRB4, the more serious the bone lesion in patients with multiple myeloma. The conditioned medium from LILRB4-WT not -KO cells could significantly promote the differentiation and maturation of osteoclasts. Xenograft, syngeneic and patient derived xenograft models furtherly confirmed that LILRB4 could mediate bone lesion of multiple myeloma. Next, cytokine array was performed to identify the differentially expressed cytokines, and RELT was identified and regulated by LILRB4. The overexpression or exogenous RELT could regenerate the bone damage in LILRB4-KO cells in vitro and in vivo. The deletion of LILRB4, anti-LILRB4 alone or in combination with bortezomib could significantly delay the progression of bone lesion of multiple myeloma. CONCLUSIONS: Our findings indicated that LILRB4 promoted the bone lesion by promoting the differentiation and mature of osteoclasts through secreting RELT, and blocking LILRB4 singling pathway could inhibit the bone lesion.


Sujet(s)
Myélome multiple , Récepteurs immunologiques , Transduction du signal , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Myélome multiple/génétique , Humains , Souris , Animaux , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Facteur de transcription NF-kappa B/métabolisme , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Lignée cellulaire tumorale , Ostéoclastes/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
13.
J Am Chem Soc ; 146(25): 17414-17427, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38865166

RÉSUMÉ

The high affinity interaction between P-selectin glycoprotein ligand-1 (PSGL-1) and P-selectin is mediated by a multimotif glycosulfopeptide (GSP) recognition domain consisting of clustered tyrosine sulfates and a Core 2 O-glycan terminated with sialyl LewisX (C2-O-sLeX). These distinct GSP motifs are much more common than previously appreciated within a wide variety of functionally important domains involved in protein-protein interactions. However, despite the potential of GSPs to serve as tools for fundamental studies and prospects for drug discovery, their utility has been limited by the absence of chemical schemes for synthesis on scale. Herein, we report the total synthesis of GSnP-6, an analogue of the N-terminal domain of PSGL-1, and potent inhibitor of P-selectin. An efficient, scalable, hydrogenolysis-free synthesis of C2-O-sLeX-Thr-COOH was identified by both convergent and orthogonal one-pot assembly, which afforded this crucial building block, ready for direct use in solid phase peptide synthesis (SPPS). C2-O-sLeX-Thr-COOH was synthesized in 10 steps with an overall yield of 23% from the 4-O,5-N oxazolidinone thiosialoside donor. This synthesis represents an 80-fold improvement in reaction yield as compared to prior reports, achieving the first gram scale synthesis of SPPS ready C2-O-sLeX-Thr-COOH and enabling the scalable synthesis of GSnP-6 for preclinical evaluation. Significantly, we established that GSnP-6 displays dose-dependent inhibition of venous thrombosis in vivo and inhibits vaso-occlusive events in a human sickle cell disease equivalent microvasculature-on-a-chip system. The insights gained in formulating this design strategy can be broadly applied to the synthesis of a wide variety of biologically important oligosaccharides and O-glycan bearing glycopeptides.


Sujet(s)
Glycopeptides , Glycoprotéines membranaires , Sélectine P , Glycopeptides/synthèse chimique , Glycopeptides/composition chimique , Glycopeptides/pharmacologie , Sélectine P/antagonistes et inhibiteurs , Sélectine P/métabolisme , Glycoprotéines membranaires/antagonistes et inhibiteurs , Glycoprotéines membranaires/métabolisme , Humains , Animaux , Souris
14.
Cell Commun Signal ; 22(1): 323, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867259

RÉSUMÉ

BACKGROUND: Mesenchymal stem cells (MSCs) are widely used in the development of therapeutic tools in regenerative medicine. However, their quality decreases during in vitro expansion because of heterogeneity and acquired cellular senescence. We investigated the potential role of podoplanin (PDPN) in minimizing cellular senescence and maintaining the stemness of tonsil-derived MSCs (TMSCs). METHODS: TMSCs were isolated from human tonsil tissues using an enzymatic method, expanded, and divided into two groups: early-passaged TMSCs, which were cultured for 3-7 passages, and late-passaged TMSCs, which were passaged more than 15 times. The TMSCs were evaluated for cellular senescence and MSC characteristics, and PDPN-positive and -negative cells were identified by fluorescence-activated cell sorting. In addition, MSC features were assessed in siRNA-mediated PDPN-depleted TMSCs. RESULTS: TMSCs, when passaged more than 15 times and becoming senescent, exhibited reduced proliferative rates, telomere length, pluripotency marker (NANOG, OCT4, and SOX2) expression, and tri-lineage differentiation potential (adipogenesis, chondrogenesis, or osteogenesis) compared to cells passaged less than five times. Furthermore, PDPN protein levels significantly decreased in a passage-dependent manner. PDPN-positive cells maintained their stemness characteristics, such as MSC-specific surface antigen (CD14, CD34, CD45, CD73, CD90, and CD105) and pluripotency marker expression, and exhibited higher tri-lineage differentiation potential than PDPN-negative cells. SiRNA-mediated silencing of PDPN led to decreased cell-cycle progression, proliferation, and migration, indicating the significance of PDPN as a preliminary senescence-related factor. These reductions directly contributed to the induction of cellular senescence via p16Ink4a/Rb pathway activation. CONCLUSION: PDPN may serve as a novel biomarker to mitigate cellular senescence in the clinical application of MSCs.


Sujet(s)
Vieillissement de la cellule , Inhibiteur p16 de kinase cycline-dépendante , Glycoprotéines membranaires , Cellules souches mésenchymateuses , Tonsille palatine , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Humains , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/génétique , Tonsille palatine/cytologie , Tonsille palatine/métabolisme , Différenciation cellulaire , Prolifération cellulaire , Transduction du signal , Cellules cultivées
15.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 66-72, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836680

RÉSUMÉ

This study aimed to investigate the mechanism of the effect of TREM2 on cognitive function in autistic mice. TREM2 overexpression and knockdown viruses were given to autism spectrum disorder (ASD) mice and BV2 microglia cell line. To assess cognitive performance, all groups of mice took part in the open field, new object recognition, Morris water maze, and three-box social experiments. Double immunofluorescence labeling demonstrated co-localization of LC3II and NeuN. Proteins from the PI3K/Akt/mTOR pathway were identified. In vivo, behavior studies revealed that TREM2 could successfully improve ASD mice's social interaction and cognitive performance. Besides, we discovered that TREM2 could increase autophagy in ASD mice. In vitro, overexpressing TREM2 reduced the expression of PI3K/AKT/mTOR pathway proteins, whereas knocking down TREM2 increased the expression of PI3K/AKT/mTOR pathway proteins. In conclusion, TREM2 could inhibit PI3K/Akt/mTOR signaling pathway, enhance autophagy, and improve the social communication ability and cognitive function of ASD mice.


Sujet(s)
Autophagie , Cognition , Glycoprotéines membranaires , Microglie , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Récepteurs immunologiques , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Cognition/physiologie , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Phosphatidylinositol 3-kinases/métabolisme , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Sérine-thréonine kinases TOR/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Souris , Microglie/métabolisme , Mâle , Trouble autistique/métabolisme , Trouble autistique/génétique , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/génétique , Trouble du spectre autistique/psychologie , Modèles animaux de maladie humaine , Comportement animal , Lignée cellulaire , Souris de lignée C57BL , Comportement social
16.
Sci Transl Med ; 16(751): eadi3259, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38865485

RÉSUMÉ

Robust structural remodeling and synaptic plasticity occurs within spinal autonomic circuitry after severe high-level spinal cord injury (SCI). As a result, normally innocuous visceral or somatic stimuli elicit uncontrolled activation of spinal sympathetic reflexes that contribute to systemic disease and organ-specific pathology. How hyperexcitable sympathetic circuitry forms is unknown, but local cues from neighboring glia likely help mold these maladaptive neuronal networks. Here, we used a mouse model of SCI to show that microglia surrounded active glutamatergic interneurons and subsequently coordinated multi-segmental excitatory synaptogenesis and expansion of sympathetic networks that control immune, neuroendocrine, and cardiovascular functions. Depleting microglia during critical periods of circuit remodeling after SCI prevented maladaptive synaptic and structural plasticity in autonomic networks, decreased the frequency and severity of autonomic dysreflexia, and prevented SCI-induced immunosuppression. Forced turnover of microglia in microglia-depleted mice restored structural and functional indices of pathological dysautonomia, providing further evidence that microglia are key effectors of autonomic plasticity. Additional data show that microglia-dependent autonomic plasticity required expression of triggering receptor expressed on myeloid cells 2 (Trem2) and α2δ-1-dependent synaptogenesis. These data suggest that microglia are primary effectors of autonomic neuroplasticity and dysautonomia after SCI in mice. Manipulating microglia may be a strategy to limit autonomic complications after SCI or other forms of neurologic disease.


Sujet(s)
Microglie , Plasticité neuronale , Traumatismes de la moelle épinière , Animaux , Microglie/anatomopathologie , Microglie/métabolisme , Traumatismes de la moelle épinière/physiopathologie , Traumatismes de la moelle épinière/anatomopathologie , Souris , Récepteurs immunologiques/métabolisme , Glycoprotéines membranaires/métabolisme , Système nerveux autonome/physiopathologie , Souris de lignée C57BL , Synapses/métabolisme , Interneurones/métabolisme
17.
Cancer Cell ; 42(6): 934-936, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38861929

RÉSUMÉ

In this issue of Cancer Cell, Zhong et al. explore the dual role of TREM2 in glioblastoma-associated myeloid cells, demonstrating its function in promoting inflammation at the tumor-neural interface and suppression within the tumor core, influenced by the local microenvironment. These findings open up promising prospects for advancements in neuro-oncological immunotherapy.


Sujet(s)
Glioblastome , Glycoprotéines membranaires , Cellules myéloïdes , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Cellules myéloïdes/immunologie , Cellules myéloïdes/anatomopathologie , Cellules myéloïdes/métabolisme , Glycoprotéines membranaires/métabolisme , Glioblastome/anatomopathologie , Glioblastome/immunologie , Glioblastome/métabolisme , Récepteurs immunologiques/métabolisme , Animaux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/métabolisme , Neurones/métabolisme , Neurones/anatomopathologie
18.
Proc Natl Acad Sci U S A ; 121(24): e2401686121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38838019

RÉSUMÉ

S-layers are crystalline arrays found on bacterial and archaeal cells. Lactobacillus is a diverse family of bacteria known especially for potential gut health benefits. This study focuses on the S-layer proteins from Lactobacillus acidophilus and Lactobacillus amylovorus common in the mammalian gut. Atomic resolution structures of Lactobacillus S-layer proteins SlpA and SlpX exhibit domain swapping, and the obtained assembly model of the main S-layer protein SlpA aligns well with prior electron microscopy and mutagenesis data. The S-layer's pore size suggests a protective role, with charged areas aiding adhesion. A highly similar domain organization and interaction network are observed across the Lactobacillus genus. Interaction studies revealed conserved binding areas specific for attachment to teichoic acids. The structure of the SlpA S-layer and the suggested incorporation of SlpX as well as its interaction with teichoic acids lay the foundation for deciphering its role in immune responses and for developing effective treatments for a variety of infectious and bacteria-mediated inflammation processes, opening opportunities for targeted engineering of the S-layer or lactobacilli bacteria in general.


Sujet(s)
Glycoprotéines membranaires , Acides teichoïques , Acides teichoïques/métabolisme , Acides teichoïques/composition chimique , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/composition chimique , Lactobacillus/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique , Protéines bactériennes/génétique , Modèles moléculaires , Lactobacillus acidophilus/métabolisme , Lactobacillus acidophilus/génétique
19.
Reproduction ; 168(2)2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38855990

RÉSUMÉ

In brief: Mammalian spermatozoa actively generate reactive oxygen species (ROS) during capacitation, a maturational process necessary for fertilization in vivo. This study shows that hypotaurine, a precursor of taurine present in the oviduct, is incorporated and concentrated in hamster sperm cells via the taurine transporter, TauT, for cytoprotection against self-produced ROS. Abstract: To achieve fertilization competence, mammalian spermatozoa undergo capacitation, during which they actively generate reactive oxygen species (ROS). Therefore, mammalian spermatozoa must protect themselves from these self-generated ROS. The mammalian oviductal fluid is rich in hypotaurine, a taurine precursor, which reportedly protects mammalian spermatozoa, including those of hamsters, from ROS; however, its precise mechanism remains unknown. This study aimed to elucidate the mechanism underlying hypotaurine-mediated protection of spermatozoa from ROS using hamsters, particularly focusing on the taurine/hypotaurine transporter TauT. The effect of hypotaurine on sperm motility and ROS levels was tested using sperm motility analysis and the CellROX dye and luminol assays. RNA sequencing analysis was performed to verify TauT expression. We found that hypotaurine was necessary for maintaining sperm motility and hyperactivated motility. Hypotaurine did not scavenge extracellular ROS but lowered intracellular ROS levels and was incorporated and concentrated in hamster spermatozoa. TauT was detected at both mRNA and protein levels. ß-Alanine blocked hypotaurine transport, increased intracellular ROS levels, and inhibited hyperactivation. Elimination of Na+ or Cl- ions inhibited hypotaurine transport and increased intracellular ROS levels. Thus, these results indicated that hamster spermatozoa incorporated and concentrated hypotaurine in sperm cells via TauT to protect themselves from self-generated ROS.


Sujet(s)
Espèces réactives de l'oxygène , Capacitation des spermatozoïdes , Mobilité des spermatozoïdes , Spermatozoïdes , Taurine , Animaux , Mâle , Taurine/analogues et dérivés , Taurine/pharmacologie , Spermatozoïdes/métabolisme , Spermatozoïdes/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Cricetinae , Mobilité des spermatozoïdes/effets des médicaments et des substances chimiques , Capacitation des spermatozoïdes/effets des médicaments et des substances chimiques , Protéines de transport membranaire/métabolisme , Protéines de transport membranaire/génétique , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Mesocricetus
20.
BMC Oral Health ; 24(1): 729, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918827

RÉSUMÉ

BACKGROUND: Despite the better prognosis associated with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC), some patients experience relapse and succumb to the disease; thus, there is a need for biomarkers identifying these patients for intensified treatment. Leucine-rich repeats and immunoglobulin-like domain (LRIG) protein 1 is a negative regulator of receptor tyrosine kinase signaling and a positive prognostic factor in OPSCC. Studies indicate that LRIG1 interacts with the LIM domain 7 protein (LMO7), a stabilizer of adherence junctions. Its role in OPSCC has not been studied before. METHODS: A total of 145 patients diagnosed with OPSCC were enrolled. Immunohistochemical LMO7 expression and staining intensity were evaluated in the tumors and correlated with known clinical and pathological prognostic factors, such as HPV status and LRIG1, CD44, Ki67, and p53 expression. RESULTS: Our results show that high LMO7 expression is associated with significantly longer overall survival (OS) (p = 0.044). LMO7 was a positive prognostic factor for OS in univariate analysis (HR 0.515, 95% CI: 0.267-0.994, p = 0.048) but not in multivariate analysis. The LMO7 expression correlated with LRIG1 expression (p = 0.048), consistent with previous findings. Interestingly, strong LRIG1 staining intensity was an independent negative prognostic factor in the HPV-driven group of tumors (HR 2.847, 95% Cl: 1.036-7.825, p = 0.043). CONCLUSIONS: We show for the first time that high LMO7 expression is a positive prognostic factor in OPSCC, and we propose that LMO7 should be further explored as a biomarker. In contrast to previous reports, LRIG1 expression was shown to be an independent negative prognostic factor in HPV-driven OPSCC.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome épidermoïde , Protéines à domaine LIM , Tumeurs de l'oropharynx , Humains , Tumeurs de l'oropharynx/virologie , Tumeurs de l'oropharynx/métabolisme , Tumeurs de l'oropharynx/anatomopathologie , Tumeurs de l'oropharynx/mortalité , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Protéines à domaine LIM/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/analyse , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/virologie , Sujet âgé , Facteurs de transcription/métabolisme , Glycoprotéines membranaires/métabolisme , Adulte , Antigène KI-67/métabolisme , Antigènes CD44/métabolisme , Antigènes CD44/analyse , Protéine p53 suppresseur de tumeur/métabolisme , Infections à papillomavirus/complications , Immunohistochimie , Sujet âgé de 80 ans ou plus , Taux de survie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...