Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.780
Filter
1.
Cell Death Dis ; 15(5): 324, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724533

ABSTRACT

Severe aplastic anemia (SAA) is a rare, fatal disease characterized by severe cytopenias and loss of hematopoietic stem cells (HSCs). Immune-mediated destruction and inflammation are known drivers of SAA, however, the underlying mechanisms driving persistent inflammation are unknown. Current treatments for SAA rely on immunosuppressive therapies or HSC transplantation, however, these treatments are not always effective. Using an established mouse model of SAA, we observed a significant increase in apoptotic cells within the bone marrow (BM) and impaired efferocytosis in SAA mice, relative to radiation controls. Single-cell transcriptomic analysis revealed heterogeneity among BM monocytes and unique populations emerged during SAA characterized by increased inflammatory signatures and significantly increased expression of Sirpa and Cd47. CD47, a "don't eat me" signal, was increased on both live and apoptotic BM cells, concurrent with markedly increased expression of signal regulatory protein alpha (SIRPα) on monocytes. Functionally, SIRPα blockade improved cell clearance and reduced accumulation of CD47-positive apoptotic cells. Lipidomic analysis revealed a reduction in the precursors of specialized pro-resolving lipid mediators (SPMs) and increased prostaglandins in the BM during SAA, indicative of impaired inflammation resolution. Specifically, 18-HEPE, a precursor of E-series resolvins, was significantly reduced in SAA-induced mice relative to radiation controls. Treatment of SAA mice with Resolvin E1 (RvE1) improved efferocytic function, BM cellularity, platelet output, and survival. Our data suggest that impaired efferocytosis and inflammation resolution contributes to SAA progression and demonstrate that SPMs, such as RvE1, offer new and/or complementary treatments for SAA that do not rely on immune suppression.


Subject(s)
Anemia, Aplastic , CD47 Antigen , Eicosapentaenoic Acid , Animals , Anemia, Aplastic/pathology , Mice , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , CD47 Antigen/metabolism , CD47 Antigen/genetics , Apoptosis/drug effects , Phagocytosis/drug effects , Disease Models, Animal , Mice, Inbred C57BL , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Monocytes/metabolism , Monocytes/drug effects , Inflammation/pathology , Male , Efferocytosis
2.
Front Immunol ; 15: 1360219, 2024.
Article in English | MEDLINE | ID: mdl-38745667

ABSTRACT

Background: Regulatory B cells (Bregs) play a pivotal role in suppressing immune responses, yet there is still a lack of cell surface markers that can rigorously identify them. In mouse models for multiple sclerosis (MS), TIM-1 or TIGIT expression on B cells is required for maintaining self-tolerance and regulating autoimmunity to the central nervous system. Here we investigated the activities of human memory B cells that differentially express TIM-1 and TIGIT to determine their potential regulatory function in healthy donors and patients with relapsing-remitting (RR) MS. Methods: FACS-sorted TIM-1+/-TIGIT+/- memory B (memB) cells co-cultured with allogenic CD4+ T cells were analyzed for proliferation and induction of inflammatory markers using flow cytometry and cytokine quantification, to determine Th1/Th17 cell differentiation. Transcriptional differences were assessed by SMARTSeq2 RNA sequencing analysis. Results: TIM-1-TIGIT- double negative (DN) memB cells strongly induce T cell proliferation and pro-inflammatory cytokine expression. The TIM-1+ memB cells enabled low levels of CD4+ T cell activation and gave rise to T cells that co-express IL-10 with IFNγ and IL-17A or FoxP3. T cells cultured with the TIM-1+TIGIT+ double positive (DP) memB cells exhibited reduced proliferation and IFNγ, IL-17A, TNFα, and GM-CSF expression, and exhibited strong regulation in Breg suppression assays. The functional activity suggests the DP memB cells are a bonafide Breg population. However, MS DP memB cells were less inhibitory than HC DP memB cells. A retrospective longitudinal study of anti-CD20 treated patients found that post-treatment DP memB cell frequency and absolute number were associated with response to therapy. Transcriptomic analyses indicated that the dysfunctional MS-derived DP memB/Breg population exhibited increased expression of genes associated with T cell activation and survival (CD80, ZNF10, PIK3CA), and had distinct gene expression compared to the TIGIT+ or TIM-1+ memB cells. Conclusion: These findings demonstrate that TIM-1/TIGIT expressing memory B cell subsets have distinct functionalities. Co-expression of TIM-1 and TIGIT defines a regulatory memory B cell subset that is functionally impaired in MS.


Subject(s)
B-Lymphocytes, Regulatory , Hepatitis A Virus Cellular Receptor 1 , Receptors, Immunologic , Humans , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Hepatitis A Virus Cellular Receptor 1/metabolism , Hepatitis A Virus Cellular Receptor 1/genetics , Female , Male , Adult , Memory B Cells/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/metabolism , Cytokines/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Lymphocyte Activation/immunology , Middle Aged , Cells, Cultured , Cell Differentiation/immunology , Immunologic Memory
3.
Invest Ophthalmol Vis Sci ; 65(5): 8, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38700874

ABSTRACT

Purpose: In the present study, we aim to elucidate the underlying molecular mechanism of endoplasmic reticulum (ER) stress induced delayed corneal epithelial wound healing and nerve regeneration. Methods: Human limbal epithelial cells (HLECs) were treated with thapsigargin to induce excessive ER stress and then RNA sequencing was performed. Immunofluorescence, qPCR, Western blot, and ELISA were used to detect the expression changes of SLIT3 and its receptors ROBO1-4. The role of recombinant SLIT3 protein in corneal epithelial proliferation and migration were assessed by CCK8 and cell scratch assay, respectively. Thapsigargin, exogenous SLIT3 protein, SLIT3-specific siRNA, and ROBO4-specific siRNA was injected subconjunctivally to evaluate the effects of different intervention on corneal epithelial and nerve regeneration. In addition, Ki67 staining was performed to evaluate the proliferation ability of epithelial cells. Results: Thapsigargin suppressed normal corneal epithelial and nerve regeneration significantly. RNA sequencing genes related to development and regeneration revealed that thapsigargin induced ER stress significantly upregulated the expression of SLIT3 and ROBO4 in corneal epithelial cells. Exogenous SLIT3 inhibited normal corneal epithelial injury repair and nerve regeneration, and significantly suppressed the proliferation and migration ability of cultured mouse corneal epithelial cells. SLIT3 siRNA inhibited ROBO4 expression and promoted epithelial wound healing under thapsigargin treatment. ROBO4 siRNA significantly attenuated the delayed corneal epithelial injury repair and nerve regeneration induced by SLIT3 treatment or thapsigargin treatment. Conclusions: ER stress inhibits corneal epithelial injury repair and nerve regeneration may be related with the upregulation of SLIT3-ROBO4 pathway.


Subject(s)
Cell Proliferation , Endoplasmic Reticulum Stress , Epithelium, Corneal , Nerve Regeneration , Receptors, Immunologic , Roundabout Proteins , Signal Transduction , Wound Healing , Animals , Humans , Mice , Blotting, Western , Cell Movement/physiology , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Enzyme-Linked Immunosorbent Assay , Epithelium, Corneal/metabolism , Limbus Corneae/cytology , Nerve Regeneration/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction/physiology , Wound Healing/physiology
4.
Arch Dermatol Res ; 316(5): 162, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734848

ABSTRACT

Psoriasis is a chronic, immune-mediated, hyperproliferative skin disease. Etiopathogenesis of psoriasis is not well understood. Plexin B2 was found to have effects on CD100-mediated T-cell morphology and expressed in the immune system. It may play a role in the pathogenesis of psoriasis. To assess the tissue level of plexin-B2 and plexin B2 related gene polymorphism which is signal regulatory protein gamma (SIRPγ-rs71212732) in psoriatic patients before and after NB-UVB, acitretin therapy alone or in combination and to detect correlation between level of tissue plexin B2 and disease severity and improvement. This single blinded randomized controlled trial was carried on 50 psoriatic patients and 50 healthy controls. Psoriasis Area and Severity Index score (PASI) was used to evaluate the disease severity. Tissue plexin-b2 level was measured using ELISA and SIRPγ-rs71212732 (T\C) was assessed using TaqMan™ assays and real-time PCR. A significant lower tissue plexin-B2 level was observed in control group (2.9 ± 0.6 pg/g) than cases (25.8 ± 2.8, pg/g) (p < 0.001). Also, a significantly higher tissue plexin-B2 level was observed in sever psoriasis (32.7 ± 3.8 pg/ml) in than moderate psoriasis (13.6 ± 2.1 pg/ml, p = 0.001). Tissue plexin B2 was positively correlated with diseases severity. Significantly higher (TC& TT) genotypes and mutant (C) allele among patients compared to the controls, p < 0.001 for all. Tissue plexin-b2 level was high in psoriasis vulgaris with positive correlation with disease severity and decreased after treatment. This may indicate a role of plexin-b2 in psoriasis vulgaris pathogenesis.


Subject(s)
Acitretin , Nerve Tissue Proteins , Psoriasis , Severity of Illness Index , Humans , Psoriasis/genetics , Psoriasis/drug therapy , Psoriasis/diagnosis , Male , Female , Adult , Nerve Tissue Proteins/genetics , Middle Aged , Acitretin/therapeutic use , Acitretin/administration & dosage , Ultraviolet Therapy/methods , Single-Blind Method , Polymorphism, Single Nucleotide , Young Adult , Skin/pathology , Skin/metabolism , Skin/drug effects , Receptors, Immunologic/genetics , Treatment Outcome , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Keratolytic Agents/therapeutic use , Keratolytic Agents/administration & dosage , Combined Modality Therapy
5.
Oncoimmunology ; 13(1): 2346359, 2024.
Article in English | MEDLINE | ID: mdl-38737794

ABSTRACT

Immune exhaustion is a hallmark of ovarian cancer. Using multiparametric flow cytometry, the study aimed to analyze protein expression of novel immunological targets on CD3+ T cells isolated from the peripheral blood (n = 20), malignant ascites (n = 16), and tumor tissue (n = 6) of patients with ovarian cancer (OVCA). The study revealed an increased proportion of effector memory CD8+ T cells in OVCA tissue and malignant ascites. An OVCA-characteristic PD-1high CD8+ T cell population was detected, which differed from PD-1lowCD8+ T cells by increased co-expression of TIGIT, CD39, and HLA-DR. In addition, these OVCA-characteristic CD8+ T cells showed reduced expression of the transcription factor TCF-1, which may also indicate reduced effector function and memory formation. On the contrary, the transcription factor TOX, which significantly regulates terminal T cell-exhaustion, was found more frequently in these cells. Further protein and gene analysis showed that CD39 and CD73 were also expressed on OVCA tumor cells isolated from solid tumors (n = 14) and malignant ascites (n = 9). In the latter compartment, CD39 and CD73 were also associated with the expression of the "don't eat me" molecule CD24 on tumor cells. Additionally, ascites-derived CD24+EpCAM+ tumor cells showed a higher frequency of CD39+ or CD73+ cells. Furthermore, CD39 expression was associated with unfavorable clinical parameters. Expression of CD39 on T cells was upregulated through CD3/CD28 stimulation and its blockade by a newly developed nanobody construct resulted in increased proliferation (eFluor), activation (CD25 and CD134), and production of cytotoxic cytokines (IFN-γ, TNF-α, and granzyme-B) of CD8+ T cells.


Subject(s)
Apyrase , CD8-Positive T-Lymphocytes , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Apyrase/metabolism , Apyrase/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Middle Aged , Ascites/immunology , Ascites/pathology , Ascites/metabolism , Antigens, CD/metabolism , Antigens, CD/genetics , Aged , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/antagonists & inhibitors , T Cell Transcription Factor 1/metabolism , T Cell Transcription Factor 1/genetics , HLA-DR Antigens/metabolism , Adult , T-Cell Exhaustion , High Mobility Group Proteins
6.
Int J Mol Sci ; 25(7)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38612763

ABSTRACT

Idiopathic intellectual disability (IID) encompasses the cases of intellectual disability (ID) without a known cause and represents approximately 50% of all cases. Neural progenitor cells (NPCs) from the olfactory neuroepithelium (NEO) contain the same information as the cells found in the brain, but they are more accessible. Some miRNAs have been identified and associated with ID of known etiology. However, in idiopathic ID, the effect of miRNAs is poorly understood. The aim of this study was to determine the miRNAs regulating the expression of mRNAs that may be involved in development of IID. Expression profiles were obtained using NPC-NEO cells from IID patients and healthy controls by microarray. A total of 796 miRNAs and 28,869 mRNAs were analyzed. Several miRNAs were overexpressed in the IID patients compared to controls. miR-25 had the greatest expression. In silico analysis showed that ROBO2 was the target for miR-25, with the highest specificity and being the most down-regulated. In vitro assay showed an increase of miR-25 expression induced a decrease in ROBO2 expression. In neurodevelopment, ROBO2 plays a crucial role in episodic learning and memory, so its down-regulation, caused by miR-25, could have a fundamental role in the intellectual disability that, until now, has been considered idiopathic.


Subject(s)
Intellectual Disability , MicroRNAs , Humans , Intellectual Disability/genetics , MicroRNAs/genetics , Brain , Down-Regulation/genetics , Learning , RNA, Messenger , Roundabout Proteins , Receptors, Immunologic/genetics
7.
Mol Immunol ; 170: 35-45, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38613944

ABSTRACT

Microglia play a pivotal role in the pathology of Alzheimer's Disease (AD), with the Triggering Receptor Expressed on Myeloid cells 2 (TREM2) central to their neuroprotective functions. The R47H variant of TREM2 has emerged as a significant genetic risk factor for AD, leading to a loss-of-function phenotype in mouse AD models. This study elucidates the roles of TREM2 in human microglia-like HMC3 cells and the regulation of these functions by SH2-containing inositol-5'-phosphatase 1 (SHIP1). Using stable cell lines expressing wild-type TREM2, the R47H variant, and TREM2-deficient lines, we found that functional TREM2 is essential for the phagocytosis of Aß, lysosomal capacity, and mitochondrial activity. Notably, the R47H variant displayed increased phagocytic activity towards apoptotic neurons. Introducing SHIP1, known to modulate TREM2 signaling in other cells, revealed its role as a negative regulator of these TREM2-mediated functions. Moreover, pharmacological inhibition of both SHIP1 and its isoform SHIP2 amplified Aß phagocytosis and lysosomal capacity, independently of TREM2 or SHIP1 expression, suggesting a potential regulatory role for SHIP2 in these functions. The absence of TREM2, combined with the presence of both SHIP isoforms, suppressed mitochondrial activity. However, pan-SHIP1/2 inhibition enhanced mitochondrial function in these cells. In summary, our findings offer a deeper understanding of the relationship between TREM2 variants and SHIP1 in microglial functions, and emphasize the therapeutic potential of targeting the TREM2 and SHIP1 pathways in microglia for neurodegenerative diseases.


Subject(s)
Membrane Glycoproteins , Microglia , Phagocytosis , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Receptors, Immunologic , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Microglia/metabolism , Humans , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Phagocytosis/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Cell Line , Mitochondria/metabolism , Animals , Lysosomes/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis/genetics , Signal Transduction , Mice
8.
ACS Nano ; 18(18): 11753-11768, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38649866

ABSTRACT

The association between dysfunctional microglia and amyloid-ß (Aß) is a fundamental pathological event and increases the speed of Alzheimer's disease (AD). Additionally, the pathogenesis of AD is intricate and a single drug may not be enough to achieve a satisfactory therapeutic outcome. Herein, we reported a facile and effective gene therapy strategy for the modulation of microglia function and intervention of Aß anabolism by ROS-responsive biomimetic exosome-liposome hybrid nanovesicles (designated as TSEL). The biomimetic nanovesicles codelivery ß-site amyloid precursor protein cleaving enzyme-1 (BACE1) siRNA (siBACE1) and TREM2 plasmid (pTREM2) gene drug efficiently penetrate the blood-brain barrier and enhance the drug accumulation at AD lesions with the help of exosomes homing ability and angiopep-2 peptides. Specifically, an upregulation of TREM2 expression can reprogram microglia from a pro-inflammatory M1 phenotype to an anti-inflammatory M2 phenotype while also restoring its capacity to phagocytose Aß and its nerve repair function. In addition, siRNA reduces the production of Aß plaques at the source by knocking out the BACE1 gene, which is expected to further enhance the therapeutic effect of AD. The in vivo study suggests that TSEL through the synergistic effect of two gene drugs can ameliorate APP/PS1 mice cognitive impairment by regulating the activated microglial phenotype, reducing the accumulation of Aß, and preventing the retriggering of neuroinflammation. This strategy employs biomimetic nanovesicles for the delivery of dual nucleic acids, achieving synergistic gene therapy for AD, thus offering more options for the treatment of AD.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Biomimetic Materials , Genetic Therapy , Alzheimer Disease/therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Animals , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Mice , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Gene Transfer Techniques , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Humans , Liposomes/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Biomimetics , Exosomes/metabolism , Exosomes/chemistry , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics
9.
Front Immunol ; 15: 1383110, 2024.
Article in English | MEDLINE | ID: mdl-38650930

ABSTRACT

Exhausted CD8 T cells (TEX) are associated with worse outcome in cancer yet better outcome in autoimmunity. Building on our past findings of increased TIGIT+KLRG1+ TEX with teplizumab therapy in type 1 diabetes (T1D), in the absence of treatment we found that the frequency of TIGIT+KLRG1+ TEX is stable within an individual but differs across individuals in both T1D and healthy control (HC) cohorts. This TIGIT+KLRG1+ CD8 TEX population shares an exhaustion-associated EOMES gene signature in HC, T1D, rheumatoid arthritis (RA), and cancer subjects, expresses multiple inhibitory receptors, and is hyporesponsive in vitro, together suggesting co-expression of TIGIT and KLRG1 may broadly define human peripheral exhausted cells. In HC and RA subjects, lower levels of EOMES transcriptional modules and frequency of TIGIT+KLRG1+ TEX were associated with RA HLA risk alleles (DR0401, 0404, 0405, 0408, 1001) even when considering disease status and cytomegalovirus (CMV) seropositivity. Moreover, the frequency of TIGIT+KLRG1+ TEX was significantly increased in RA HLA risk but not non-risk subjects treated with abatacept (CTLA4Ig). The DR4 association and selective modulation with abatacept suggests that therapeutic modulation of TEX may be more effective in DR4 subjects and TEX may be indirectly influenced by cellular interactions that are blocked by abatacept.


Subject(s)
Abatacept , Alleles , Arthritis, Rheumatoid , CD8-Positive T-Lymphocytes , Receptors, Immunologic , Humans , Abatacept/therapeutic use , Abatacept/pharmacology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/genetics , Male , Female , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , Adult , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , HLA Antigens/genetics , HLA Antigens/immunology , Middle Aged , Antirheumatic Agents/therapeutic use , Genetic Predisposition to Disease , T-Cell Exhaustion
10.
Clin Transl Med ; 14(4): e1665, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38649789

ABSTRACT

BACKGROUND: White matter injury (WMI) is an important pathological process after traumatic brain injury (TBI). The correlation between white matter functions and the myeloid cells expressing triggering receptor-2 (TREM2) has been convincingly demonstrated. Moreover, a recent study revealed that microglial sterol metabolism is crucial for early remyelination after demyelinating diseases. However, the potential roles of TREM2 expression and microglial sterol metabolism in WMI after TBI have not yet been explored. METHODS: Controlled cortical injury was induced in both wild-type (WT) and TREM2 depletion (TREM2 KO) mice to simulate clinical TBI. COG1410 was used to upregulate TREM2, while PLX5622 and GSK2033 were used to deplete microglia and inhibit the liver X receptor (LXR), respectively. Immunofluorescence, Luxol fast blue staining, magnetic resonance imaging, transmission electron microscopy, and oil red O staining were employed to assess WMI after TBI. Neurological behaviour tests and electrophysiological recordings were utilized to evaluate cognitive functions following TBI. Microglial cell sorting and transcriptomic sequencing were utilized to identify alterations in microglial sterol metabolism-related genes, while western blot was conducted to validate the findings. RESULTS: TREM2 expressed highest at 3 days post-TBI and was predominantly localized to microglial cells within the white matter. Depletion of TREM2 worsened aberrant neurological behaviours, and this phenomenon was mediated by the exacerbation of WMI, reduced renewal of oligodendrocytes, and impaired phagocytosis ability of microglia after TBI. Subsequently, the upregulation of TREM2 alleviated WMI, promoted oligodendrocyte regeneration, and ultimately facilitated the recovery of neurological behaviours after TBI. Finally, the expression of DHCR24 increased in TREM2 KO mice after TBI. Interestingly, TREM2 inhibited DHCR24 and upregulated members of the LXR pathway. Moreover, LXR inhibition could partially reverse the effects of TREM2 upregulation on electrophysiological activities. CONCLUSIONS: We demonstrate that TREM2 has the potential to alleviate WMI following TBI, possibly through the DHCR24/LXR pathway in microglia.


Subject(s)
Brain Injuries, Traumatic , Membrane Glycoproteins , Microglia , Receptors, Immunologic , White Matter , Animals , Male , Mice , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/genetics , Disease Models, Animal , Liver X Receptors/metabolism , Liver X Receptors/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , White Matter/metabolism , White Matter/pathology
11.
Int J Biol Sci ; 20(6): 1992-2007, 2024.
Article in English | MEDLINE | ID: mdl-38617547

ABSTRACT

Objective: Osteoarthritis (OA) is the most prominent chronic arthritic disease, affecting over 3 billion people globally. Synovial macrophages, as immune cells, play an essential role in cartilage damage in OA. Therefore, regulating macrophages is crucial for controlling the pathological changes in OA. Triggering receptor expressed on myeloid cells 2 (TREM2), as expressed on immune cell surfaces, such as macrophages and dendritic cells, has suppressed inflammation and regulated M2 macrophage polarization but demonstrated an unknown role in synovial macrophage polarization in OA. This study aimed to investigate TREM2 expression downregulation in OA mice macrophages. Furthermore, the expression trend of TREM2 was associated with polarization-related molecule expression in macrophages of OA mice. Results: We used TREM2 knockout (TREM2-KO) mice to observe that TREM2 deficiency significantly exacerbated the joint inflammation response in OA mice, thereby accelerating disease progression. Separating macrophages and chondrocytes from TREM2-KO mice and co-cultivating them significantly increased chondrocyte apoptosis and inhibited chondrocyte proliferation. Further, TREM2 deficiency also significantly enhanced phosphatidylinositol 3-kinase(PI3K)/AKT signaling pathway activation, increasing nuclear factor kappa light chain enhancer of activated B cells (NF-κB) signaling and C-X-C Motif Chemokine Ligand 3 (CXCL3) expression. Furthermore, NF-κB signaling pathway inhibition significantly suppressed arthritis inflammation in OA mice, thereby effectively alleviating TREM2 deficiency-related adverse effects on chondrocytes. Notably, knocking down CXCL3 of TREM2-KO mice macrophages significantly inhibits inflammatory response and promotes chondrocyte proliferation. Intravenous recombinant TREM2 protein (soluble TREM2, sTREM2) injection markedly promotes macrophage polarization from M1 to M2 and improves the joint tissue pathology and inflammatory response of OA. Conclusion: Our study reveals that TREM2 promotes macrophage polarization from M1 to M2 during OA by NF-κB/CXCL3 axis regulation, thereby improving the pathological state of OA.


Subject(s)
NF-kappa B , Osteoarthritis , Animals , Mice , Chemokines, CXC , Inflammation , Membrane Glycoproteins/genetics , Osteoarthritis/genetics , Phosphatidylinositol 3-Kinases , Receptors, Immunologic/genetics , Signal Transduction/genetics
12.
FASEB J ; 38(8): e23590, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38656553

ABSTRACT

Studies have suggested that microglial IL-6 modulates inflammatory pain; however, the exact mechanism of action remains unclear. We therefore hypothesized that PKCε and MEG2 competitively bind to STAT3 and contribute to IL-6-mediated microglial hyperalgesia during inflammatory pain. Freund's complete adjuvant (FCA) and lipopolysaccharide (LPS) were used to induce hyperalgesia model mice and microglial inflammation. Mechanical allodynia was evaluated using von Frey tests in vivo. The interaction among PKCε, MEG2, and STAT3 was determined using ELISA and immunoprecipitation assay in vitro. The PKCε, MEG2, t-STAT3, pSTAT3Tyr705, pSTAT3Ser727, IL-6, GLUT3, and TREM2 were assessed by Western blot. IL-6 promoter activity and IL-6 concentration were examined using dual luciferase assays and ELISA. Overexpression of PKCε and MEG2 promoted and attenuated inflammatory pain, accompanied by an increase and decrease in IL-6 expression, respectively. PKCε displayed a stronger binding ability to STAT3 when competing with MEG2. STAT3Ser727 phosphorylation increased STAT3 interaction with both PKCε and MEG2. Moreover, LPS increased PKCε, MEG2, pSTAT3Tyr705, pSTAT3Ser727, IL-6, and GLUT3 levels and decreased TREM2 during microglia inflammation. IL-6 promoter activity was enhanced or inhibited by PKCε or MEG2 in the presence of STAT3 and LPS stimulation, respectively. In microglia, overexpression of PKCε and/or MEG2 resulted in the elevation of tSTAT3, pSTAT3Tyr705, pSTAT3Ser727, IL-6, and TREM2, and the reduction of GLUT3. PKCε is more potent than MEG2 when competitively binding to STAT3, displaying dual modulatory effects of IL-6 production, thus regulating the GLUT3 and TREM2 in microglia during inflammatory pain sensation.


Subject(s)
Hyperalgesia , Inflammation , Interleukin-6 , Microglia , Protein Kinase C-epsilon , STAT3 Transcription Factor , Animals , Male , Mice , Freund's Adjuvant , Hyperalgesia/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Interleukin-6/genetics , Lipopolysaccharides/toxicity , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Microglia/metabolism , Pain/metabolism , Phosphorylation , Protein Binding , Protein Kinase C-epsilon/metabolism , Protein Kinase C-epsilon/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , STAT3 Transcription Factor/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
13.
Sci Rep ; 14(1): 9458, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658633

ABSTRACT

Male sex is a risk factor for colorectal cancer (CRC) with higher illness burden and earlier onset. Thus, we hypothesized that loss of chromosome Y (LOY) in the tumor micro-environment (TME) might be involved in oncogenesis. Previous studies show that LOY in circulating leukocytes of aging men was associated with shorter survival and non-hematological cancer, as well as higher LOY in CD4 + T-lymphocytes in men with prostate cancer vs. controls. However, nothing is known about LOY in leukocytes infiltrating TME and we address this aspect here. We studied frequency and functional effects of LOY in blood, TME and non-tumorous tissue. Regulatory T-lymphocytes (Tregs) in TME had the highest frequency of LOY (22%) in comparison to CD4 + T-lymphocytes and cytotoxic CD8 + T-lymphocytes. LOY score using scRNA-seq was also linked to higher expression of PDCD1, TIGIT and IKZF2 in Tregs. PDCD1 and TIGIT encode immune checkpoint receptors involved in the regulation of Tregs function. Our study sets the direction for further functional research regarding a probable role of LOY in intensifying features related to the suppressive phenotype of Tregs in TME and consequently a possible influence on immunotherapy response in CRC patients.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , T-Lymphocytes, Regulatory , Tumor Microenvironment , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Tumor Microenvironment/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Male , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Aged , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Middle Aged , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Female , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism
14.
Metabolism ; 155: 155911, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38609037

ABSTRACT

BACKGROUND: The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing year by year and has become one of the leading causes of end-stage liver disease worldwide. Triggering Receptor Expressed on Myeloid Cells 2 (Trem2) has been confirmed to play an essential role in the progression of MASLD, but its specific mechanism still needs to be clarified. This study aims to explore the role and mechanism of Trem2 in MASLD. METHODS: Human liver tissues were obtained from patients with MASLD and controls. Myeloid-specific knockout mice (Trem2mKO) and myeloid-specific overexpression mice (Trem2TdT) were fed a high-fat diet, either AMLN or CDAHFD, to establish the MASLD model. Relevant signaling molecules were assessed through lipidomics and RNA-seq analyses after that. RESULTS: Trem2 is upregulated in human MASLD/MASH-associated macrophages and is associated with hepatic steatosis and inflammation progression. Hepatic steatosis and inflammatory responses are exacerbated with the knockout of myeloid Trem2 in MASLD mice, while mice overexpressing Trem2 exhibit the opposite phenomenon. Mechanistically, Trem2mKO can aggravate macrophage pyroptosis through the PI3K/AKT signaling pathway and amplify the resulting inflammatory response. At the same time, Trem2 promotes the inflammation resolution phenotype transformation of macrophages through TGFß1, thereby promoting tissue repair. CONCLUSIONS: Myeloid Trem2 ameliorates the progression of Metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. We believe targeting myeloid Trem2 could represent a potential avenue for treating MASLD.


Subject(s)
Disease Progression , Fatty Liver , Inflammation , Macrophages , Membrane Glycoproteins , Mice, Knockout , Pyroptosis , Receptors, Immunologic , Animals , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice , Humans , Macrophages/metabolism , Inflammation/metabolism , Inflammation/pathology , Pyroptosis/physiology , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/genetics , Male , Mice, Inbred C57BL , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Metabolic Diseases/genetics , Liver/metabolism , Liver/pathology
15.
Cancer Immunol Res ; 12(5): 575-591, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38588410

ABSTRACT

Poliovirus receptor-related 2 (PVRL2, also known as nectin-2 or CD112) is believed to act as an immune checkpoint protein in cancer; however, most insight into its role is inferred from studies on its known receptor, poliovirus receptor (PVR)-related immunoglobulin domain protein (PVRIG, also known as CD112R). Here, we study PVRL2 itself. PVRL2 levels were found to be high in tumor cells and tumor-derived exosomes. Deletion of PVRL2 in multiple syngeneic mouse models of cancer showed a dramatic reduction in tumor growth that was immune dependent. This effect was even greater than that seen with deletion of PD-L1. PVRL2 was shown to function by suppressing CD8+ T and natural killer cells in the tumor microenvironment. The loss of PVRL2 suppressed tumor growth even in the absence of PVRIG. In contrast, PVRIG loss showed no additive effect in the absence of PVRL2. T-cell immunoreceptor with Ig and ITIM domains (TIGIT) blockade combined with PVRL2 deletion resulted in a near complete block in tumor growth. This effect was not recapitulated by the combined deletion of PVRL2 with its paralog, PVR, which is the ligand for TIGIT. These data uncover PVRL2 as a distinct inhibitor of the antitumor immune response with functions beyond that of its known receptor PVRIG. Moreover, the data provide a strong rationale for combinatorial targeting of PVRL2 and TIGIT for cancer immunotherapy.


Subject(s)
Nectins , Receptors, Cell Surface , Receptors, Immunologic , Tumor Microenvironment , Animals , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Nectins/metabolism , Mice , Humans , Tumor Microenvironment/immunology , Cell Line, Tumor , Signal Transduction , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism
16.
J Transl Med ; 22(1): 395, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685028

ABSTRACT

BACKGROUND: Current cancer therapies often fall short in addressing the complexities of malignancies, underscoring the urgent need for innovative treatment strategies. RNA interference technology, which specifically suppresses gene expression, offers a promising new approach in the fight against tumors. Recent studies have identified a novel immunostimulatory small-interfering RNA (siRNA) with a unique sequence (sense strand, 5'-C; antisense strand, 3'-GGG) capable of activating the RIG-I/IRF3 signaling pathway. This activation induces the release of type I and III interferons, leading to an effective antiviral immune response. However, this class of immunostimulatory siRNA has not yet been explored in cancer therapy. METHODS: IsiBCL-2, an innovative immunostimulatory siRNA designed to suppress the levels of B-cell lymphoma 2 (BCL-2), contains a distinctive motif (sense strand, 5'-C; antisense strand, 3'-GGG). Glioblastoma cells were subjected to 100 nM isiBCL-2 treatment in vitro for 48 h. Morphological changes, cell viability (CCK-8 assay), proliferation (colony formation assay), migration/invasion (scratch test and Transwell assay), apoptosis rate, reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) were evaluated. Western blotting and immunofluorescence analyses were performed to assess RIG-I and MHC-I molecule levels, and ELISA was utilized to measure the levels of cytokines (IFN-ß and CXCL10). In vivo heterogeneous tumor models were established, and the anti-tumor effect of isiBCL-2 was confirmed through intratumoral injection. RESULTS: IsiBCL-2 exhibited significant inhibitory effects on glioblastoma cell growth and induced apoptosis. BCL-2 mRNA levels were significantly decreased by 67.52%. IsiBCL-2 treatment resulted in an apoptotic rate of approximately 51.96%, accompanied by a 71.76% reduction in MMP and a 41.87% increase in ROS accumulation. Western blotting and immunofluorescence analyses demonstrated increased levels of RIG-I, MAVS, and MHC-I following isiBCL-2 treatment. ELISA tests indicated a significant increase in IFN-ß and CXCL10 levels. In vivo studies using nude mice confirmed that isiBCL-2 effectively impeded the growth and progression of glioblastoma tumors. CONCLUSIONS: This study introduces an innovative method to induce innate signaling by incorporating an immunostimulatory sequence (sense strand, 5'-C; antisense strand, 3'-GGG) into siRNA, resulting in the formation of RNA dimers through Hoogsteen base-pairing. This activation triggers the RIG-I signaling pathway in tumor cells, causing further damage and inducing a potent immune response. This inventive design and application of immunostimulatory siRNA offer a novel perspective on tumor immunotherapy, holding significant implications for the field.


Subject(s)
Apoptosis , Glioma , RNA, Small Interfering , Humans , Animals , Cell Line, Tumor , Glioma/therapy , Glioma/pathology , Glioma/genetics , RNA, Small Interfering/metabolism , Mice, Nude , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , Cell Proliferation , Cell Movement , Xenograft Model Antitumor Assays , Mice , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Reactive Oxygen Species/metabolism , Neoplasm Invasiveness , Cell Survival
17.
Mol Neurodegener ; 19(1): 37, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38654375

ABSTRACT

BACKGROUND: Microglia play important roles in maintaining brain homeostasis and neurodegeneration. The discovery of genetic variants in genes predominately or exclusively expressed in myeloid cells, such as Apolipoprotein E (APOE) and triggering receptor expressed on myeloid cells 2 (TREM2), as the strongest risk factors for Alzheimer's disease (AD) highlights the importance of microglial biology in the brain. The sequence, structure and function of several microglial proteins are poorly conserved across species, which has hampered the development of strategies aiming to modulate the expression of specific microglial genes. One way to target APOE and TREM2 is to modulate their expression using antisense oligonucleotides (ASOs). METHODS: In this study, we identified, produced, and tested novel, selective and potent ASOs for human APOE and TREM2. We used a combination of in vitro iPSC-microglia models, as well as microglial xenotransplanted mice to provide proof of activity in human microglial in vivo. RESULTS: We proved their efficacy in human iPSC microglia in vitro, as well as their pharmacological activity in vivo in a xenografted microglia model. We demonstrate ASOs targeting human microglia can modify their transcriptional profile and their response to amyloid-ß plaques in vivo in a model of AD. CONCLUSIONS: This study is the first proof-of-concept that human microglial can be modulated using ASOs in a dose-dependent manner to manipulate microglia phenotypes and response to neurodegeneration in vivo.


Subject(s)
Alzheimer Disease , Microglia , Oligonucleotides, Antisense , Microglia/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Humans , Oligonucleotides, Antisense/pharmacology , Animals , Mice , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Induced Pluripotent Stem Cells/metabolism , Gene Expression Regulation/drug effects , Disease Models, Animal
18.
Nat Neurosci ; 27(5): 886-900, 2024 May.
Article in English | MEDLINE | ID: mdl-38539015

ABSTRACT

Microglia are central players in Alzheimer's disease pathology but analyzing microglial states in human brain samples is challenging due to genetic diversity, postmortem delay and admixture of pathologies. To circumvent these issues, here we generated 138,577 single-cell expression profiles of human stem cell-derived microglia xenotransplanted in the brain of the AppNL-G-F model of amyloid pathology and wild-type controls. Xenografted human microglia adopt a disease-associated profile similar to that seen in mouse microglia, but display a more pronounced human leukocyte antigen or HLA state, likely related to antigen presentation in response to amyloid plaques. The human microglial response also involves a pro-inflammatory cytokine/chemokine cytokine response microglia or CRM response to oligomeric Aß oligomers. Genetic deletion of TREM2 or APOE as well as APOE polymorphisms and TREM2R47H expression in the transplanted microglia modulate these responses differentially. The expression of other Alzheimer's disease risk genes is differentially regulated across the distinct cell states elicited in response to amyloid pathology. Thus, we have identified multiple transcriptomic cell states adopted by human microglia in a multipronged response to Alzheimer's disease-related pathology, which should be taken into account in translational studies.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Microglia , Receptors, Immunologic , Transcriptome , Humans , Microglia/metabolism , Microglia/pathology , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Animals , Amyloid beta-Peptides/metabolism , Mice , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Mice, Transgenic , Heterografts , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Brain/metabolism , Brain/pathology
19.
Discov Med ; 36(182): 621-631, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38531803

ABSTRACT

BACKGROUND: Diffuse large B-cell lymphoma (DLBCL) poses a significant threat to the quality of life for people worldwide. Regrettably, effective treatment strategies for this disease remain elusive in clinical practice due to the unclear understanding of its molecular mechanisms. Therefore, this study was devised to address these issues and identify novel diagnostic, therapeutic, and prognostic biomarkers for DLBCL. METHODS: Gene expression and clinical data for DLBCL patients were retrieved from The Cancer Genome Atlas (TCGA) database, and relevant clinical data, tumor mutational burden (TMB), and gene expression levels were extracted. Bioinformatics analysis was conducted to screen for differentially expressed genes (DEGs). The prognostic significance of flotillin-2 (FLOT2) was assessed using Kaplan-Meier survival analysis. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses were employed to evaluate mRNA and protein levels of the genes. Cell proliferation, apoptosis, and invasion were assessed using cell counting kit-8 (CCK-8) assay, flow cytometry analysis, and Transwell assay, respectively. RESULTS: Our bioinformatics analysis revealed that FLOT2 was significantly overexpressed in DLBCL tissues compared to normal tissues, a finding corroborated by subsequent immunohistochemistry staining, qRT-PCR, and Western blot analyses. To elucidate its biological functions, shRNAs targeting FLOT2 were transfected into DLBCL cell lines (LY-3 and U2932), resulting in suppressed cell proliferation and invasion, while promoting apoptosis. Furthermore, a positive correlation between TMB and FLOT2 expression in DLBCL was observed. Subsequently, quanTIseq was utilized to calculate the immune score and assess FLOT2 gene expression. In DLBCL, FLOT2 gene expression was found to be associated with T cell CD4+ (non-regulatory) (p < 0.01), monocytes (p < 0.05), and uncharacterized cells (p < 0.05). Regarding immune checkpoint markers, including the cluster of differentiation 274 (CD274), cytotoxic T lymphocyte-associated antigen-4 (CTLA4), hepatitis A virus cellular receptor 2 (HAVCR2), lymphocyte activation gene-3 (LAG3), programmed cell death protein 1 (PDCD1), programmed cell death 1 ligand 2 (PDCD1LG2), Siglec-15 (SIGLEC15), and T cell immunoreceptor with Ig and ITIM domains (TIGIT), our analysis indicated that in DLBCL, FLOT2 exhibited a relationship only with TIGIT (p < 0.05). CONCLUSIONS: In summary, FLOT2 functions as an oncogene and is linked to DLBCL prognosis and the tumor microenvironment. Targeting FLOT2 deletion emerges as a novel strategy to impede DLBCL aggressiveness by inhibiting cell proliferation and invasion, ultimately inducing apoptotic cell death.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Membrane Proteins , Quality of Life , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Biomarkers, Tumor/analysis , Epigenesis, Genetic , Receptors, Immunologic/genetics , Tumor Microenvironment
20.
Cell Rep Med ; 5(3): 101450, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38508139

ABSTRACT

CD47 is a ligand of SIRPα, an inhibitory receptor expressed by macrophages, dendritic cells, and natural killer (NK) cells, and, therefore, transgenic overexpression of CD47 is considered an effective approach to inhibiting transplant rejection. However, the detrimental effect of CD47 signaling is overlooked when exploring this approach. Here, we construct a mutant CD47 by replacing the transmembrane and intracellular domains with a membrane anchor (CD47-IgV). In both human and mouse cells, CD47-IgV is efficiently expressed on the cell surface and protects against phagocytosis in vitro and in vivo but does not induce cell death or inhibit angiogenesis. Furthermore, hematopoietic stem cells expressing transgenic CD47-IgV show no detectable alterations in engraftment or differentiation. This study provides a potentially effective means of achieving transgenic CD47 expression that may help to produce gene-edited pigs for xenotransplantation and hypoimmunogenic pluripotent stem cells for regenerative medicine.


Subject(s)
Angiogenesis , CD47 Antigen , Animals , Humans , Mice , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cell Death , Phagocytosis/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...