Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.300
Filter
1.
Exp Clin Transplant ; 22(7): 540-550, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39223812

ABSTRACT

OBJECTIVES: Chronic rejection remains the leading cause of progressive decline in graft function. Accumulating evidence indicates that macrophages participate in chronic rejection dependent on CD40-CD40L. The FOS family members are critical in inflammatory and immune responses. However, the mechanisms underlying the role of FOS family members in chronic rejection remain unclear. In this study, we aimed to elucidate the role and underlying mechanisms of FOS-positive macrophages regulated by CD40 that mediate chronic allograft rejection. MATERIALS AND METHODS: We downloaded publicly accessible chronic rejection kidney transplant single-cell sequencing datasets from the gene expression omnibus database. Differentially expressed genes between the CD40hi and CD40low macrophage chronic rejection groups were analyzed. We established a chronic rejection mouse model by using CTLA-4-Ig. We treated bone marrow-derived macrophages with an anti-CD40 antibody. We assessed expression of the FOS family by flow cytometry, real-time quantitative polymerase chain reaction, Western blotting, and immunofluorescence. We identified altered signaling pathways by using RNA sequencing analysis. We detected DNA specifically bound to transcription factors by using ChIP-sequencing, with detection of the degree of graft fibrosis and survival. RESULTS: FOS was highly expressed on CD40hi macrophages in patients with chronic transplantrejection. Mechanistically, we showed that CD40 activated NF-κB2 translocation into the nucleus to upregulate c-Fos and FosB expression, thus promoting chronic rejection of cardiac transplant.We showed thatNF-κB2 regulated c-Fos and FosB expression by binding to the c-fos and fosb promoter regions. Inhibition of c-Fos/activator protein-1 decreased graft fibrosis and prolonged graft survival. CONCLUSIONS: CD40 may activate transcription factor NF-κB2 translocation into the nucleus of macrophages to upregulate c-Fos and FosB expression, thus promoting chronic rejection of cardiac transplant. Inhibition of c-Fos/activator protein-1 decreased grafts fibrosis and prolonged graft survival.


Subject(s)
CD40 Antigens , Disease Models, Animal , Graft Rejection , Heart Transplantation , Macrophages , Proto-Oncogene Proteins c-fos , Signal Transduction , Animals , Humans , Male , Mice , CD40 Antigens/metabolism , CD40 Antigens/genetics , Cells, Cultured , Chronic Disease , Databases, Genetic , Fibrosis , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/genetics , Graft Survival , Heart Transplantation/adverse effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , Transcription Factor AP-1/metabolism
2.
Neurosurg Rev ; 47(1): 571, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39242402

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most aggressive and deadly forms of brain cancer, which has a very complex tumor microenvironment (TME) promoting tumor growth, immune evasion, and resistance to therapy. The main players within this environment are represented by cytokines such as Interleukin-4, Interleukin-6, and Interleukin-13, along with the costimulatory molecule CD40. The paper draws back the curtain on the complex interactions played out by these molecules in contributing to the formation of a TME within GBM. IL-4 and IL-13 induce an immunosuppressive environment through the polarization of tumor-associated macrophages (TAMs) into a pro-tumoral M2 phenotype. In contrast, IL-6 takes part in the activation of the JAK-STAT3 pathway, enhancing survival and proliferation of tumor cells. In this context, CD40 either induces anti-tumor immunity through APC activation or facilitates tumors by angiogenesis and survival pathways. The synergistic actions of these molecules create feedback loops that keep up the malignancy of GBM and present a big problem for therapy. Knowledge of these interactions opens new ways for the development of multi-targeted therapeutic strategies at the other end. This may result in the interruption of the tumor-supportive environment in GBM, reducing tumor growth and improving patient outcomes by targeting IL-4, IL-6, IL-13, and CD40 simultaneously.


Subject(s)
Brain Neoplasms , CD40 Antigens , Glioblastoma , Interleukin-13 , Interleukin-4 , Interleukin-6 , Tumor Microenvironment , Humans , CD40 Antigens/metabolism , Clinical Trials as Topic , Interleukin-13/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism
3.
Int Immunopharmacol ; 140: 112785, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39088915

ABSTRACT

OBJECTIVE: To observe the effects and mechanisms of Maresin2 on the function of DCs(Dendritic cells). METHOD: The levels of IL-6, IL-12, TNF-α and IL-1ß secreted by BMDCs (Bone marrow-derived Dendritic cells) after Maresin2 treatment were detected by ELISA. At the same time, the expressions of costimulatory molecules CD40 and CD86 on the surface, the ability of phagocytosis of ovalbumin(OVA) antigen, and antigen presentation function in BMDCs were analyzed by flow cytometry. Finally, MAPK and NF-κB pathway signaling phosphorylation in Maresin2-treated BMDCs were detected by western blot. RESULTS: The secretion levels of IL-6, IL-12, TNF-α and IL-1ß were significantly decreased in the Maresin2 treatment group after LPS treatment (P < 0.05). The expression levels of CD86 and CD40 were significantly decreased after Maresin2 treatment (P < 0.05). Maresin2 enhanced the phagocytosis ability of ovalbumin(OVA) (P < 0.05), but the ability of antigen presentation of BMDCs with the treatment of Maresin2 changed slightly (P > 0.05). Phosphorylation of p38, JNK, p65, ikka/ß and ERK peaked at 15 min in the LPS group, while phosphorylation of p-p38 and p-ERK weakened 30 min and 60 min after treatment with Maresin2. CONCLUSIONS: Maresin2 inhibits inflammatory cytokine secretion but enhances phagocytosis via the MAPK/NF-κB pathway in BMDCs, which may contribute to negatively regulating inflammation.


Subject(s)
Cytokines , Dendritic Cells , NF-kappa B , Phagocytosis , Signal Transduction , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , NF-kappa B/metabolism , Mice , Cytokines/metabolism , Signal Transduction/drug effects , Phagocytosis/drug effects , Cells, Cultured , Ovalbumin/immunology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/immunology , Mice, Inbred C57BL , Cell Differentiation/drug effects , CD40 Antigens/metabolism , Antigen Presentation/drug effects , Docosahexaenoic Acids
4.
Cell Rep ; 43(9): 114664, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39178113

ABSTRACT

Trained immunity is characterized by histone modifications and metabolic changes in innate immune cells following exposure to inflammatory signals, leading to heightened responsiveness to secondary stimuli. Although our understanding of the molecular regulation of trained immunity has increased, the role of adaptive immune cells herein remains largely unknown. Here, we show that T cells modulate trained immunity via cluster of differentiation 40-tissue necrosis factor receptor-associated factor 6 (CD40-TRAF6) signaling. CD40-TRAF6 inhibition modulates functional, transcriptomic, and metabolic reprogramming and modifies histone 3 lysine 4 trimethylation associated with trained immunity. Besides in vitro studies, we reveal that single-nucleotide polymorphisms in the proximity of CD40 are linked to trained immunity responses in vivo and that combining CD40-TRAF6 inhibition with cytotoxic T lymphocyte antigen 4-immunoglobulin (CTLA4-Ig)-mediated co-stimulatory blockade induces long-term graft acceptance in a murine heart transplantation model. Combined, our results reveal that trained immunity is modulated by CD40-TRAF6 signaling between myeloid and adaptive immune cells and that this can be leveraged for therapeutic purposes.


Subject(s)
CD40 Antigens , Mice, Inbred C57BL , Signal Transduction , TNF Receptor-Associated Factor 6 , CD40 Antigens/metabolism , Animals , TNF Receptor-Associated Factor 6/metabolism , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Humans , Male , Heart Transplantation , Trained Immunity
5.
PLoS Negl Trop Dis ; 18(8): e0012417, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39159266

ABSTRACT

Visceral leishmaniasis (VL) is a neglected tropical disease caused by parasites from the Leishmania (L.) donovani complex. VL is characterised by uncontrolled parasite replication in spleen, liver and bone marrow, and by an impaired immune response and high systemic levels of inflammation. Monocytes have been poorly characterised in VL patients. The aim of this study was to evaluate the expression levels of markers involved in the regulation of T cell responses on different subsets of monocytes from the blood of VL patients and healthy non-endemic controls (HNEC). Monocytes can broadly be divided into three subsets: classical, intermediate and non-classical monocytes. Our results show that the percentages of all three subsets stayed similar at the time of VL diagnosis (ToD) and at the end of anti-leishmanial treatment (EoT). We first looked at co-stimulatory receptors: the expression levels of CD40 were significantly increased on classical and intermediate, but not non-classical monocytes, at ToD as compared to EoT and HNEC. CD80 expression levels were also increased on intermediate monocytes at ToD as compared to EoT and HNEC, and on classical monocytes only as compared to HNEC. The levels of CD86 were similar at EoT and ToD and in HNEC on classical and intermediate monocytes, but significantly higher at EoT on non-classical monocytes. We also looked at an inhibitory molecule, PD-L1. Our results show that the expression levels of PD-L1 were significantly higher on all three monocyte subsets at ToD as compared to HNEC, and to EoT on classical and intermediate monocytes. These results show that monocytes from the blood of VL patients upregulate both co-stimulatory and inhibitory receptors and that their expression levels are restored at EoT.


Subject(s)
CD40 Antigens , Leishmaniasis, Visceral , Monocytes , Humans , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/blood , Monocytes/immunology , Male , Female , Adult , CD40 Antigens/metabolism , Young Adult , Middle Aged , B7-1 Antigen/metabolism , Adolescent , B7-2 Antigen/metabolism , Leishmania donovani/immunology
6.
Nat Commun ; 15(1): 6971, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39138218

ABSTRACT

Ligation of the B cell antigen receptor (BCR) initiates humoral immunity. However, BCR signaling without appropriate co-stimulation commits B cells to death rather than to differentiation into immune effector cells. How BCR activation depletes potentially autoreactive B cells while simultaneously primes for receiving rescue and differentiation signals from cognate T lymphocytes remains unknown. Here, we use a mass spectrometry-based proteomic approach to identify cytosolic/nuclear shuttling elements and uncover transcription factor EB (TFEB) as a central BCR-controlled rheostat that drives activation-induced apoptosis, and concurrently promotes the reception of co-stimulatory rescue signals by supporting B cell migration and antigen presentation. CD40 co-stimulation prevents TFEB-driven cell death, while enhancing and prolonging TFEB's nuclear residency, which hallmarks antigenic experience also of memory B cells. In mice, TFEB shapes the transcriptional landscape of germinal center B cells. Within the germinal center, TFEB facilitates the dark zone entry of light-zone-residing centrocytes through regulation of chemokine receptors and, by balancing the expression of Bcl-2/BH3-only family members, integrates antigen-induced apoptosis with T cell-provided CD40 survival signals. Thus, TFEB reprograms antigen-primed germinal center B cells for cell fate decisions.


Subject(s)
Apoptosis , B-Lymphocytes , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , CD40 Antigens , Germinal Center , Receptors, Antigen, B-Cell , Animals , Germinal Center/immunology , Germinal Center/cytology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Mice , CD40 Antigens/metabolism , CD40 Antigens/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Mice, Inbred C57BL , Lymphocyte Activation/immunology , Cell Differentiation/immunology , Signal Transduction , Antigen Presentation/immunology
7.
COPD ; 21(1): 2389909, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39143749

ABSTRACT

The involvement of Group 3 innate lymphoid cells (ILC3s) and dendritic cells (DCs) in chronic lung inflammation has been increasingly regarded as the key to understand the inflammatory mechanisms of smoke-related chronic obstructive pulmonary disease (COPD). However, the mechanism underlying the engagement of both remains unclear. Our study aimed to explore NCR-ILC3 differentiation in the lungs of mice exposed to cigarette smoke (CS) and to further investigate whether DCs activated by CS exposure contribute to the differentiation of ILCs into NCR-ILC3s. The study involved both in vivo and in vitro experiments. In the former, the frequencies of lung NCR-ILC3s and NKp46-IL-17A+ ILCs and the expression of DCs, CD40, CD86, IL-23, and IL-1ß quantified by flow cytometry were compared between CS-exposed mice and air-exposed mice. In the latter, NKp46-IL-17A+ ILC frequencies quantified by flow cytometry were compared after two cocultures, one involving lung CD45+Lin-CD127+ ILCs sorted from air-exposed mice and DCs sifted by CD11c magnetic beads from CS-exposed mice and another including identical CD45+Lin-CD127+ ILCs and DCs from air-exposed mice. The results indicated significant increases in the frequencies of NCR-ILC3s and NKp46-IL-17A+ ILCs; in the expression of DCs, CD40, CD86, IL-23, and IL-1ß in CS-exposed mice; and in the frequency of NKp46-IL-17A+ ILCs after the coculture with DCs from CS-exposed mice. In conclusion, CS exposure increases the frequency of lung ILCs and NCR-ILC3s. CS-induced DC activation enhances the differentiation of ILCs into NCR-ILC3s, which likely acts as a mediating step in the involvement of NCR-ILC3s in chronic lung inflammation.


Subject(s)
Cell Differentiation , Dendritic Cells , Interleukin-17 , Interleukin-1beta , Lung , Natural Cytotoxicity Triggering Receptor 1 , Animals , Dendritic Cells/immunology , Mice , Lung/immunology , Lung/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Interleukin-23/metabolism , B7-2 Antigen/metabolism , Mice, Inbred C57BL , Smoke/adverse effects , Pulmonary Disease, Chronic Obstructive/immunology , CD40 Antigens/metabolism , Cigarette Smoking/adverse effects , Immunity, Innate , Antigens, Ly/metabolism , Coculture Techniques , Male
8.
Nat Commun ; 15(1): 6923, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39134540

ABSTRACT

The combination of radiotherapy/chemoradiotherapy and immune checkpoint blockade can result in poor outcomes in patients with locally advanced head and neck squamous cell carcinoma (HNSCC). Here, we show that combining ATR inhibition (ATRi) with radiotherapy (RT) increases the frequency of activated NKG2A+PD-1+ T cells in animal models of HNSCC. Compared with the ATRi/RT treatment regimen alone, the addition of simultaneous NKG2A and PD-L1 blockade to ATRi/RT, in the adjuvant, post-radiotherapy setting induces a robust antitumour response driven by higher infiltration and activation of cytotoxic T cells in the tumour microenvironment. The efficacy of this combination relies on CD40/CD40L costimulation and infiltration of activated, proliferating memory CD8+ and CD4+ T cells with persistent or new T cell receptor (TCR) signalling, respectively. We also observe increased richness in the TCR repertoire and emergence of numerous and large TCR clonotypes that cluster based on antigen specificity in response to NKG2A/PD-L1/ATRi/RT. Collectively, our data point towards potential combination approaches for the treatment of HNSCC.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , B7-H1 Antigen , Immunotherapy , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment , Animals , Female , Humans , Mice , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/radiation effects , CD40 Antigens/metabolism , CD40 Antigens/immunology , CD40 Antigens/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Head and Neck Neoplasms/radiotherapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Squamous Cell Carcinoma of Head and Neck/therapy , Squamous Cell Carcinoma of Head and Neck/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/radiation effects
9.
Int Immunopharmacol ; 139: 112654, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-38996777

ABSTRACT

Hypertension causes platelet activation and adhesion in the brain resulting in glial activation and neuroinflammation. Further, activation of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor (ACE2/Ang (1-7)/MasR) axis of central Renin-Angiotensin System (RAS), is known to reduce glial activation and neuroinflammation, thereby exhibiting anti-hypertensive and anti-neuroinflammatory properties. Therefore, in the present study, the role of ACE2/Ang (1-7)/MasR axis was studied on platelet-induced glial activation and neuroinflammation using Diminazene Aceturate (DIZE), an ACE2 activator, in astrocytes and microglial cells as well as in rat model of hypertension. We found that the ACE2 activator DIZE, independently of its BP-lowering properties, efficiently prevented hypertension-induced glial activation, neuroinflammation, and platelet CD40-CD40L signaling via upregulation of ACE2/Ang (1-7)/MasR axis. Further, DIZE decreased platelet deposition in the brain by reducing the expression of adhesion molecules on the brain endothelium. Activation of ACE2 also reduced hypertension-induced endothelial dysfunction by increasing eNOS bioavailability. Interestingly, platelets isolated from hypertensive rats or activated with ADP had significantly increased sCD40L levels and induced significantly more glial activation than platelets from DIZE treated group. Therefore, injection of DIZE pre-treated ADP-activated platelets into normotensive rats strongly reduced glial activation compared to ADP-treated platelets. Moreover, CD40L-induced glial activation, CD40 expression, and NFкB-NLRP3 inflammatory signaling are reversed by DIZE. Furthermore, the beneficial effects of ACE2 activation, DIZE was found to be significantly blocked by MLN4760 (ACE2 inhibitor) as well as A779 (MasR antagonist) treatments. Hence, our study demonstrated that ACE2 activation reduced the platelet CD40-CD40L induced glial activation and neuroinflammation, hence imparted neuroprotection.


Subject(s)
Angiotensin-Converting Enzyme 2 , CD40 Ligand , Diminazene , Disease Models, Animal , Hypertension , Peptidyl-Dipeptidase A , Signal Transduction , Animals , Diminazene/analogs & derivatives , Diminazene/pharmacology , Diminazene/therapeutic use , Angiotensin-Converting Enzyme 2/metabolism , Male , Signal Transduction/drug effects , Hypertension/drug therapy , CD40 Ligand/metabolism , Peptidyl-Dipeptidase A/metabolism , Rats , Blood Platelets/drug effects , Blood Platelets/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Proto-Oncogene Mas , Neuroglia/drug effects , Neuroglia/metabolism , Peptide Fragments , Angiotensin I , Cells, Cultured , Microglia/drug effects , Microglia/metabolism , Brain/drug effects , Brain/metabolism , Rats, Wistar , Renin-Angiotensin System/drug effects , Receptors, G-Protein-Coupled/metabolism , CD40 Antigens/metabolism , Humans , Platelet Activation/drug effects , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use
10.
Immunity ; 57(8): 1848-1863.e7, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38889716

ABSTRACT

Expression of the transcriptional regulator ZFP318 is induced in germinal center (GC)-exiting memory B cell precursors and memory B cells (MBCs). Using a conditional ZFP318 fluorescence reporter that also enables ablation of ZFP318-expressing cells, we found that ZFP318-expressing MBCs were highly enriched with GC-derived cells. Although ZFP318-expressing MBCs constituted only a minority of the antigen-specific MBC compartment, their ablation severely impaired recall responses. Deletion of Zfp318 did not alter the magnitude of primary responses but markedly reduced MBC participation in recall. CD40 ligation promoted Zfp318 expression, whereas B cell receptor (BCR) signaling was inhibitory. Enforced ZFP318 expression enhanced recall performance of MBCs that otherwise responded poorly. ZFP318-deficient MBCs expressed less mitochondrial genes, had structurally compromised mitochondria, and were susceptible to reactivation-induced cell death. The abundance of ZFP318-expressing MBCs, instead of the number of antigen-specific MBCs, correlated with the potency of prime-boost vaccination. Therefore, ZFP318 controls the MBC recallability and represents a quality checkpoint of humoral immune memory.


Subject(s)
Germinal Center , Immunologic Memory , Memory B Cells , Mitochondria , Animals , Mitochondria/metabolism , Mitochondria/immunology , Mice , Immunologic Memory/genetics , Immunologic Memory/immunology , Memory B Cells/immunology , Memory B Cells/metabolism , Germinal Center/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/genetics , Gene Expression Regulation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/metabolism , Transcription Factors/genetics , Signal Transduction/immunology , CD40 Antigens/metabolism , CD40 Antigens/genetics , CD40 Antigens/immunology , Immunity, Humoral , Transcription, Genetic , Membrane Proteins , Mitochondrial Proteins
11.
Biomaterials ; 311: 122688, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38943821

ABSTRACT

CD40 agonist antibodies (αCD40) have shown promising anti-tumor response in both preclinical and early clinical studies. However, its systemic administration is associated with immune- and hepato-toxicities which hampers its clinical usage. In addition, αCD40 showed low tumor retention and induced PD-L1 expression which makes tumor microenvironment (TME) immunosuppressive. To overcome these issues, in this study, we have developed a multifunctional Immunosome where αCD40 is conjugated on the surface and RRX-001, a small molecule immunomodulator was encapsulated inside it. Immunosomes showed higher tumor accumulation till 96 h of administration and displayed sustained release of αCD40 in vivo. Immunosomes significantly delayed tumor growth and showed tumor free survival in mice bearing GL-261 glioblastoma by increasing the population of CD45+CD8+ T cells, CD45+CD20+ B cells, CD45+CD11c+ DCs and F4/80+CD86+ cells in TME. Immunosome significantly reduced the population of T-regulatory cells, M2 macrophage, and MDSCs and lowered the PD-L1 expression. Moreover, Immunosomes significantly enhanced the levels of Th1 cytokines (IFN-γ, IL-6, IL-2) over Th2 cytokines (IL-4 and IL-10) which supported anti-tumor response. Most interestingly, Immunosomes averted the in vivo toxicities associated with free αCD40 by lowering the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), IL-6, IL-1α and reduced the degree of liver damage. In addition, Immunosomes treated long-term surviving mice showed tumor specific immune memory response which prevented tumor growth upon rechallenge. Our results suggested that this novel formulation can be further explored in clinics to improve in vivo anti-tumor efficacy of αCD40 with long-lasting tumor specific immunity while reducing the associated toxicities.


Subject(s)
CD40 Antigens , Glioblastoma , Tumor Microenvironment , Animals , Tumor Microenvironment/drug effects , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/immunology , CD40 Antigens/metabolism , Mice , Cell Line, Tumor , Mice, Inbred C57BL , Female , Cytokines/metabolism , Humans
12.
ACS Nano ; 18(19): 12194-12209, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38689426

ABSTRACT

In situ vaccines (ISVs) utilize the localized delivery of chemotherapeutic agents or radiotherapy to stimulate the release of endogenous antigens from tumors, thereby eliciting systemic and persistent immune activation. Recently, a bioinspired ISV strategy has attracted tremendous attention due to its features such as an immune adjuvant effect and genetic plasticity. M13 bacteriophages are natural nanomaterials with intrinsic immunogenicity, genetic flexibility, and cost-effectiveness for large-scale production, demonstrating the potential for application in cancer vaccines. In this study, we propose an ISV based on the engineered M13 bacteriophage targeting CD40 (M13CD40) for dendritic cell (DC)-targeted immune stimulation, named H-GM-M13CD40. We induce immunogenic cell death and release tumor antigens through local delivery of (S)-10-hydroxycamptothecin (HCPT), followed by intratumoral injection of granulocyte-macrophage colony stimulating factor (GM-CSF) and M13CD40 to enhance DC recruitment and activation. We demonstrate that this ISV strategy can result in significant accumulation and activation of DCs at the tumor site, reversing the immunosuppressive tumor microenvironment. In addition, H-GM-M13CD40 can synergize with the PD-1 blockade and induce abscopal effects in cold tumor models. Overall, our study verifies the immunogenicity of the engineered M13CD40 bacteriophage and provides a proof of concept that the engineered M13CD40 phage can function as an adjuvant for ISVs.


Subject(s)
Bacteriophage M13 , Cancer Vaccines , Dendritic Cells , Tumor Microenvironment , Cancer Vaccines/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Animals , Bacteriophage M13/immunology , Bacteriophage M13/chemistry , Mice , Dendritic Cells/immunology , CD40 Antigens/immunology , CD40 Antigens/metabolism , Mice, Inbred C57BL , Female , Cell Line, Tumor , Granulocyte-Macrophage Colony-Stimulating Factor , Antigens, Neoplasm/immunology , Humans
13.
ACS Appl Mater Interfaces ; 16(22): 28184-28192, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38770711

ABSTRACT

B cells, despite their several unique functionalities, remain largely untapped for use as an adoptive cell therapy and are limited to in vitro use for antibody production. B cells can be easily sourced, they possess excellent lymphoid-homing capabilities, and they can act as antigen-presenting cells (APCs), offering an alternative to dendritic cells (DCs), which have shown limited efficacy in the clinical setting. Soluble factors such as IL-4 and anti-CD40 antibody can enhance the activation, survival, and antigen-presenting capabilities of B cells; however, it is difficult to attain sufficiently high concentrations of these biologics to stimulate B cells in vivo. Micropatches as Cell Engagers (MACE) are polymeric microparticles, surface functionalized with anti-CD40 and anti-IgM, which can attach to B cells and simultaneously engage multiple B-cell receptors (BCR) and CD40 receptors. Stimulation of these receptors through MACE, unlike free antibodies, enhanced the display of costimulatory molecules on the B-cell surface, increased B-cell viability, and improved antigen presentation by B cells to T cells in vitro. B-cell activation by MACE further synergized with soluble IL-4 and anti-CD40. MACE also elicited T-cell chemokine secretion by B cells. Upon intravenous adoptive transfer, MACE-bound B cells homed to the spleen and lymph nodes, key sites for antigen presentation to T cells. Adoptive transfer of MACE-B cells pulsed with the CD4+ and CD8+ epitopes of ovalbumin significantly delayed tumor progression in a murine subcutaneous EG7-OVA tumor model, demonstrating the functional benefit conferred to B cells by MACE.


Subject(s)
B-Lymphocytes , CD40 Antigens , Polymers , Animals , B-Lymphocytes/immunology , Mice , CD40 Antigens/metabolism , CD40 Antigens/immunology , Polymers/chemistry , Receptors, Antigen, B-Cell/metabolism , Humans , T-Lymphocytes/immunology , Interleukin-4 , Mice, Inbred C57BL
14.
Int Immunopharmacol ; 134: 112100, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38728877

ABSTRACT

The parasite Leishmania resides as amastigotes within the macrophage parasitophorous vacuoles inflicting the disease Leishmaniasis. Leishmania selectively modulates mitogen-activated protein kinase (MAPK) phosphorylation subverting CD40-triggered anti-leishmanial functions of macrophages. The mechanism of any pathogen-derived molecule induced host MAPK modulation remains poorly understood. Herein, we show that of the fifteen MAPKs, LmjMAPK4 expression is higher in virulent L. major. LmjMAPK4- detected in parasitophorous vacuoles and cytoplasm- binds MEK-1/2, but not MKK-3/6. Lentivirally-overexpressed LmjMAPK4 augments CD40-activated MEK-1/2-ERK-1/2-MKP-1, but inhibits MKK3/6-p38MAPK-MKP-3, phosphorylation. A rationally-identified LmjMAPK4 inhibitor reinstates CD40-activated host-protective anti-leishmanial functions in L. major-infected susceptible BALB/c mice. These results identify LmjMAPK4 as a MAPK modulator at the host-pathogen interface and establish a pathogen-intercepted host receptor signaling as a scientific rationale for identifying drug targets.


Subject(s)
CD40 Antigens , Leishmania major , Leishmaniasis, Cutaneous , Macrophages , Mice, Inbred BALB C , Signal Transduction , Animals , Leishmania major/immunology , Leishmania major/physiology , CD40 Antigens/metabolism , Mice , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Macrophages/immunology , Macrophages/parasitology , Humans , Female , Phosphorylation , Host-Parasite Interactions/immunology , MAP Kinase Signaling System/immunology
15.
Immunol Lett ; 268: 106882, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38810887

ABSTRACT

Anti-CD40 antibodies (Abs) have been shown to induce antitumor T-cell responses. We reported that the engineered agonistic anti-CD40 Ab (5C11, IgG4 isotype) recognized human CD40 antigen expressed on a human B lymphoblastoid cell line as well as on splenic cells isolated from humanized CD40 mice. Of note, a single high dosage of 5C11 was able to prohibit tumor growth in parallel with an increase in the population of infiltrated CD8+ T cells. Furthermore, the antitumor effects of 5C11 were enhanced in the presence of ß-glucan along with an increase in the population of infiltrated CD8+ T cells. In addition, the numbers of CD86+ TAMs and neutrophils were elevated in the combination of 5C11 and ß-glucan compared with either 5C11 or ß-glucan alone. Furthermore, the abundance of Faecalibaculum, one of the probiotics critical for tumor suppression, was obviously increased in the combination of 5C11 and ß-glucan-treated mice. These data reveal a novel mechanism of tumor suppression upon the combination treatment of 5C11 and ß-glucan and propose that the combination treatment of agonistic anti-human CD40 antibody 5C11 and ß-glucan could be a promising therapeutic strategy for cancer patients.


Subject(s)
CD40 Antigens , beta-Glucans , Animals , CD40 Antigens/agonists , CD40 Antigens/immunology , CD40 Antigens/metabolism , beta-Glucans/pharmacology , Mice , Humans , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology , Drug Synergism
16.
Medicine (Baltimore) ; 103(14): e37718, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38579073

ABSTRACT

The interaction between CD40 and CD40 ligand (CD40L) a crucial co-stimulatory signal for activating adaptive immune cells, has a noteworthy role in atherosclerosis. It is well-known that atherosclerosis is linked to immune inflammation in blood vessels. In atherosclerotic lesions, there is a multitude of proinflammatory cytokines, adhesion molecules, and collagen, as well as smooth muscle cells, macrophages, and T lymphocytes, particularly the binding of CD40 and CD40L. Therefore, research on inhibiting the CD40-CD40L system to prevent atherosclerosis has been ongoing for more than 30 years. However, it's essential to note that long-term direct suppression of CD40 or CD40L could potentially result in immunosuppression, emphasizing the critical role of the CD40-CD40L system in atherosclerosis. Thus, specifically targeting the CD40-CD40L interaction on particular cell types or their downstream signaling pathways may be a robust strategy for mitigating atherosclerosis, reducing potential side effects. This review aims to summarize the potential utility of the CD40-CD40L system as a viable therapeutic target for atherosclerosis.


Subject(s)
Atherosclerosis , CD40 Ligand , Humans , Atherosclerosis/drug therapy , Atherosclerosis/immunology , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/metabolism , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/metabolism , Cytokines/metabolism , Interleukin-2/metabolism , Macrophages/metabolism
17.
Biochem Biophys Res Commun ; 714: 149969, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38657446

ABSTRACT

CD40 is a member of the tumor necrosis factor receptor superfamily, and it is widely expressed on immune and non-immune cell types. The interaction between CD40 and the CD40 ligand (CD40L) plays an essential function in signaling, and the CD40/CD40L complex works as an immune checkpoint molecule. CD40 has become a therapeutic target, and a variety of agonistic/antagonistic anti-CD40 monoclonal antibodies (mAbs) have been developed. To better understand the mode of action of anti-CD40 mAbs, we determined the X-ray crystal structures of dacetuzumab (agonist) and bleselumab (antagonist) in complex with the extracellular domain of human CD40, respectively. The structure reveals that dacetuzumab binds to CD40 on the top of cysteine-rich domain 1 (CRD1), which is the domain most distant from the cell surface, and it does not compete with CD40L binding. The binding interface of bleselumab spread between CRD2 and CRD1, overlapping with the binding surface of the ligand. Our results offer important insights for future structural and functional studies of CD40 and provide clues to understanding the mechanism of biological response. These data can be applied to developing new strategies for designing antibodies with more therapeutic efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized , CD40 Antigens , Humans , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Binding Sites , CD40 Antigens/chemistry , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/chemistry , CD40 Ligand/metabolism , CD40 Ligand/immunology , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Conformation
18.
Basic Res Cardiol ; 119(4): 1-18, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38554187

ABSTRACT

CD40L-CD40-TRAF signaling plays a role in atherosclerosis progression and affects the pathogenesis of coronary heart disease (CHD). We tested the hypothesis that CD40L-CD40-TRAF signaling is a potential therapeutic target in hyperlipidemia, diabetes, and hypertension. In mouse models of hyperlipidemia plus diabetes (db/db mice) or hypertension (1 mg/kg/d angiotensin-II for 7 days), TRAF6 inhibitor treatment (2.5 mg/kg/d for 7 or 14 days) normalized markers of oxidative stress and inflammation. As diabetes and hypertension are important comorbidities aggravating CHD, we explored whether the CD40L-CD40-TRAF signaling cascade and their associated inflammatory pathways are expressed in CHD patients suffering from comorbidities. Therefore, we analyzed vascular bypass material (aorta or internal mammary artery) and plasma from patients with CHD with diabetes and/or hypertension. Our Olink targeted plasma proteomic analysis using the IMMUNO-ONCOLOGY panel revealed a pattern of step-wise increase for 13/92 markers of low-grade inflammation with significant changes. CD40L or CD40 significantly correlated with 38 or 56 other inflammatory targets. In addition, specific gene clusters that correlate with the comorbidities were identified in isolated aortic mRNA of CHD patients through RNA-sequencing. These signaling clusters comprised CD40L-CD40-TRAF, immune system, hemostasis, muscle contraction, metabolism of lipids, developmental biology, and apoptosis. Finally, immunological analysis revealed key markers correlated with comorbidities in CHD patients, such as CD40L, NOX2, CD68, and 3-nitrotyrosine. These data indicate that comorbidities increase inflammatory pathways in CHD, and targeting these pathways will be beneficial in reducing cardiovascular events in CHD patients with comorbidities.


Subject(s)
CD40 Antigens , CD40 Ligand , Hypertension , Signal Transduction , Humans , Animals , CD40 Ligand/metabolism , Hypertension/immunology , Hypertension/metabolism , CD40 Antigens/metabolism , Male , Inflammation/metabolism , Inflammation/immunology , Mice , Mice, Inbred C57BL , Female , Middle Aged , TNF Receptor-Associated Factor 6/metabolism , Aged , Coronary Disease/immunology , Coronary Disease/metabolism
19.
Am J Pathol ; 194(7): 1230-1247, 2024 07.
Article in English | MEDLINE | ID: mdl-38548267

ABSTRACT

Hepatocellular carcinoma (HCC) is associated with increased soluble CD40 levels. This study aimed to investigate CD40's role in liver tumor progression. CD40 levels were examined in HCC patient tissues and various HCC cell lines, and their interaction with CD4+T cells was studied. RNA sequencing analysis was performed to explore the mechanisms of CD40 induction. Poorly differentiated HCC tumor tissues exhibited high membrane-bound CD40 expression, in contrast to nontumor areas. Poorly differentiated HCC cell lines showed high expression of membrane-bound CD40 with low CD40 promoter methylation, which was the opposite of that observed in the well-differentiated HCC cell lines. Solely modulating CD40 expression in HCC cells exerted no direct consequences on cell growth or appearance. Interestingly, the human hepatoma cell line HLF co-cultured with activated (CD40 ligand+) CD4+ T cells had increased CD40 levels and a modest 3.2% dead cells. The percentage of dead cells increased to 10.9% and underwent preneutralizing CD40 condition, whereas preblocking both CD40 and integrin α5ß1 concomitantly caused only 1.9% cell death. RNA sequencing of co-cultured HLFs with activated CD4+ T cells revealed the up-regulation of interferon and immune-response pathways. Increased interferon-γ levels in the activated T-cell media stimulated the Janus kinase/signal transducer and activator of transcription 3 pathway, resulting in increased CD40 expression in HLF. Collectively, CD40 expression in poorly differentiated HCC cells prevented cell death by interacting with CD40 ligand in activated T cells. Targeting CD40 may represent a promising anticancer therapy.


Subject(s)
Apoptosis , CD40 Antigens , CD40 Ligand , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/genetics , CD40 Ligand/metabolism , CD40 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Line, Tumor
20.
Cells ; 13(5)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38474393

ABSTRACT

CD40 induces pro-inflammatory responses in endothelial and Müller cells and is required for the development of diabetic retinopathy (DR). CD40 is upregulated in these cells in patients with DR. CD40 upregulation is a central feature of CD40-driven inflammatory disorders. What drives CD40 upregulation in the diabetic retina remains unknown. We examined the role of advanced glycation end products (AGEs) in CD40 upregulation in endothelial cells and Müller cells. Human endothelial cells and Müller cells were incubated with unmodified or methylglyoxal (MGO)-modified fibronectin. CD40 expression was assessed by flow cytometry. The expression of ICAM-1 and CCL2 was examined by flow cytometry or ELISA after stimulation with CD154 (CD40 ligand). The expression of carboxymethyl lysine (CML), fibronectin, and laminin as well as CD40 in endothelial and Müller cells from patients with DR was examined by confocal microscopy. Fibronectin modified by MGO upregulated CD40 in endothelial and Müller cells. CD40 upregulation was functionally relevant. MGO-modified fibronectin enhanced CD154-driven upregulation of ICAM-1 and CCL2 in endothelial and Müller cells. Increased CD40 expression in endothelial and Müller cells from patients with DR was associated with increased CML expression in fibronectin and laminin. These findings identify AGEs as inducers of CD40 upregulation in endothelial and Müller cells and enhancers of CD40-dependent pro-inflammatory responses. CD40 upregulation in these cells is associated with higher CML expression in fibronectin and laminin in patients with DR. This study revealed that CD40 and AGEs, two important drivers of DR, are interconnected.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Humans , Diabetic Retinopathy/metabolism , Intercellular Adhesion Molecule-1/metabolism , Fibronectins/metabolism , Ependymoglial Cells/metabolism , Endothelial Cells/metabolism , Magnesium Oxide/metabolism , Retina/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Laminin/metabolism , Glycation End Products, Advanced/metabolism , Diabetes Mellitus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL