Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Immunol Cell Biol ; 102(7): 605-617, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38804132

ABSTRACT

M1/M2 macrophage polarization plays an important role in regulating the balance of the microenvironment within tissues. Moreover, macrophage polarization involves the reprogramming of metabolism, such as glucose and lipid metabolism. Transcriptional coactivator B-cell lymphoma-3 (Bcl-3) is an atypical member of the IκB family that controls inflammatory factor levels in macrophages by regulating nuclear factor kappa B pathway activation. However, the relationship between Bcl-3 and macrophage polarization and metabolism remains unclear. In this study, we show that the knockdown of Bcl-3 in macrophages can regulate glycolysis-related gene expression by promoting the activation of the nuclear factor kappa B pathway. Furthermore, the loss of Bcl-3 was able to promote the interferon gamma/lipopolysaccharide-induced M1 macrophage polarization by accelerating glycolysis. Taken together, these results suggest that Bcl-3 may be a candidate gene for regulating M1 polarization in macrophages.


Subject(s)
B-Cell Lymphoma 3 Protein , Glycolysis , Macrophages , NF-kappa B , Animals , Mice , B-Cell Lymphoma 3 Protein/metabolism , Cell Polarity/genetics , Gene Expression Regulation , Interferon-gamma/metabolism , Lipopolysaccharides , Macrophage Activation , Macrophages/metabolism , Macrophages/immunology , Mice, Inbred C57BL , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction
2.
PLoS Pathog ; 18(11): e1010502, 2022 11.
Article in English | MEDLINE | ID: mdl-36318581

ABSTRACT

The atypical IκB family member Bcl3 associates with p50/NF-κB1 or p52/NF-κB2 homodimers in the nucleus, and positively or negatively modulates transcription in a context-dependent manner. In mice lacking Bcl3 globally or specifically in CD11c+ cells, we previously reported that Toxoplasma gondii infection is uniformly fatal and is associated with an impaired Th1 immune response. Since Bcl3 expression in dendritic cells (DC) is pivotal for antigen presentation and since classical DCs (cDC) are major antigen presenting cells, we investigated the role of Bcl3 specifically in cDCs in vivo by crossing Zbtb46 cre mice with Bcl3flx/flx mice. Bcl3flx/flx Zbtb46 cre mice were as susceptible to lethal T. gondii infection as total Bcl3-/- mice and generated poor Th1 immune responses. Consistent with this, compared to wildtype controls, splenic Xcr1+ Bcl3-deficient cDC1 cells were defective in presenting Ova antigen to OT-I cells both for Ova257-264 peptide and after infection with Ovalbumin-expressing T. gondii. Moreover, splenic CD4+ and CD8+ T cells from infected Bcl3flx/flx Zbtb46 cre mice exhibited decreased T. gondii-specific priming as revealed by both reduced cytokine production and reduced T. gondii-specific tetramer staining. In vitro differentiation of cDCs from bone marrow progenitors also revealed Bcl3-dependent cDC-specific antigen-presentation activity. Consistent with this, splenocyte single cell RNA seq (scRNAseq) in infected mice revealed Bcl3-dependent expression of genes involved in antigen processing in cDCs. We also identified by scRNAseq, a unique Bcl3-dependent hybrid subpopulation of Zbtb46+ DCs co-expressing the monocyte/macrophage transcription factor Lysozyme M. This subpopulation exhibited Bcl3-dependent expansion after infection. Likewise, by flow cytometry we identified two T. gondii-induced hybrid subpopulations of Bcl3-dependent cDC1 and cDC2 cells both expressing monocyte/macrophage markers, designated as icDC1 and icDC2. Together, our results indicate that Bcl3 in classical DCs is a major determinant of protective T cell responses and survival in T. gondii-infection.


Subject(s)
B-Cell Lymphoma 3 Protein , Toxoplasma , Toxoplasmosis , Animals , Mice , CD8-Positive T-Lymphocytes , Dendritic Cells , Mice, Inbred C57BL , NF-kappa B/metabolism , Toxoplasma/metabolism , Toxoplasmosis/metabolism , B-Cell Lymphoma 3 Protein/metabolism
3.
BMC Immunol ; 24(1): 35, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37794349

ABSTRACT

BACKGROUND: Bcl-3 is a member of the IκB protein family and an essential modulator of NF-κB activity. It is well established that Bcl-3 is critical for the normal development, survival and differentiation of adaptive immune cells, especially T cells. However, the regulation of immune cell function by Bcl-3 through metabolic pathways has rarely been studied. RESULTS: In this study, we explored the role of Bcl-3 in the metabolism and function of T cells via the mTOR pathway. We verified that the proliferation of Bcl-3-deficient Jurkat T cells was inhibited, but their activation was promoted, and Bcl-3 depletion regulated cellular energy metabolism by reducing intracellular ATP and ROS production levels and mitochondrial membrane potential. Bcl-3 also regulates cellular energy metabolism in naive CD4+ T cells. In addition, the knockout of Bcl-3 altered the expression of mTOR, Akt, and Raptor, which are metabolism-related genes, in Jurkat cells. CONCLUSIONS: This finding indicates that Bcl-3 may mediate the energy metabolism of T cells through the mTOR pathway, thereby affecting their function. Overall, we provide novel insights into the regulatory role of Bcl-3 in T-cell energy metabolism for the prevention and treatment of immune diseases.


Subject(s)
Apoptosis , B-Cell Lymphoma 3 Protein , NF-kappa B , T-Lymphocytes , Humans , Cell Survival , Energy Metabolism , NF-kappa B/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , B-Cell Lymphoma 3 Protein/metabolism , T-Lymphocytes/metabolism
4.
PLoS Pathog ; 17(1): e1009249, 2021 01.
Article in English | MEDLINE | ID: mdl-33508001

ABSTRACT

Bcl-3 is an atypical member of the IκB family that acts in the nucleus to modulate transcription of many NF-κB targets in a highly context-dependent manner. Accordingly, complete Bcl-3-/- mice have diverse defects in both innate and adaptive immune responses; however, direct effects of Bcl-3 action in individual immune cell types have not been clearly defined. Here, we document a cell-autonomous role for Bcl-3 in CD8+ T cell differentiation during the response to lymphocytic choriomeningitis virus infection. Single-cell RNA-seq and flow cytometric analysis of virus-specific Bcl3-/- CD8+ T cells revealed that differentiation was skewed towards terminal effector cells at the expense of memory precursor effector cells (MPECs). Accordingly, Bcl3-/- CD8+ T cells exhibited reduced memory cell formation and a defective recall response. Conversely, Bcl-3-overexpression in transgenic CD8+ T cells enhanced MPEC formation but reduced effector cell differentiation. Together, our results establish Bcl-3 as an autonomous determinant of memory/terminal effector cell balance during CD8+ T cell differentiation in response to acute viral infection. Our results provide proof-of-principle for targeting Bcl-3 pharmacologically to optimize adaptive immune responses to infectious agents, cancer cells, vaccines and other stimuli that induce CD8+ T cell differentiation.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , NF-kappa B/immunology , Animals , B-Cell Lymphoma 3 Protein/genetics , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Female , Flow Cytometry , Male , Mice , Mice, Transgenic , Sequence Analysis, RNA , Single-Cell Analysis
5.
Breast Cancer Res ; 24(1): 40, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35681213

ABSTRACT

BACKGROUND: The Bcl-3 protein is an atypical member of the inhibitor of -κB family that has dual roles as a transcriptional repressor and a coactivator for dimers of NF-κB p50 and p52. Bcl-3 is expressed in mammary adenocarcinomas and can promote tumorigenesis and survival signaling and has a key role in tumor metastasis. In this study, we have investigated the role of Bcl-3 in the normal mammary gland and impact on tumor pathology. METHODS: We utilized bcl-3-/- mice to study mammary gland structure in virgins and during gestation, lactation and early involution. Expression of involution-associated genes and proteins and putative Bcl-3 target genes was examined by qRT-PCR and immunoblot analysis. Cell autonomous branching morphogenesis and collagen I invasion properties of bcl-3-/- organoids were tested in 3D hydrogel cultures. The role of Bcl-3 in tumorigenesis and tumor pathology was also assessed using a stochastic carcinogen-induced mammary tumor model. RESULTS: Bcl-3-/- mammary glands demonstrated reduced branching complexity in virgin and pregnant mice. This defect was recapitulated in vitro where significant defects in bud formation were observed in bcl-3-/- mammary organoid cultures. Bcl-3-/- organoids showed a striking defect in protrusive collective fibrillary collagen I invasion associated with reduced expression of Fzd1 and Twist2. Virgin and pregnant bcl-3-/- glands showed increased apoptosis and rapid increases in lysosomal cell death and apoptosis after forced weaning compared to WT mice. Bcl-2 and Id3 are strongly induced in WT but not bcl-3-/- glands in early involution. Tumors in WT mice were predominately adenocarcinomas with NF-κB activation, while bcl-3-/- lesions were largely squamous lacking NF-κB and with low Bcl-2 expression. CONCLUSIONS: Collectively, our results demonstrate that Bcl-3 has a key function in mammary gland branching morphogenesis, in part by regulation of genes involved in extracellular matrix invasion. Markedly reduced levels of pro-survival proteins expression in bcl-3 null compared to WT glands 24 h post-weaning indicate that Bcl-3 has a role in moderating the rate of early phase involution. Lastly, a reduced incidence of bcl-3-/- mammary adenocarcinomas versus squamous lesions indicates that Bcl-3 supports the progression of epithelial but not metaplastic cancers.


Subject(s)
Adenocarcinoma , B-Cell Lymphoma 3 Protein , Breast Neoplasms , Carcinoma, Squamous Cell , Mammary Glands, Animal , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/genetics , B-Cell Lymphoma 3 Protein/metabolism , Breast Neoplasms/pathology , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/pathology , Collagen/metabolism , Epithelial Cells/metabolism , Female , Lactation , Mammary Glands, Animal/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Pregnancy , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
6.
Gastroenterology ; 161(1): 318-332.e9, 2021 07.
Article in English | MEDLINE | ID: mdl-33819482

ABSTRACT

BACKGROUND & AIMS: The existence of different subtypes of pancreatic ductal adenocarcinoma (PDAC) and their correlation with patient outcome have shifted the emphasis on patient classification for better decision-making algorithms and personalized therapy. The contribution of mechanisms regulating the cancer stem cell (CSC) population in different subtypes remains unknown. METHODS: Using RNA-seq, we identified B-cell CLL/lymphoma 3 (BCL3), an atypical nf-κb signaling member, as differing in pancreatic CSCs. To determine the biological consequences of BCL3 silencing in vivo and in vitro, we generated bcl3-deficient preclinical mouse models as well as murine cell lines and correlated our findings with human cell lines, PDX models, and 2 independent patient cohorts. We assessed the correlation of bcl3 expression pattern with clinical parameters and subtypes. RESULTS: Bcl3 was significantly down-regulated in human CSCs. Recapitulating this phenotype in preclinical mouse models of PDAC via BCL3 genetic knockout enhanced tumor burden, metastasis, epithelial to mesenchymal transition, and reduced overall survival. Fluorescence-activated cell sorting analyses, together with oxygen consumption, sphere formation, and tumorigenicity assays, all indicated that BCL3 loss resulted in CSC compartment expansion promoting cellular dedifferentiation. Overexpression of BCL3 in human PDXs diminished tumor growth by significantly reducing the CSC population and promoting differentiation. Human PDACs with low BCL3 expression correlated with increased metastasis, and BCL3-negative tumors correlated with lower survival and nonclassical subtypes. CONCLUSIONS: We demonstrate that bcl3 impacts pancreatic carcinogenesis by restraining CSC expansion and by curtailing an aggressive and metastatic tumor burden in PDAC across species. Levels of BCL3 expression are a useful stratification marker for predicting subtype characterization in PDAC, thereby allowing for personalized therapeutic approaches.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Animals , B-Cell Lymphoma 3 Protein/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/secondary , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Energy Metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction , Tumor Burden , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 116(51): 25828-25838, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31772019

ABSTRACT

Proinflammatory responses induced by Toll-like receptors (TLRs) are dependent on the activation of the NF-ĸB and mitogen-activated protein kinase (MAPK) pathways, which coordinate the transcription and synthesis of proinflammatory cytokines. We demonstrate that BCL-3, a nuclear IĸB protein that regulates NF-ĸB, also controls TLR-induced MAPK activity by regulating the stability of the TPL-2 kinase. TPL-2 is essential for MAPK activation by TLR ligands, and the rapid proteasomal degradation of active TPL-2 is a critical mechanism limiting TLR-induced MAPK activity. We reveal that TPL-2 is a nucleocytoplasmic shuttling protein and identify the nucleus as the primary site for TPL-2 degradation. BCL-3 interacts with TPL-2 and promotes its degradation by promoting its nuclear localization. As a consequence, Bcl3-/- macrophages have increased TPL-2 stability following TLR stimulation, leading to increased MAPK activity and MAPK-dependent responses. Moreover, BCL-3-mediated regulation of TPL-2 stability sets the MAPK activation threshold and determines the amount of TLR ligand required to initiate the production of inflammatory cytokines. Thus, the nucleus is a key site in the regulation of TLR-induced MAPK activity. BCL-3 links control of the MAPK and NF-ĸB pathways in the nucleus, and BCL-3-mediated TPL-2 regulation impacts on the cellular decision to initiate proinflammatory cytokine production in response to TLR activation.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Cell Nucleus/metabolism , I-kappa B Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins/metabolism , Toll-Like Receptors/metabolism , Animals , B-Cell Lymphoma 3 Protein/genetics , Cytokines/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , RAW 264.7 Cells
8.
Biol Chem ; 402(2): 207-219, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33544468

ABSTRACT

This study was designed to illustrate the function and role of PCAT1 in CCA. The relative expression was confirmed by RT-qPCR and western blot. The biological function of PCAT1 was evaluated by CCK8, EdU, colony formation, wound healing, transwell, and subcutaneous tumor formation assays. Protein levels of EMT markers were measured by western blot. The binding relationship was predicted by JASPAR and starBase. The binding of YY1 to PCAT1 promoter was assessed by ChIP and luciferase reporter. The binding capacity between miR-216a-3p and PCAT1 as well as BCL3 was assessed by luciferase reporter and AGO2-RIP assays. In this study, we found that PCAT1 was up-regulated in CCA tissues and cells, and the PCAT1 overexpression was associated with poor prognosis. Moreover, PCAT1 was assessed as an independent risk factor of prognosis for CCA patients. Amplified PCAT1 was found to promote tumor proliferation, migration, invasion and EMT process, whereas PCAT1 knockdown inhibited these malignant phenotypes. Mechanistically, PCAT1 was predominantly localized in the cytoplasm and competitively bound miR-216a-3p to increase BCL3 expression. In addition, PCAT1 was activated by transcription factor YY1. This study revealed that PCAT1 acted as an oncogene in CCA, and the YY1/PCAT1/miR-216a-3p/BCL3 axis exhibited critical functions in CCA progression.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Up-Regulation , YY1 Transcription Factor/metabolism , B-Cell Lymphoma 3 Protein/genetics , Bile Duct Neoplasms/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Cholangiocarcinoma/pathology , Female , Humans , Male , MicroRNAs/genetics , Middle Aged , RNA, Long Noncoding/genetics , YY1 Transcription Factor/genetics
9.
J Cell Mol Med ; 24(1): 202-213, 2020 01.
Article in English | MEDLINE | ID: mdl-31668012

ABSTRACT

Ovarian cancer is a common malignancy among women with some clinically approved diagnostic coding gene biomarkers. However, long non-coding RNAs (lncRNAs) have been indicated to play an important role in controlling tumorigenesis of ovarian cancer. Hereby, the aim of the study was to uncover the function of lncRNA LINC00176 in the development and progression of ovarian cancer by regulating ceruloplasmin (CP). Bioinformatics prediction in combination with RT-qPCR analysis for the expression pattern of LINC00176 revealed that LINC00176 was highly expressed in ovarian cancer tissues as well as in ovarian cancer cell lines, respectively. LINC00176 was predominantly localized in the nucleus. Delivery of si-LINC00176, oe-LINC00176, si-BCL3 and si-CP plasmids was conducted to explore the effects of LINC00176 on ovarian cancer. Promoted proliferation, migration and invasion along with reduced apoptosis were observed in cells treated with oe-LINC00176, while si-BCL3 and si-CP were able to block the promoting effects. Investigations with regard to the correlation between LINC00176 and promoter region of CP turned out to be positive via B-cell CLL/lymphoma 3 (BCL3) by means of dual-luciferase reporter gene assay, ChIP and RIP assays. Furthermore, oncogenic properties of the LINC00176/BCL3/CP axis were also demonstrated by tumour formation in vivo generated upon injecting cells in nude mice. Our results demonstrate that restored LINC00176 initiates tumorigenesis in ovarian cancer by increasing CP expression via recruiting BCL3, the mechanism of which represented a potential and promising therapeutic target for the disease.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Biomarkers, Tumor/metabolism , Ceruloplasmin/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/pathology , RNA, Long Noncoding/genetics , Animals , Apoptosis , B-Cell Lymphoma 3 Protein/genetics , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Ceruloplasmin/genetics , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Prognosis , RNA, Long Noncoding/antagonists & inhibitors , RNA, Small Interfering , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Immunol Cell Biol ; 97(2): 134-141, 2019 02.
Article in English | MEDLINE | ID: mdl-30428148

ABSTRACT

Macrophages play a central role in innate immunity as the first line of defense against pathogen infection. Upon exposure to inflammatory stimuli, macrophages rapidly respond and subsequently undergo metabolic reprogramming to substantially produce cellular metabolites such as itaconate. As a derivate of the tricarboxylic acid cycle, itaconate is derived from the decarboxylation of cis-aconitate mediated by immunoresponsive gene 1 in the mitochondrial matrix. It is well known that itaconate has a direct antimicrobial effect by inhibiting isocitrate lyase. Strikingly, two recent studies published in Nature showed that itaconate markedly decreases the production of proinflammatory mediators in lipopolysaccharide-treated macrophages and ameliorates sepsis and psoriasis in animal models, revealing a novel biological action of itaconate beyond its regular roles in antimicrobial defense. The mechanism for this anti-inflammatory effect has been proposed to involve the inhibition of succinate dehydrogenase, blockade of IκBζ translation and activation of Nrf2. These intriguing discoveries provide a new explanation for how macrophages are switched from a pro- to an anti-inflammatory state to limit the damage and facilitate tissue repair under proinflammatory conditions. Thus, the emerging effect of itaconate as a crucial determinant of macrophage inflammation has important implications in further understanding cellular immunometabolism and developing future therapeutics for the treatment of inflammatory diseases. In this review, we focus on the roles of itaconate in controlling the inflammatory response during macrophage activation, providing a rationale for future investigation and therapeutic intervention.


Subject(s)
Inflammation/metabolism , Macrophage Activation , Macrophages/metabolism , Succinates/metabolism , Animals , B-Cell Lymphoma 3 Protein/metabolism , Humans , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Succinate Dehydrogenase/metabolism
11.
Neuroscience ; 537: 105-115, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38006962

ABSTRACT

Intracerebral hemorrhage (ICH) is a severe disease with high mortality. Recently, the role of BCL-3 in ICH has started to gain attention, but its mechanism remains unclear. A collagenase injection method was used to establish an ICH model in rats, and the expression of BCL-3 were detected. Rat brain microvascular endothelial cells (rBMECs) were isolated and induced with Hemin to establish an in vitro ICH model. The expression of BCL-3 was assessed, followed by detection of cell apoptosis. In the cell model, the recruitment, polarization, and pro-inflammatory features of the microglia (MGs) were assessed after co-cultured with rBMECs. Finally, in the ICH animal model, after knockdown of BCL-3, comprehensive evaluations of inflammatory responses in brain tissue, polarization and recruitment of microglia, and apoptosis were conducted. Results revealed an upregulated expression of BCL-3 in brain tissue of the ICH animal model. In Hemin-treated rBMECs, an upward trend in BCL-3 expression was observed, accompanied by an increase of cell apoptosis. After co-culturing with the in vitro model, microglia exhibited enhanced M1 polarization and intensified inflammatory responses. However, when BCL-3 expression was inhibited in the in vitro model, a reversal occurred in the polarization tendency and inflammatory responses of microglia. Additionally, after knockdown of BCL-3 in the animal model, notable improvements occurred in M1 polarization, infiltration of macrophages, and inflammatory reactions in the brain tissue. Therefore, BCL-3 modulates the inflammatory response after ICH occurrence through the BMECs/MGs microenvironment. Additionally, BCL-3 might be a potential therapeutic target for ICH management.


Subject(s)
Endothelial Cells , Hemin , Animals , Rats , Cerebral Hemorrhage/metabolism , Endothelial Cells/metabolism , Hemin/metabolism , Inflammation/metabolism , Microglia/metabolism , B-Cell Lymphoma 3 Protein/metabolism
12.
Mucosal Immunol ; 17(4): 673-691, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38663461

ABSTRACT

Peripherally-induced regulatory T cells (pTregs) expressing the retinoic acid receptor-related orphan-receptor gamma t (RORγt) are indispensable for intestinal immune homeostasis. Nuclear factor kappa family members regulate the differentiation of thymic Tregs and promote their survival in the periphery. However, the Treg intrinsic molecular mechanisms controlling the size of the pTregs in the intestine and associated lymphoid organs remain unclear. Here, we provide direct evidence that B-cell lymphoma 3 (Bcl3) limits the development of pTregs in a T cell-intrinsic manner. Moreover, the absence of Bcl3 allowed for the formation of an unusual intestinal Treg population co-expressing the transcription factors Helios and RORγt. The expanded RORγt+ Treg populations in the absence of Bcl3 displayed an activated phenotype and secreted high levels of the anti-inflammatory cytokines interleukin (IL)-10 and transforming growth factor beta. They were fully capable of suppressing effector T cells in a transfer colitis model despite an intrinsic bias to trans-differentiate toward T helper 17-like cells. Finally, we provide a Bcl3-dependent gene signature in pTregs including altered responsiveness to the cytokines IL-2, IL-6, and tumor necrosis factor alpha. Our results demonstrate that Bcl3 acts as a molecular switch to limit the expansion of different intestinal Treg subsets and may thus serve as a novel therapeutic target for inflammatory bowel disease by restoring intestinal immune tolerance.


Subject(s)
B-Cell Lymphoma 3 Protein , Cell Differentiation , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3 , T-Lymphocytes, Regulatory , Animals , B-Cell Lymphoma 3 Protein/metabolism , B-Cell Lymphoma 3 Protein/genetics , T-Lymphocytes, Regulatory/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Colitis/immunology , Colitis/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestines/immunology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cells, Cultured , Th17 Cells/immunology
13.
Cell Death Dis ; 15(9): 690, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39327470

ABSTRACT

The t(14;19)(q32;q13) is a rare recurring translocation found in B-cell lymphoproliferative malignancies, involving the Bcl-3 gene. This chromosomal translocation is often found in patients under the age of 50 and causes a more progressive disease. The Bcl-3 gene encodes a protein belonging to the IκB family of proteins, which tightly regulates NFκB signaling by acting as an activator or repressor of transcription. Previously, we developed a second-generation Bcl-3 inhibitor that could directly interfere with Bcl-3 signaling pathway, resulting in reduced melanoma cell proliferation, invasion, and migration. The present study aimed to investigate the effect of a Bcl-3 inhibitor on B-cell lymphoma and leukemia cells. It was found that treatment of cells with this inhibitor caused a decrease in cell proliferation and cell survival. Furthermore, Bcl-3 inhibition in B-cell malignant cells resulted in the loss of mitochondrial membrane potential and functionality, as well as the increased expression of cleaved caspase 3, indicating that cell death occurs through the intrinsic apoptotic pathway. This observation is further supported by reduced expression of cIAP1 protein 1 (cIAP1) upon treatment of cancer cells. Given the current lack of clinical advancements targeting Bcl-3 in oncology, this opens a novel avenue for the development and investigation of highly specific therapeutic interventions against B-cell malignancies.


Subject(s)
Apoptosis , B-Cell Lymphoma 3 Protein , Cell Proliferation , Humans , B-Cell Lymphoma 3 Protein/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/drug therapy , Membrane Potential, Mitochondrial/drug effects , Signal Transduction/drug effects , Cell Survival/drug effects , Caspase 3/metabolism
14.
Cell Death Dis ; 13(5): 510, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35641486

ABSTRACT

Acute liver failure (ALF) is a rare entity but exhibits a high mortality. The mechanisms underlying ALF are not completely understood. The present study explored the role of the hepatic B cell leukemia-3 (Bcl-3), a transcriptional regulator of nuclear factor-kappa B (NF-κB), in two independent models of ALF. We employed a recently developed transgenic mouse model in a C57BL6/J background comparing wild-type (WT) and transgenic littermates with hepatocyte-specific overexpression of Bcl-3 (Bcl-3Hep) in the ALF model of d-galactosamine (d-GalN) and lipopolysaccharide (LPS). Additionally, the apoptosis-inducing CD95 (FAS/APO-1)-ligand was explored. Bcl-3Hep mice exhibited a significant protection from ALF with decreased serum transaminases, decreased activation of the apoptotic caspases 8, 9, and 3, lower rates of oxidative stress, B-cell lymphoma 2 like 1 (BCL2L1/BCL-XL) degradation and accompanying mitochondrial cytochrome c release, and ultimately a decreased mortality rate from d-GalN/LPS compared to WT mice. d-GalN/LPS treatment resulted in a marked inflammatory cytokine release and stimulated the activation of signal transducer and activator of transcription (STAT) 3, c-Jun N-terminal kinases (JNK) and extracellular signal-regulated kinase (ERK) signaling comparably in the hepatic compartment of Bcl-3Hep and WT mice. However, in contrast to the WT, Bcl-3Hep mice showed a diminished rate of IkappaB kinase-beta (IKK-ß) degradation, persistent receptor interacting protein kinase (RIPK) 1 function and thus prolonged cytoprotective nuclear factor-kappa B (NF-κB) p65 signaling through increased p65 stability and enhanced transcription. Likewise, Bcl-3 overexpression in hepatocytes protected from ALF with massive hepatocyte apoptosis induced by the anti-FAS antibody Jo2. The protection was also linked to IKK-ß stabilization. Overall, our study showed that Bcl-3 rendered hepatocytes more resistant to hepatotoxicity induced by d-GalN/LPS and FAS-ligand. Therefore, Bcl-3 appears to be a critical regulator of the dynamics in ALF through IKK-ß.


Subject(s)
B-Cell Lymphoma 3 Protein , Liver Failure, Acute , Receptors, Death Domain , Animals , Apoptosis/physiology , B-Cell Lymphoma 3 Protein/metabolism , Galactosamine/metabolism , Hepatocytes/metabolism , I-kappa B Kinase/metabolism , Ligands , Lipopolysaccharides , Liver Failure, Acute/genetics , Liver Failure, Acute/metabolism , Mice , NF-kappa B/metabolism , Receptors, Death Domain/metabolism , bcl-X Protein/metabolism
15.
Cell Death Differ ; 29(6): 1176-1186, 2022 06.
Article in English | MEDLINE | ID: mdl-34853447

ABSTRACT

Tumor necrosis factor-α (TNF) is described as a main regulator of cell survival and apoptosis in multiple types of cells, including hepatocytes. Dysregulation in TNF-induced apoptosis is associated with many autoimmune diseases and various liver diseases. Here, we demonstrated a crucial role of Bcl-3, an IκB family member, in regulating TNF-induced hepatic cell death. Specifically, we found that the presence of Bcl-3 promoted TNF-induced cell death in the liver, while Bcl-3 deficiency protected mice against TNF/D-GalN induced hepatoxicity and lethality. Consistently, Bcl-3-depleted hepatic cells exhibited decreased sensitivity to TNF-induced apoptosis when stimulated with TNF/CHX. Mechanistically, the in vitro results showed that Bcl-3 interacted with the deubiquitinase CYLD to synergistically switch the ubiquitination status of RIP1 and facilitate the formation of death-inducing Complex II. This complex further resulted in activation of the caspase cascade to induce apoptosis. By revealing this novel role of Bcl-3 in regulating TNF-induced hepatic cell death, this study provides a potential therapeutic target for liver diseases caused by TNF-related apoptosis.


Subject(s)
B-Cell Lymphoma 3 Protein , GTPase-Activating Proteins , Hepatocytes , Tumor Necrosis Factor-alpha , Animals , Apoptosis/physiology , B-Cell Lymphoma 3 Protein/metabolism , Caspases/metabolism , GTPase-Activating Proteins/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/cytology , Liver/drug effects , Liver/metabolism , Mice , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination
16.
Sci Rep ; 11(1): 5665, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33707466

ABSTRACT

The alkylating agent, temozolomide (TMZ), is the most commonly used chemotherapeutic for the treatment of glioblastoma (GBM). The anti-glioma effect of TMZ involves a complex response that includes G2-M cell cycle arrest and cyclin-dependent kinase 1 (CDK1) activation. While CDK1 phosphorylation is a well-described consequence of TMZ treatment, we find that TMZ also robustly induces CDK1 expression. Analysis of this pathway demonstrates that CDK1 is regulated by NF-κB via a putative κB-site in its proximal promoter. CDK1 was induced in a manner dependent on mature p50 and the atypical inhibitor κB protein, BCL-3. Treatment with TMZ induced binding of NF-κB to the κB-site as assessed by gel shift analysis and chromatin immunoprecipitation. Examination of a CDK1 promoter-reporter demonstrated the functional relevance of the κB-site and underlined the requirement of p50 and BCL-3 for activation. Targeted knockdown of CDK1 or chemical inhibition with the selective CDK1 inhibitor, RO-3306, potentiated the cytotoxic effect of TMZ. These results identify CDK1 as an NF-κB target gene regulated by p50 and BCL-3 and suggest that targeting CDK1 may be a strategy to improve the efficacy of TMZ against GBM.


Subject(s)
Brain Neoplasms/metabolism , CDC2 Protein Kinase/metabolism , Glioblastoma/metabolism , NF-kappa B/metabolism , Temozolomide/pharmacology , B-Cell Lymphoma 3 Protein/metabolism , Base Sequence , Binding Sites , Brain Neoplasms/genetics , Brain Neoplasms/pathology , CDC2 Protein Kinase/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Promoter Regions, Genetic/genetics
17.
Gene ; 769: 145240, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33068678

ABSTRACT

Serotonin (5-HT) is a monoamine and it could regulate cell growth by its receptors working on signaling pathways. 5-HTP is the precursor of 5-HT that help 5-HT synthesis. B cell leukemia/lymphoma 3 (Bcl-3) involved in cell death and proliferation through mitogen activated protein kinase (MAPK) pathway. However, there is little information about the effects of MAPK/Bcl-3 on apoptosis of goat mammary gland epithelial cells (GMECs). The aim of this study is to explore the interaction among 5-HTP, MAPK and Bcl-3 in GMEC apoptosis. In this study, 5-HTP treatment decreased cell apoptosis and promoted phosphorylation of ERK1/2 in GMEC. We also found that the activation and inhibition of ERK1/2 could affect GMEC apoptosis. The Annexin V-FITC/PI staining and western blotting results suggested that 5-HTP decreased GMEC apoptosis through ERK1/2 signaling pathway. And the results of RT-qPCR and western blotting demonstrated that both 5-HTP and ERK1/2 positively regulated Bcl-3 expression. Sum up all the results, we could draw the conclusion that 5-HTP decreased GMEC apoptosis through MAPK/ERK/Bcl-3 pathway.


Subject(s)
5-Hydroxytryptophan/pharmacology , Apoptosis/drug effects , B-Cell Lymphoma 3 Protein/metabolism , Epithelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mammary Glands, Animal/drug effects , Animals , Cell Proliferation/drug effects , Cells, Cultured , Goats , Mammary Glands, Animal/cytology , Phosphorylation , Signal Transduction/drug effects
18.
Signal Transduct Target Ther ; 5(1): 52, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32355204

ABSTRACT

Wnt/ß-catenin signaling plays a critical role in colorectal cancer (CRC) tumorigenesis and the homeostasis of colorectal cancer stem cells (CSCs), but its molecular mechanism remains unclear. B-cell lymphoma 3 (Bcl-3), a member of the IκB family, is overexpressed in CRC and promotes tumorigenicity. Here, we report a novel function of Bcl-3 in maintaining colorectal CSC homeostasis by activating Wnt/ß-catenin signaling. Silencing Bcl-3 suppresses the self-renewal capacity of colorectal CSCs and sensitizes CRC cells to chemotherapeutic drugs through a decrease in Wnt/ß-catenin signaling. Moreover, our data show that Bcl-3 is a crucial component of Wnt/ß-catenin signaling and is essential for ß-catenin transcriptional activity in CRC cells. Interestingly, Wnt3a increases the level and nuclear translocation of Bcl-3, which binds directly to ß-catenin and enhances the acetylation of ß-catenin at lysine 49 (Ac-K49-ß-catenin) and transcriptional activity. Bcl-3 depletion decreases the Ac-K49-ß-catenin level by increasing the level of histone deacetylase 1 to remove acetyl groups from ß-catenin, thus interrupting Wnt/ß-catenin activity. In CRC clinical specimens, Bcl-3 expression negatively correlates with the overall survival of CRC patients. A significantly positive correlation was found between the expression of Bcl-3 and Ac-K49-ß-catenin. Collectively, our data reveal that Bcl-3 plays a crucial role in CRC chemoresistance and colorectal CSC maintenance via its modulation of the Ac-K49-ß-catenin, which serves as a promising therapeutic target for CRC.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Colorectal Neoplasms/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Acetylation , B-Cell Lymphoma 3 Protein/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Humans , Male , Survival Rate , beta Catenin/genetics
19.
Cell Cycle ; 19(9): 1036-1047, 2020 05.
Article in English | MEDLINE | ID: mdl-32240036

ABSTRACT

Previous studies show that Long non-coding RNAs (LncRNAs) are involved in the regulation of various human diseases. This study aimed to reveal how LncRNA CRNDE regulated vascular smooth muscle cells (VSMCs) proliferation and apoptosis in abdominal aortic aneurysms (AAA). Here, we found CRNDE was down-regulated in AAA tissues and AngII-stimulated VSMCs. The overexpression of CRNDE promoted VSMCs proliferation and inhibited cell apoptosis. The interaction between CRNDE and Bcl-3 or Bcl-3 and Smad3 was verified. The interference with Bcl-3 or CRNDE reduced Smad3 stability or promoted Smad3 ubiquitination. After pcDNA-CRNDE or pcDNA-CRNDE+si-Bcl-3 was transfected into VSMCs and stimulated with AngII, CRNDE affected VSMCs proliferation and apoptosis via regulating Smad3 via Bcl-3. Vivo experiments showed the overexpression of CRNDE repressed AAA growth. Therefore, we concluded that CRNDE was down-regulated in AAA tissues and AngII-stimulated VSMCs. Furthermore, the overexpression of CRNDE promoted VSMCs proliferation and repressed cell apoptosis in AAA by up-regulating Smad3 via Bcl-3.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Apoptosis/genetics , B-Cell Lymphoma 3 Protein/metabolism , Cell Proliferation/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Smad3 Protein/metabolism , Animals , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , B-Cell Lymphoma 3 Protein/genetics , Cells, Cultured , Disease Models, Animal , Down-Regulation , Genetic Vectors/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , RNA, Long Noncoding/genetics , Transfection , Up-Regulation/genetics
20.
Int J Oncol ; 56(5): 1304-1313, 2020 05.
Article in English | MEDLINE | ID: mdl-32319612

ABSTRACT

First discovered as an oncogene in leukaemia, recent reports highlight an emerging role for the proto­oncogene BCL­3 in solid tumours. Importantly, BCL­3 expression is upregulated in >30% of colorectal cancer cases and is reported to be associated with a poor prognosis. However, the mechanism by which BCL­3 regulates tumorigenesis in the large intestine is yet to be fully elucidated. In the present study, it was shown for the first time that knocking down BCL­3 expression suppressed cyclooxygenase­2 (COX­2)/prostaglandin E2 (PGE2) signalling in colorectal cancer cells, a pathway known to drive several of the hallmarks of cancer. RNAi­mediated suppression of BCL­3 expression decreased COX­2 expression in colorectal cancer cells both at the mRNA and protein level. This reduction in COX­2 expression resulted in a significant and functional reduction (30­50%) in the quantity of pro­tumorigenic PGE2 produced by the cancer cells, as shown by enzyme linked immunoassays and medium exchange experiments. In addition, inhibition of BCL­3 expression also significantly suppressed cytokine­induced (TNF­α or IL­1ß) COX­2 expression. Taken together, the results of the present study identified a novel role for BCL­3 in colorectal cancer and suggested that expression of BCL­3 may be a key determinant in the COX­2­meditated response to inflammatory cytokines in colorectal tumour cells. These results suggest that targeting BCL­3 to suppress PGE2 synthesis may represent an alternative or complementary approach to using non­steroidal anti­inflammatory drugs [(NSAIDs), which inhibit cyclooxygenase activity and suppress the conversion of arachidonic acid to prostaglandin], for prevention and/or recurrence in PGE2­driven tumorigenesis.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Colorectal Neoplasms/metabolism , Cyclooxygenase 2/metabolism , Up-Regulation , B-Cell Lymphoma 3 Protein/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Interleukin-1beta/pharmacology , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL