Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50.352
Filter
1.
Sci Rep ; 14(1): 16181, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003361

ABSTRACT

Quorum sensing (QS) is a communication form between bacteria via small signal molecules that enables global gene regulation as a function of cell density. We applied a microfluidic mother machine to study the kinetics of the QS response of Pseudomonas aeruginosa bacteria to additions and withdrawals of signal molecules. We traced the fast buildup and the subsequent considerably slower decay of a population-level and single-cell-level QS response. We applied a mathematical model to explain the results quantitatively. We found significant heterogeneity in QS on the single-cell level, which may result from variations in quorum-controlled gene expression and protein degradation. Heterogeneity correlates with cell lineage history, too. We used single-cell data to define and quantitatively characterize the population-level quorum state. We found that the population-level QS response is well-defined. The buildup of the quorum is fast upon signal molecule addition. At the same time, its decay is much slower following signal withdrawal, and the quorum may be maintained for several hours in the absence of the signal. Furthermore, the quorum sensing response of the population was largely repeatable in subsequent pulses of signal molecules.


Subject(s)
Bacterial Proteins , Pseudomonas aeruginosa , Quorum Sensing , Single-Cell Analysis , Trans-Activators , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Gene Expression Regulation, Bacterial , Signal Transduction , Kinetics
2.
Sci Signal ; 17(845): eadg4124, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012937

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic herpesvirus that is linked directly to the development of Kaposi's sarcoma. KSHV establishes a latent infection in B cells, which can be reactivated to initiate lytic replication, producing infectious virions. Using pharmacological and genetic silencing approaches, we showed that the voltage-gated K+ channel Kv1.3 in B cells enhanced KSHV lytic replication. The KSHV replication and transcription activator (RTA) protein increased the abundance of Kv1.3 and led to enhanced K+ channel activity and hyperpolarization of the B cell membrane. Enhanced Kv1.3 activity promoted intracellular Ca2+ influx, leading to the Ca2+-driven nuclear localization of KSHV RTA and host nuclear factor of activated T cells (NFAT) proteins and subsequently increased the expression of NFAT1 target genes. KSHV lytic replication and infectious virion production were inhibited by Kv1.3 blockers or silencing. These findings highlight Kv1.3 as a druggable host factor that is key to the successful completion of KSHV lytic replication.


Subject(s)
Herpesvirus 8, Human , Kv1.3 Potassium Channel , NFATC Transcription Factors , Virus Replication , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , Kv1.3 Potassium Channel/metabolism , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/antagonists & inhibitors , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , B-Lymphocytes/virology , B-Lymphocytes/metabolism , Calcium/metabolism , Sarcoma, Kaposi/virology , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/genetics
3.
Adv Exp Med Biol ; 1459: 143-156, 2024.
Article in English | MEDLINE | ID: mdl-39017843

ABSTRACT

The development of highly specialized blood cells from hematopoietic stem cells (HSCs) in the bone marrow (BM) is dependent upon a stringently orchestrated network of stage- and lineage-restricted transcription factors (TFs). Thus, the same stem cell can give rise to various types of differentiated blood cells. One of the key regulators of B-lymphocyte development is early B-cell factor 1 (EBF1). This TF belongs to a small, but evolutionary conserved, family of proteins that harbor a Zn-coordinating motif and an IPT/TIG (immunoglobulin-like, plexins, transcription factors/transcription factor immunoglobulin) domain, creating a unique DNA-binding domain (DBD). EBF proteins play critical roles in diverse developmental processes, including body segmentation in the Drosophila melanogaster embryo, and retina formation in mice. While several EBF family members are expressed in neuronal cells, adipocytes, and BM stroma cells, only B-lymphoid cells express EBF1. In the absence of EBF1, hematopoietic progenitor cells (HPCs) fail to activate the B-lineage program. This has been attributed to the ability of EBF1 to act as a pioneering factor with the ability to remodel chromatin, thereby creating a B-lymphoid-specific epigenetic landscape. Conditional inactivation of the Ebf1 gene in B-lineage cells has revealed additional functions of this protein in relation to the control of proliferation and apoptosis. This may explain why EBF1 is frequently targeted by mutations in human leukemia cases. This chapter provides an overview of the biochemical and functional properties of the EBF family proteins, with a focus on the roles of EBF1 in normal and malignant B-lymphocyte development.


Subject(s)
B-Lymphocytes , Cell Lineage , Trans-Activators , Animals , Humans , Trans-Activators/genetics , Trans-Activators/metabolism , B-Lymphocytes/metabolism , Cell Lineage/genetics , Hematopoietic Stem Cells/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics
4.
ISME J ; 18(1)2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38959853

ABSTRACT

Effector proteins secreted by bacteria that infect mammalian and plant cells often subdue eukaryotic host cell defenses by simultaneously affecting multiple targets. However, instances when a bacterial effector injected in the competing bacteria sabotage more than a single target have not been reported. Here, we demonstrate that the effector protein, LtaE, translocated by the type IV secretion system from the soil bacterium Lysobacter enzymogenes into the competing bacterium, Pseudomonas protegens, affects several targets, thus disabling the antibacterial defenses of the competitor. One LtaE target is the transcription factor, LuxR1, that regulates biosynthesis of the antimicrobial compound, orfamide A. Another target is the sigma factor, PvdS, required for biosynthesis of another antimicrobial compound, pyoverdine. Deletion of the genes involved in orfamide A and pyoverdine biosynthesis disabled the antibacterial activity of P. protegens, whereas expression of LtaE in P. protegens resulted in the near-complete loss of the antibacterial activity against L. enzymogenes. Mechanistically, LtaE inhibits the assembly of the RNA polymerase complexes with each of these proteins. The ability of LtaE to bind to LuxR1 and PvdS homologs from several Pseudomonas species suggests that it can sabotage defenses of various competitors present in the soil or on plant matter. Our study thus reveals that the multi-target effectors have evolved to subdue cell defenses not only in eukaryotic hosts but also in bacterial competitors.


Subject(s)
Bacterial Proteins , Lysobacter , Pseudomonas , Type IV Secretion Systems , Pseudomonas/genetics , Pseudomonas/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Lysobacter/genetics , Lysobacter/metabolism , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , Gene Expression Regulation, Bacterial , Oligopeptides/metabolism , Oligopeptides/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sigma Factor/genetics , Sigma Factor/metabolism
5.
Nat Commun ; 15(1): 5746, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982056

ABSTRACT

Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) demonstrates that synergistic lethality is driven by Candida-induced upregulation of functional S. aureus α-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken revealing that zcf13Δ/Δ fails to drive augmented α-toxin or lethal synergism during co-infection. A combination of transcriptional and phenotypic profiling approaches shows that ZCF13 regulates genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments reveal that ribose inhibits the staphylococcal agr quorum sensing system and concomitantly represses toxicity. Unlike wild-type C. albicans, zcf13Δ/Δ did not effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13Δ/Δ mutant fully restores pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.


Subject(s)
Candida albicans , Candidiasis , Coinfection , Fungal Proteins , Staphylococcal Infections , Staphylococcus aureus , Candida albicans/metabolism , Candida albicans/pathogenicity , Candida albicans/genetics , Animals , Coinfection/microbiology , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/metabolism , Staphylococcus aureus/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Candidiasis/microbiology , Mice , Fungal Proteins/metabolism , Fungal Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Intraabdominal Infections/microbiology , Female , Transcription Factors/metabolism , Transcription Factors/genetics , Quorum Sensing/genetics , Virulence , Gene Expression Regulation, Fungal , Disease Models, Animal , Trans-Activators/metabolism , Trans-Activators/genetics
6.
Nat Commun ; 15(1): 5693, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972954

ABSTRACT

Leukemias with ambiguous lineage comprise several loosely defined entities, often without a clear mechanistic basis. Here, we extensively profile the epigenome and transcriptome of a subgroup of such leukemias with CpG Island Methylator Phenotype. These leukemias exhibit comparable hybrid myeloid/lymphoid epigenetic landscapes, yet heterogeneous genetic alterations, suggesting they are defined by their shared epigenetic profile rather than common genetic lesions. Gene expression enrichment reveals similarity with early T-cell precursor acute lymphoblastic leukemia and a lymphoid progenitor cell of origin. In line with this, integration of differential DNA methylation and gene expression shows widespread silencing of myeloid transcription factors. Moreover, binding sites for hematopoietic transcription factors, including CEBPA, SPI1 and LEF1, are uniquely inaccessible in these leukemias. Hypermethylation also results in loss of CTCF binding, accompanied by changes in chromatin interactions involving key transcription factors. In conclusion, epigenetic dysregulation, and not genetic lesions, explains the mixed phenotype of this group of leukemias with ambiguous lineage. The data collected here constitute a useful and comprehensive epigenomic reference for subsequent studies of acute myeloid leukemias, T-cell acute lymphoblastic leukemias and mixed-phenotype leukemias.


Subject(s)
CpG Islands , DNA Methylation , Epigenesis, Genetic , Gene Regulatory Networks , Humans , DNA Methylation/genetics , CpG Islands/genetics , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Gene Expression Regulation, Leukemic , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin/metabolism , Chromatin/genetics , Male , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Female , Hematopoiesis/genetics , Child , Transcriptome , Proto-Oncogene Proteins , Trans-Activators
7.
Front Immunol ; 15: 1421012, 2024.
Article in English | MEDLINE | ID: mdl-38979414

ABSTRACT

Objective: This study revealed a core regulator and common upstream mechanisms for the multifaceted pathological processes of age-related macular degeneration (AMD) and provided proof-of-concept for this new therapeutic target. Methods: Comprehensive gene expression analysis was performed using RNA sequencing of eye cup from old mice as well as laser-induced choroidal neovascularization (CNV) mouse model. Through integrative analysis and protein-protein interaction (PPI) analysis, common pathways and key transcription factor was identified simultaneously engaged in age-related retinal degeneration and CNV, the two typical pathological process of AMD. Subsequently, the expression changes of Spi1, the key regulator, as well as the alternation of the downstream mechanisms were validated in both models through qRT-PCR, Elisa, flow cytometry and immunofluorescence. Further, we assessed the impact of Spi1 knockdown in vitro and in vivo using gene intervention vectors carried by adeno-associated virus or lentivirus to test its potential as a therapeutic target. Results: Compared to corresponding controls, we found 1,939 and 1,319 genes differentially expressed in eye cups of old and CNV mice respectively. The integrative analysis identified a total of 275 overlapping DEGs, of which 150 genes were co-upregulated. PPI analysis verified a central transcription factor, SPI1. The significant upregulation of Spi1 expression was then validated in both models, accompanied by macrophage polarization towards the M1 phenotype. Finally, SPI1 suppression significantly inhibited M1 polarization of BMDMs and attenuated neovascularization in CNV mice. Conclusion: This study demonstrates that SPI1 exerts a pivotal role in AMD by regulation of macrophage polarization and innate immune response, offering promise as an innovative target for treating AMD.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Macrophages , Macular Degeneration , Trans-Activators , Animals , Macular Degeneration/immunology , Macular Degeneration/metabolism , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Macrophages/immunology , Macrophages/metabolism , Choroidal Neovascularization/immunology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Mice, Inbred C57BL , Macrophage Activation/genetics , Humans , Gene Expression Profiling , Male
8.
BMC Oral Health ; 24(1): 782, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997708

ABSTRACT

BACKGROUND: Originating from odontogenic tissue, Odontogenic cysts are pathological cavities lined with epithelial cells and surrounded by fibrous connective tissue. This study investigated expression of CITED1 protein in different types of odontogenic cysts. MATERIAL AND METHOD: 40 keratocysts, 40 radicular cysts, and 40 dentigerous cysts were excised and processed for routine paraffin wax embedding protocol. Macroscopic and panoramic radiographies images were used for diagnosis. Demographical properties and dental parameters were recorded. Cystic tissues were stained with hematoxylin-eosin dye and CITED1 antibody. Semi-quantitative analysis was performed for immune staining. The protein-protein interaction network, hub gene detection and KEGG analysis were conducted using Cytoscape software. RESULT: Odontogenic keratocysts was imaged with 6-8 layered epithelial cells and fibrous cyst walls with inflammatory cells. Radicular cysts had stratified squamous epithelium with varying thickness, ciliated cells, and Rushton hyaline bodies. Dentigerous cysts presented hyperplastic non-keratinized epithelium, fibrous tissue, rete ridges, and inflammatory cells. CITED1 immunoexpression was highest in odontogenic keratocysts, followed by radicular cysts, and lowest in dentigerous cysts. Nuclear and cytoplasmic CITED1 expression was significantly elevated in odontogenic keratocysts compared to radicular and dentigerous cysts. The top five targets of CITED1 were identified, primarily showing enrichment in hormone and cancer related pathways. CONCLUSIONS: Positive CITED1 expression in all three types of odontogenic cysts suggest a potential role for CITED1 in the pathogenesis of odontogenic cysts, particularly in keratocysts. Further investigations are needed to elucidate the exact mechanisms underlying the differential expression of CITED1 and its implications for the development and progression of odontogenic cysts.


Subject(s)
Odontogenic Cysts , Humans , Odontogenic Cysts/pathology , Odontogenic Cysts/metabolism , Male , Trans-Activators , Female , Adult , Dentigerous Cyst/pathology , Dentigerous Cyst/diagnostic imaging , Radicular Cyst/pathology , Radicular Cyst/diagnostic imaging , Middle Aged , Adolescent
9.
Nat Commun ; 15(1): 5894, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003281

ABSTRACT

Remarkable advances in protocol development have been achieved to manufacture insulin-secreting islets from human pluripotent stem cells (hPSCs). Distinct from current approaches, we devised a tunable strategy to generate islet spheroids enriched for major islet cell types by incorporating PDX1+ cell budding morphogenesis into staged differentiation. In this process that appears to mimic normal islet morphogenesis, the differentiating islet spheroids organize with endocrine cells that are intermingled or arranged in a core-mantle architecture, accompanied with functional heterogeneity. Through in vitro modelling of human pancreas development, we illustrate the importance of PDX1 and the requirement for EphB3/4 signaling in eliciting cell budding morphogenesis. Using this new approach, we model Mitchell-Riley syndrome with RFX6 knockout hPSCs illustrating unexpected morphogenesis defects in the differentiation towards islet cells. The tunable differentiation system and stem cell-derived islet models described in this work may facilitate addressing fundamental questions in islet biology and probing human pancreas diseases.


Subject(s)
Cell Differentiation , Homeodomain Proteins , Islets of Langerhans , Morphogenesis , Pluripotent Stem Cells , Spheroids, Cellular , Trans-Activators , Humans , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Receptors, Eph Family/metabolism , Receptors, Eph Family/genetics
10.
J Med Virol ; 96(7): e29805, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39011773

ABSTRACT

Heterogeneous nuclear protein U (HNRNPU) plays a pivotal role in innate immunity by facilitating chromatin opening to activate immune genes during host defense against viral infection. However, the mechanism by which HNRNPU is involved in Hepatitis B virus (HBV) transcription regulation through mediating antiviral immunity remains unknown. Our study revealed a significant decrease in HNRNPU levels during HBV transcription, which depends on HBx-DDB1-mediated degradation. Overexpression of HNRNPU suppressed HBV transcription, while its knockdown effectively promoted viral transcription, indicating HNRNPU as a novel host restriction factor for HBV transcription. Mechanistically, HNRNPU inhibits HBV transcription by activating innate immunity through primarily the positive regulation of the interferon-stimulating factor 2'-5'-oligoadenylate synthetase 3, which mediates an ribonuclease L-dependent mechanism to enhance innate immune responses. This study offers new insights into the host immune regulation of HBV transcription and proposes potential targets for therapeutic intervention against HBV infection.


Subject(s)
2',5'-Oligoadenylate Synthetase , Hepatitis B virus , Immunity, Innate , Transcription, Genetic , Humans , Hepatitis B virus/immunology , Hepatitis B virus/genetics , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Hep G2 Cells , Hepatitis B/immunology , Hepatitis B/virology , Hepatitis B/genetics , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/immunology , Trans-Activators
11.
Nat Commun ; 15(1): 5809, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987584

ABSTRACT

Human papillomaviruses (HPVs) cause most cervical cancers and an increasing number of anogenital and oral carcinomas, with most cases caused by HPV16 or HPV18. HPV hijacks host signalling pathways to promote carcinogenesis. Understanding these interactions could permit identification of much-needed therapeutics for HPV-driven malignancies. The Hippo signalling pathway is important in HPV+ cancers, with the downstream effector YAP playing a pro-oncogenic role. In contrast, the significance of its paralogue TAZ remains largely uncharacterised in these cancers. We demonstrate that TAZ is dysregulated in a HPV-type dependent manner by a distinct mechanism to that of YAP and controls proliferation via alternative cellular targets. Analysis of cervical cancer cell lines and patient biopsies revealed that TAZ expression was only significantly increased in HPV18+ and HPV18-like cells and TAZ knockdown reduced proliferation, migration and invasion only in HPV18+ cells. RNA-sequencing of HPV18+ cervical cells revealed that YAP and TAZ have distinct targets, suggesting they promote carcinogenesis by different mechanisms. Thus, in HPV18+ cancers, YAP and TAZ play non-redundant roles. This analysis identified TOGARAM2 as a previously uncharacterised TAZ target and demonstrates its role as a key effector of TAZ-mediated proliferation, migration and invasion in HPV18+ cancers.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Proliferation , Hippo Signaling Pathway , Human papillomavirus 18 , Papillomavirus Infections , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , Uterine Cervical Neoplasms , YAP-Signaling Proteins , Humans , Female , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , YAP-Signaling Proteins/metabolism , Papillomavirus Infections/virology , Papillomavirus Infections/metabolism , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Human papillomavirus 18/genetics , Human papillomavirus 18/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Carcinogenesis/genetics
12.
Ren Fail ; 46(2): 2373276, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38967134

ABSTRACT

BACKGROUND: Podocytes, as intrinsic renal cells, can also express MHC-II and costimulatory molecules under inflammatory conditions, suggesting that they may act as antigen-presenting cells (APCs) to activate immune cell responses and then lead to immune-mediated renal injury. They are already recognized as main targets in the pathogenic mechanism of hepatitis B virus (HBV)-associated glomerulonephritis (HBV-GN). Previous studies also have indicated that inflammatory cells infiltration and immune-mediated tissue injury are evident in the kidney samples of patients with HBV-GN. However, the role of podocytes immune disorder in the pathogenic mechanism of HBV-GN remains unclear. METHODS: Renal function and inflammatory cells infiltration were measured in HBV transgenic (HBV-Tg) mice. In vitro, podocytes/CD4+ T cells or macrophages co-culture system was established. Then, the expression of HBx, CD4, and CD68 was determined by immunohistochemistry, while the expression of MHC-II, CD40, and CD40L was determined by immunofluorescence. Co-stimulatory molecules expression was examined by flow cytometry. The levels of inflammatory factors were detected by ELISA. RESULTS: In vivo, renal function was obviously impaired in HBV-Tg mice. HBx was significantly upregulated and immune cells infiltrated in the glomerulus of HBV-Tg mice. Expression of MHC-II and costimulatory molecule CD40 increased in the podocytes of HBV-Tg mice; CD4+ T cells exhibited increased CD40L expression in glomerulus. In vitro, CD40 expression was markedly elevated in HBx-podocytes. In co-culture systems, HBx-podocytes stimulated CD4+ T cells activation and caused the imbalance between IFN-γ and IL-4. HBx-podocytes also enhanced the adhesion ability of macrophages and induced the release of proinflammatory mediators. CONCLUSION: Taken together, these podocyte-related immune disorder may be involved in the pathogenic mechanism of HBV-GN.


Subject(s)
Glomerulonephritis , Hepatitis B virus , Mice, Transgenic , Podocytes , Trans-Activators , Viral Regulatory and Accessory Proteins , Animals , Podocytes/immunology , Podocytes/pathology , Podocytes/metabolism , Mice , Trans-Activators/metabolism , Trans-Activators/genetics , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Glomerulonephritis/virology , Hepatitis B virus/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Hepatitis B/immunology , Hepatitis B/complications , Humans , Coculture Techniques , Male , Disease Models, Animal , Mice, Inbred C57BL
13.
Nat Commun ; 15(1): 5765, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982067

ABSTRACT

The ATP-dependent RNA helicase UPF1 plays a crucial role in various mRNA degradation pathways, most importantly in nonsense-mediated mRNA decay (NMD). Here, we show that UPF1 is upregulated during the early stages of B cell development and is important for early B cell development in the bone marrow. B-cell-specific Upf1 deletion in mice severely impedes the early to late LPre-B cell transition, in which VH-DHJH recombination occurs at the Igh gene. Furthermore, UPF1 is indispensable for VH-DHJH recombination, without affecting DH-JH recombination. Intriguingly, the genetic pre-arrangement of the Igh gene rescues the differentiation defect in early LPre-B cells under Upf1 deficient conditions. However, differentiation is blocked again following Ig light chain recombination, leading to a failure in development into immature B cells. Notably, UPF1 interacts with and regulates the expression of genes involved in immune responses, cell cycle control, NMD, and the unfolded protein response in B cells. Collectively, our findings underscore the critical roles of UPF1 during the early LPre-B cell stage and beyond, thus orchestrating B cell development.


Subject(s)
B-Lymphocytes , Cell Differentiation , Nonsense Mediated mRNA Decay , RNA Helicases , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/cytology , Mice , RNA Helicases/metabolism , RNA Helicases/genetics , Mice, Knockout , Mice, Inbred C57BL , Trans-Activators/metabolism , Trans-Activators/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Unfolded Protein Response/genetics , Humans , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light Chains/genetics
14.
BMC Med Genomics ; 17(1): 176, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956580

ABSTRACT

BACKGROUND: Intellectual disability (ID) is a neurodevelopmental condition affecting around 2% of children and young adults worldwide, characterized by deficits in intellectual functioning and adaptive behavior. Genetic factors contribute to the development of ID phenotypes, including mutations and structural changes in chromosomes. Pathogenic variants in the HCFC1 gene cause X-linked mental retardation syndrome, also known as Siderius type X-linked mental retardation. The MN1 gene is necessary for palate development, and mutations in this gene result in a genetic condition called CEBALID syndrome. METHODS: Exome sequencing was used to identify the disease-causing variants in two affected families, A and B, from various regions of Pakistan. Affected individuals in these two families presented ID, developmental delay, and behavioral abnormalities. The validation and co-segregation analysis of the filtered variant was carried out using Sanger sequencing. RESULTS: In an X-linked family A, a novel hemizygous missense variant (c.5705G > A; p.Ser1902Asn) in the HCFC1 gene (NM_005334.3) was identified, while in family B exome sequencing revealed a heterozygous nonsense variant (c.3680 G > A; p. Trp1227Ter) in exon-1 of the MN1 gene (NM_032581.4). Sanger sequencing confirmed the segregation of these variants with ID in each family. CONCLUSIONS: The investigation of two Pakistani families revealed pathogenic genetic variants in the HCFC1 and MN1 genes, which cause ID and expand the mutational spectrum of these genes.


Subject(s)
Host Cell Factor C1 , Intellectual Disability , Pedigree , Humans , Pakistan , Male , Intellectual Disability/genetics , Female , Host Cell Factor C1/genetics , Tumor Suppressor Proteins/genetics , Trans-Activators/genetics , Child , Exome Sequencing , Child, Preschool
15.
Nat Commun ; 15(1): 5515, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951495

ABSTRACT

Like many other viruses, KSHV has two life cycle modes: the latent phase and the lytic phase. The RTA protein from KSHV is essential for lytic reactivation, but how this protein's activity is regulated is not fully understood. Here, we report that linear ubiquitination regulates the activity of RTA during KSHV lytic reactivation and de novo infection. Overexpressing OTULIN inhibits KSHV lytic reactivation, whereas knocking down OTULIN or overexpressing HOIP enhances it. Intriguingly, we found that RTA is linearly polyubiquitinated by HOIP at K516 and K518, and these modifications control the RTA's nuclear localization. OTULIN removes linear polyubiquitin chains from cytoplasmic RTA, preventing its nuclear import. The RTA orthologs encoded by the EB and MHV68 viruses are also linearly polyubiquitinated and regulated by OTULIN. Our study establishes that linear polyubiquitination plays a critically regulatory role in herpesvirus infection, adding virus infection to the list of biological processes known to be controlled by linear polyubiquitination.


Subject(s)
Herpesvirus 8, Human , Immediate-Early Proteins , Trans-Activators , Ubiquitination , Virus Replication , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , HEK293 Cells , Trans-Activators/metabolism , Trans-Activators/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Virus Activation , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Cell Nucleus/metabolism
16.
Cancer Med ; 13(14): e7471, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39015025

ABSTRACT

BACKGROUND: ZNF384-fusion (Z-fusion) genes were recently identified in B-cell acute lymphoblastic leukemia (B-ALL) and are frequent in Japanese adult patients. The frequency is about 20% in those with Philadelphia chromosome-negative B-ALL. ZNF384 is a transcription factor and Z-fusion proteins have increased transcriptional activity; however, the detailed mechanisms of leukemogenesis of Z-fusion proteins have yet to be clarified. METHODS: We established three transfectants of cell lines expressing different types of Z-fusion proteins, and analyzed their gene expression profile (GEP) by RNA-seq. We also analyzed the GEP of clinical ALL samples using our previous RNA-seq data of 323 Japanese ALL patients. We selected upregulated genes in both Z-fusion gene-expressing transfectants and Z-fusion gene-positive ALL samples, and investigated the binding of Z-fusion proteins to regulatory regions of the candidate genes by ChIP-qPCR. RESULTS: We selected six commonly upregulated genes. After the investigation by ChIP-qPCR, we finally identified CREB5 and RGS1 as direct and common target genes. RGS1 is an inhibitor of CXCL12-CXCR4 signaling that is required for the homing of hematopoietic progenitor cells to the bone marrow microenvironment and development of B cells. Consistent with this, Z-fusion gene transfectants showed impaired migration toward CXCL12. CONCLUSIONS: We identified CREB5 and RGS1 as direct and common transcriptional targets of Z-fusion proteins. The present results provide novel insight into the aberrant transcriptional regulation by Z-fusion proteins.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Oncogene Proteins, Fusion , RGS Proteins , Humans , RGS Proteins/genetics , RGS Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cell Line, Tumor , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Gene Expression Regulation, Leukemic , Trans-Activators
17.
Nat Commun ; 15(1): 5514, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951492

ABSTRACT

HIV-1 Vpr promotes efficient spread of HIV-1 from macrophages to T cells by transcriptionally downmodulating restriction factors that target HIV-1 Envelope protein (Env). Here we find that Vpr induces broad transcriptomic changes by targeting PU.1, a transcription factor necessary for expression of host innate immune response genes, including those that target Env. Consistent with this, we find silencing PU.1 in infected macrophages lacking Vpr rescues Env. Vpr downmodulates PU.1 through a proteasomal degradation pathway that depends on physical interactions with PU.1 and DCAF1, a component of the Cul4A E3 ubiquitin ligase. The capacity for Vpr to target PU.1 is highly conserved across primate lentiviruses. In addition to impacting infected cells, we find that Vpr suppresses expression of innate immune response genes in uninfected bystander cells, and that virion-associated Vpr can degrade PU.1. Together, we demonstrate Vpr counteracts PU.1 in macrophages to blunt antiviral immune responses and promote viral spread.


Subject(s)
HIV-1 , Immunity, Innate , Macrophages , Proto-Oncogene Proteins , Trans-Activators , vpr Gene Products, Human Immunodeficiency Virus , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , vpr Gene Products, Human Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/genetics , HIV-1/physiology , HIV-1/immunology , Trans-Activators/metabolism , Trans-Activators/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , HIV Infections/immunology , HIV Infections/virology , HIV Infections/genetics , HEK293 Cells , Virion/metabolism , Protein Serine-Threonine Kinases
18.
Subcell Biochem ; 104: 17-31, 2024.
Article in English | MEDLINE | ID: mdl-38963481

ABSTRACT

The copper efflux regulator (CueR) is a classical member of the MerR family of metalloregulators and is common in gram-negative bacteria. Through its C-terminal effector-binding domain, CueR senses cytoplasmic copper ions to regulate the transcription of genes contributing to copper homeostasis, an essential process for survival of all cells. In this chapter, we review the regulatory roles of CueR in the model organism Escherichia coli and the mechanisms for CueR in copper binding, DNA recognition, and interplay with RNA polymerase in regulating transcription. In light of biochemical and structural analyses, we provide molecular details for how CueR represses transcription in the absence of copper ions, how copper ions mediate CueR conformational change to form holo CueR, and how CueR bends and twists promoter DNA to activate transcription. We also characterize the functional domains and key residues involved in these processes. Since CueR is a representative member of the MerR family, elucidating its regulatory mechanisms could help to understand the CueR-like regulators in other organisms and facilitate the understanding of other metalloregulators in the same family.


Subject(s)
Copper , Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Bacterial , Copper/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Transcription, Genetic , Promoter Regions, Genetic , Trans-Activators
19.
Adv Exp Med Biol ; 1459: 53-77, 2024.
Article in English | MEDLINE | ID: mdl-39017839

ABSTRACT

BOB.1/OBF.1 is a transcriptional coactivator involved in octamer-dependent transcription. Thereby, BOB.1/OBF.1 is involved in the transcriptional regulation of genes important for lymphocyte physiology. BOB.1/OBF.1-deficient mice reveal multiple B- and T-cell developmental defects. The most prominent defect of these mice is the complete absence of germinal centers (GCs) resulting in severely impaired T-cell-dependent immune responses. In humans, BOB.1/OBF.1 is associated with several autoimmune and inflammatory diseases but also linked to liquid and solid tumors. Although its role for B-cell development is relatively well understood, its exact role for the GC reaction and T-cell biology has long been unclear. Here, the contribution of BOB.1/OBF.1 for B-cell maturation is summarized, and recent findings regarding its function in GC B- as well as in various T-cell populations are discussed. Finally, a detailed perspective on how BOB.1/OBF.1 contributes to different pathologies is provided.


Subject(s)
Adaptive Immunity , B-Lymphocytes , T-Lymphocytes , Trans-Activators , Animals , Humans , Adaptive Immunity/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Trans-Activators/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Mice
20.
Sci Transl Med ; 16(756): eadi9548, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39018368

ABSTRACT

Immune rejection remains the major obstacle to long-term survival of allogeneic lung transplants. The expression of major histocompatibility complex molecules and minor histocompatibility antigens triggers allogeneic immune responses that can lead to allograft rejection. Transplant outcomes therefore depend on long-term immunosuppression, which is associated with severe side effects. To address this problem, we investigated the effect of genetically engineered transplants with permanently down-regulated swine leukocyte antigen (SLA) expression to prevent rejection in a porcine allogeneic lung transplantation (LTx) model. Minipig donor lungs with unmodified SLA expression (control group, n = 7) or with modified SLA expression (treatment group, n = 7) were used to evaluate the effects of SLA knockdown on allograft survival and on the nature and strength of immune responses after terminating an initial 4-week period of immunosuppression after LTx. Genetic engineering to down-regulate SLA expression was achieved during ex vivo lung perfusion by lentiviral transduction of short hairpin RNAs targeting mRNAs encoding ß2-microglobulin and class II transactivator. Whereas all grafts in the control group were rejected within 3 months, five of seven animals in the treatment group maintained graft survival without immunosuppression during the 2-year monitoring period. Compared with controls, SLA-silenced lung recipients had lower donor-specific antibodies and proinflammatory cytokine concentrations in the serum. Together, these data demonstrate a survival benefit of SLA-down-regulated lung transplants in the absence of immunosuppression.


Subject(s)
Gene Knockdown Techniques , Graft Survival , Histocompatibility Antigens Class I , Immunosuppression Therapy , Lung Transplantation , Animals , Swine , Graft Survival/immunology , Histocompatibility Antigens Class I/metabolism , Graft Rejection/immunology , Swine, Miniature , Histocompatibility Antigens Class II/metabolism , Transplantation, Homologous , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Lung/metabolism , Nuclear Proteins , Trans-Activators
SELECTION OF CITATIONS
SEARCH DETAIL
...