Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25.546
Filtrar
1.
Nat Commun ; 15(1): 5515, 2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38951495

RESUMO

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.


Assuntos
Herpesvirus Humano 8 , Proteínas Imediatamente Precoces , Transativadores , Ubiquitinação , Replicação Viral , Herpesvirus Humano 8/fisiologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/genética , Células HEK293 , Transativadores/metabolismo , Transativadores/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Ativação Viral , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Núcleo Celular/metabolismo
2.
Front Immunol ; 15: 1421012, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38979414

RESUMO

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.


Assuntos
Neovascularização de Coroide , Modelos Animais de Doenças , Macrófagos , Degeneração Macular , Transativadores , Animais , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Degeneração Macular/genética , Degeneração Macular/patologia , Camundongos , Macrófagos/imunologia , Macrófagos/metabolismo , Neovascularização de Coroide/imunologia , Neovascularização de Coroide/genética , Neovascularização de Coroide/metabolismo , Transativadores/genética , Transativadores/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Camundongos Endogâmicos C57BL , Ativação de Macrófagos/genética , Humanos , Perfilação da Expressão Gênica , Masculino
3.
Ren Fail ; 46(2): 2373276, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38967134

RESUMO

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.


Assuntos
Glomerulonefrite , Vírus da Hepatite B , Camundongos Transgênicos , Podócitos , Transativadores , Proteínas Virais Reguladoras e Acessórias , Animais , Podócitos/imunologia , Podócitos/patologia , Podócitos/metabolismo , Camundongos , Transativadores/metabolismo , Transativadores/genética , Glomerulonefrite/imunologia , Glomerulonefrite/patologia , Glomerulonefrite/virologia , Vírus da Hepatite B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Hepatite B/imunologia , Hepatite B/complicações , Humanos , Técnicas de Cocultura , Masculino , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
4.
Nat Commun ; 15(1): 5765, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982067

RESUMO

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.


Assuntos
Linfócitos B , Diferenciação Celular , Degradação do RNAm Mediada por Códon sem Sentido , RNA Helicases , Animais , Linfócitos B/metabolismo , Linfócitos B/citologia , Camundongos , RNA Helicases/metabolismo , RNA Helicases/genética , Camundongos Knockout , Camundongos Endogâmicos C57BL , Transativadores/metabolismo , Transativadores/genética , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/metabolismo , Resposta a Proteínas não Dobradas/genética , Humanos , Cadeias Leves de Imunoglobulina/metabolismo , Cadeias Leves de Imunoglobulina/genética
5.
Elife ; 122024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39046443

RESUMO

The role of processing bodies (P-bodies) in tumorigenesis and tumor progression is not well understood. Here, we showed that the oncogenes YAP/TAZ promote P-body formation in a series of cancer cell lines. Mechanistically, both transcriptional activation of the P-body-related genes SAMD4A, AJUBA, and WTIP and transcriptional suppression of the tumor suppressor gene PNRC1 are involved in enhancing the effects of YAP/TAZ on P-body formation in colorectal cancer (CRC) cells. By reexpression of PNRC1 or knockdown of P-body core genes (DDX6, DCP1A, and LSM14A), we determined that disruption of P-bodies attenuates cell proliferation, cell migration, and tumor growth induced by overexpression of YAP5SA in CRC. Analysis of a pancancer CRISPR screen database (DepMap) revealed co-dependencies between YAP/TEAD and the P-body core genes and correlations between the mRNA levels of SAMD4A, AJUBA, WTIP, PNRC1, and YAP target genes. Our study suggests that the P-body is a new downstream effector of YAP/TAZ, which implies that reexpression of PNRC1 or disruption of P-bodies is a potential therapeutic strategy for tumors with active YAP.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Carcinogênese , Transativadores , Fatores de Transcrição , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP , Humanos , Proteínas de Sinalização YAP/metabolismo , Proteínas de Sinalização YAP/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional/metabolismo , Transativadores/metabolismo , Transativadores/genética , Animais , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Camundongos , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular , Proteínas com Domínio LIM
6.
Nat Commun ; 15(1): 5809, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38987584

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proliferação de Células , Via de Sinalização Hippo , Papillomavirus Humano 18 , Infecções por Papillomavirus , Proteínas Serina-Treonina Quinases , Transdução de Sinais , Fatores de Transcrição , Neoplasias do Colo do Útero , Proteínas de Sinalização YAP , Humanos , Feminino , Neoplasias do Colo do Útero/virologia , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Linhagem Celular Tumoral , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Sinalização YAP/metabolismo , Infecções por Papillomavirus/virologia , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/patologia , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional/metabolismo , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Transativadores/metabolismo , Transativadores/genética , Carcinogênese/genética
7.
ISME J ; 18(1)2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38959853

RESUMO

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.


Assuntos
Proteínas de Bactérias , Lysobacter , Pseudomonas , Sistemas de Secreção Tipo IV , Pseudomonas/genética , Pseudomonas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Lysobacter/genética , Lysobacter/metabolismo , Sistemas de Secreção Tipo IV/genética , Sistemas de Secreção Tipo IV/metabolismo , Regulação Bacteriana da Expressão Gênica , Oligopeptídeos/metabolismo , Oligopeptídeos/genética , Transativadores/genética , Transativadores/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fator sigma/genética , Fator sigma/metabolismo
8.
Nat Commun ; 15(1): 5894, 2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-39003281

RESUMO

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.


Assuntos
Diferenciação Celular , Proteínas de Homeodomínio , Ilhotas Pancreáticas , Morfogênese , Células-Tronco Pluripotentes , Esferoides Celulares , Transativadores , Humanos , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Transativadores/metabolismo , Transativadores/genética , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais , Receptores da Família Eph/metabolismo , Receptores da Família Eph/genética
9.
Front Immunol ; 15: 1426620, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39035010

RESUMO

The nucleotide-binding and oligomerization domain-like receptors (NLRs) NLR family CARD domain-containing protein 5 (NLRC5) and Class II Major Histocompatibility Complex Transactivator (CIITA) are transcriptional regulators of major histocompatibility complex (MHC) class I and class II genes, respectively. MHC molecules are central players in our immune system, allowing the detection of hazardous 'non-self' antigens and, thus, the recognition and elimination of infected or transformed cells from the organism. Recently, CIITA and NLRC5 have emerged as regulators of selected genes of the butyrophilin (BTN) family that interestingly are located in the extended MHC locus. BTNs are transmembrane proteins exhibiting structural similarities to B7 family co-modulatory molecules. The family member BTN2A2, which indeed contributes to the control of T cell activation, was found to be transcriptionally regulated by CIITA. NLRC5 emerged instead as an important regulator of the BTN3A1, BTN3A2, and BTN3A3 genes. Together with BTN2A1, BTN3As regulate non-conventional Vγ9Vδ2 T cell responses triggered by selected metabolites of microbial origin or accumulating in hematologic cancer cells. Even if endogenous metabolites conform to the canonical definition of 'self', metabolically abnormal cells can represent a danger for the organism and should be recognized and controlled by immune system cells. Collectively, new data on the role of NLRC5 in the expression of BTN3As link the mechanisms regulating canonical 'non-self' presentation and those marking cells with abnormal metabolic configurations for immune recognition, an evolutionary parallel that we discuss in this perspective review.


Assuntos
Butirofilinas , Peptídeos e Proteínas de Sinalização Intracelular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Butirofilinas/metabolismo , Butirofilinas/genética , Butirofilinas/imunologia , Animais , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transativadores/genética , Transativadores/metabolismo , Regulação da Expressão Gênica , Ativação Linfocitária/imunologia , Antígenos CD
10.
Nat Commun ; 15(1): 5514, 2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38951492

RESUMO

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.


Assuntos
HIV-1 , Imunidade Inata , Macrófagos , Proteínas Proto-Oncogênicas , Transativadores , Produtos do Gene vpr do Vírus da Imunodeficiência Humana , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , HIV-1/fisiologia , HIV-1/imunologia , Transativadores/metabolismo , Transativadores/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , Infecções por HIV/genética , Células HEK293 , Vírion/metabolismo , Proteínas Serina-Treonina Quinases
11.
Nat Commun ; 15(1): 5746, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982056

RESUMO

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.


Assuntos
Candida albicans , Candidíase , Coinfecção , Proteínas Fúngicas , Infecções Estafilocócicas , Staphylococcus aureus , Candida albicans/metabolismo , Candida albicans/patogenicidade , Candida albicans/genética , Animais , Coinfecção/microbiologia , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/metabolismo , Staphylococcus aureus/genética , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/metabolismo , Candidíase/microbiologia , Camundongos , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Infecções Intra-Abdominais/microbiologia , Feminino , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Percepção de Quorum/genética , Virulência , Regulação Fúngica da Expressão Gênica , Modelos Animais de Doenças , Transativadores/metabolismo , Transativadores/genética
12.
Adv Exp Med Biol ; 1459: 53-77, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39017839

RESUMO

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.


Assuntos
Imunidade Adaptativa , Linfócitos B , Linfócitos T , Transativadores , Animais , Humanos , Imunidade Adaptativa/genética , Linfócitos B/imunologia , Linfócitos B/metabolismo , Transativadores/genética , Transativadores/metabolismo , Transativadores/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Camundongos
13.
Sci Rep ; 14(1): 16181, 2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-39003361

RESUMO

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.


Assuntos
Proteínas de Bactérias , Pseudomonas aeruginosa , Percepção de Quorum , Análise de Célula Única , Transativadores , Pseudomonas aeruginosa/fisiologia , Pseudomonas aeruginosa/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Transativadores/metabolismo , Transativadores/genética , Regulação Bacteriana da Expressão Gênica , Transdução de Sinais , Cinética
14.
Sci Signal ; 17(845): eadg4124, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39012937

RESUMO

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.


Assuntos
Herpesvirus Humano 8 , Canal de Potássio Kv1.3 , Fatores de Transcrição NFATC , Replicação Viral , Herpesvirus Humano 8/fisiologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição NFATC/genética , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/genética , Transativadores/metabolismo , Transativadores/genética , Linfócitos B/virologia , Linfócitos B/metabolismo , Cálcio/metabolismo , Sarcoma de Kaposi/virologia , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/genética
15.
Adv Exp Med Biol ; 1459: 143-156, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39017843

RESUMO

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.


Assuntos
Linfócitos B , Linhagem da Célula , Transativadores , Animais , Humanos , Transativadores/genética , Transativadores/metabolismo , Linfócitos B/metabolismo , Linhagem da Célula/genética , Células-Tronco Hematopoéticas/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética
16.
Sci Rep ; 14(1): 13321, 2024 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858497

RESUMO

Myocardin-related transcription factors (MRTFs: myocardin/MYOCD, MRTF-A/MRTFA, and MRTF-B/MRTFB) suppress production of pro-inflammatory cytokines and chemokines in human smooth muscle cells (SMCs) through sequestration of RelA in the NF-κB complex, but additional mechanisms are likely involved. The cGAS-STING pathway is activated by double-stranded DNA in the cytosolic compartment and acts through TANK-binding kinase 1 (TBK1) to spark inflammation. The present study tested if MRTFs suppress inflammation also by targeting cGAS-STING signaling. Interrogation of a transcriptomic dataset where myocardin was overexpressed using a panel of 56 cGAS-STING cytokines showed the panel to be repressed. Moreover, MYOCD, MRTFA, and SRF associated negatively with the panel in human arteries. RT-qPCR in human bronchial SMCs showed that all MRTFs reduced pro-inflammatory cytokines on the panel. MRTFs diminished phosphorylation of TBK1, while STING phosphorylation was marginally affected. The TBK1 inhibitor amlexanox, but not the STING inhibitor H-151, reduced the anti-inflammatory effect of MRTF-A. Co-immunoprecipitation and proximity ligation assays supported binding between MRTF-A and TBK1 in SMCs. MRTFs thus appear to suppress cellular inflammation in part by acting on the kinase TBK1. This may defend SMCs against pro-inflammatory insults in disease.


Assuntos
Inflamação , Miócitos de Músculo Liso , Proteínas Nucleares , Proteínas Serina-Treonina Quinases , Transativadores , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Miócitos de Músculo Liso/metabolismo , Transativadores/metabolismo , Transativadores/genética , Inflamação/metabolismo , Inflamação/patologia , Transdução de Sinais , Citocinas/metabolismo , Fosforilação , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Células Cultivadas
17.
BMC Microbiol ; 24(1): 207, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858621

RESUMO

BACKGROUND: Quorum sensing (QS) is a cell density-based intercellular communication system that controls virulence gene expression and biofilm formation. In Pseudomonas aeruginosa (P. aeruginosa), the LasR system sits at the top of the QS hierarchy and coordinates the expression of a series of important traits. However, the role of lasR in phage infection remains unclear. This study aims to investigate the role of lasR QS in phage infection. METHODS: The P. aeruginosa phage was isolated from sewage, and its biological characteristics and whole genome were analyzed. The adsorption receptor was identified via a phage adsorption assay. Following lasR gene knockout, the adsorption rate and bactericidal activity of phage were analyzed. Finally, real-time quantitative polymerase chain reaction (RT-qPCR) was conducted to explore how lasR promoting phage infection. RESULTS: The lytic phage vB_Pae_PLY was isolated and lipopolysaccharide (LPS) was identified as its adsorption receptor. The adsorption rate and bactericidal activity of vB_Pae_PLY were reduced after lasR knockout. RT-qPCR results showed that the expression of galU, a key gene involved in LPS synthesis, was down-regulated, and several genes related to type IV pili (T4P) were also down-regulated in the lasR mutant PaΔlasR. CONCLUSIONS: The study showed that QS lasR may promote phage vB_Pae_PLY infection by involving in the synthesis of LPS and T4P. This study provides an example of QS in promoting phage infection and deepens the understanding of phage-bacteria interactions.


Assuntos
Proteínas de Bactérias , Pseudomonas aeruginosa , Percepção de Quorum , Transativadores , Pseudomonas aeruginosa/virologia , Pseudomonas aeruginosa/genética , Percepção de Quorum/genética , Transativadores/genética , Transativadores/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fagos de Pseudomonas/genética , Fagos de Pseudomonas/fisiologia , Esgotos/virologia , Esgotos/microbiologia , Regulação Bacteriana da Expressão Gênica , Lipopolissacarídeos/metabolismo , Técnicas de Inativação de Genes
18.
Arch Pharm Res ; 47(6): 558-570, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38874747

RESUMO

Metabolic dysfunction-associated steatotic liver disease (MASLD) is becoming an increasingly pressing global health challenge, with increasing mortality rates showing an upward trend. Two million deaths occur annually from cirrhosis and liver cancer together each year. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key effectors of the Hippo signaling pathway, critically regulate tissue homeostasis and disease progression in the liver. While initial studies have shown that YAP expression is normally restricted to cholangiocytes in healthy livers, the activation of YAP/TAZ is observed in other hepatic cells during chronic liver disease. The disease-driven dysregulation of YAP/TAZ appears to be a critical element in the MASLD progression, contributing to hepatocyte dysfunction, inflammation, and fibrosis. In this study, we focused on the complex roles of YAP/TAZ in MASLD and explored how the YAP/TAZ dysregulation of YAP/TAZ drives steatosis, inflammation, fibrosis, and cirrhosis. Finally, the cell-type-specific functions of YAP/TAZ in different types of hepatic cells, such as hepatocytes, hepatic stellate cells, hepatic macrophages, and biliary epithelial cells are discussed, highlighting the multifaceted impact of YAP/TAZ on liver physiology and pathology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Progressão da Doença , Fatores de Transcrição , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP , Humanos , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional/metabolismo , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Transativadores/metabolismo , Transdução de Sinais
19.
Front Biosci (Schol Ed) ; 16(2): 10, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38939973

RESUMO

The ETS transcription factor PU.1 plays an essential role in blood cell development. Its precise expression pattern is governed by cis-regulatory elements (CRE) acting at the chromatin level. CREs mediate the fine-tuning of graded levels of PU.1, deviations of which can cause acute myeloid leukemia. In this review, we perform an in-depth analysis of the regulation of PU.1 expression in normal and malignant hematopoiesis. We elaborate on the role of trans-acting factors and the biomolecular interplays in mediating local chromatin dynamics. Moreover, we discuss the current understanding of CRE bifunctionality exhibiting enhancer or silencer activities in different blood cell lineages and future directions toward gene-specific chromatin-targeted therapeutic development.


Assuntos
Hematopoese , Proteínas Proto-Oncogênicas , Transativadores , Humanos , Hematopoese/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/genética , Transativadores/metabolismo , Linhagem da Célula , Animais , Transcrição Gênica , Regulação da Expressão Gênica , Leucemia Mieloide Aguda/genética , Cromatina/metabolismo , Cromatina/genética
20.
Sci Rep ; 14(1): 13608, 2024 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-38871849

RESUMO

Transplantation of stem cell-derived ß-cells is a promising therapeutic advancement in the treatment of type 1 diabetes mellitus. A current limitation of this approach is the long differentiation timeline that generates a heterogeneous population of pancreatic endocrine cells. To address this limitation, an inducible lentiviral overexpression system of mature ß-cell markers was introduced into human induced-pluripotent stem cells (hiPSCs). Following the selection of the successfully transduced hiPSCs, the cells were treated with doxycycline in the pancreatic progenitor induction medium to support their transition toward the pancreatic lineage. Cells cultured with doxycycline presented the markers of interest, NGN3, PDX1, and MAFA, after five days of culture, and glucose-stimulated insulin secretion assays demonstrated that the cells were glucose-responsive in a monolayer culture. When cultured as a spheroid, the markers of interest and insulin secretion in a static glucose-stimulated insulin secretion assay were maintained; however, insulin secretion upon consecutive glucose challenges was limited. Comparison to human fetal and adult donor tissues identified that although the hiPSC-derived spheroids present similar markers to adult insulin-producing cells, they are functionally representative of fetal development. Together, these results suggest that with optimization of the temporal expression of these markers, forward programming of hiPSCs towards insulin-producing cells could be a possible alternative for islet transplantation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular , Proteínas de Homeodomínio , Células-Tronco Pluripotentes Induzidas , Células Secretoras de Insulina , Fatores de Transcrição Maf Maior , Proteínas do Tecido Nervoso , Transativadores , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citologia , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Transativadores/metabolismo , Transativadores/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição Maf Maior/metabolismo , Fatores de Transcrição Maf Maior/genética , Insulina/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Secreção de Insulina/efeitos dos fármacos , Células Cultivadas , Doxiciclina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA