Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 51(19): 10519-10535, 2023 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-37739427

RESUMO

Homologous recombination (HR) requires bidirectional end resection initiated by a nick formed close to a DNA double-strand break (DSB), dysregulation favoring error-prone DNA end-joining pathways. Here we investigate the role of the ATAD5, a PCNA unloading protein, in short-range end resection, long-range resection not being affected by ATAD5 deficiency. Rapid PCNA loading onto DNA at DSB sites depends on the RFC PCNA loader complex and MRE11-RAD50-NBS1 nuclease complexes bound to CtIP. Based on our cytological analyses and on an in vitro system for short-range end resection, we propose that PCNA unloading by ATAD5 is required for the completion of short-range resection. Hampering PCNA unloading also leads to failure to remove the KU70/80 complex from the termini of DSBs hindering DNA repair synthesis and the completion of HR. In line with this model, ATAD5-depleted cells are defective for HR, show increased sensitivity to camptothecin, a drug forming protein-DNA adducts, and an augmented dependency on end-joining pathways. Our study highlights the importance of PCNA regulation at DSB for proper end resection and HR.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , DNA/metabolismo , Reparo do DNA por Junção de Extremidades , Endodesoxirribonucleases/metabolismo , Recombinação Homóloga/genética , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Humanos
2.
NAR Cancer ; 5(3): zcad042, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37554969

RESUMO

Targeting BRCA1- and BRCA2-deficient tumors through synthetic lethality using poly(ADP-ribose) polymerase inhibitors (PARPi) has emerged as a successful strategy for cancer therapy. PARPi monotherapy has shown excellent efficacy and safety profiles in clinical practice but is limited by the need for tumor genome mutations in BRCA or other homologous recombination genes as well as the rapid emergence of resistance. In this study, we identified 2-chloro-N,N-diethylethanamine hydrochloride (CDEAH) as a small molecule that selectively kills PARP1- and xeroderma pigmentosum A-deficient cells. CDEAH is a monofunctional alkylating agent that preferentially alkylates guanine nucleobases, forming DNA adducts that can be removed from DNA by either a PARP1-dependent base excision repair or nucleotide excision repair. Treatment of PARP1-deficient cells leads to the formation of strand breaks, an accumulation of cells in S phase and activation of the DNA damage response. Furthermore, CDEAH selectively inhibits PARP1-deficient xenograft tumor growth compared to isogenic PARP1-proficient tumors. Collectively, we report the discovery of an alkylating agent inducing DNA damage that requires PARP1 activity for repair and acts synergistically with PARPi.

3.
Nucleic Acids Res ; 51(11): 5584-5602, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37140056

RESUMO

DNA double-strand break (DSB) repair via homologous recombination is initiated by end resection. The extent of DNA end resection determines the choice of the DSB repair pathway. Nucleases for end resection have been extensively studied. However, it is still unclear how the potential DNA structures generated by the initial short resection by MRE11-RAD50-NBS1 are recognized and recruit proteins, such as EXO1, to DSB sites to facilitate long-range resection. We found that the MSH2-MSH3 mismatch repair complex is recruited to DSB sites through interaction with the chromatin remodeling protein SMARCAD1. MSH2-MSH3 facilitates the recruitment of EXO1 for long-range resection and enhances its enzymatic activity. MSH2-MSH3 also inhibits access of POLθ, which promotes polymerase theta-mediated end-joining (TMEJ). Collectively, we present a direct role of MSH2-MSH3 in the initial stages of DSB repair by promoting end resection and influencing the DSB repair pathway by favoring homologous recombination over TMEJ.


Assuntos
Reparo do DNA , Exodesoxirribonucleases , Proteína 2 Homóloga a MutS , Proteína 3 Homóloga a MutS , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Exodesoxirribonucleases/metabolismo , Recombinação Homóloga , Proteína 2 Homóloga a MutS/metabolismo , Humanos , Linhagem Celular , DNA Helicases/metabolismo , Proteína 3 Homóloga a MutS/metabolismo
4.
Nucleic Acids Res ; 51(5): 2257-2269, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36805268

RESUMO

DNA polymerase θ (POLQ) is a unique DNA polymerase that is able to perform microhomology-mediated end-joining as well as translesion synthesis (TLS) across an abasic (AP) site and thymine glycol (Tg). However, the biological significance of the TLS activity is currently unknown. Herein we provide evidence that the TLS activity of POLQ plays a critical role in repairing complex DNA double-strand breaks (DSBs) induced by high linear energy transfer (LET) radiation. Radiotherapy with high LET radiation such as carbon ions leads to more deleterious biological effects than corresponding doses of low LET radiation such as X-rays. High LET-induced DSBs are considered to be complex, carrying additional DNA damage such as AP site and Tg in close proximity to the DSB sites. However, it is not clearly understood how complex DSBs are processed in mammalian cells. We demonstrated that genetic disruption of POLQ results in an increase of chromatid breaks and enhanced cellular sensitivity following treatment with high LET radiation. At the biochemical level, POLQ was able to bypass an AP site and Tg during end-joining and was able to anneal two single-stranded DNA tails when DNA lesions were located outside the microhomology. This study offers evidence that POLQ is directly involved in the repair of complex DSBs.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Polimerase Dirigida por DNA , Animais , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Reparo do DNA , DNA/genética , Reparo do DNA por Junção de Extremidades , Mamíferos/genética
5.
Nucleic Acids Res ; 51(2): 631-649, 2023 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-36594163

RESUMO

TRAIP is a key factor involved in the DNA damage response (DDR), homologous recombination (HR) and DNA interstrand crosslink (ICL) repair. However, the exact functions of TRAIP in these processes in mammalian cells are not fully understood. Here we identify the zinc finger protein 212, ZNF212, as a novel binding partner for TRAIP and find that ZNF212 colocalizes with sites of DNA damage. The recruitment of TRAIP or ZNF212 to sites of DNA damage is mutually interdependent. We show that depletion of ZNF212 causes defects in the DDR and HR-mediated repair in a manner epistatic to TRAIP. In addition, an epistatic analysis of Zfp212, the mouse homolog of human ZNF212, in mouse embryonic stem cells (mESCs), shows that it appears to act upstream of both the Neil3 and Fanconi anemia (FA) pathways of ICLs repair. We find that human ZNF212 interacted directly with NEIL3 and promotes its recruitment to ICL lesions. Collectively, our findings identify ZNF212 as a new factor involved in the DDR, HR-mediated repair and ICL repair though direct interaction with TRAIP.


Assuntos
Reparo do DNA , Anemia de Fanconi , Animais , Camundongos , Humanos , Reparo do DNA/genética , Dano ao DNA , Replicação do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genômica , Anemia de Fanconi/genética , Mamíferos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas do Tecido Nervoso/genética
6.
Nat Commun ; 13(1): 6732, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36347866

RESUMO

Aminoacyl-tRNA synthetases (ARSs) have evolved to acquire various additional domains. These domains allow ARSs to communicate with other cellular proteins in order to promote non-translational functions. Vertebrate cytoplasmic isoleucyl-tRNA synthetases (IARS1s) have an uncharacterized unique domain, UNE-I. Here, we present the crystal structure of the chicken IARS1 UNE-I complexed with glutamyl-tRNA synthetase 1 (EARS1). UNE-I consists of tandem ubiquitin regulatory X (UBX) domains that interact with a distinct hairpin loop on EARS1 and protect its neighboring proteins in the multi-synthetase complex from degradation. Phosphomimetic mutation of the two serine residues in the hairpin loop releases IARS1 from the complex. IARS1 interacts with BRCA1 in the nucleus, regulates its stability by inhibiting ubiquitylation via the UBX domains, and controls DNA repair function.


Assuntos
Aminoacil-tRNA Sintetases , Isoleucina-tRNA Ligase , Isoleucina-tRNA Ligase/química , Aminoacil-tRNA Sintetases/metabolismo , Glutamato-tRNA Ligase/química , RNA de Transferência/metabolismo
7.
Cancer Med ; 11(21): 4005-4020, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35352878

RESUMO

Cobll1 affects blast crisis (BC) progression and tyrosine kinase inhibitor (TKI) resistance in chronic myeloid leukemia (CML). PACSIN2, a novel Cobll1 binding protein, activates TKI-induced apoptosis in K562 cells, and this activation is suppressed by Cobll1 through the interaction between PACSIN2 and Cobll1. PACSIN2 also binds and inhibits SH3BP1 which activates the downstream Rac1 pathway and induces TKI resistance. PACSIN2 competitively interacts with Cobll1 or SH3BP1 with a higher affinity for Cobll1. Cobll1 preferentially binds to PACSIN2, releasing SH3BP1 to promote the SH3BP1/Rac1 pathway and suppress TKI-mediated apoptosis and eventually leading to TKI resistance. Similar interactions among Cobll1, PACSIN2, and SH3BP1 control hematopoiesis during vertebrate embryogenesis. Clinical analysis showed that most patients with CML have Cobll1 and SH3BP1 expression at the BC phase and BC patients with Cobll1 and SH3BP1 expression showed severe progression with a higher blast percentage than those without any Cobll1, PACSIN2, or SH3BP1 expression. Our study details the molecular mechanism of the Cobll1/PACSIN2/SH3BP1 pathway in regulating drug resistance and BC progression in CML.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Ativadoras de GTPase , Leucemia Mielogênica Crônica BCR-ABL Positiva , Fatores de Transcrição , Humanos , Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose , Crise Blástica , Resistência a Medicamentos , Resistencia a Medicamentos Antineoplásicos , Proteínas Ativadoras de GTPase/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Inibidores de Proteínas Quinases/farmacologia , Fatores de Transcrição/genética
8.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35217600

RESUMO

An ideal cancer therapeutic strategy involves the selective killing of cancer cells without affecting the surrounding normal cells. However, researchers have failed to develop such methods for achieving selective cancer cell death because of shared features between cancerous and normal cells. In this study, we have developed a therapeutic strategy called the cancer-specific insertions-deletions (InDels) attacker (CINDELA) to selectively induce cancer cell death using the CRISPR-Cas system. CINDELA utilizes a previously unexplored idea of introducing CRISPR-mediated DNA double-strand breaks (DSBs) in a cancer-specific fashion to facilitate specific cell death. In particular, CINDELA targets multiple InDels with CRISPR-Cas9 to produce many DNA DSBs that result in cancer-specific cell death. As a proof of concept, we demonstrate here that CINDELA selectively kills human cancer cell lines, xenograft human tumors in mice, patient-derived glioblastoma, and lung patient-driven xenograft tumors without affecting healthy human cells or altering mouse growth.


Assuntos
Sistemas CRISPR-Cas , Mutação INDEL , Neoplasias/genética , Animais , Morte Celular/genética , Quebras de DNA de Cadeia Dupla , Xenoenxertos , Humanos , Camundongos
9.
Nucleic Acids Res ; 49(20): 11746-11764, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34718749

RESUMO

Reactive oxygen species (ROS) generate oxidized bases and single-strand breaks (SSBs), which are fixed by base excision repair (BER) and SSB repair (SSBR), respectively. Although excision and repair of damaged bases have been extensively studied, the function of the sliding clamp, proliferating cell nuclear antigen (PCNA), including loading/unloading, remains unclear. We report that, in addition to PCNA loading by replication factor complex C (RFC), timely PCNA unloading by the ATPase family AAA domain-containing protein 5 (ATAD5)-RFC-like complex is important for the repair of ROS-induced SSBs. We found that PCNA was loaded at hydrogen peroxide (H2O2)-generated direct SSBs after the 3'-terminus was converted to the hydroxyl moiety by end-processing enzymes. However, PCNA loading rarely occurred during BER of oxidized or alkylated bases. ATAD5-depleted cells were sensitive to acute H2O2 treatment but not methyl methanesulfonate treatment. Unexpectedly, when PCNA remained on DNA as a result of ATAD5 depletion, H2O2-induced repair DNA synthesis increased in cancerous and normal cells. Based on higher H2O2-induced DNA breakage and SSBR protein enrichment by ATAD5 depletion, we propose that extended repair DNA synthesis increases the likelihood of DNA polymerase stalling, shown by increased PCNA monoubiquitination, and consequently, harmful nick structures are more frequent.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Reparo do DNA por Junção de Extremidades , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , ATPases Associadas a Diversas Atividades Celulares/genética , Proteínas de Ligação a DNA/genética , Células HEK293 , Células HeLa , Humanos , Peróxido de Hidrogênio/toxicidade , Estresse Oxidativo , Antígeno Nuclear de Célula em Proliferação/genética
10.
Nucleic Acids Res ; 49(10): 5605-5622, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33963872

RESUMO

Proper activation of DNA repair pathways in response to DNA replication stress is critical for maintaining genomic integrity. Due to the complex nature of the replication fork (RF), problems at the RF require multiple proteins, some of which remain unidentified, for resolution. In this study, we identified the N-methyl-D-aspartate receptor synaptonuclear signaling and neuronal migration factor (NSMF) as a key replication stress response factor that is important for ataxia telangiectasia and Rad3-related protein (ATR) activation. NSMF localizes rapidly to stalled RFs and acts as a scaffold to modulate replication protein A (RPA) complex formation with cell division cycle 5-like (CDC5L) and ATR/ATR-interacting protein (ATRIP). Depletion of NSMF compromised phosphorylation and ubiquitination of RPA2 and the ATR signaling cascade, resulting in genomic instability at RFs under DNA replication stress. Consistently, NSMF knockout mice exhibited increased genomic instability and hypersensitivity to genotoxic stress. NSMF deficiency in human and mouse cells also caused increased chromosomal instability. Collectively, these findings demonstrate that NSMF regulates the ATR pathway and the replication stress response network for genome maintenance and cell survival.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a RNA/metabolismo , Proteína de Replicação A/metabolismo , Fatores de Transcrição/fisiologia , Animais , Replicação do DNA , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout
11.
Genome Res ; 30(9): 1306-1316, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32887690

RESUMO

The higher-order structural organization and dynamics of the chromosomes play a central role in gene regulation. To explore this structure-function relationship, it is necessary to directly visualize genomic elements in living cells. Genome imaging based on the CRISPR system is a powerful approach but has limited applicability due to background signals and nonspecific aggregation of fluorophores within nuclei. To address this issue, we developed a novel visualization scheme combining tripartite fluorescent proteins with the SunTag system and demonstrated that it strongly suppressed background fluorescence and amplified locus-specific signals, allowing long-term tracking of genomic loci. We integrated the multicomponent CRISPR system into stable cell lines to allow quantitative and reliable analysis of dynamic behaviors of genomic loci. Due to the greatly elevated signal-to-background ratio, target loci with only small numbers of sequence repeats could be successfully tracked, even under a conventional fluorescence microscope. This feature enables the application of CRISPR-based imaging to loci throughout the genome and opens up new possibilities for the study of nuclear processes in living cells.


Assuntos
Sistemas CRISPR-Cas , Proteínas de Fluorescência Verde/genética , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Estruturas Genéticas , Técnicas Genéticas , Proteínas de Fluorescência Verde/química , Células HEK293 , Humanos , Processamento de Imagem Assistida por Computador
12.
Nat Commun ; 10(1): 5718, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31844045

RESUMO

Maintaining stability of replication forks is important for genomic integrity. However, it is not clear how replisome proteins contribute to fork stability under replication stress. Here, we report that ATAD5, a PCNA unloader, plays multiple functions at stalled forks including promoting its restart. ATAD5 depletion increases genomic instability upon hydroxyurea treatment in cultured cells and mice. ATAD5 recruits RAD51 to stalled forks in an ATR kinase-dependent manner by hydroxyurea-enhanced protein-protein interactions and timely removes PCNA from stalled forks for RAD51 recruitment. Consistent with the role of RAD51 in fork regression, ATAD5 depletion inhibits slowdown of fork progression and native 5-bromo-2'-deoxyuridine signal induced by hydroxyurea. Single-molecule FRET showed that PCNA itself acts as a mechanical barrier to fork regression. Consequently, DNA breaks required for fork restart are reduced by ATAD5 depletion. Collectively, our results suggest an important role of ATAD5 in maintaining genome integrity during replication stress.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Replicação do DNA/genética , Proteínas de Ligação a DNA/metabolismo , Instabilidade Genômica/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Rad51 Recombinase/metabolismo , ATPases Associadas a Diversas Atividades Celulares/genética , Bromodesoxiuridina/metabolismo , Linhagem Celular Tumoral , Quebras de DNA/efeitos dos fármacos , Reparo do DNA , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência , Técnicas de Silenciamento de Genes , Instabilidade Genômica/efeitos dos fármacos , Células HEK293 , Humanos , Hidroxiureia/farmacologia , Ligação Proteica/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Imagem Individual de Molécula
13.
Cell Rep ; 29(13): 4632-4645.e5, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31875566

RESUMO

Proliferating cell nuclear antigen (PCNA) is a DNA clamp essential for DNA replication. During DNA synthesis, PCNA is continuously loaded onto and unloaded from DNA. PCNA recruits various proteins to nascent DNA to facilitate chromosome duplication. Therefore, timely PCNA unloading is crucial for high-fidelity DNA replication. The ATAD5-RFC-like complex (ATAD5-RLC) unloads PCNA from replicated DNA. It is unclear how ATAD5-RLC activity is regulated to prevent premature PCNA unloading. Here, we find that BRD4, an acetyl-histone-binding chromatin reader, inhibits the PCNA-unloading activity of ATAD5-RLC. The BRD4 ET domain interacts with a region upstream of the ATAD5 PCNA-unloading domain. BRD4-ATAD5 binds to acetyl-histones in nascent chromatin. BRD4 release from chromatin correlates with PCNA unloading. Disruption of the interaction between BRD4 and acetyl-histones or between BRD4 and ATAD5 reduces the PCNA amount on chromatin. In contrast, the overexpression of BRD4 increases the amount of chromatin-bound PCNA. Thus, acetyl-histone-bound BRD4 fine-tunes PCNA unloading from nascent DNA.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fatores de Transcrição/metabolismo , ATPases Associadas a Diversas Atividades Celulares/química , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Acetilação , Motivos de Aminoácidos , Sequência de Aminoácidos , DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Células HeLa , Histonas/metabolismo , Humanos , Mitose , Fosforilação , Ligação Proteica , Domínios Proteicos
14.
iScience ; 19: 177-190, 2019 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-31376680

RESUMO

Polyubiquitination of proliferating cell nuclear antigen (PCNA) regulates the error-free template-switching mechanism for the bypass of DNA lesions during DNA replication. PCNA polyubiquitination is critical for the maintenance of genomic integrity; however, the underlying mechanism is poorly understood. Here, we demonstrate that tonicity-responsive enhancer-binding protein (TonEBP) regulates PCNA polyubiquitination in response to DNA damage. TonEBP was recruited to DNA damage sites with bulky adducts and sequentially recruited E3 ubiquitin ligase SHPRH, followed by deubiquitinase USP1, to DNA damage sites, in correlation with the dynamics of PCNA polyubiquitination. Similarly, TonEBP was found to be required for replication fork protection in response to DNA damage. The Rel-homology domain of TonEBP, which encircles DNA, was essential for the interaction with SHPRH and USP1, PCNA polyubiquitination, and cell survival after DNA damage. The present findings suggest that TonEBP is an upstream regulator of PCNA polyubiquitination and of the DNA damage bypass pathway.

15.
Nat Commun ; 10(1): 2420, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31160570

RESUMO

Replication-Factor-C (RFC) and RFC-like complexes (RLCs) mediate chromatin engagement of the proliferating cell nuclear antigen (PCNA). It remains controversial how RFC and RLCs cooperate to regulate PCNA loading and unloading. Here, we show the distinct PCNA loading or unloading activity of each clamp loader. ATAD5-RLC possesses the potent PCNA unloading activity. ATPase motif and collar domain of ATAD5 are crucial for the unloading activity. DNA structures did not affect PCNA unloading activity of ATAD5-RLC. ATAD5-RLC could unload ubiquitinated PCNA. Through single molecule measurements, we reveal that ATAD5-RLC unloaded PCNA through one intermediate state before ATP hydrolysis. RFC loaded PCNA through two intermediate states on DNA, separated by ATP hydrolysis. Replication proteins such as Fen1 could inhibit the PCNA unloading activity of Elg1-RLC, a yeast homolog of ATAD5-RLC in vitro. Our findings provide molecular insights into how PCNA is released from chromatin to finalize DNA replication/repair.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteína de Replicação C/metabolismo , Adenosina Trifosfatases , Trifosfato de Adenosina/metabolismo , Proteínas de Transporte/metabolismo , Cromatina/metabolismo , Endonucleases Flap/metabolismo , Humanos , Hidrólise , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/metabolismo
16.
Clin Exp Vaccine Res ; 3(2): 227-34, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25003097

RESUMO

PURPOSE: Protein cages are promising nanoplatform candidates for efficient delivery systems due to their homogenous size and structure with high biocompatibility and biodegradability. In this study, we investigate the potential of lumazine synthase protein cage as an antigen delivery system to dendritic cells (DCs), which induce antigen-specific T cell proliferation. MATERIALS AND METHODS: Ovalbumin (OVA) peptides OT-1 (SIINFEKL) and OT-2 (ISQAVHAAHAEINEAGR) were genetically inserted to lumazine synthase and each protein cage was over-expressed in Escherichia coli as a soluble protein. The efficiency of antigen delivery and the resulting antigen-specific T cell proliferation by DCs was examined in vitro as well as in vivo. RESULTS: We successfully generated and characterized OVA peptides carrying lumazine synthase protein cages. The OT-1 and OT-2 peptides carried by lumazine synthases were efficiently delivered and processed by DCs in vitro as well as in vivo, and induced proliferation of OT-1-specific CD8(+)T cells and OT-2-specific CD4(+)T cells. CONCLUSION: Our data demonstrate the potential of lumazine synthase protein cage being used as a novel antigen delivery system for DC-based vaccine development in future clinical applications.

17.
Nanomedicine ; 10(3): 561-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24262997

RESUMO

We utilized ferritin protein cage nanoparticles (FPCN) as antigen delivery nanoplatforms for DC-based vaccine development and investigated DC-mediated antigen-specific immune responses. Antigenic peptides, OT-1 (SIINFEKL) or OT-2 (ISQAVHAAHAEINEAGR) which are derived from ovalbumin, were genetically introduced either onto the exterior surface or into the interior cavity of FPCN. FPCN carrying antigenic peptides (OT-1-FPCN and OT-2-FPCN) were effectively delivered to DCs and processed within endosomes. Delivered antigenic peptides, OT-1 or OT-2, to DCs successfully induced antigen-specific CD8(+) or CD4(+) T cell proliferations both in vitro and in vivo. Naïve mice immunized with OT-1-FPCN efficiently differentiated OT-1 specific CD8(+) T cells into functional effector cytotoxic T cells resulting in selective killing of antigen-specific target cells. Effective differentiation of proliferated OT-2 specific CD4(+) T cells into functional CD4(+) Th1 and Th2 cells was confirmed with the productions of IFN-γ/IL-2 and IL-10/IL-13 cytokines, respectively. FROM THE CLINICAL EDITOR: In this study, the authors utilized ferritin protein cage nanoparticles as antigen delivery nanoplatforms for dendritic cell-based vaccine development and investigated DC-mediated antigen-specific immune responses using strong model antigens derived from ovalbumin, suggesting potential future clinical applicability of this or similar techniques.


Assuntos
Antígenos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Células Dendríticas/imunologia , Ferritinas/química , Nanopartículas/química , Ovalbumina/administração & dosagem , Sequência de Aminoácidos , Animais , Antígenos/química , Antígenos/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Células Cultivadas , Citocinas/imunologia , Células Dendríticas/citologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Ovalbumina/química , Ovalbumina/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Células Th1/citologia , Células Th1/imunologia , Células Th2/citologia , Células Th2/imunologia
18.
Exp Mol Med ; 37(6): 619-23, 2005 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-16391523

RESUMO

A C6 beta-chemokine, CKbeta8-1, suppressed the colony formation of CD34+ cells of human cord blood (CB). Molecular mechanisms involved in CKbeta8-1-medicated suppression of colony formation of CD34+ cells are not known. To address this issue, the level of various G1/S cell cycle regulating proteins in CKbeta8-1-treated CD34+ cells were compared with those in untreated CD34+ cells. CKbeta8-1 did not significantly alter the expression of the G1/S cycle regulation proteins (cyclin D1, D3, and E), CDK inhibitor (p27and Rb), and other cell proliferation regulation protein (p53) in CB CD34+ cells. Here we describe an in vitro system in which CB CD34+ cells were committed to a multipotent progenitor lineage of colony forming units-granulocyte/macrophage (CFU-GM) by a simple combination of recombinant human (rh) GM-CSF and rhIL-3. In this culture system, we found that cyclin E protein appeared later and disappeared faster in the CKbeta8-1-treated cells than in the control cells during CFU-GM lineage development. These findings suggested that cyclin E may play a role in suppressing the colony formation of CFU-GM by CKbeta8-1.


Assuntos
Quimiocinas CC/farmacologia , Ciclina E/metabolismo , Sangue Fetal/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Granulócitos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Antígenos CD34/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem da Célula , Células Cultivadas , Fase G1/efeitos dos fármacos , Granulócitos/citologia , Granulócitos/metabolismo , Substâncias de Crescimento/farmacologia , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
19.
Mol Cells ; 15(2): 176-80, 2003 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-12803479

RESUMO

Several recently identified chemokines, Lkn-1, CKbeta8-1, MRP-2, and Mu C10 (MRP-1), are classified as C6 beta-chemokines. All of these chemokines have been found to suppress colony formation by bone marrow (BM) myeloid progenitors. Since cord blood (CB), like BM, contains CD34-positive cells, we examined the effects of these chemokines on CD34+ cells isolated from human CB. Lkn-1 and CKbeta8-1 suppressed colony formation by multi-potential granulocyte erythroid mega-karyocyte macrophages (CFU-GEMM), granulocyte-macrophages (CFU-GM), and erythroid (BFU-E) cells among the CD34+ cells from CB. CC chemokine receptor 1 (CCR1) that is known to be a receptor for Lkn-1 and CKbeta8-1 in neutrophils, monocytes, and lymphocytes, was also present on the surface of CD34+ cells from CB. Taken together these results suggest that Lkn-1 and CKbeta8-1 are active in inhibiting myeloid progenitor cells from both BM and CB. Macrophage inflammatory protein related protein-2 (mMRP-2) and Mu C10 (mMRP-1), which are murine C6 beta-chemokines, also inhibited colony formation by CB CD34+ cells. The inhibitory activity of these chemokines suggests that they may protect hematopoietic progenitors from the cytotoxic effects of the antiblastic drugs used in cancer therapy.


Assuntos
Diferenciação Celular/fisiologia , Quimiocinas CC/metabolismo , Sangue Fetal/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Antígenos CD34/metabolismo , Medula Óssea/fisiologia , Humanos , Proteínas Inflamatórias de Macrófagos/metabolismo , Camundongos , Proteína 2 Associada à Farmacorresistência Múltipla
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...