Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Sci Rep ; 13(1): 4648, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36944687

RESUMO

SARS-CoV-2 continues to circulate in the human population necessitating regular booster immunization for its long-term control. Ideally, vaccines should ideally not only protect against symptomatic disease, but also prevent transmission via asymptomatic shedding and cover existing and future variants of the virus. This may ultimately only be possible through induction of potent and long-lasting immune responses in the nasopharyngeal tract, the initial entry site of SARS-CoV-2. To this end, we have designed a vaccine based on recombinantly expressed receptor binding domain (RBD) of SARS-CoV-2, fused to the C-terminus of C. perfringens enterotoxin, which is known to target Claudin-4, a matrix molecule highly expressed on mucosal microfold (M) cells of the nasal and bronchial-associated lymphoid tissues. To further enhance immune responses, the vaccine was adjuvanted with a novel toll-like receptor 3/RIG-I agonist (Riboxxim™), consisting of synthetic short double stranded RNA. Intranasal prime-boost immunization of mice induced robust mucosal and systemic anti-SARS-CoV-2 neutralizing antibody responses against SARS-CoV-2 strains Wuhan-Hu-1, and several variants (B.1.351/beta, B.1.1.7/alpha, B.1.617.2/delta), as well as systemic T-cell responses. A combination vaccine with M-cell targeted recombinant HA1 from an H1N1 G4 influenza strain also induced mucosal and systemic antibodies against influenza. Taken together, the data show that development of an intranasal SARS-CoV-2 vaccine based on recombinant RBD adjuvanted with a TLR3 agonist is feasible, also as a combination vaccine against influenza.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Influenza Humana , Animais , Humanos , Camundongos , Adjuvantes Imunológicos , Adjuvantes Farmacêuticos , Anticorpos Neutralizantes , Anticorpos Antivirais , Clostridium perfringens , COVID-19/prevenção & controle , Vacinas contra COVID-19/imunologia , Mucosa Gástrica , Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Células M , SARS-CoV-2 , Receptor 3 Toll-Like
2.
Cardiovasc Res ; 119(4): 1030-1045, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-36464772

RESUMO

AIMS: The loss of vascular wall cells in allotransplanted arteries is the initial event leading to transplant arteriosclerosis (TA) and ensuing loss of allograft function. Pharmacological agents able to prevent TA are currently lacking. We previously showed that selective inhibition of the immunoproteasome prevented the chronic rejection of renal allografts. However, the role and mechanisms of selective inhibition of a single immunoproteasome subunit to prevent immune-mediated vascular allograft rejection and TA is not clear. METHODS AND RESULTS: The effect and potential mechanism of combined or individual inhibition of peptidolytically active immunoproteasome LMP7 (ß5i) and LMP2 (ß1i) subunits on immune rejection-mediated TA was investigated using the epoxyketone inhibitor ONX 0914, and the recently developed LMP7-selective inhibitor KZR-329 and LMP2-selective inhibitor KZR-504 in a rat aorta transplantation model. We find that co-inhibition of LMP7 and LMP2 in allogeneic recipients significantly suppressed T-cell activation and function by expressing inhibitory surface markers and then activating inhibitory signals. Moreover, co-inhibition of LMP7 and LMP2 substantially reduced the number of immunoglobulin G-secreting cells and plasma cells and production of alloantibodies through activating the unfolded protein response and incapacitating the survival niche of plasma cells in the bone marrow. Consequentially, the accumulation of inflammatory cytokines, complement, and antibodies is reduced and the apoptosis of vascular wall cells decreased in aortic allografts via LMP7 and LMP2 co-inhibition with ONX 0914 treatment or combined KZR-329 and KZR-504 treatment. However, neither individual inhibition of LMP7 by KZR-329 nor individual inhibition of LMP2 by KZR-504 showed suppression of immune rejection and TA. CONCLUSIONS: We define a critical role of LMP7 and LMP2 in TA and strongly propose co-inhibition of both immunoproteasome subunits as promising therapeutic approach to suppress TA and allograft rejection.


Assuntos
Arteriosclerose , Rim , Ratos , Animais , Rim/metabolismo , Citocinas/metabolismo , Rejeição de Enxerto/prevenção & controle
3.
Front Immunol ; 13: 870720, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35711460

RESUMO

Background: Allergic asthma is a chronic disease and medical treatment often fails to fully control the disease in the long term, leading to a great need for new therapeutic approaches. Immunoproteasome inhibition impairs T helper cell function and is effective in many (auto-) inflammatory settings but its effect on allergic airway inflammation is unknown. Methods: Immunoproteasome expression was analyzed in in vitro polarized T helper cell subsets. To study Th2 cells in vivo acute allergic airway inflammation was induced in GATIR (GATA-3-vYFP reporter) mice using ovalbumin and house dust mite extract. Mice were treated with the immunoproteasome inhibitor ONX 0914 or vehicle during the challenge phase and the induction of airway inflammation was analyzed. Results: In vitro polarized T helper cell subsets (Th1, Th2, Th17, and Treg) express high levels of immunoproteasome subunits. GATIR mice proved to be a useful tool for identification of Th2 cells. Immunoproteasome inhibition reduced the Th2 response in both airway inflammation models. Furthermore, T cell activation and antigen-specific cytokine secretion was impaired and a reduced infiltration of eosinophils and professional antigen-presenting cells into the lung and the bronchoalveolar space was observed in the ovalbumin model. Conclusion: These results show the importance of the immunoproteasome in Th2 cells and airway inflammation. Our data provides first insight into the potential of using immunoproteasome inhibition to target the aberrant Th2 response, e.g. in allergic airway inflammation.


Assuntos
Asma , Células Th2 , Animais , Asma/tratamento farmacológico , Asma/metabolismo , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Camundongos , Ovalbumina/farmacologia , Células Th17 , Células Th2/metabolismo
4.
Eur J Immunol ; 52(9): 1510-1522, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35733374

RESUMO

Polymyositis (PM) is a chronic autoimmune inflammatory myopathy resulting in muscle weakness. The limited approved therapies and their poor efficacy contribute to its comorbidity. We investigated the therapeutic use of ONX 0914 and KZR-616, selective inhibitors of the immunoproteasome, in C protein-induced myositis (CIM), a mouse model of PM that closely resembles the human disease. Diseased mice (day 13 postimmunization) were treated with 10 mg/kg ONX 0914, KZR-616, or vehicle on alternate days until day 28. Endpoints included muscle strength assessed by a grip strength meter, serum creatine kinase activity, histology, and immunohistochemistry analysis. Treatment with ONX 0914 or KZR-616 prevented the loss of grip strength in mice after CIM induction, while vehicle-treated animals displayed progressive muscle weakness. Immunoproteasome inhibition lowered PM-associated leukocyte infiltration of the muscle and prevented increased serum creatine kinase levels. LMP7-deficient mice were resistant to CIM induction, as they showed no alterations in grip strength or creatine kinase (CK) levels or muscular alterations. In conclusion, selective inhibition of the immunoproteasome displays therapeutic efficacy in a preclinical mouse model of PM with suppression of muscle inflammation and preservation of muscle strength. Positive results from this study support the rationale for using KZR-616 in clinical studies.


Assuntos
Debilidade Muscular , Polimiosite , Animais , Creatina Quinase/uso terapêutico , Humanos , Camundongos , Morfolinas , Debilidade Muscular/tratamento farmacológico , Polimiosite/tratamento farmacológico , Polimiosite/patologia , Complexo de Endopeptidases do Proteassoma
5.
Eur J Pharm Sci ; 175: 106209, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35580737

RESUMO

The essential role of tissue-resident memory T cells (TRM cells) in offering protection from recurring infections and malignant tumors is becoming increasingly clear. Due to their presence in many barrier tissues, TRM cells are ideally located to rapidly respond to re-encountered pathogens. Moreover, a host of studies has shown that the quantity of TRM cells correlates with increased survival rates in cancer patients. Therefore, vaccination strategies which induce a strong and sustained TRM cell response are particularly promising. In this study we show that this response can be induced by employing a prime-boost vaccination strategy using biodegradable poly (D,L-lactide-co-glycolide) microspheres (PLGA MS). A subcutaneous prime immunization followed by an intranasal boost immunization led to a strong TRM cell response in the lungs of mice 6 days after the boost vaccination. Although numbers subsequently declined, TRM cells were still detectable 60 days after vaccination. Functionally, we observed that immunized mice were protected from lung metastasis formation and tumor growth in a B16Bl6 melanoma model. Furthermore, the TRM cells induced by PLGA MS immunization provided protection in an infectious model using a recombinant influenza A virus (IAV). Taken together, these results show that the ability of PLGA MS to induce a strong TRM cell response further supports their use as a potent vaccine.


Assuntos
Memória Imunológica , Neoplasias , Animais , Linfócitos T CD8-Positivos , Pulmão/patologia , Camundongos , Neoplasias/patologia , Vacinação/métodos
6.
J Vis Exp ; (180)2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35188118

RESUMO

Renal allograft rejection limits the long-term survival of patients after renal transplantation. Rat orthotopic renal transplantation is an essential model to investigate the mechanism of renal allograft rejection in pre-clinical studies and could aid in the development of novel approaches to improve the long-term survival of renal allografts. Donor kidney implantation in rat orthotopic renal transplantation is commonly performed by end-to-side anastomosis to recipients' aorta and inferior vena cava. In this model, the donor's kidney was implanted using end-to-end anastomosis to the recipients' renal artery and renal vein. The donor's ureter was anastomosed to the recipient's bladder in an end-to-side 'tunnel' method. This model contributes to better healing of ureter-bladder anastomosis and increases the recipients' survival by avoiding interference with blood supply and venous reflux of the lower body. This model can be used to investigate the mechanisms of acute and chronic immune and pathologic rejection of renal allografts. Here, the study describes the detailed protocols of this orthotopic renal transplantation between rats.


Assuntos
Transplante de Rim , Aloenxertos , Anastomose Cirúrgica/métodos , Animais , Rejeição de Enxerto , Humanos , Rim/irrigação sanguínea , Rim/cirurgia , Transplante de Rim/efeitos adversos , Transplante de Rim/métodos , Ratos , Veias Renais/cirurgia , Veia Cava Inferior/cirurgia
7.
Cells ; 10(11)2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34831438

RESUMO

Numerous cellular processes are controlled by the proteasome, a multicatalytic protease in the cytosol and nucleus of all eukaryotic cells, through regulated protein degradation. The immunoproteasome is a special type of proteasome which is inducible under inflammatory conditions and constitutively expressed in hematopoietic cells. MECL-1 (ß2i), LMP2 (ß1i), and LMP7 (ß5i) are the proteolytically active subunits of the immunoproteasome (IP), which is known to shape the antigenic repertoire presented on major histocompatibility complex (MHC) class I molecules. Furthermore, the immunoproteasome is involved in T cell expansion and inflammatory diseases. In recent years, targeting the immunoproteasome in cancer, autoimmune diseases, and transplantation proved to be therapeutically effective in preclinical animal models. However, the prime function of standard proteasomes and immunoproteasomes is the control of protein homeostasis in cells. To maintain protein homeostasis in cells, proteasomes remove proteins which are not properly folded, which are damaged by stress conditions such as reactive oxygen species formation, or which have to be degraded on the basis of regular protein turnover. In this review we summarize the latest insights on how the immunoproteasome influences protein homeostasis.


Assuntos
Complexo de Endopeptidases do Proteassoma/imunologia , Proteostase , Animais , Humanos , Modelos Biológicos , Oxirredução , Proteólise , Ubiquitinação
8.
Nat Commun ; 12(1): 2935, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34006895

RESUMO

With emerging supremacy, cancer immunotherapy has evolved as a promising therapeutic modality compared to conventional antitumor therapies. Cancer immunotherapy composed of biodegradable poly(lactic-co-glycolic acid) (PLGA) particles containing antigens and toll-like receptor ligands induces vigorous antitumor immune responses in vivo. Here, we demonstrate the supreme adjuvant effect of the recently developed and pharmaceutically defined double-stranded (ds)RNA adjuvant Riboxxim especially when incorporated into PLGA particles. Encapsulation of Riboxxim together with antigens potently activates murine and human dendritic cells, and elevated tumor-specific CD8+ T cell responses are superior to those obtained using classical dsRNA analogues. This PLGA particle vaccine affords primary tumor growth retardation, prevention of metastases, and prolonged survival in preclinical tumor models. Its advantageous therapeutic potency was further enhanced by immune checkpoint blockade that resulted in reinvigoration of cytotoxic T lymphocyte responses and tumor ablation. Thus, combining immune checkpoint blockade with immunotherapy based on Riboxxim-bearing PLGA particles strongly increases its efficacy.


Assuntos
Vacinas Anticâncer/imunologia , Proteína DEAD-box 58/imunologia , Inibidores de Checkpoint Imunológico/imunologia , Imunoterapia/métodos , Neoplasias Experimentais/terapia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/imunologia , Receptores Imunológicos/imunologia , Receptor 3 Toll-Like/imunologia , Animais , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Células Cultivadas , Proteína DEAD-box 58/metabolismo , Sinergismo Farmacológico , Feminino , Humanos , Inibidores de Checkpoint Imunológico/administração & dosagem , Ligantes , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Nanopartículas/química , Nanopartículas/ultraestrutura , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Receptores Imunológicos/metabolismo , Células THP-1 , Receptor 3 Toll-Like/metabolismo , Resultado do Tratamento
9.
J Immunol ; 206(8): 1697-1708, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33731337

RESUMO

The prime function of proteasomes is the control of protein homeostasis in cells (i.e., the removal of proteins that are not properly folded, damaged by stress conditions like reactive oxygen species formation, or degraded on the basis of regular protein turnover). During viral infection, the standard proteasome is replaced by the so-called immunoproteasome (IP) in an IFN-γ-dependent manner. It has been proposed that the IP is required to protect cell viability under conditions of IFN-induced oxidative stress. In this study, we investigated the requirement for IP to cope with the enhanced need for protein degradation during lymphocytic choriomeningitis virus (LCMV) infection in mice lacking the IP subunit LMP7. We found that IP are upregulated in the liver but not in the spleen during LCMV infection, although the total proteasome content was not altered. The expression of standard proteasome subunits is not induced in LMP7-deficient mice, indicating that enhanced proteasomal activity is not required during viral infection. Furthermore, ubiquitin accumulation, apoptosis induction, and viral titers were similar in LCMV-infected mice lacking LMP7 compared with wild-type mice. Taken together, these data indicate that the IP is not required to regulate protein homeostasis during LCMV infection.


Assuntos
Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Linfócitos T/imunologia , Animais , Células Cultivadas , Homeostase , Interferon gama/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/genética , Proteólise , Regulação para Cima
10.
Cell Rep ; 34(11): 108857, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33730565

RESUMO

Parkin is an E3 ubiquitin ligase belonging to the RING-between-RING family. Mutations in the Parkin-encoding gene PARK2 are associated with familial Parkinson's disease. Here, we investigate the interplay between Parkin and the inflammatory cytokine-induced ubiquitin-like modifier FAT10. FAT10 targets hundreds of proteins for degradation by the 26S proteasome. We show that FAT10 gets conjugated to Parkin and mediates its degradation in a proteasome-dependent manner. Parkin binds to the E2 enzyme of FAT10 (USE1), auto-FAT10ylates itself, and facilitates FAT10ylation of the Parkin substrate Mitofusin2 in vitro and in cells, thus identifying Parkin as a FAT10 E3 ligase. On mitochondrial depolarization, FAT10ylation of Parkin inhibits its activation and ubiquitin-ligase activity causing impairment of mitophagy progression and aggravation of rotenone-mediated death of dopaminergic neuronal cells. In conclusion, FAT10ylation inhibits Parkin and mitophagy rendering FAT10 a likely inflammation-induced exacerbating factor and potential drug target for Parkinson's disease.


Assuntos
Mitofagia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinas/metabolismo , Morte Celular , Citosol/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Células HEK293 , Células HeLa , Humanos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Transporte Proteico , Proteólise , Espécies Reativas de Oxigênio/metabolismo , Especificidade por Substrato , Ubiquitinação
11.
Biochem Biophys Res Commun ; 554: 56-62, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-33774280

RESUMO

The endoplasmic reticulum (ER) is an organelle with high protein density and therefore prone to be damaged by protein aggregates. One proposed preventive measure is a pre-emptive quality control pathway that attenuates ER import during protein folding stress. ER resident proteins are targeted into the ER via signal peptides cleaved rapidly upon ER insertion by the ER signal peptidase. Here we show that the ER insertion and cleavage of the ER-targeting peptide of the prostate carcinoma antigen prostate stem cell antigen (PSCA) is retarded and strongly reduced when the proteasome is inhibited or genetically silenced. Also overexpression of the C-terminally extended ubiquitin variant Ub2-UBB+1 or oxidative stress attenuated signal peptide processing. Proteasome inhibition likewise protracted ER signal processing of the ER targeted hormone leptin and the MHC class I molecule H-2Dd. These findings, which are consistent with a pre-emptive ER quality control pathway, may explain why an immunodominant MHC class I peptide ligand of PSCA spanning its ER signal peptidase cleavage site is efficiently generated in the cytoplasm from PSCA precursors that fail to reach the ER.


Assuntos
Antígenos de Neoplasias/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Retículo Endoplasmático/metabolismo , Proteínas de Neoplasias/metabolismo , Estresse Oxidativo/fisiologia , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Células Cultivadas , Retículo Endoplasmático/patologia , Proteínas Ligadas por GPI/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Leptina/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Sinais Direcionadores de Proteínas , Serina Endopeptidases/metabolismo
12.
Eur J Immunol ; 51(1): 138-150, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32686110

RESUMO

The IFN stimulated gene 15 (ISG15) encodes a 15-kDa ubiquitin-like protein, that is induced by type I IFNs and is conjugated to the bulk of newly synthesized polypeptides at the ribosome. ISG15 functions as an antiviral molecule possibly by being covalently conjugated to viral proteins and disturbing virus particle assembly. Here, we have investigated the effect of ISGylation on degradation and antigen presentation of viral and cellular proteins. ISGylation did not induce proteasomal degradation of bulk ISG15 target proteins neither after overexpressing ISG15 nor after induction by IFN-ß. The MHC class I cell surface expression of splenocytes derived from ISG15-deficient mice or mice lacking the catalytic activity of the major de-ISGylating enzyme USP18 was unaltered as compared to WT mice. Fusion of ubiquitin or FAT10 to the long-lived nucleoprotein (NP) of lymphocytic choriomeningitis virus accelerated the proteasomal degradation of NP while fusion to ISG15 did not detectably speed up NP degradation. Nevertheless, MHC-I restricted presentation of two epitopes of NP were markedly enhanced when it was fused to ISG15 similarly to fusion with ubiquitin or FAT10. Thus, we provide evidence that ISG15 can enhance the presentation of antigens on MHC-I most likely by promoting co-translational antigen processing.


Assuntos
Apresentação de Antígeno/imunologia , Citocinas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Ubiquitinas/imunologia , Animais , Citocinas/deficiência , Citocinas/genética , Citocinas/metabolismo , Células HEK293 , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/imunologia , Proteínas do Nucleocapsídeo/metabolismo , Complexo de Endopeptidases do Proteassoma/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Modificação Traducional de Proteínas/imunologia , Proteólise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitina Tiolesterase/deficiência , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/imunologia , Ubiquitinas/deficiência , Ubiquitinas/genética , Ubiquitinas/metabolismo
13.
Genes Immun ; 21(5): 273-287, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32839530

RESUMO

The proteasome is a multicatalytic protease in the cytosol and nucleus of all eukaryotic cells that controls numerous cellular processes through regulated protein degradation. Proteasome inhibitors have significantly improved the survival of multiple myeloma patients. However, clinically approved proteasome inhibitors have failed to show efficacy against solid tumors, neither alone nor in combination with other therapies. Targeting the immunoproteasome with selective inhibitors has been therapeutically effective in preclinical models for several autoimmune diseases and colon cancer. Moreover, immunoproteasome inhibitors prevented the chronic rejection of allogeneic organ transplants. In recent years, it has become apparent that inhibition of one single active center of the proteasome is insufficient to achieve therapeutic benefits. In this review we summarize the latest insights how targeting multiple catalytically active proteasome subunits can interfere with disease progression in autoimmunity, growth of solid tumors, and allograft rejection.


Assuntos
Antineoplásicos/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Doença Enxerto-Hospedeiro/tratamento farmacológico , Imunossupressores/uso terapêutico , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/uso terapêutico , Animais , Antineoplásicos/química , Humanos , Imunossupressores/química , Complexo de Endopeptidases do Proteassoma/química , Inibidores de Proteassoma/química
14.
J Biol Chem ; 295(42): 14402-14418, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-32817338

RESUMO

The retina-specific chaperone aryl hydrocarbon interacting protein-like 1 (AIPL1) is essential for the correct assembly of phosphodiesterase 6 (PDE6), which is a pivotal effector enzyme for phototransduction and vision because it hydrolyzes cGMP. AIPL1 interacts with the cytokine-inducible ubiquitin-like modifier FAT10, which gets covalently conjugated to hundreds of proteins and targets its conjugation substrates for proteasomal degradation, but whether FAT10 affects PDE6 function or turnover is unknown. Here, we show that FAT10 mRNA is expressed in human retina and identify rod PDE6 as a retina-specific substrate of FAT10 conjugation. We found that AIPL1 stabilizes the FAT10 monomer and the PDE6-FAT10 conjugate. Additionally, we elucidated the functional consequences of PDE6 FAT10ylation. On the one hand, we demonstrate that FAT10 targets PDE6 for proteasomal degradation by formation of a covalent isopeptide linkage. On the other hand, FAT10 inhibits PDE6 cGMP hydrolyzing activity by noncovalently interacting with the PDE6 GAFa and catalytic domains. Therefore, FAT10 may contribute to loss of PDE6 and, as a consequence, degeneration of retinal cells in eye diseases linked to inflammation and inherited blindness-causing mutations in AIPL1.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Retina/metabolismo , Ubiquitinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Motivos de Aminoácidos , Animais , Domínio Catalítico , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/química , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Complexo de Endopeptidases do Proteassoma/química , Inibidores de Proteassoma/farmacologia , Ligação Proteica , Proteólise/efeitos dos fármacos , RNA Mensageiro/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Ubiquitina/metabolismo , Ubiquitinas/química , Ubiquitinas/genética
15.
J Cell Sci ; 133(14)2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32719056

RESUMO

Human leukocyte antigen (HLA)-F adjacent transcript 10 (FAT10) also called ubiquitin D (UBD) is a member of the ubiquitin-like modifier (ULM) family. The FAT10 gene is localized in the MHC class I locus and FAT10 protein expression is mainly restricted to cells and organs of the immune system. In all other cell types and tissues, FAT10 expression is highly inducible by the pro-inflammatory cytokines interferon (IFN)-γ and tumor necrosis factor (TNF). Besides ubiquitin, FAT10 is the only ULM which directly targets its substrates for degradation by the 26S proteasome. This poses the question as to why two ULMs sharing the proteasome-targeting function have evolved and how they differ from each other. This Review summarizes the current knowledge of the special structure of FAT10 and highlights its differences from ubiquitin. We discuss how these differences might result in differential outcomes concerning proteasomal degradation mechanisms and non-covalent target interactions. Moreover, recent insights about the structural and functional impact of FAT10 interacting with specific non-covalent interaction partners are reviewed.


Assuntos
Complexo de Endopeptidases do Proteassoma , Ubiquitina , Humanos , Complexo de Endopeptidases do Proteassoma/genética , Fator de Necrose Tumoral alfa , Ubiquitinas/genética
16.
J Cell Sci ; 133(14)2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32546531

RESUMO

Dendritic cell (DC) aggresome-like induced structures (DALIS) are protein aggregates of polyubiquitylated proteins that form transiently during DC maturation. DALIS scatter randomly throughout the cytosol and serve as antigen storage sites synchronising DC maturation and antigen presentation. Maturation of DCs is accompanied by the induction of the ubiquitin-like modifier FAT10 (also known as UBD), which localises to aggresomes, structures that are similar to DALIS. FAT10 is conjugated to substrate proteins and serves as a signal for their rapid and irreversible degradation by the 26S proteasome similar to, yet independently of ubiquitin, thereby contributing to antigen presentation. Here, we have investigated whether FAT10 is involved in the formation and turnover of DALIS, and whether proteins accumulating in DALIS can be modified through conjunction to FAT10 (FAT10ylated). We found that FAT10 localises to DALIS in maturing DCs and that this localisation occurs independently of its conjugation to substrates. Additionally, we investigated the DALIS turnover in FAT10-deficient and -proficient DCs, and observed FAT10-mediated disassembly of DALIS. Thus, we report further evidence that FAT10 is involved in antigen processing, which may provide a functional rationale as to why FAT10 is selectively induced upon DC maturation.


Assuntos
Apresentação de Antígeno , Células Dendríticas , Diferenciação Celular , Corpos de Inclusão , Ubiquitina , Ubiquitinas/genética
17.
Biomolecules ; 10(6)2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32586037

RESUMO

The revelation that the human major histocompatibility complex (MHC) class I locus encodes a ubiquitin-like protein designated HLA-F adjacent transcript 10 (FAT10) or ubiquitin D (UBD) has attracted increasing attention to the function of this protein. Interestingly, the pro-inflammatory cytokines interferon (IFN)-γ and tumor necrosis factor (TNF) α synergize to strongly induce FAT10 expression, thereby suggesting a role of FAT10 in the immune response. Recent reports that FAT10 downregulates type I interferon production while it upregulates IFN-γ pose mechanistic questions on how FAT10 differentially regulates interferon induction. Several covalent and non-covalent binding partners of FAT10 involved in signal transduction pathways leading to IFN synthesis have been identified. After introducing FAT10, we review here recent insights into how FAT10 affects proteins in the interferon pathways, like the virus-responsive pattern recognition receptor RIG-I, the ubiquitin ligase ZNF598, and the deubiquitylating enzyme OTUB1. Moreover, we outline the consequences of FAT10 deficiency on interferon synthesis and viral expansion in mice and human cells. We discuss the need for covalent isopeptide linkage of FAT10 to the involved target proteins and the concomitant targeting for proteasomal degradation. After years of investigating the elusive biological functions of this fascinating ubiquitin-like modifier, we review the emerging evidence for a novel role of FAT10 in interferon regulation.


Assuntos
Interferon gama/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitinas/metabolismo , Animais , Humanos , Ubiquitinas/deficiência
18.
ACS Cent Sci ; 6(2): 241-246, 2020 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-32123742

RESUMO

We have developed a syringolin-based chemical probe and explored its utility for the profiling of metabolite extracts as potent inhibitors of the 20S proteasome. Activity-guided fractionation by competitive labeling allowed us to isolate and identify glidobactin A and C as well as luminmycin A from a Burkholderiales strain. The natural products exhibited unique subunit specificities for the proteolytic subunits of human and mouse constitutive and immunoproteasome in the lower nanomolar range. In particular, glidobactin C displayed an unprecedented ß2/ß5 coinhibition profile with single-digit nanomolar potency in combination with sufficiently high cell permeability. These properties render glidobactin C a promising live cell proteasome inhibitor with potent activity against human breast cancer cell lines and comparably low immunotoxicity.

19.
Nat Commun ; 10(1): 4452, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31575873

RESUMO

The covalent attachment of the cytokine-inducible ubiquitin-like modifier HLA-F adjacent transcript 10 (FAT10) to hundreds of substrate proteins leads to their rapid degradation by the 26 S proteasome independently of ubiquitylation. Here, we identify another function of FAT10, showing that it interferes with the activation of SUMO1/2/3 in vitro and down-regulates SUMO conjugation and the SUMO-dependent formation of promyelocytic leukemia protein (PML) bodies in cells. Mechanistically, we show that FAT10 directly binds to and impedes the activity of the heterodimeric SUMO E1 activating enzyme AOS1/UBA2 by competing very efficiently with SUMO for activation and thioester formation. Nevertheless, activation of FAT10 by AOS1/UBA2 does not lead to covalent conjugation of FAT10 with substrate proteins which relies on its cognate E1 enzyme UBA6. Hence, we report that one ubiquitin-like modifier (FAT10) inhibits the conjugation and function of another ubiquitin-like modifier (SUMO) by impairing its activation.


Assuntos
Proteína da Leucemia Promielocítica/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Proteína SUMO-1/metabolismo , Ubiquitinas/metabolismo , Regulação para Baixo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Recombinantes , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fatores de Transcrição/metabolismo , Enzimas Ativadoras de Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/metabolismo , Ubiquitinação , Ubiquitinas/genética
20.
J Immunol ; 203(7): 1776-1785, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31484727

RESUMO

MECL-1 (ß2i), LMP2 (ß1i), and LMP7 (ß5i) are the proteolytically active subunits of the immunoproteasome (IP), a special type of proteasome mainly expressed in hematopoietic cells. Targeting the IP in autoimmune diseases proved to be therapeutically effective in preclinical mouse models. In endotoxin-stimulated human PBMCs, IP inhibition reduces the secretion of several proinflammatory cytokines, with the suppression of IL-23 being the most prominent. In this study, we investigated why the production of IL-23, a key mediator of inflammation in autoimmunity, is blocked when the IP is inhibited in LPS-stimulated human PBMCs. CD14+ monocytes could be identified as the main producers of IL-23 in LPS-stimulated PBMCs. We found that IP inhibition with the irreversible LMP7/LMP2 inhibitor ONX 0914 induced apoptosis in CD14+ monocytes, whereas CD4+, CD3+, CD19+, and CD56+ cells remained unaffected. A high expression of IPs renders monocytes susceptible to IP inhibition, leading to an accumulation of polyubiquitylated proteins and the induction of the unfolded protein response. Similar to IP inhibition, inducers of the unfolded protein response selectively kill CD14+ monocytes in human PBMCs. The blockage of the translation in CD14+ monocytes protects these cells from ONX 0914-induced cell death, indicating that the IP is required to maintain protein turnover in monocytes. Taken together, our data reveal why IP inhibition is particularly effective in the suppression of IL-23-driven autoimmunity.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Interleucina-23/imunologia , Receptores de Lipopolissacarídeos/imunologia , Monócitos/imunologia , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/imunologia , Inibidores de Proteassoma/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Regulação Enzimológica da Expressão Gênica/imunologia , Humanos , Lipopolissacarídeos/toxicidade , Monócitos/patologia , Biossíntese de Proteínas/imunologia , Proteólise/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...