Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166340, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35032596

RESUMO

BACKGROUND: Visual outcome of patients with neovascular age-related macular degeneration has significantly improved during the last years following the introduction of anti-vascular endothelial growth factor (VEGF) therapy. However, about one third of patients show persistent exudation and decreasing visual acuity despite recurrent anti-VEGF treatment, which implies a role of other, still unknown proangiogenic mediators. METHODS: The present study applied transcriptional profiling of human and mouse (C57BL/6J wildtype) choroidal neovascularization (CNV) membranes each with reference to healthy control tissue to identify yet unrecognized mediators of CNV formation. Key factors were further investigated by immunohistochemistry as well as by intravitreal inhibition experiments and multiplex protein assays in the laser-induced CNV mouse model. FINDINGS: Transcriptional profiles of CNV membranes were characterized by enhanced activation of blood vessel development, cytoskeletal organization, and cytokine production, with angiogenesis and wound healing processes predominating in humans and activation of immune processes in mice. Besides several species-specific factors, 95 phylogenetically conserved CNV-associated genes were detected, among which fibroblast growth factor inducible-14 (FN14), a member of the tumor necrosis factor (TNF) receptor family, was identified as a key player of CNV formation. Blocking the pathway by intravitreal injection of a FN14 decoy receptor modulated the cytokine profile - most notably IL-6 - and led to a significant reduction of CNV size in vivo. INTERPRETATION: This study characterizes the transcriptome of human and mouse CNV membranes in an unprejudiced manner and identifies FN14 as a phylogenetically conserved mediator of CNV formation and a promising new therapeutic target for neovascular AMD. FUNDING: This study was funded by the Helmut Ecker Foundation and the Volker Homann Foundation.


Assuntos
Corioide/metabolismo , Neovascularização de Coroide/metabolismo , Degeneração Macular/patologia , Receptor de TWEAK/metabolismo , Transcriptoma , Animais , Lâmina Basilar da Corioide/metabolismo , Estudos de Casos e Controles , Corioide/patologia , Neovascularização de Coroide/etiologia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Lasers/efeitos adversos , Ligantes , Degeneração Macular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Filogenia , Receptor de TWEAK/antagonistas & inibidores , Receptor de TWEAK/classificação , Receptor de TWEAK/genética , Regulação para Cima
2.
Sci Signal ; 11(541)2018 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-30065029

RESUMO

Members of the family of nuclear factor κB (NF-κB) transcription factors are critical for multiple cellular processes, including regulating innate and adaptive immune responses, cell proliferation, and cell survival. Canonical NF-κB complexes are retained in the cytoplasm by the inhibitory protein IκBα, whereas noncanonical NF-κB complexes are retained by p100. Although activation of canonical NF-κB signaling through the IκBα kinase complex is well studied, few regulators of the NF-κB-inducing kinase (NIK)-dependent processing of noncanonical p100 to p52 and the subsequent nuclear translocation of p52 have been identified. We discovered a role for cyclin-dependent kinase 12 (CDK12) in transcriptionally regulating the noncanonical NF-κB pathway. High-content phenotypic screening identified the compound 919278 as a specific inhibitor of the lymphotoxin ß receptor (LTßR), and tumor necrosis factor (TNF) receptor superfamily member 12A (FN14)-dependent nuclear translocation of p52, but not of the TNF-α receptor-mediated nuclear translocation of p65. Chemoproteomics identified CDK12 as the target of 919278. CDK12 inhibition by 919278, the CDK inhibitor THZ1, or siRNA-mediated knockdown resulted in similar global transcriptional changes and prevented the LTßR- and FN14-dependent expression of MAP3K14 (which encodes NIK) as well as NIK accumulation by reducing phosphorylation of the carboxyl-terminal domain of RNA polymerase II. By coupling a phenotypic screen with chemoproteomics, we identified a pathway for the activation of the noncanonical NF-κB pathway that could serve as a therapeutic target in autoimmunity and cancer.


Assuntos
Antineoplásicos/farmacologia , Quinases Ciclina-Dependentes/metabolismo , Regulação Neoplásica da Expressão Gênica , NF-kappa B/metabolismo , Osteossarcoma/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Ciclinas/genética , Ciclinas/metabolismo , Perfilação da Expressão Gênica , Ensaios de Triagem em Larga Escala , Humanos , Indóis/farmacologia , Receptor beta de Linfotoxina/antagonistas & inibidores , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Subunidade p52 de NF-kappa B/genética , Subunidade p52 de NF-kappa B/metabolismo , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Propionatos/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteoma , Transdução de Sinais , Receptor de TWEAK/antagonistas & inibidores , Receptor de TWEAK/genética , Receptor de TWEAK/metabolismo , Células Tumorais Cultivadas , Quinase Induzida por NF-kappaB
3.
J Cell Mol Med ; 22(9): 4344-4353, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29971943

RESUMO

Atrial myocyte hypertrophy is one of the most important substrates in the development of atrial fibrillation (AF). The TWEAK/Fn14 axis is a positive regulator of cardiac hypertrophy in cardiomyopathy. This study therefore investigated the effects of Fn14 on atrial hypertrophy and underlying cellular mechanisms using HL-1 atrial myocytes. In patients with AF, Fn14 protein levels were higher in atrial myocytes from atrial appendages, and expression of TWEAK was increased in peripheral blood mononuclear cells, while TWEAK serum levels were decreased. In vitro, Fn14 expression was up-regulated in response to TWEAK treatment in HL-1 atrial myocytes. TWEAK increased the expression of ANP and Troponin T, and Fn14 knockdown counteracted the effect. Inhibition of JAK2, STAT3 by specific siRNA attenuated TWEAK-induced HL-1 atrial myocytes hypertrophy. In conclusion, TWEAK/Fn14 axis mediates HL-1 atrial myocytes hypertrophy partly through activation of the JAK2/STAT3 pathway.


Assuntos
Fibrilação Atrial/genética , Cardiomegalia/genética , Citocina TWEAK/genética , Janus Quinase 2/genética , Miócitos Cardíacos/metabolismo , Fator de Transcrição STAT3/genética , Receptor de TWEAK/genética , Idoso , Animais , Fibrilação Atrial/metabolismo , Fibrilação Atrial/patologia , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Estudos de Casos e Controles , Citocina TWEAK/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Miócitos Cardíacos/patologia , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Receptor de TWEAK/antagonistas & inibidores , Receptor de TWEAK/metabolismo , Troponina T/genética , Troponina T/metabolismo
4.
Cell Death Dis ; 9(8): 801, 2018 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-30038329

RESUMO

Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a multifunctional cytokine that acts through its receptor fibroblast growth factor-inducible 14 (Fn14). Recent studies demonstrated that the TWEAK/Fn14 signals participate in the development of psoriasis. The purpose of this study was to further explore the effect of Fn14 inhibition on experimental psoriasis. Psoriasis-like skin disease was induced in the wild-type and Fn14-knockout BALB/c mice. We found that Fn14 deficiency ameliorates psoriasis-like lesion in this model, accompanied by less inflammatory cell infiltration and proinflammatory cytokine production in lesional skin. The cutaneous expression of TNF receptor type 2 also decreased in the Fn14-deficient mice. Moreover, the topical application of TWEAK exacerbated psoriatic lesion in the wild-type but not in the Fn14-deficient mice. Furthermore, TWEAK promoted the expression of interleukin 8, keratin 17, and epidermal growth factor receptor (EGFR) but inhibited the expression of involucrin in psoriatic keratinocytes in vitro. Interestingly, such effect of TWEAK was abrogated by an EGFR inhibitor (erlotinib). TWEAK also enhances the proliferation and interleukin-6 production of dermal microvascular endothelial cells under psoriatic condition. In conclusion, TWEAK/Fn14 signals contribute to the development of psoriasis, and involves the modulation of resident cells and the transduction of the EGFR pathway. Fn14 inhibition might be a novel therapeutic strategy for patients with psoriasis.


Assuntos
Psoríase/patologia , Receptor de TWEAK/genética , Animais , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL5/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Cloridrato de Erlotinib/farmacologia , Cloridrato de Erlotinib/uso terapêutico , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Queratina-17/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Psoríase/tratamento farmacológico , Psoríase/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Pele/metabolismo , Pele/patologia , Receptor de TWEAK/antagonistas & inibidores , Receptor de TWEAK/metabolismo , Regulação para Cima/efeitos dos fármacos
5.
MAbs ; 10(1): 62-70, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29190188

RESUMO

Discovery of the upregulation of fibroblast growth factor-inducible-14 (Fn14) receptor following tissue injury has prompted investigation into biotherapeutic targeting of the Fn14 receptor for the treatment of conditions such as chronic kidney diseases. In the development of monoclonal antibody (mAb) therapeutics, there is an increasing trend to use biomeasures combined with mechanistic pharmacokinetic/pharmacodynamic (PK/PD) modeling to enable decision making in early discovery. With the aim of guiding preclinical efforts on designing an antibody with optimized properties, we developed a mechanistic site-of-action (SoA) PK/PD model for human application. This model incorporates experimental biomeasures, including concentration of soluble Fn14 (sFn14) in human plasma and membrane Fn14 (mFn14) in human kidney tissue, and turnover rate of human sFn14. Pulse-chase studies using stable isotope-labeled amino acids and mass spectrometry indicated the sFn14 half-life to be approximately 5 hours in healthy volunteers. The biomeasures (concentration, turnover) of sFn14 in plasma reveals a significant hurdle in designing an antibody against Fn14 with desired characteristics. The projected dose (>1 mg/kg/wk for 90% target coverage) derived from the human PK/PD model revealed potential high and frequent dosing requirements under certain conditions. The PK/PD model suggested a unique bell-shaped relationship between target coverage and antibody affinity for anti-Fn14 mAb, which could be applied to direct the antibody engineering towards an optimized affinity. This investigation highlighted potential applications, including assessment of PK/PD risks during early target validation, human dose prediction and drug candidate optimization.


Assuntos
Anticorpos Monoclonais/farmacocinética , Desenvolvimento de Medicamentos/métodos , Nefropatias/tratamento farmacológico , Rim/efeitos dos fármacos , Modelos Biológicos , Receptor de TWEAK/antagonistas & inibidores , Anticorpos Monoclonais/efeitos adversos , Esquema de Medicação , Cálculos da Dosagem de Medicamento , Estudos de Viabilidade , Humanos , Rim/imunologia , Rim/metabolismo , Nefropatias/sangue , Nefropatias/imunologia , Medição de Risco , Fatores de Risco , Receptor de TWEAK/sangue , Receptor de TWEAK/imunologia
6.
World J Gastroenterol ; 23(13): 2294-2307, 2017 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-28428709

RESUMO

AIM: To clarify the roles of TWEAK and its receptor Fn14 in 5-fluorouracil (5-FU)-induced diarrhea. METHODS: Diarrhea was induced in wild-type (WT), Fn14 knockout (KO), and IL-13 receptor (IL-13R)α1 KO BALB/c mice using a single injection of 5-FU. Histological analysis, cytokine analysis, and flow cytometry was performed on ileal tissues and cells. Murine colon carcinoma-bearing mice were co-treated with an anti-TWEAK antibody and 5-FU. Embryonic fibroblast response to cytokines was also analyzed. RESULTS: 5-FU induced high Fn14 expression in epithelial cells. The severity of 5-FU-induced diarrhea was lower in Fn14 KO mice compared with WT mice. Administration of anti-TWEAK antibody reduced 5-FU-induced diarrhea without affecting the antitumor effects of 5-FU in vivo. 5-FU-induced expression of IL-13, IL-17A, TNF-α, and IFN-γ in the ileum was Fn14 dependent. The severity of 5-FU-induced diarrhea was lower in IL-13Rα1 KO mice, indicating major role for IL-13 signaling via IL-13Rα1 in pathogenesis. We found that IL-13Rα2, an IL-13 neutralizing/cell protective receptor, was strongly induced by IL-33 in vitro and in vivo. IL-13Rα2 was upregulated in the ileum of 5-FU-treated Fn14 KO mice. Thus, the deletion of Fn14 upregulated IL-13Rα2 expression, which reduced IL-13 expression and activity. CONCLUSION: Disruption of the TWEAK/Fn14 pathway affects several interconnected pathways, including those associated with IL-13, IL-33, and IL-13Rα2, to attenuate 5-FU-induced intestinal side effects.


Assuntos
Antimetabólitos Antineoplásicos/efeitos adversos , Citocina TWEAK/metabolismo , Diarreia/induzido quimicamente , Fluoruracila/efeitos adversos , Receptor de TWEAK/metabolismo , Animais , Linhagem Celular Tumoral , Citocina TWEAK/antagonistas & inibidores , Diarreia/metabolismo , Diarreia/prevenção & controle , Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Interleucina-33/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptor de TWEAK/antagonistas & inibidores
7.
Mol Neurobiol ; 54(9): 7497-7506, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-27822714

RESUMO

The aim of our research is to investigate the regulatory role of TNF-like weak inducer of apoptosis- fibroblast growth factor-inducible 14 (TWEAK-Fn14) pathway in nuclear factor-kappa B (NF-κB) expression and neurogenesis status after spinal cord injury (SCI). We constructed a mice model of spinal cord injury and injected different lentiviral vectors which were transfected with TWEAK, TWEAK small interfering RNA (siRNA) and Fn14 siRNA into different groups of mice. Locomotor functional recovery status of the hind limb in mice was assessed using the Basso, Beattie and Bresnahan (BBB) test. Apoptosis status in the injured area was examined via TDT-mediated dUTP-biotin nick end-labeling (TUNEL) staining, the expression of GAP-43 in injured spinal cord was quantified by immunohistochemistry and expressions of TWEAK, Fn14, NF-κB, TNF-α, and IL-1ß were evaluated by either western blot or ELISA. The expressions of TWEAK, Fn14, and NF-κB in the model group were significantly higher compared with those in the control group. Furthermore, the TWEAK group in which TWEAK was overexpressed exhibited significantly higher expressions of TWEAK, Fn14, and NF-κB, TNF-α and IL-1ß in relation to those in the model group (P < 0.05 for all). Moreover, the transfection of Fn14 siRNA antagonized the above effect of TWEAK transfection on injured mice. On the other hand, the TWEAK siRNA group in which the expression of TWEAK was inhibited exhibited significantly lower expressions of TWEAK, Fn14, NF-κB, TNF-α, and IL-1ß (P < 0.05 for all). Moreover, the transfection of TWEAK siRNA enhanced the locomotor functional recovery status in injured mice and suppressed the apoptosis of injured areas (P < 0.05 for all). In conclusion, stimulating the TWEAK-Fn14 pathway may elevate the expression of NF-κB, thereby slow the function recovery of SCI mice whereas inhibiting the TWEAK-Fn14 pathway may improve the neurogenesis status in mice with spinal cord injuries.


Assuntos
NF-kappa B/fisiologia , Neurogênese/fisiologia , Traumatismos da Medula Espinal/metabolismo , Receptor de TWEAK/biossíntese , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismos da Medula Espinal/patologia , Receptor de TWEAK/antagonistas & inibidores
8.
Curr Opin Clin Nutr Metab Care ; 19(4): 316-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27254081

RESUMO

PURPOSE OF REVIEW: Although cancer cachexia is a very significant condition that is present in up to 80% of cancer cases, the cause of the condition has remained a puzzle. Cancer cachexia is a condition which is mainly characterised by muscle wasting, mobilization of fat reserves, and overall metabolic disturbance. This review aims to highlight some of the recent findings in cancer cachexia research. RECENT RESEARCH: It has been recently demonstrated that the expression of a single receptor, fibroblast growth factor-inducible 14, on a tumour can initiate cachexia and that this can be completely ablated by treatment with an antibody against this receptor. Also recently described was the role of parathyroid hormone receptor-binding proteins in causing cachexia through a mechanism where white adipose tissue is replaced with brown adipose tissue. In parallel, work done in drosophila suggests that the impaired insulin signalling is a direct cause of cancer cachexia through the release of an insulin growth factor binding protein that inhibits insulin and insulin-like growth factor 1 signalling. SUMMARY: Successful therapies are urgently needed to combat cancer cachexia in the clinic. Recent research is making progress toward discovering the underlying molecular causes of the condition, which could lead to new therapeutic approaches and in the future contribute to more positive clinical outcomes for cancer sufferers.


Assuntos
Caquexia/etiologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Neoplasias/fisiopatologia , Receptor de TWEAK/metabolismo , Adipogenia/efeitos dos fármacos , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Antineoplásicos/uso terapêutico , Caquexia/metabolismo , Caquexia/prevenção & controle , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptor de TWEAK/antagonistas & inibidores , Receptor de TWEAK/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA