Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cancer Sci ; 115(2): 357-368, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38148492

RESUMEN

Combination immunotherapy with multiple immune checkpoint inhibitors (ICIs) has been approved for various types of malignancies, including malignant pleural mesothelioma (MPM). Podoplanin (PDPN), a transmembrane sialomucin-like glycoprotein, has been investigated as a diagnostic marker and therapeutic target for MPM. We previously generated and developed a PDPN-targeting Ab reagent with high Ab-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). However, the effects of anti-PDPN Abs on various tumor-infiltrating immune cells and their synergistic effects with ICIs have remained unclear. In the present study, we established a novel rat-mouse chimeric anti-mouse PDPN IgG2a mAb (PMab-1-mG2a ) and its core-fucose-deficient Ab (PMab-1-mG2a -f) to address these limitations. We identified the ADCC and CDC activity of PMab-1-mG2a -f against the PDPN-expressing mesothelioma cell line AB1-HA. The antitumor effect of monotherapy with PMab-1-mG2a -f was not sufficient to overcome tumor progression in AB1-HA-bearing immunocompetent mice. However, PMab-1-mG2a -f enhanced the antitumor effects of CTLA-4 blockade. Combination therapy with anti-PDPN Ab and anti-CTLA-4 Ab increased tumor-infiltrating natural killer (NK) cells. The depletion of NK cells inhibited the synergistic effects of PMab-1-mG2a -f and CTLA-4 blockade in vivo. These findings indicated the essential role of NK cells in novel combination immunotherapy targeting PDPN and shed light on the therapeutic strategy in advanced MPM.


Asunto(s)
Mesotelioma Maligno , Mesotelioma , Ratas , Ratones , Animales , Cricetinae , Anticuerpos Monoclonales/uso terapéutico , Antígeno CTLA-4 , Glicoproteínas de Membrana , Mesotelioma/patología , Células Asesinas Naturales/metabolismo , Cricetulus , Células CHO
2.
Am J Respir Crit Care Med ; 207(11): 1498-1514, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36917778

RESUMEN

Rationale: Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease characterized by progressive lung scarring. IPF-related pulmonary vascular remodeling and pulmonary hypertension (PH) result in a particularly poor prognosis. Objectives: To study the pathogenesis of vascular remodeling in fibrotic lungs and its contribution to progression of fibrosis. Methods: We used an experimental model of lung fibrosis associated with PH by transient overexpression of active TGF-ß1 (transforming growth factor-ß1). Samples from patients with fibrotic lung diseases were analyzed in depth using immunostaining, gene expression, and gene mutations. Measurements and Main Results: We found a reduction in endothelial cells (ECs) and activation of vascular smooth muscle cells (VSMCs) in fibrotic lungs. Coculturing fibroblasts with VSMCs or ECs from fibrotic lungs induced fibrotic phenotypes in fibroblasts. IPF fibroblasts induced EC death and activation of VSMCs in coculture systems. Decreased concentrations of BMPR2 (bone morphogenic protein receptor 2) and its signaling were observed in ECs and VSMCs from fibrotic lungs in both rats and humans. On fibroblasts treated with media from VSMCs, BMPR2 suppression in VSMCs led to fibrogenic effects. Tacrolimus activated BMPR2 signaling and attenuated fibrosis and PH in rodent lungs. Whole-exome sequencing revealed rare mutations in PH-related genes, including BMPR2, in patients with IPF undergoing transplantation. A unique missense BMPR2 mutation (p.Q721R) was discovered to have dysfunctional effects on BMPR2 signaling. Conclusions: Endothelial dysfunction and vascular remodeling in PH secondary to pulmonary fibrosis enhance fibrogenesis through impaired BMPR2 signaling. Tacrolimus may have value as a treatment of advanced IPF and concomitant PH. Genetic abnormalities may determine the development of PH in advanced IPF.


Asunto(s)
Hipertensión Pulmonar , Fibrosis Pulmonar Idiopática , Humanos , Ratas , Animales , Remodelación Vascular , Células Endoteliales/metabolismo , Tacrolimus , Pulmón/patología , Fibrosis Pulmonar Idiopática/patología , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Fibroblastos/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética
3.
Cancer Sci ; 114(2): 384-398, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36285504

RESUMEN

Myeloid-derived suppressor cells (MDSCs) have been known to play a pivotal role in the induction of immune tolerance, which limits the benefits of immune checkpoint inhibitors (ICIs). Recent studies revealed that several chemotherapeutic agents decreased tumor-infiltrating MDSCs. Therefore, combination therapy with cytotoxic chemotherapeutic agents and ICIs was approved for first-line treatment for lung cancer. However, the impact of chemotherapeutic agents on MDSCs and an optimal partner of ICIs has not been fully investigated in thoracic tumors, including lung cancer and malignant pleural mesothelioma. In the present study, we found that treatment with 5-FU and its oral formulation, S-1, suppressed tumor progression and inhibited the accumulation of MDSCs in thoracic tumor-bearing mice. Tumor-infiltrating T cells and dendritic cells were significantly expanded in S-1-treated mice. 5-FU suppressed the ability of tumor cells to recruit MDSCs, while it did not suppress the survival and differentiation of mouse MDSCs in vitro. We also revealed that 5-FU or S-1 significantly downregulated the expression of tumor-derived Bv8 and S100A8. The knockdown of Bv8 or S100A8 in tumor cells suppressed tumor growth and MDSC recruitment in vivo. Furthermore, in comparison with pemetrexed, administration of S-1 improved the synergistic therapeutic efficacy of anti-PD-1 antibodies with or without carboplatin. Our findings revealed a novel mechanism wherein S-1 primed a favorable tumor microenvironment to provide the rationale for combination therapy with S-1 and ICIs as the optimal therapy for thoracic cancer.


Asunto(s)
Neoplasias Pulmonares , Células Supresoras de Origen Mieloide , Neoplasias Torácicas , Ratones , Animales , Calgranulina A , Linfocitos T , Neoplasias Torácicas/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Microambiente Tumoral , Línea Celular Tumoral
4.
Respir Res ; 24(1): 148, 2023 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-37269004

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a fatal fibrotic lung disease with few effective therapeutic options. Recently, drug repositioning, which involves identifying novel therapeutic potentials for existing drugs, has been popularized as a new approach for the development of novel therapeutic reagents. However, this approach has not yet been fully utilized in the field of pulmonary fibrosis. METHODS: The present study identified novel therapeutic options for pulmonary fibrosis using a systematic computational approach for drug repositioning based on integration of public gene expression signatures of drug and diseases (in silico screening approach). RESULTS: Among the top compounds predicted to be therapeutic for IPF by the in silico approach, we selected BI2536, a polo-like kinase (PLK) 1/2 inhibitor, as a candidate for treating pulmonary fibrosis using an in silico analysis. However, BI2536 accelerated mortality and weight loss rate in an experimental mouse model of pulmonary fibrosis. Because immunofluorescence staining revealed that PLK1 expression was dominant in myofibroblasts while PLK2 expression was dominant in lung epithelial cells, we next focused on the anti-fibrotic effect of the selective PLK1 inhibitor GSK461364. Consequently, GSK461364 attenuated pulmonary fibrosis with acceptable mortality and weight loss in mice. CONCLUSIONS: These findings suggest that targeting PLK1 may be a novel therapeutic approach for pulmonary fibrosis by inhibiting lung fibroblast proliferation without affecting lung epithelial cells. In addition, while in silico screening is useful, it is essential to fully determine the biological activities of candidates by wet-lab validation studies.


Asunto(s)
Reposicionamiento de Medicamentos , Fibrosis Pulmonar Idiopática , Ratones , Animales , Tiofenos/uso terapéutico , Bencimidazoles/metabolismo , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Pulmón/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Fibroblastos/metabolismo , Bleomicina/farmacología
5.
Oncologist ; 27(7): e554-e560, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35325241

RESUMEN

BACKGROUND: Rash eruptions are a common side-effect of pemetrexed, for which the administration of 8 mg/day of dexamethasone for 3 days from the day preceding pemetrexed administration is recommended. This study aimed to prospectively assess the effectiveness of prophylactic administration of low-dose dexamethasone for pemetrexed-induced rashes. METHODS: This single-arm, phase II study recruited patients with non-squamous non-small cell lung cancer and malignant pleural mesothelioma scheduled to receive chemotherapy including pemetrexed. Patients received 2 mg of dexamethasone daily from days 2 to 6 after chemotherapy with pemetrexed. The primary endpoint was the 3-week incidence of rash eruptions. RESULTS: Twenty-five patients were enrolled between September 2017 and May 2019. The incidence of rash after 3 weeks was 16.7%. Rashes erupted mainly on the upper half of the body, such as the chest and neck, and were of grades 1 and 2 in 2 patients each. No rashes of grade 3 or higher were observed, and there were no adverse events associated with additional corticosteroids. CONCLUSION: Prophylactic administration of low-dose dexamethasone for 5 days from the day after pemetrexed administration resulted in a milder incidence and severity of rash. These findings may provide a standard preventative strategy for pemetrexed-induced rashes. (Trial identifier: UMIN000025666).


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Dexametasona , Exantema , Neoplasias Pulmonares , Mesotelioma Maligno , Pemetrexed , Corticoesteroides/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/complicaciones , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino , Dexametasona/uso terapéutico , Exantema/inducido químicamente , Exantema/tratamiento farmacológico , Exantema/prevención & control , Humanos , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/tratamiento farmacológico , Mesotelioma Maligno/complicaciones , Mesotelioma Maligno/tratamiento farmacológico , Pemetrexed/efectos adversos
6.
Thorax ; 76(9): 895-906, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33859055

RESUMEN

RATIONALE: Extracellular vesicles (EVs) are small lipid vesicles, and EV-coupled microRNAs (miRNAs) are important modulators of biological processes. Fibrocytes are circulating bone marrow-derived cells that migrate into the injured lungs and contribute to fibrogenesis. The question of whether EV-coupled miRNAs derived from fibrocytes are able to regulate pulmonary fibrosis has not been addressed yet. METHODS: Pulmonary fibrosis was induced in rats by intratracheal administration of an adenoviral gene vector encoding active transforming growth factor-ß1 (TGF-ß1) or control vector. Primary fibrocytes and fibroblasts were cultured from rat lungs and were sorted by anti-CD45 magnetic beads. Human circulating fibrocytes and fibrocytes in bronchoalveolar lavage fluid (BALF) were isolated by fibronectin-coated dishes. Fibrocytes were cultured on different stiffness plates or decellularised lung scaffolds. We also determined the effects of extracellular matrix (ECM) and recombinant TGF-ß1 on the cellular and EV-coupled miRNA expression of fibrocytes. RESULTS: The EVs of fibrocytes derived from fibrotic lungs significantly upregulated the expression of col1a1 of fibroblasts. Culturing on rigid plates or fibrotic decellularised lung scaffolds increased miR-21-5 p expression compared with soft plates or normal lung scaffolds. Dissolved ECM collected from fibrotic lungs and recombinant TGF-ß1 increased miR-21-5 p expression on fibrocytes, and these effects were attenuated on soft plates. Fibrocytes from BALF collected from fibrotic interstitial pneumonia patients showed higher miR-21-5 p expression than those from other patients. CONCLUSIONS: Our results indicate that ECM contributes to fibrogenesis through biomechanical and biochemical effects on miRNA expression in fibrocytes.


Asunto(s)
Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , MicroARNs/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Vesículas Extracelulares/metabolismo , Humanos , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba
7.
Am J Respir Cell Mol Biol ; 63(3): 317-326, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32338990

RESUMEN

FGFs (fibroblast growth factors) are major factors associated with the pathogenesis of pulmonary fibrosis. On the one hand, nintedanib, a tyrosine kinase inhibitor targeting several growth factor receptors, including the FGF receptor (FGFR), has been approved for the treatment of idiopathic pulmonary fibrosis. On the other hand, recent reports suggest that FGFs are required for epithelial recovery. In this study, we focused on FGF signaling to both fibroblasts and alveolar epithelial cells (AECs), and we examined the effect of a pan-FGFR blocker on experimental pulmonary fibrosis in mice. The effects of BGJ398, a pan-FGFR inhibitor, on the migration and proliferation of fibroblasts and AECs were assessed using Transwell migration or [3H]thymidine incorporation assays. The expression of FGFR was analyzed using IB or flow cytometry. We also investigated the effect of BGJ398 on pulmonary fibrosis induced by bleomycin in mice. Both lung fibroblasts and AECs expressed FGFRs. BGJ398 significantly inhibited the proliferation and migration of lung fibroblasts stimulated with FGF2. BGJ398 also reduced the proliferation of AECs in response to FGF2. Although the administration of BGJ398 ameliorated pulmonary fibrosis in bleomycin-treated mice, it increased mortality resulting from alveolar injury and inhibition of AEC regeneration. These data suggest that the total inhibition of FGFR signaling can suppress lung fibrosis by inhibiting fibroblast activities, although alveolar injury is simultaneously caused.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Indoles/farmacología , Pulmón/efectos de los fármacos , Receptores de Factores de Crecimiento de Fibroblastos/efectos de los fármacos , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Ratones , Fosforilación , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Am J Respir Cell Mol Biol ; 60(4): 478-487, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30540913

RESUMEN

The signaling pathways of growth factors, including platelet-derived growth factor, can be considered specific targets for overcoming the poor prognosis of idiopathic pulmonary fibrosis. Nintedanib, the recently approved multiple kinase inhibitor, has shown promising antifibrotic effects in patients with idiopathic pulmonary fibrosis; however, its efficacy is still limited, and in some cases, treatment discontinuation is necessary owing to toxicities such as gastrointestinal disorders. Therefore, more effective agents with less toxicity are still needed. TAS-115 is a novel multiple tyrosine kinase inhibitor that preferably targets platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor, and c-FMS in addition to other molecules. In this study, we evaluated the antifibrotic effect of TAS-115 on pulmonary fibrosis in vitro and in vivo. TAS-115 inhibited the phosphorylation of PDGFR on human lung fibroblast cell line MRC-5 cells and suppressed their platelet-derived growth factor-induced proliferation and migration. Furthermore, TAS-115 inhibited the phosphorylation of c-FMS, a receptor of macrophage colony-stimulating factor, in murine bone marrow-derived macrophages and decreased the production of CCL2, another key molecule for inducing pulmonary fibrosis, under the stimulation of macrophage colony-stimulating factor. Importantly, the inhibitory effects of TAS-115 on both PDGFR and c-FMS were 3- to 10-fold higher than those of nintedanib. In a mouse model of bleomycin-induced pulmonary fibrosis, TAS-115 significantly inhibited the development of pulmonary fibrosis and the collagen deposition in bleomycin-treated lungs. These data suggest that strong inhibition of PDGFR and c-FMS by TAS-115 may be a promising strategy for overcoming the intractable pathogenesis of pulmonary fibrosis.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Quinolinas/farmacología , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Tiourea/análogos & derivados , Animales , Bleomicina/toxicidad , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Tiourea/farmacología
9.
Thorax ; 74(5): 455-465, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30808717

RESUMEN

BACKGROUND: The role of mast cells accumulating in idiopathic pulmonary fibrosis (IPF) lungs is unknown. OBJECTIVES: We investigated the effect of fibrotic extracellular matrix (ECM) on mast cells in experimental and human pulmonary fibrosis. RESULTS: In IPF lungs, mast cell numbers were increased and correlated with disease severity (control vs 60%90% vs 60%90% vs FVC<60%, mean difference=-268.6, 95% CI of difference -441.0 to -96.17, p=0.0007). Plasma tryptase levels were increased in IPF and negatively correlated with FVC (control vs FVC<60%, mean difference=-17.12, 95% CI of difference -30.02 to -4.22, p=0.006: correlation curves R=-0.045, p=0.025). In a transforming growth factor (TGF)-ß1-induced pulmonary fibrosis model, chymase-positive and tryptase-positive mast cells accumulated in fibrotic lung. Lung tissue was decellularised and reseeded with bone marrow or peritoneum-derived mast cells; cells on fibrotic ECM released more TGF-ß1 compared with normal ECM (active TGF-ß1: bone marrow-derived mast cell (BMMC)-DL vs BMMC-TGF-ß1 p=0.0005, peritoneal mast cell (PMC)-DL vs PMC-TGF-ß1 p=0.0003, total TGF-ß1: BMMC-DL vs BMMC-TGF-ß1 p=0.013, PMC-DL vs PMC-TGF-ß1 p=0.001). Mechanical stretch of lungs caused mast cell degranulation; mast cell stabilisers inhibited degranulation (histamine: cont vs doxantrazole p=0.004, ß-hexosaminidase: cont vs doxantrazole, mean difference=1.007, 95% CI of difference 0.2700 to 1.744, p=0.007) and TGF-ß1 activation (pSmad2/Smad2: cont vs dox p=0.006). Cromoglycate attenuated pulmonary fibrosis in rats (collagen: phosphate-buffered saline (PBS) vs cromoglycate p=0.036, fibrotic area: PBS vs cromoglycate p=0.031). CONCLUSION: This study suggests that mast cells may contribute to the progression of pulmonary fibrosis.


Asunto(s)
Degranulación de la Célula , Pulmón/patología , Mastocitos/fisiología , Fibrosis Pulmonar/metabolismo , Estrés Mecánico , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Pulmón/metabolismo , Fibrosis Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal
10.
Exp Lung Res ; 45(7): 188-199, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31298961

RESUMEN

Purpose/Aim of the Study: Wnt/ß-catenin signaling was reported to be activated in pulmonary fibrosis, and was focused on as a target for antifibrotic therapy. However, the mechanism how the inhibition of Wnt/ß-catenin signaling ameliorate pulmonary fibrosis has not been fully elucidated. The purpose of this study is to explore the target cells of Wnt/ß-catenin inhibition in pulmonary fibrosis and to examine the antifibrotic effect of the novel inhibitor PRI-724 specifically disrupting the interaction of ß-catenin and CBP. Materials and Methods: The effect of C-82, an active metabolite of PRI-724, on the expression of TGF-ß1 and α-smooth muscle actin (SMA) was examined on fibroblasts and macrophages. We also examined the effects of PRI-724 in mouse model of bleomycin-induced pulmonary fibrosis. Results: The activation and increased accumulation of ß-catenin in the canonical pathway were detected in lung fibroblasts as well as macrophages stimulated by Wnt3a using Western blotting. Treatment with C-82 reduced CBP protein and increased p300 protein binding to ß-catenin in the nucleus of lung fibroblasts. In addition, C-82 inhibited the expression of SMA in lung fibroblasts treated with TGF-ß, indicating the inhibition of myofibroblast differentiation. In the fibrotic lungs induced by bleomycin, ß-catenin was stained strongly in macrophages, but the staining of ß-catenin in alveolar epithelial cells and fibroblasts was weak. The administration of PRI-724 ameliorated pulmonary fibrosis induced by bleomycin in mice when administered with a late, but not an early, treatment schedule. Analysis of bronchoalveolar fluid (BALF) showed a decreased number of alveolar macrophages. In addition, the level of TGF-ß1 in BALF was decreased in mice treated with PRI-724. C-82 also inhibited the production of TGF-ß1 by alveolar macrophages. Conclusions: These results suggest that the ß-catenin/CBP inhibitor PRI-724 is a potent antifibrotic agent that acts by modulating the activity of macrophages in the lungs.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Fibrosis Pulmonar/tratamiento farmacológico , Pirimidinonas/uso terapéutico , beta Catenina/antagonistas & inhibidores , Animales , Bleomicina , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Evaluación Preclínica de Medicamentos , Fibroblastos/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Pirimidinonas/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
11.
Nature ; 499(7456): 97-101, 2013 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-23803760

RESUMEN

Obesity has become more prevalent in most developed countries over the past few decades, and is increasingly recognized as a major risk factor for several common types of cancer. As the worldwide obesity epidemic has shown no signs of abating, better understanding of the mechanisms underlying obesity-associated cancer is urgently needed. Although several events were proposed to be involved in obesity-associated cancer, the exact molecular mechanisms that integrate these events have remained largely unclear. Here we show that senescence-associated secretory phenotype (SASP) has crucial roles in promoting obesity-associated hepatocellular carcinoma (HCC) development in mice. Dietary or genetic obesity induces alterations of gut microbiota, thereby increasing the levels of deoxycholic acid (DCA), a gut bacterial metabolite known to cause DNA damage. The enterohepatic circulation of DCA provokes SASP phenotype in hepatic stellate cells (HSCs), which in turn secretes various inflammatory and tumour-promoting factors in the liver, thus facilitating HCC development in mice after exposure to chemical carcinogen. Notably, blocking DCA production or reducing gut bacteria efficiently prevents HCC development in obese mice. Similar results were also observed in mice lacking an SASP inducer or depleted of senescent HSCs, indicating that the DCA-SASP axis in HSCs has key roles in obesity-associated HCC development. Moreover, signs of SASP were also observed in the HSCs in the area of HCC arising in patients with non-alcoholic steatohepatitis, indicating that a similar pathway may contribute to at least certain aspects of obesity-associated HCC development in humans as well. These findings provide valuable new insights into the development of obesity-associated cancer and open up new possibilities for its control.


Asunto(s)
Senescencia Celular , Ácido Desoxicólico/metabolismo , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Células Estrelladas Hepáticas/metabolismo , Neoplasias Hepáticas/metabolismo , Obesidad/metabolismo , Animales , Antibacterianos/farmacología , Bacterias/metabolismo , Ácidos y Sales Biliares/metabolismo , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/prevención & control , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Citocinas/metabolismo , Daño del ADN/efectos de los fármacos , Ácido Desoxicólico/sangre , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Hígado Graso/complicaciones , Hígado Graso/patología , Tracto Gastrointestinal/efectos de los fármacos , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Interleucina-1beta/deficiencia , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Obesidad/inducido químicamente , Fenotipo , Factores de Riesgo
12.
Am J Respir Cell Mol Biol ; 58(4): 461-470, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29115860

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by excessive deposition of extracellular matrix (ECM) in the lung parenchyma. The abnormal ECM deposition slowly overtakes normal lung tissue, disturbing gas exchange and leading to respiratory failure and death. ECM cross-linking and subsequent stiffening is thought to be a major contributor of disease progression and also promotes the activation of transforming growth factor (TGF)-ß1, one of the main profibrotic growth factors. Lysyl oxidase-like (LOXL) 1 belongs to the cross-linking enzyme family and has been shown to be up-regulated in active fibrotic regions of bleomycin-treated mice and patients with IPF. We demonstrate in this study that LOXL1-deficient mice are protected from experimental lung fibrosis induced by overexpression of TGF-ß1 using adenoviral (Ad) gene transfer (AdTGF-ß1). The lack of LOXL1 prevented accumulation of insoluble cross-linked collagen in the lungs, and therefore limited lung stiffness after AdTGF-ß1. In addition, we applied mechanical stretch to lung slices from LOXL1+/+ and LOXL1-/- mice treated with AdTGF-ß1. Lung stiffness (Young's modulus) of LOXL1-/- lung slices was significantly lower compared with LOXL1+/+ lung slices. Moreover, the release of activated TGF-ß1 after mechanical stretch was significantly lower in LOXL1-/- mice compared with LOXL1+/+ mice after AdTGF-ß1. These data support the concept that cross-linking enzyme inhibition represents an interesting therapeutic target for drug development in IPF.


Asunto(s)
Adenoviridae/genética , Aminoácido Oxidorreductasas/deficiencia , Colágeno/metabolismo , Técnicas de Transferencia de Gen , Fibrosis Pulmonar Idiopática/prevención & control , Pulmón/enzimología , Factor de Crecimiento Transformador beta1/genética , Adenoviridae/metabolismo , Aminoácido Oxidorreductasas/genética , Animales , Modelos Animales de Enfermedad , Módulo de Elasticidad , Fibrosis Pulmonar Idiopática/enzimología , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Pulmón/fisiopatología , Rendimiento Pulmonar , Mecanotransducción Celular , Ratones Noqueados , Receptores de Estiramiento Pulmonares/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Regulación hacia Arriba
13.
Eur Respir J ; 52(2)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29976656

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive disease with an unknown cause. Two drugs, nintedanib and pirfenidone, have been shown to slow, but not stop, disease progression. Pulmonary hypertension (PH) is a frequent complication in IPF patients and is associated with poor prognosis. Macitentan is a dual endothelin receptor antagonist that is approved for pulmonary arterial hypertension treatment. We hypothesised that using macitentan to treat animals with pulmonary fibrosis induced by adenoviral vector encoding biologically active transforming growth factor-ß1 (AdTGF-ß1) would improve the PH caused by chronic lung disease and would limit the progression of fibrosis.Rats (Sprague Dawley) which received AdTGF-ß1 were treated by daily gavage of macitentan (100 mg·kg-1·day-1), pirfenidone (0.5% food admix) or a combination from day 14 to day 28. Pulmonary artery pressure (PAP) was measured before the rats were killed, and fibrosis was subsequently evaluated by morphometric measurements and hydroxyproline analysis.AdTGF-ß1 induced pulmonary fibrosis associated with significant PH. Macitentan reduced the increase in PAP and both macitentan and pirfenidone stopped fibrosis progression from day 14 to day 28. Macitentan protected endothelial cells from myofibroblast differentiation and apoptosis whereas pirfenidone only protected against fibroblast-to-myofibroblast differentiation. Both drugs induced apoptosis of differentiated myofibroblasts in vitro and in vivoOur results demonstrate that dual endothelin receptor antagonism was effective in both PH and lung fibrosis whereas pirfenidone only affected fibrosis.


Asunto(s)
Hipertensión Pulmonar/tratamiento farmacológico , Miofibroblastos/efectos de los fármacos , Fibrosis Pulmonar/patología , Pirimidinas/farmacología , Sulfonamidas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Progresión de la Enfermedad , Femenino , Humanos , Hipertensión Pulmonar/inducido químicamente , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Masculino , Miofibroblastos/metabolismo , Fibrosis Pulmonar/inducido químicamente , Piridonas/farmacología , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/farmacología
14.
Eur Respir J ; 51(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29386344

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive disease of the lung parenchyma, causing significant morbidity through worsening dyspnoea and overall functional decline. IPF is characterised by apoptosis-resistant myofibroblasts, which are a major source for the excessive production of extracellular matrix (ECM) overtaking normal lung tissue. We sought to study the role of heat shock protein (HSP) isoforms HSP90α and HSP90ß, whose distinct roles in lung fibrogenesis remain elusive.We determined the level of circulating HSP90α in IPF patients (n=31) and age-matched healthy controls (n=9) by ELISA. The release of HSP90α and HSP90ß was evaluated in vitro in primary IPF and control lung fibroblasts and ex vivo after mechanical stretch on fibrotic lung slices from rats receiving adenovector-mediated transforming growth factor-ß1.We demonstrate that circulating HSP90α is upregulated in IPF patients in correlation with disease severity. The release of HSP90α is enhanced by the increase in mechanical stress of the fibrotic ECM. This increase in extracellular HSP90α signals through low-density lipoprotein receptor-related protein 1 (LRP1) to promote myofibroblast differentiation and persistence. In parallel, we demonstrate that the intracellular form of HSP90ß stabilises LRP1, thus amplifying HSP90α extracellular action.We believe that the specific inhibition of extracellular HSP90α is a promising therapeutic strategy to reduce pro-fibrotic signalling in IPF.


Asunto(s)
Matriz Extracelular/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Pulmón/patología , Miofibroblastos/metabolismo , Animales , Estudios de Casos y Controles , Células Cultivadas , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Glicoproteínas de Membrana , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Regulación hacia Arriba
15.
Respir Res ; 18(1): 172, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28915889

RESUMEN

BACKGROUND: Nintedanib, a tyrosine kinase inhibitor that is specific for platelet-derived growth factor receptors (PDGFR), fibroblast growth factor receptors (FGFR), and vascular endothelial growth factor receptors (VEGFR), has recently been approved for idiopathic pulmonary fibrosis. Fibrocytes are bone marrow-derived progenitor cells that produce growth factors and contribute to fibrogenesis in the lungs. However, the effects of nintedanib on the functions of fibrocytes remain unclear. METHODS: Human monocytes were isolated from the peripheral blood of healthy volunteers. The expression of growth factors and their receptors in fibrocytes was analyzed using ELISA and Western blotting. The effects of nintedanib on the ability of fibrocytes to stimulate lung fibroblasts were examined in terms of their proliferation. The direct effects of nintedanib on the differentiation and migration of fibrocytes were also assessed. We investigated whether nintedanib affected the accumulation of fibrocytes in mouse lungs treated with bleomycin. RESULTS: Human fibrocytes produced PDGF, FGF2, and VEGF-A. Nintedanib and specific inhibitors for each growth factor receptor significantly inhibited the proliferation of lung fibroblasts stimulated by the supernatant of fibrocytes. Nintedanib inhibited the migration and differentiation of fibrocytes induced by growth factors in vitro. The number of fibrocytes in the bleomycin-induced lung fibrosis model was reduced by the administration of nintedanib, and this was associated with anti-fibrotic effects. CONCLUSIONS: These results support the role of fibrocytes as producers of and responders to growth factors, and suggest that the anti-fibrotic effects of nintedanib are at least partly mediated by suppression of fibrocyte function.


Asunto(s)
Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Indoles/uso terapéutico , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Indoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
J Immunol ; 190(12): 6239-49, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23690472

RESUMEN

Podoplanin (Aggrus), which is a type I transmembrane sialomucin-like glycoprotein, is highly expressed in malignant pleural mesothelioma (MPM). We previously reported the generation of a rat anti-human podoplanin Ab, NZ-1, which inhibited podoplanin-induced platelet aggregation and hematogenous metastasis. In this study, we examined the antitumor effector functions of NZ-1 and NZ-8, a novel rat-human chimeric Ab generated from NZ-1 including Ab-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity against MPM in vitro and in vivo. Immunostaining with NZ-1 showed the expression of podoplanin in 73% (11 out of 15) of MPM cell lines and 92% (33 out of 36) of malignant mesothelioma tissues. NZ-1 could induce potent ADCC against podoplanin-positive MPM cells mediated by rat NK (CD161a(+)) cells, but not murine splenocytes or human mononuclear cells. Treatment with NZ-1 significantly reduced the growth of s.c. established tumors of MPM cells (ACC-MESO-4 or podoplanin-transfected MSTO-211H) in SCID mice, only when NZ-1 was administered with rat NK cells. In in vivo imaging, NZ-1 efficiently accumulated to xenograft of MPM, and its accumulation continued for 3 wk after systemic administration. Furthermore, NZ-8 preferentially recognized podoplanin expressing in MPM, but not in normal tissues. NZ-8 could induce higher ADCC mediated by human NK cells and complement-dependent cytotoxicity as compared with NZ-1. Treatment with NZ-8 and human NK cells significantly inhibited the growth of MPM cells in vivo. These results strongly suggest that targeting therapy to podoplanin with therapeutic Abs (i.e., NZ-8) derived from NZ-1 might be useful as a novel immunotherapy against MPM.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunoterapia/métodos , Glicoproteínas de Membrana/inmunología , Mesotelioma/inmunología , Neoplasias Pleurales/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones SCID , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Am J Respir Cell Mol Biol ; 51(6): 793-801, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24885373

RESUMEN

Circulating fibrocytes have been reported to migrate into the injured lungs, and contribute to fibrogenesis via CXCL12-CXCR4 axis. In contrast, we report that imatinib mesylate prevented bleomycin (BLM)-induced pulmonary fibrosis in mice by inhibiting platelet-derived growth factor receptor (PDGFR), even when it was administered only in the early phase. The goal of this study was to test the hypothesis that platelet-derived growth factor (PDGF) might directly contribute to the migration of fibrocytes to the injured lungs. PDGFR expression in fibrocytes was examined by flow cytometry and RT-PCR. The migration of fibrocytes was evaluated by using a chemotaxis assay for human fibrocytes isolated from peripheral blood. The numbers of fibrocytes triple-stained for CD45, collagen-1, and CXCR4 were also examined in lung digests of BLM-treated mice. PDGFR mRNA levels in fibrocytes isolated from patients with idiopathic pulmonary fibrosis were investigated by real-time PCR. Fibrocytes expressed both PDGFR-α and -ß, and migrated in response to PDGFs. PDGFR inhibitors (imatinib, PDGFR-blocking antibodies) suppressed fibrocyte migration in vitro, and reduced the number of fibrocytes in the lungs of BLM-treated mice. PDGF-BB was a stronger chemoattractant than the other PDGFs in vitro, and anti-PDGFR-ß-blocking antibody decreased the numbers of fibrocytes in the lungs compared with anti-PDGFR-α antibody in vivo. Marked expression of PDGFR-ß was observed in fibrocytes from patients with idiopathic pulmonary fibrosis compared with healthy subjects. These results suggest that PDGF directly functions as a strong chemoattractant for fibrocytes. In particular, the PDGF-BB-PDGFR-ß biological axis might play a critical role in fibrocyte migration into the fibrotic lungs.


Asunto(s)
Factor de Crecimiento Derivado de Plaquetas/fisiología , Fibrosis Pulmonar/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Benzamidas/administración & dosificación , Estudios de Casos y Controles , Quimiotaxis , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Mesilato de Imatinib , Inyecciones Intraperitoneales , Ratones Endogámicos C57BL , Piperazinas/administración & dosificación , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Pirimidinas/administración & dosificación , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Transducción de Señal
18.
Lung ; 192(5): 711-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24880792

RESUMEN

PURPOSE: Idiopathic pleuroparenchymal fibroelastosis (IPPFE) is a recently reported rare disease entity characterized by fibrotic thickening of the pleural and subpleural parenchyma predominantly in the upper lobes in idiopathic interstitial pneumonias (IIPs). Because the clinical features of this rare disease are not fully elucidated, we examined the clinical characteristics of IPPFE, especially for serum interstitial biomarkers, surfactant protein-D (SP-D), and Krebs von den Lungen-6 (KL-6). METHODS AND RESULTS: Four consecutive cases of IPPFE who fulfilled the diagnostic criteria were studied. All cases were more than 60 years of age, and were classified as underweight by body mass index. A severe restrictive ventilatory defect was found in all cases on admission. High-resolution computed tomography showed intense pleural thickening associated with fibrosis predominant in upper lobes. Histopathological findings were also confirmed in three out of four cases. Interestingly, the serum level of SP-D was markedly elevated in all cases, while KL-6 was within normal range in three out of four cases. As compared with major IIPs such as idiopathic pulmonary fibrosis and fibrotic nonspecific interstitial pneumonia, IPPFE significantly showed higher frequency of cases with a unique pattern of serum biomarkers, which is characterized by an elevated level of SP-D with a normal range of KL-6. CONCLUSIONS: In IPPFE, SP-D might tend to be elevated, while KL-6 was within a normal range. Further study is required to determine the pathogenesis and clinical significance of the elevated SP-D in IPPFE.


Asunto(s)
Neumonías Intersticiales Idiopáticas/sangre , Mucina-1/sangre , Enfermedades Pleurales/sangre , Proteína D Asociada a Surfactante Pulmonar/sangre , Anciano , Autopsia , Biomarcadores/sangre , Biopsia , Resultado Fatal , Humanos , Neumonías Intersticiales Idiopáticas/diagnóstico , Neumonías Intersticiales Idiopáticas/fisiopatología , Neumonías Intersticiales Idiopáticas/terapia , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Enfermedades Pleurales/diagnóstico , Enfermedades Pleurales/fisiopatología , Enfermedades Pleurales/terapia , Ventilación Pulmonar , Estudios Retrospectivos , Espirometría , Factores de Tiempo , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Regulación hacia Arriba
19.
J Med Invest ; 71(1.2): 9-22, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38735730

RESUMEN

Patients with interstitial lung disease (ILD), especially those with idiopathic pulmonary fibrosis, are at increased risk of developing lung cancer (LC). Pharmacotherapy for advanced LC has dramatically progressed in recent years;however, management of LC with pre-existing ILD (LC-ILD) is challenging due to serious concerns about the risk of acute exacerbation of ILD (AE-ILD). As patients with LC-ILD have been excluded from most prospective clinical trials of advanced LC, optimal pharmacotherapy remains to be elucidated. Although the antitumor activity of first-line platinum-based cytotoxic chemotherapy appears to be similar in advanced LC patients with or without ILD, its impact on the survival of patients with LC-ILD is limited. Immune checkpoint inhibitors may hold promise for long-term survival, but many challenges remain, including safety and appropriate patient selection. Further understanding the predictive factors for AE-ILD after receiving pharmacotherapy in LC-ILD may lead to appropriate patient selection and lower treatment risk. The aim of this review was to summarize the current evidence related to pharmacotherapy for advanced LC-ILD and discuss emerging areas of research. J. Med. Invest. 71 : 9-22, February, 2024.


Asunto(s)
Enfermedades Pulmonares Intersticiales , Neoplasias Pulmonares , Humanos , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Enfermedades Pulmonares Intersticiales/etiología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/complicaciones , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Antineoplásicos/uso terapéutico , Antineoplásicos/efectos adversos
20.
Respir Med Case Rep ; 50: 102040, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38803368

RESUMEN

A 71-year-old male visited a hospital with a chief complaint of exertional dyspnea. A chest CT revealed multiple nodular lesions on the parietal pleura. Thoracoscopic pleural biopsy was performed resulting in a diagnosis of pleural mesothelioma with epithelioid type. When chemotherapy was initially initiated, his serum level of Krebs von den Lungen-6 (KL-6) was high. However, once chemotherapy was started, the serum KL-6 level gradually decreased with tumor shrinkage. Immunohistochemical staining revealed the expression of KL-6 from the tumor cells. This is the first report of KL-6 production directly from tumor cells in epithelial-type pleural mesothelioma.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA