Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Pharmacol Ther ; 253: 108578, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38103794

RESUMO

The treatment of interstitial lung diseases, including idiopathic pulmonary fibrosis (IPF), remains challenging as current available antifibrotic agents are not effective in halting disease progression. Connective tissue growth factor (CTGF), also known as cellular communication factor 2 (CCN2), is a member of the CCN family of proteins that regulates cell signaling through cell surface receptors such as integrins, the activity of cytokines/growth factors, and the turnover of extracellular matrix (ECM) proteins. Accumulating evidence indicates that CTGF plays a crucial role in promoting lung fibrosis through multiple processes, including inducing transdifferentiation of fibroblasts to myofibroblasts, epithelial-mesenchymal transition (EMT), and cooperating with other fibrotic mediators such as TGF-ß. Increased expression of CTGF has been observed in fibrotic lungs and inhibiting CTGF signaling has been shown to suppress lung fibrosis in several animal models. Thus, the CTGF signaling pathway is emerging as a potential therapeutic target in IPF and other pulmonary fibrotic conditions. This review provides a comprehensive overview of the current evidence on the pathogenic role of CTGF in pulmonary fibrosis and discusses the current therapeutic agents targeting CTGF using a systematic review approach.


Assuntos
Fator de Crescimento do Tecido Conjuntivo , Fibrose Pulmonar Idiopática , Animais , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibrose , Fibroblastos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/patologia , Fator de Crescimento Transformador beta1 , Pulmão/metabolismo
2.
Biochem Pharmacol ; 211: 115501, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36921632

RESUMO

Idiopathic pulmonary fibrosis (IPF) is the representative phenotype of interstitial lung disease where severe scarring develops in the lung interstitium. Although antifibrotic treatments are available and have been shown to slow the progression of IPF, improved therapeutic options are still needed. Recent data indicate that macrophages play essential pro-fibrotic roles in the pathogenesis of pulmonary fibrosis. Historically, macrophages have been classified into two functional subtypes, "M1″ and "M2," and it is well described that "M2″ or "alternatively activated" macrophages contribute to fibrosis via the production of fibrotic mediators, such as TGF-ß, CTGF, and CCL18. However, highly plastic macrophages may possess distinct functions and phenotypes in the fibrotic lung environment. Thus, M2-like macrophages in vitro and pro-fibrotic macrophages in vivo are not completely identical cell populations. Recent developments in transcriptome analysis, including single-cell RNA sequencing, have attempted to depict more detailed phenotypic characteristics of pro-fibrotic macrophages. This review will outline the role and characterization of pro-fibrotic macrophages in fibrotic lung diseases and discuss the possibility of treating lung fibrosis by preventing or reprogramming the polarity of macrophages. We also utilized a systematic approach to review the literature and identify novel and promising therapeutic agents that follow this treatment strategy.


Assuntos
Fibrose Pulmonar Idiopática , Doenças Pulmonares Intersticiais , Humanos , Doenças Pulmonares Intersticiais/tratamento farmacológico , Doenças Pulmonares Intersticiais/patologia , Pulmão/patologia , Macrófagos/patologia , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , Fibrose
3.
Am J Respir Crit Care Med ; 207(11): 1498-1514, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36917778

RESUMO

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.


Assuntos
Hipertensão Pulmonar , Fibrose Pulmonar Idiopática , Humanos , Ratos , Animais , Remodelação Vascular , Células Endoteliais/metabolismo , Tacrolimo , Pulmão/patologia , Fibrose Pulmonar Idiopática/patologia , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fibroblastos/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética
4.
JAMA Intern Med ; 182(12): 1248-1259, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36251286

RESUMO

Importance: Particulate matter 2.5 µm or less in diameter (PM2.5) is associated with adverse outcomes for patients with idiopathic pulmonary fibrosis, but its association with other fibrotic interstitial lung diseases (fILDs) and the association of PM2.5 composition with adverse outcomes remain unclear. Objective: To investigate the association of PM2.5 exposure with mortality and lung function among patients with fILD. Design, Setting, and Participants: In this multicenter, international, prospective cohort study, patients were enrolled in the Simmons Center for Interstitial Lung Disease Registry at the University of Pittsburgh in Pittsburgh, Pennsylvania; 42 sites of the Pulmonary Fibrosis Foundation Registry; and 8 sites of the Canadian Registry for Pulmonary Fibrosis. A total of 6683 patients with fILD were included (Simmons, 1424; Pulmonary Fibrosis Foundation, 1870; and Canadian Registry for Pulmonary Fibrosis, 3389). Data were analyzed from June 1, 2021, to August 2, 2022. Exposures: Exposure to PM2.5 and its constituents was estimated with hybrid models, combining satellite-derived aerosol optical depth with chemical transport models and ground-based PM2.5 measurements. Main Outcomes and Measures: Multivariable linear regression was used to test associations of exposures 5 years before enrollment with baseline forced vital capacity and diffusion capacity for carbon monoxide. Multivariable Cox models were used to test associations of exposure in the 5 years before censoring with mortality, and linear mixed models were used to test associations of exposure with a decrease in lung function. Multiconstituent analyses were performed with quantile-based g-computation. Cohort effect estimates were meta-analyzed. Models were adjusted for age, sex, smoking history, race, a socioeconomic variable, and site (only for Pulmonary Fibrosis Foundation and Canadian Registry for Pulmonary Fibrosis cohorts). Results: Median follow-up across the 3 cohorts was 2.9 years (IQR, 1.5-4.5 years), with death for 28% of patients and lung transplant for 10% of patients. Of the 6683 patients in the cohort, 3653 were men (55%), 205 were Black (3.1%), and 5609 were White (84.0%). Median (IQR) age at enrollment across all cohorts was 66 (58-73) years. A PM2.5 exposure of 8 µg/m3 or more was associated with a hazard ratio for mortality of 4.40 (95% CI, 3.51-5.51) in the Simmons cohort, 1.71 (95% CI, 1.32-2.21) in the Pulmonary Fibrosis Foundation cohort, and 1.45 (95% CI, 1.18-1.79) in the Canadian Registry for Pulmonary Fibrosis cohort. Increasing exposure to sulfate, nitrate, and ammonium PM2.5 constituents was associated with increased mortality across all cohorts, and multiconstituent models demonstrated that these constituents tended to be associated with the most adverse outcomes with regard to mortality and baseline lung function. Meta-analyses revealed consistent associations of exposure to sulfate and ammonium with mortality and with the rate of decrease in forced vital capacity and diffusion capacity of carbon monoxide and an association of increasing levels of PM2.5 multiconstituent mixture with all outcomes. Conclusions and Relevance: This cohort study found that exposure to PM2.5 was associated with baseline severity, disease progression, and mortality among patients with fILD and that sulfate, ammonium, and nitrate constituents were associated with the most harm, highlighting the need for reductions in human-derived sources of pollution.


Assuntos
Poluentes Atmosféricos , Poluição do Ar , Compostos de Amônio , Fibrose Pulmonar , Idoso , Feminino , Humanos , Masculino , Poluentes Atmosféricos/efeitos adversos , Poluentes Atmosféricos/análise , Poluição do Ar/efeitos adversos , Compostos de Amônio/análise , Canadá/epidemiologia , Monóxido de Carbono/análise , Estudos de Coortes , Exposição Ambiental/efeitos adversos , Pulmão , Nitratos/análise , Material Particulado/efeitos adversos , Material Particulado/análise , Estudos Prospectivos , Fibrose Pulmonar/induzido quimicamente , Sulfatos/análise , Pessoa de Meia-Idade
5.
ERJ Open Res ; 8(4)2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36299359

RESUMO

Background: This multicentre, international, prospective cohort study evaluated whether patients with pulmonary sarcoidosis living in neighbourhoods with greater material and social disadvantage experience worse clinical outcomes. Methods: The area deprivation index and the Canadian Index of Multiple Deprivation evaluate neighbourhood-level disadvantage in the US and Canada, with higher scores reflecting greater disadvantage. Multivariable linear regression evaluated associations of disadvantage with baseline forced vital capacity (FVC) or diffusing capacity of the lung for carbon monoxide (D LCO) and linear mixed effects models for associations with rate of FVC or D LCO decline, and competing hazards models were used for survival analyses in the US cohort, evaluating competing outcomes of death or lung transplantation. Adjustments were made for age at diagnosis, sex, race and smoking history. Results: We included 477 US and 122 Canadian patients with sarcoidosis. Higher disadvantage was not associated with survival or baseline FVC. The highest disadvantage quartile was associated with lower baseline D LCO in the US cohort (ß = -6.80, 95% CI -13.16 to -0.44, p=0.04), with similar findings in the Canadian cohort (ß = -7.47, 95% CI -20.28 to 5.33, p=0.25); with more rapid decline in FVC and D LCO in the US cohort (FVC ß = -0.40, 95% CI -0.70 to -0.11, p=0.007; D LCO ß = -0.59, 95% CI -0.95 to -0.23, p=0.001); and with more rapid FVC decline in the Canadian cohort (FVC ß = -0.80, 95% CI -1.37 to -0.24, p=0.003). Conclusion: Patients with sarcoidosis living in high disadvantage neighbourhoods experience worse baseline lung function and more rapid lung function decline, highlighting the need for better understanding of how neighbourhood-level factors impact individual patient outcomes.

6.
Am J Respir Cell Mol Biol ; 66(3): 260-270, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34797990

RESUMO

Idiopathic pulmonary fibrosis is a fatal lung disease characterized by progressive and excessive accumulation of myofibroblasts and in the lung. Connective-tissue growth factor (CTGF) exacerbates pulmonary fibrosis in radiation-induced lung fibrosis, and in this study, we demonstrate upregulation of CTGF in a rat lung fibrosis model induced by an adenovirus vector encoding active TGF-ß1 (AdTGF-ß1). We show that CTGF is also upregulated in patients with idiopathic pulmonary fibrosis. Expression of CTGF was upregulated in vascular smooth muscle cells cultured from fibrotic lungs on Days 7 and 14 as well as endothelial cells sorted from fibrotic lungs on Days 14 and 28. These findings suggest contributions of different cells in maintaining the fibrotic phenotype during fibrogenesis. Treatment of fibroblasts with recombinant CTGF along with TGF-ß increases profibrotic markers in fibroblasts, confirming the synergistic effect of recombinant CTGF with TGF-ß in inducing pulmonary fibrosis. Also, the fibrotic extracellular matrix upregulated CTGF expression, compared with the normal extracellular matrix, suggesting that not only profibrotic mediators but also a profibrotic environment contributes to fibrogenesis. We also showed that pamrevlumab, a CTGF inhibitory antibody, partially attenuates fibrosis in the model. These results suggest that pamrevlumab could be an option for treatment of pulmonary fibrosis.


Assuntos
Fator de Crescimento do Tecido Conjuntivo , Fibrose Pulmonar Idiopática , Fator de Crescimento Transformador beta1 , Animais , Anticorpos Monoclonais Humanizados , Fator de Crescimento do Tecido Conjuntivo/genética , Células Endoteliais/metabolismo , Fibrose , Fibrose Pulmonar Idiopática/genética , Ratos , Fator de Crescimento Transformador beta1/farmacologia
7.
Am J Respir Crit Care Med ; 205(4): 459-467, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34818133

RESUMO

Rationale: Fibrotic interstitial lung disease (fILD) is a group of pathologic entities characterized by scarring of the lungs and high morbidity and mortality. Research investigating how socioeconomic and residential factors impact outcomes in patients with fILD is lacking. Objectives: To determine the association between neighborhood-level disadvantage and presentation severity, disease progression, lung transplantation, and mortality in patients with fILD from the United States and Canada. Methods: We performed a multicenter, international, prospective cohort study of 4,729 patients with fILD from one U.S. and eight Canadian ILD registry sites. Neighborhood-level disadvantage was measured by the area deprivation index in the United States and the Canadian Index of Multiple Deprivation in Canada. Measurements and Main Results: In the U.S. but not in the Canadian cohort, patients with fILD living in neighborhoods with the greatest disadvantage (top quartile) experience the highest risk of mortality (hazard ratio = 1.51, P = 0.002), and in subgroups of patients with idiopathic pulmonary fibrosis, the top quartile of disadvantage experienced the lowest odds of lung transplantation (odds ratio = 0.46, P = 0.04). Greater disadvantage was associated with reduced baseline DLCO in both cohorts, but it was not associated with baseline FVC or FVC or DLCO decline in either cohort. Conclusions: Patients with fILD who live in areas with greater neighborhood-level disadvantage in the United States experience higher mortality, and patients with idiopathic pulmonary fibrosis experience lower odds of lung transplantation. These disparities are not seen in Canadian patients, which may indicate differences in access to care between the United States and Canada.


Assuntos
Disparidades nos Níveis de Saúde , Disparidades em Assistência à Saúde , Fibrose Pulmonar Idiopática , Doenças Pulmonares Intersticiais , Características de Residência , Privação Social , Determinantes Sociais da Saúde , Idoso , Canadá/epidemiologia , Progressão da Doença , Feminino , Disparidades em Assistência à Saúde/economia , Disparidades em Assistência à Saúde/estatística & dados numéricos , Humanos , Fibrose Pulmonar Idiopática/diagnóstico , Fibrose Pulmonar Idiopática/economia , Fibrose Pulmonar Idiopática/mortalidade , Fibrose Pulmonar Idiopática/cirurgia , Doenças Pulmonares Intersticiais/diagnóstico , Doenças Pulmonares Intersticiais/economia , Doenças Pulmonares Intersticiais/mortalidade , Doenças Pulmonares Intersticiais/cirurgia , Transplante de Pulmão/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Gravidade do Paciente , Prognóstico , Estudos Prospectivos , Fatores de Risco , Estados Unidos/epidemiologia
8.
Expert Opin Ther Targets ; 25(11): 939-948, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34784834

RESUMO

INTRODUCTION: Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with high associated morbidity and mortality. The therapeutic landscape has significantly changed in the last 20 years with two drugs currently approved that have demonstrated the ability to slow disease progression. Despite these developments, survival in IPF is limited, so there is a major interest in therapeutic targets which could serve to open up new therapeutic avenues. AREAS COVERED: We review the most recent information regarding drug targets and therapies currently being investigated in preclinical and early-stage clinical trials. EXPERT OPINION: The complex pathogenesis of IPF and variability in disease course and response to therapy highlights the importance of a precision approach to therapy. Novel technologies including transcriptomics and the use of serum biomarkers, will become essential tools to guide future drug development and therapeutic decision making particularly as it pertains to combination therapy.


Assuntos
Fibrose Pulmonar Idiopática , Biomarcadores , Progressão da Doença , Desenvolvimento de Medicamentos , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico
9.
Am J Respir Cell Mol Biol ; 64(2): 235-246, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33253593

RESUMO

Pulmonary fibrosis is a progressive lung disease characterized by myofibroblast accumulation and excessive extracellular matrix deposition. We sought to investigate the role of FKBP13 (13-kD FK506-binding protein), an endoplasmic reticulum-resident molecular chaperone, in various forms of pulmonary fibrosis. We first characterized the gene and protein expression of FKBP13 in lung biopsy specimens from 24 patients with idiopathic pulmonary fibrosis and 17 control subjects. FKBP13 expression was found to be elevated in the fibrotic regions of idiopathic pulmonary fibrosis lung tissues and correlated with declining forced vital capacity and dyspnea severity. FKBP13 expression was also increased in lung biopsy specimens of patients with hypersensitivity pneumonitis, rheumatoid arthritis, and sarcoidosis-associated interstitial lung disease. We next evaluated the role of this protein using FKBP13-/- mice in a bleomycin model of pulmonary fibrosis. Animals were assessed for lung function and histopathology at different stages of lung injury including the inflammatory (Day 7), fibrotic (Day 21), and resolution (Day 50) phases. FKBP13-/- mice showed increased infiltration of inflammatory cells and cytokines at Day 7, increased lung elastance and fibrosis at Day 21, and impaired resolution of fibrosis at Day 50. These changes were associated with an increased number of cells that stained positive for TUNEL and cleaved caspase 3 in the FKBP13-/- lungs, indicating a heightened cellular sensitivity to bleomycin. Our findings suggest that FKBP13 is a potential biomarker for severity of interstitial lung diseases and that it has a biologically relevant role in protecting mice against bleomycin-induced injury, inflammation, and fibrosis.


Assuntos
Doenças Pulmonares Intersticiais/metabolismo , Doenças Pulmonares Intersticiais/patologia , Proteínas de Ligação a Tacrolimo/metabolismo , Regulação para Cima/fisiologia , Animais , Biomarcadores/metabolismo , Biópsia/métodos , Bleomicina/efeitos adversos , Citocinas/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Feminino , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Inflamação/metabolismo , Inflamação/patologia , Pulmão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Regulação para Cima/efeitos dos fármacos
10.
Eur Respir J ; 55(6)2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32165401

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a complex disease of unknown aetiology, which makes drug development challenging. Single administration of bleomycin directly to the lungs of mice is a widely used experimental model for studying pulmonary fibrogenesis and evaluating the effect of therapeutic antifibrotic strategies. The model works by inducing an early inflammatory phase, which transitions into fibrosis after 5-7 days. This initial inflammation makes therapeutic timing crucial. To accurately assess antifibrotic efficacy, the intervention should inhibit fibrosis without impacting early inflammation.Studies published between 2008 and 2019 using the bleomycin model to investigate pulmonary fibrosis were retrieved from PubMed, and study characteristics were analysed. Intervention-based studies were classified as either preventative (starting <7 days after bleomycin installation) or therapeutic (>7 days). In addition, studies were cross-referenced with current major clinical trials to assess the availability of preclinical rationale.A total of 976 publications were evaluated. 726 investigated potential therapies, of which 443 (61.0%) were solely preventative, 166 (22.9%) were solely therapeutic and 105 (14.5%) were both. Of the 443 preventative studies, only 70 (15.8%) characterised inflammation during the model's early inflammatory phase. In the reported 145 IPF clinical trials investigating 93 compounds/combinations, only 25 (26.9%) interventions had any preclinical data on bleomycin available on PubMed.Since 2008, we observed a shift (from <5% to 37.4%) in the number of studies evaluating drugs in the therapeutic setting in the bleomycin model. While this shift is encouraging, further characterisation of early inflammation and appropriate preclinical therapeutic testing are still needed. This will facilitate fruitful drug development in IPF, and more therapeutic strategies for patients with this devastating disease.


Assuntos
Bleomicina , Modelos Animais de Doenças , Fibrose Pulmonar Idiopática , Animais , Fibrose , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/patologia , Pulmão/patologia , Camundongos
11.
Immunol Cell Biol ; 97(2): 203-217, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30298952

RESUMO

Although recent evidence has shown that IL-6 is involved in enhanced alternative activation of macrophages toward a profibrotic phenotype, the mechanisms leading to their increased secretory capacity are not fully understood. Here, we investigated the effect of IL-6 on endoplasmic reticulum (ER) expansion and alternative activation of macrophages in vitro. An essential mediator in this ER expansion process is the IRE1 pathway, which possesses a kinase and endoribonuclease domain to cleave XBP1 into a spliced bioactive molecule. To investigate the IRE1-XBP1 expansion pathway, IL-4/IL-13 and IL-4/IL-13/IL-6-mediated alternative programming of murine bone marrow-derived and human THP1 macrophages were assessed by arginase activity in cell lysates, CD206 and arginase-1 expression by flow cytometry, and secreted CCL18 by ELISA, respectively. Ultrastructural intracellular morphology and ER biogenesis were examined by transmission electron microscopy and immunofluorescence. Transcription profiling of 128 genes were assessed by NanoString and Pharmacological inhibition of the IRE1-XBP1 arm was achieved using STF-083010 and was verified by RT-PCR. The addition of IL-6 to the conventional alternative programming cocktail IL-4/IL-13 resulted in increased ER and mitochondrial expansion, profibrotic profiles and unfolded protein response-mediated induction of molecular chaperones. IRE1-XBP1 inhibition substantially reduced the IL-6-mediated hyperpolarization and normalized the above effects. In conclusion, the addition of IL-6 enhances ER expansion and the profibrotic capacity of IL-4/IL-13-mediated activation of macrophages. Therapeutic strategies targeting IL-6 or the IRE1-XBP1 axis may be beneficial to prevent the profibrotic capacity of macrophages.


Assuntos
Retículo Endoplasmático , Endorribonucleases/metabolismo , Interleucina-3/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Fatores Ativadores de Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/ultraestrutura , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/fisiologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/imunologia , Humanos , Interleucina-4/farmacologia , Interleucina-6/farmacologia , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Células THP-1
12.
Expert Opin Investig Drugs ; 28(1): 19-28, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30513000

RESUMO

INTRODUCTION: Idiopathic pulmonary fibrosis (IPF) is an age-associated, progressive, and irreversible fatal interstitial lung disease. Although many drugs have failed in clinical trials, these failures improved the understanding of the pathogenesis of IPF. Currently, there are two drugs approved for IPF that slow the progression of the disease. However, the prognosis for patients with IPF remains poor and the search continues for drugs that inhibit the pathogenic pathways active in IPF to further reduce or even halt the progression of the disease. AREAS COVERED: We highlight the recent information on the therapeutic targets currently explored in early stage clinical trials and discuss the potential for new therapy and the limitation of basic research in the treatment of IPF. EXPERT OPINION: A key challenge in the coming years will lie in deciding which compounds to combine and how to evaluate combination therapies in clinical trials. The drugs most likely to provide additive efficacy when used in combination with one of the approved therapies are those with alternative, complementary, or synergistic mechanisms of action.


Assuntos
Desenvolvimento de Medicamentos/métodos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Terapia de Alvo Molecular , Fatores Etários , Animais , Ensaios Clínicos como Assunto , Progressão da Doença , Quimioterapia Combinada , Humanos , Fibrose Pulmonar Idiopática/fisiopatologia , Prognóstico
13.
Sci Rep ; 7(1): 13281, 2017 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-29038604

RESUMO

Although recent evidence indicates that gp130 cytokines, Oncostatin M (OSM) and IL-6 are involved in alternative programming of macrophages, their role in lung fibrogenesis is poorly understood. Here, we investigated the effect of transient adenoviral overexpression of OSM or IL-6 in mice during bleomycin-induced lung fibrosis. Lung fibrosis and M2-like macrophage accumulation were assessed by immunohistochemistry, western blotting, gene expression and flow cytometry. Ex-vivo isolated alveolar and bone marrow-derived macrophages were examined for M2-like programming and signalling. Airway physiology measurements at day 21 demonstrated that overexpression of OSM or IL-6 exacerbated bleomycin-induced lung elastance, consistent with histopathological assessment of extracellular matrix and myofibroblast accumulation. Flow cytometry analysis at day 7 showed increased numbers of M2-like macrophages in lungs of mice exposed to bleomycin and OSM or IL-6. These macrophages expressed the IL-6Rα, but were deficient for OSMRß, suggesting that IL-6, but not OSM, may directly induce alternative macrophage activation. In conclusion, the gp130 cytokines IL-6 and OSM contribute to the accumulation of profibrotic macrophages and enhancement of bleomycin-induced lung fibrosis. This study suggests that therapeutic strategies targeting these cytokines or their receptors may be beneficial to prevent the accumulation of M2-like macrophages and the progression of fibrotic lung disease.


Assuntos
Bleomicina/efeitos adversos , Expressão Gênica , Interleucina-6/genética , Macrófagos/metabolismo , Oncostatina M/genética , Fibrose Pulmonar/etiologia , Animais , Biomarcadores , Feminino , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Lectinas Tipo C/metabolismo , Pulmão , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos Alveolares , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Modelos Biológicos , Oncostatina M/metabolismo , Fenótipo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Receptores de Superfície Celular/metabolismo
14.
Pulm Pharmacol Ther ; 25(4): 263-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21951688

RESUMO

Fibrocytes are bone marrow-derived mesenchymal cell precursors, defined primarily by their ability to co-express markers of both haematopoietic (e.g. CD45 or CXCR4) and stromal (e.g. collagen) lineages. Fibrocytes in culture also have ultrastructural cell surface features that distinguish them from other leukocytes. Extensive efforts have helped to characterise fibrocytes phenotypically and functionally, but it is still unclear exactly how these cells contribute to tissue repair and/or pathologic fibrosis. Nevertheless, the varied levels of fibrocytes in blood have raised considerable interest as a biomarker of disease activity, such as chronic lung diseases, including pulmonary fibrosis, asthma and pulmonary hypertension. These cells also may become a novel therapeutic target for these difficult to treat disorders. This review will briefly summarize the current knowledge about fibrocytes in human lung disease and in animal disease models and highlight areas of consensus as well as issues that remain controversial to date.


Assuntos
Fibroblastos/metabolismo , Pneumopatias/fisiopatologia , Asma/fisiopatologia , Biomarcadores , Diferenciação Celular , Doença Crônica , Humanos , Hipertensão Pulmonar/fisiopatologia , Pulmão/metabolismo , Células-Tronco Mesenquimais , Monócitos/metabolismo
15.
IDrugs ; 13(5): 332-45, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20432191

RESUMO

Tyrosine kinase receptors have important signaling functions in various physiological and pathological pathways. The recognition of their involvement in tumor angiogenesis, which is the main event of tumor progression, opened a new era in the discovery of anticancer drugs. Developers soon grasped that by targeting several tyrosine kinase receptors at once, so-called multitarget tyrosine kinase inhibitors, a drug could dramatically affect the progression of cancer and decrease resistance. Several antiangiogenic, multitarget tyrosine kinase inhibitors, such as sorafenib and sunitinib, are already marketed, while many more are undergoing clinical trials for a range of cancer types. Boehringer Ingelheim Corp is developing intedanib (BIBF-1120), a triple kinase inhibitor blocking VEGFR, PDGFR and FGFR for the treatment of several malignancies and idiopathic pulmonary fibrosis. The preliminary data for intedanib appears at least as good as that for other antiangiogenic tyrosine kinase inhibitors or other antiangiogenic approaches that are not targeting the tyrosine kinases. The sustained inhibition of VEGFR phosphorylation (> 24 h), the fast in vivo clearance and clinical efficacy against a broad range of malignancies appear to be the major advantages of intedanib. Furthermore, the existing data suggest an excellent safety profile. At the time of publication, intedanib had reached phase III trials for the treatment of NSCLC and ovarian cancer.


Assuntos
Inibidores da Angiogênese/farmacologia , Indóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Sistemas de Liberação de Medicamentos , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/fisiopatologia , Indóis/uso terapêutico , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
16.
Kidney Int ; 77(4): 319-28, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19956083

RESUMO

Transition of peritoneal mesothelial cells to a mesenchymal phenotype plays an integral role in the angiogenic and fibrotic changes seen in the peritoneum of patients receiving long-term peritoneal dialysis. While signaling by transforming growth factor (TGF)-beta through Smad proteins likely causes these changes, it is possible that non-Smad pathways may also play a role. Here, we found that Smad3-deficient mice were protected from peritoneal fibrosis and angiogenesis caused by adenovirus-mediated gene transfer of active TGF-beta1 to mesothelial cells; however, mesothelial transition occurred in this setting, suggesting involvement of non-Smad mechanisms. The phosphatidyl inositol 3 kinase (PI3K) target, Akt, was upregulated in both Smad-deficient and wild-type mice after exposure to TGF-beta1. In vivo inhibition of the mammalian target of rapamycin (mTOR) by rapamycin completely abrogated the transition response in Smad3-deficient but not in wild-type mice. Rapamycin blocked nuclear localization of beta-catenin independent of glycogen synthase kinase 3beta activity. Further, in Smad3-deficient mice rapamycin reduced the expression of alpha-smooth muscle actin, which is an epithelial-to-mesenchymal transition-associated gene. Hence, we conclude that TGF-beta1 causes peritoneal injury through Smad-dependent and Smad-independent pathways; the latter involves redundant mechanisms inhibited by rapamycin, suggesting that suppression of both pathways may be necessary to abrogate mesothelial transition.


Assuntos
Fibrose Peritoneal/etiologia , Peritônio/patologia , Proteína Smad3/fisiologia , Animais , Diferenciação Celular , Células Epiteliais , Mesoderma/citologia , Camundongos , Transdução de Sinais
17.
Int J Biochem Cell Biol ; 41(12): 2477-84, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19666134

RESUMO

Given that extracellular ATP is markedly elevated in inflammation and is known to modulate fibroblast function, we examined the effects of exogenously added ATP on Ca(2+)-handling and gene expression in human pulmonary fibroblasts. Cells were loaded with the Ca(2+)-indicator dye fluo-4 and studied using confocal fluorimetry. Standard RT-PCR was used to probe gene expression. ATP (10(-5)M) evoked recurring Ca(2+)-waves which were completely occluded by cyclopiazonic acid (depletes the internal Ca(2+)-store) or the phospholipase inhibitor U73122. Pretreatment with ryanodine (10(-5)M), however, had no effect on the ATP-evoked responses. Regarding the receptor through which ATP acted, we found the ATP-response to be mimicked by UTP or ADP but not by adenosine or alpha,beta-methylene-ATP, and to be blocked by the purinergic receptor blocker PPADS. The ATP-evoked response was greater and longer lasting within the nucleus than in the non-nuclear portion of the cytosol. RT-PCR showed that ATP also rapidly and dramatically increased gene expression of P2Y(4) receptors, the cytokine TGF-beta (an important modulator of wound repair) and two matrix proteins (collagen A1 and fibronectin) approximately 4-5 times above baseline: this increase was not significantly affected by ryanodine but was abolished by PPADS. We conclude that, in human pulmonary fibroblasts, ATP acts upon P2Y receptors to liberate internal Ca(2+) through ryanodine-insensitive channels, leading to a Ca(2+)-wave which courses throughout the cell and modulates gene expression.


Assuntos
Trifosfato de Adenosina/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Fibroblastos/metabolismo , Compostos de Anilina , Cafeína/farmacologia , Núcleo Celular/metabolismo , Células Cultivadas , Colágeno/biossíntese , Colágeno/genética , Estrenos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibronectinas/biossíntese , Fibronectinas/genética , Humanos , Indóis/farmacologia , Pulmão/citologia , Antagonistas do Receptor Purinérgico P2 , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacologia , Pirrolidinonas/farmacologia , Rianodina/farmacologia , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/genética , Fosfolipases Tipo C/antagonistas & inibidores , Xantenos
18.
J Transl Med ; 6: 16, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18402687

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a treatment resistant disease with poor prognosis. Numerous compounds have been demonstrated to efficiently prevent pulmonary fibrosis (PF) in animal models but only a few were successful when given to animals with established fibrosis. Major concerns of current PF models are spontaneous resolution and high variability of fibrosis, and the lack of assessment methods that can allow to monitor the effect of drugs in individual animals over time. We used a model of experimental PF in rats and compare parameters obtained in living animals with conventional assessment tools that require removal of the lungs. METHODS: PF was induced in rats by adenoviral gene transfer of transforming growth factor-beta. Morphological and functional changes were assessed for up to 56 days by micro-CT, lung compliance (measured via a mechanical ventilator) and VO2max and compared to histomorphometry and hydroxyproline content. RESULTS: Standard histological and collagen assessment confirmed the persistent fibrotic phenotype as described before. The histomorphological scores correlated both to radiological (r2 = 0.29, p < 0.01) and functional changes (r2 = 0.51, p < 0.0001). VO2max did not correlate with fibrosis. CONCLUSION: The progression of pulmonary fibrosis can be reliably assessed and followed in living animals over time using invasive, non-terminal compliance measurements and micro-CT. This approach directly translates to the management of patients with IPF and allows to monitor therapeutic effects in drug intervention studies.


Assuntos
Modelos Animais de Doenças , Pulmão/patologia , Fibrose Pulmonar/patologia , Adenoviridae/genética , Animais , Colágeno/metabolismo , Progressão da Doença , Feminino , Técnicas de Transferência de Genes , Hidroxiprolina/metabolismo , Pulmão/diagnóstico por imagem , Ratos , Ratos Sprague-Dawley , Testes de Função Respiratória , Fatores de Tempo , Tomografia Computadorizada por Raios X , Fatores de Crescimento Transformadores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA