Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38559095

RESUMO

Pulmonary fibrosis is a devastating disease with no effective treatments to cure, stop or reverse the unremitting, fatal fibrosis. A critical barrier to treating this disease is the lack of understanding of the pathways leading to fibrosis as well as those regulating the resolution of fibrosis. Fibrosis is the pathologic side of normal tissue repair that results when the normal wound healing programs go awry. Successful resolution of tissue injury requires several highly coordinated pathways, and this research focuses on the interplay between these overlapping pathways: immune effectors, inflammatory mediators and fibroproliferation in the resolution of fibrosis. Previously we have successfully prevented, mitigated, and even reversed established fibrosis using vaccinia vaccination immunotherapy in two models of murine lung fibrosis. The mechanism by which vaccinia reverses fibrosis is by vaccine induced lung specific Th1 skewed tissue resident memory (TRMs) in the lung. In this study, we isolated a population of vaccine induced TRMs - CD49a+ CD4+ T cells - that are both necessary and sufficient to reverse established pulmonary fibrosis. Using adoptive cellular therapy, we demonstrate that intratracheal administration of CD49a+ CD4+ TRMs into established fibrosis, reverses the fibrosis histologically, by promoting a decrease in collagen, and functionally, by improving lung function, without the need for vaccination. Furthermore, co-culture of in vitro derived CD49+ CD4+ human TRMs with human fibroblasts from individuals with idiopathic pulmonary fibrosis (IPF) results in the down regulation of IPF fibroblast collagen production. Lastly, we demonstrate in human IPF lung histologic samples that CD49a+ CD4+ TRMs, which can down regulate human IPF fibroblast function, fail to increase in the IPF lungs, thus potentially failing to promote resolution. Thus, we define a novel unappreciated role for tissue resident memory T cells in regulating established lung fibrosis to promote resolution of fibrosis and re-establish lung homeostasis. We demonstrate that immunotherapy, in the form of adoptive transfer of CD49a+ CD4+ TRMs into the lungs of mice with established fibrosis, not only stops progression of the fibrosis but more importantly reverses the fibrosis. These studies provide the insight and preclinical rationale for a novel paradigm shifting approach of using cellular immunotherapy to treat lung fibrosis.

2.
J Clin Invest ; 131(21)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34491913

RESUMO

Peripheral nerves have the capacity for regeneration, but the rate of regeneration is so slow that many nerve injuries lead to incomplete recovery and permanent disability for patients. Macrophages play a critical role in the peripheral nerve response to injury, contributing to both Wallerian degeneration and nerve regeneration, and their function has recently been shown to be dependent on intracellular metabolism. To date, the impact of their intracellular metabolism on peripheral nerve regeneration has not been studied. We examined conditional transgenic mice with selective ablation in macrophages of solute carrier family 16, member 1 (Slc16a1), which encodes monocarboxylate transporter 1 (MCT1), and found that MCT1 contributed to macrophage metabolism, phenotype, and function, specifically in regard to phagocytosis and peripheral nerve regeneration. Adoptive cell transfer of wild-type macrophages ameliorated the impaired nerve regeneration in macrophage-selective MCT1-null mice. We also developed a mouse model that overexpressed MCT1 in macrophages and found that peripheral nerves in these mice regenerated more rapidly than in control mice. Our study provides further evidence that MCT1 has an important biological role in macrophages and that manipulations of macrophage metabolism can enhance recovery from peripheral nerve injuries, for which there are currently no approved medical therapies.


Assuntos
Macrófagos/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Regeneração Nervosa , Traumatismos dos Nervos Periféricos/metabolismo , Nervo Isquiático , Simportadores/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Transportadores de Ácidos Monocarboxílicos/genética , Traumatismos dos Nervos Periféricos/genética , Nervo Isquiático/lesões , Nervo Isquiático/fisiologia , Simportadores/genética
3.
J Immunol ; 207(3): 913-922, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34290107

RESUMO

Metabolic programming is integrally linked to immune cell function. Nowhere is this clearer than in the differentiation of macrophages. Proinflammatory M1 macrophages primarily use glycolysis as a rapid energy source but also to generate antimicrobial compounds, whereas alternatively activated M2 macrophages primarily rely on oxidative phosphorylation for the longevity required for proper wound healing. mTOR signaling has been demonstrated to be a key regulator of immune cell metabolism and function. mTORC2 signaling is required for the generation of M2 macrophages, whereas the role of mTORC1 signaling, a key regulator of glycolysis, has been controversial. By using genetic deletion of mTORC1 signaling in C57BL/6 mouse macrophages, we observed enhanced M1 macrophage function in vitro and in vivo. Surprisingly, this enhancement occurred despite a significant defect in M1 macrophage glycolytic metabolism. Mechanistically, enhanced M1 function occurred because of inhibition of the class III histone deacetylases the sirtuins, resulting in enhanced histone acetylation. Our findings provide a counterpoint to the paradigm that enhanced immune cell function must occur in the presence of increased cellular metabolism and identifies a potential, pharmacologic target for the regulation of inflammatory responses.


Assuntos
Inflamação/imunologia , Macrófagos/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Acetilação , Animais , Células Cultivadas , Reprogramação Celular , Citocinas/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Sirtuínas/metabolismo , Células Th1/imunologia
4.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L204-L212, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34009049

RESUMO

The quantification of airway compliance (Caw) is essential to the study of airway alterations in disease models. However, the required measurements of airway pressure and volume are difficult to acquire in mice. We hypothesized that the inflation limb of full-range pressure-volume (PV) curves could be used to quantify Caw, as it contains a segment where only the airway tree is distended. The study objective was to assess the feasibility of the approach by analysis of full-range PV curves previously collected in three mouse models: an elastase model of emphysema, a genetic model spontaneously developing emphysema (leukotriene C4 synthase knockout; LTC4S-KO), and a bleomycin model of lung fibrosis. Attempts to validate results included Caw change relative to respiratory system compliance (ΔCaw/ΔC), the minute work of breathing (mWOB), and the elastance at 20.5 Hz (Ers_20.5) from prior respiratory mechanics measurements in the same subjects. Caw was estimated at 3% of total compliance in healthy mice or 2.3 ± 1 µL/cmH2O (n = 17). The technique detected changes in models of respiratory obstructive and restrictive diseases relative to control mice as well as differences in the two emphysema models studied. The changes in Caw were consistent with those seen in ΔCaw/ΔC, mWOB, or Ers_20.5, with some variations according to the model, as well as with results reported in the literature in humans and mice. Direct Caw measurements in subjects as small as mice could prove useful to further characterize other respiratory disease models associated with airway remodeling or to assess treatment effects.


Assuntos
Resistência das Vias Respiratórias , Bleomicina/toxicidade , Enfisema Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , Transtornos Respiratórios/complicações , Animais , Antibióticos Antineoplásicos/toxicidade , Feminino , Complacência Pulmonar , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Enfisema Pulmonar/etiologia , Fibrose Pulmonar/induzido quimicamente , Mecânica Respiratória
5.
J Clin Invest ; 131(2)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33463535

RESUMO

Idiopathic pulmonary fibrosis (IPF) affects hundreds of thousands of people worldwide, reducing their quality of life and leading to death from respiratory failure within years of diagnosis. Treatment options remain limited, with only two FDA-approved drugs available in the United States, neither of which reverse the lung damage caused by the disease or prolong the life of individuals with IPF. The only cure for IPF is lung transplantation. In this review, we discuss recent major advances in our understanding of the role of the immune system in IPF that have revealed immune dysregulation as a critical driver of disease pathophysiology. We also highlight ways in which an improved understanding of the immune system's role in IPF may enable the development of targeted immunomodulatory therapies that successfully halt or potentially even reverse lung fibrosis.


Assuntos
Fibrose Pulmonar Idiopática/imunologia , Fatores Imunológicos/uso terapêutico , Transplante de Pulmão , Pulmão/imunologia , Humanos , Fibrose Pulmonar Idiopática/terapia , Estados Unidos
6.
J Clin Invest ; 130(7): 3865-3884, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32324593

RESUMO

Myeloid cells comprise a major component of the tumor microenvironment (TME) that promotes tumor growth and immune evasion. By employing a small-molecule inhibitor of glutamine metabolism, not only were we able to inhibit tumor growth, but we markedly inhibited the generation and recruitment of myeloid-derived suppressor cells (MDSCs). Targeting tumor glutamine metabolism led to a decrease in CSF3 and hence recruitment of MDSCs as well as immunogenic cell death, leading to an increase in inflammatory tumor-associated macrophages (TAMs). Alternatively, inhibiting glutamine metabolism of the MDSCs themselves led to activation-induced cell death and conversion of MDSCs to inflammatory macrophages. Surprisingly, blocking glutamine metabolism also inhibited IDO expression of both the tumor and myeloid-derived cells, leading to a marked decrease in kynurenine levels. This in turn inhibited the development of metastasis and further enhanced antitumor immunity. Indeed, targeting glutamine metabolism rendered checkpoint blockade-resistant tumors susceptible to immunotherapy. Overall, our studies define an intimate interplay between the unique metabolism of tumors and the metabolism of suppressive immune cells.


Assuntos
Imunidade Celular , Macrófagos/imunologia , Células Supressoras Mieloides/imunologia , Neoplasias Experimentais/imunologia , Microambiente Tumoral/imunologia , Animais , Feminino , Glutamina/imunologia , Imunoterapia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Supressoras Mieloides/patologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia
7.
Nat Rev Drug Discov ; 18(9): 669-688, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31363227

RESUMO

Metabolic programming is emerging as a critical mechanism to alter immune cell activation, differentiation and function. Targeting metabolism does not completely suppress or activate the immune system but selectively regulates immune responses. The different metabolic requirements of the diverse cells that constitute an immune response provide a unique opportunity to separate effector functions from regulatory functions. Likewise, cells can be metabolically reprogrammed to promote either their short-term effector functions or long-term memory capacity. Studies in the growing field of immunometabolism support a paradigm of 'cellular selectivity based on demand', in which generic inhibitors of ubiquitous metabolic processes selectively affect cells with the greatest metabolic demand and have few effects on other cells of the body. Targeting metabolism, rather than particular cell types or cytokines, in metabolically demanding processes such as autoimmunity, graft rejection, cancer and uncontrolled inflammation could lead to successful strategies in controlling the pathogenesis of these complex disorders.


Assuntos
Autoimunidade/imunologia , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/metabolismo , Imunomodulação/efeitos dos fármacos , Metabolismo/efeitos dos fármacos , Neoplasias/imunologia , Neoplasias/metabolismo , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Modelos Biológicos
8.
Nat Commun ; 10(1): 1128, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30850660

RESUMO

Scleroderma is an autoimmune rheumatic disorder accompanied by severe fibrosis in skin and other internal organs. During scleroderma progression, resident fibroblasts undergo activation and convert to α-smooth muscle actin (α-SMA) expressing myofibroblasts (MFBs) with increased capacity to synthesize collagens and fibrogenic components. Accordingly, MFBs are a major therapeutic target for fibrosis in scleroderma and treatment with blocking MFBs could produce anti-fibrotic effects. TLY012 is an engineered human TNF-related apoptosis-inducing ligand (TRAIL) which induces selective apoptosis in transformed cells expressing its cognate death receptors (DRs). Here we report that TLY012 selectively blocks activation of dermal fibroblasts and induces DR-mediated apoptosis in α-SMA+ MFBs through upregulated DR5 during its activation. In vivo, TLY012 reverses established skin fibrosis to near-normal skin architecture in mouse models of scleroderma. Thus, the TRAIL pathway plays a critical role in tissue remodeling and targeting upregulated DR5 in α-SMA+ MFBs is a viable therapy for fibrosis in scleroderma.


Assuntos
Actinas/genética , Derme/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Escleroderma Sistêmico/tratamento farmacológico , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Actinas/metabolismo , Adulto , Idoso , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Diferenciação Celular , Colágeno/genética , Colágeno/metabolismo , Derme/metabolismo , Derme/patologia , Modelos Animais de Doenças , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Engenharia de Proteínas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/imunologia , Escleroderma Sistêmico/patologia , Transdução de Sinais
9.
Physiol Rep ; 7(5): e14019, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30821123

RESUMO

Despite recent advances, acute respiratory distress syndrome (ARDS) remains a severe and often fatal disease for which there is no therapy able to reduce the underlying excessive lung inflammation or enhance resolution of injury. Metabolic programming plays a critical role in regulating inflammatory responses. Due to their high metabolic needs, neutrophils, macrophages, and lymphocytes rely upon glutamine metabolism to support activation and function. Additionally, during times of physiologic stress, nearly all cells, including fibroblasts and epithelial cells, require glutamine metabolism. We hypothesized that inhibiting glutamine metabolism reduces lung inflammation and promotes resolution of acute lung injury. Lung injury was induced by instilling lipopolysaccharide (LPS) intratracheally. To inhibit glutamine metabolism, we administered a glutamine analogue, 6-diazo-5-oxo-L-norleucine (DON) that binds to glutamine-utilizing enzymes and transporters, after injury was well established. Treatment with DON led to less lung injury, fewer lung neutrophils, lung inflammatory and interstitial macrophages, and lower levels of proinflammatory cytokines and chemokines at 5 and/or 7 days after injury. Additionally, DON led to earlier expression of the growth factor amphiregulin and more rapid recovery of LPS-induced weight loss. Thus, DON reduced lung inflammation and promoted resolution of injury. These data contribute to our understanding of how glutamine metabolism regulates lung inflammation and repair, and identifies a novel target for future therapies for ARDS and other inflammatory lung diseases.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Anti-Inflamatórios/farmacologia , Antimetabólitos/farmacologia , Diazo-Oxo-Norleucina/farmacologia , Glutamina/metabolismo , Pulmão/efeitos dos fármacos , Pneumonia/prevenção & controle , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Anfirregulina/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/efeitos dos fármacos , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Pneumonia/patologia , Fatores de Tempo
10.
Respir Med ; 130: 52-54, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29206633

RESUMO

Interstitial lung disease (ILD) is common in patients with autoimmune myositis but factors that determine susceptibility are unknown. Familial and sporadic idiopathic pulmonary fibrosis (IPF) are strongly associated with a single nucleotide polymorphism in the promoter region of MUC5B (rs35705950). We sought to determine the relationship between MUC5B polymorphism expression and myositis-ILD. The MUC5B minor allele frequency (MAF) was examined in 402 European American participants; 60 with idiopathic interstitial pneumonia (IIP), 208 with myositis-ILD, and 134 unaffected controls. The MUC5B minor allele frequency was 26%, 8%, and 7% in those with non-myositis ILD, myositis-ILD, and unaffected controls, respectively. The MUC5B variant was associated with IIP (OR 4.10; p < 0.001). The MUC5B polymorphism was not significantly associated with myositis-ILD (OR 1.08; p = 0.80)]. We found MUC5B MAFs in our IIP cohort similar to published frequencies for subjects with familial and sporadic IPF. Overall, the MUC5B promoter variant does not appear to contribute to ILD risk in myositis patients.


Assuntos
Dermatomiosite/genética , Pneumonias Intersticiais Idiopáticas/genética , Doenças Pulmonares Intersticiais/genética , Mucina-5B/genética , Miosite/genética , Cromossomos Humanos Par 11 , Dermatomiosite/imunologia , Frequência do Gene , Humanos , Fibrose Pulmonar Idiopática/genética , Doenças Pulmonares Intersticiais/diagnóstico , Miosite/imunologia , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Risco , População Branca
11.
Cell Rep ; 20(10): 2439-2454, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28877476

RESUMO

Tissue-resident macrophages play critical roles in sentinel and homeostatic functions as well as in promoting inflammation and immunity. It has become clear that the generation of these cells is highly dependent upon tissue-specific cues derived from the microenvironment that, in turn, regulate unique differentiation programs. Recently, a role for GATA6 has emerged in the differentiation programming of resident peritoneal macrophages. We identify a critical role for mTOR in integrating cues from the tissue microenvironment in regulating differentiation and metabolic reprogramming. Specifically, inhibition of mTORC2 leads to enhanced GATA6 expression in a FOXO1 dependent fashion. Functionally, inhibition of mTORC2 promotes peritoneal resident macrophage generation in the resolution phase during zymosan-induced peritonitis. Also, mTORC2-deficient peritoneal resident macrophages displayed increased functionality and metabolic reprogramming. Notably, mTORC2 activation distinguishes tissue-resident macrophage proliferation and differentiation from that of M2 macrophages. Overall, our data implicate a selective role for mTORC2 in the differentiation of tissue-resident macrophages.


Assuntos
Macrófagos Peritoneais/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Peritonite/metabolismo , Animais , Feminino , Citometria de Fluxo , Proteína Forkhead Box O1/metabolismo , Fator de Transcrição GATA6/metabolismo , Immunoblotting , Macrófagos/metabolismo , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Camundongos , Fagocitose/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Zimosan/toxicidade
12.
Am J Physiol Endocrinol Metab ; 312(5): E381-E393, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28223293

RESUMO

Fatty acid oxidation in macrophages has been suggested to play a causative role in high-fat diet-induced metabolic dysfunction, particularly in the etiology of adipose-driven insulin resistance. To understand the contribution of macrophage fatty acid oxidation directly to metabolic dysfunction in high-fat diet-induced obesity, we generated mice with a myeloid-specific knockout of carnitine palmitoyltransferase II (CPT2 Mϕ-KO), an obligate step in mitochondrial long-chain fatty acid oxidation. While fatty acid oxidation was clearly induced upon IL-4 stimulation, fatty acid oxidation-deficient CPT2 Mϕ-KO bone marrow-derived macrophages displayed canonical markers of M2 polarization following IL-4 stimulation in vitro. In addition, loss of macrophage fatty acid oxidation in vivo did not alter the progression of high-fat diet-induced obesity, inflammation, macrophage polarization, oxidative stress, or glucose intolerance. These data suggest that although IL-4-stimulated alternatively activated macrophages upregulate fatty acid oxidation, fatty acid oxidation is dispensable for macrophage polarization and high-fat diet-induced metabolic dysfunction. Macrophage fatty acid oxidation likely plays a correlative, rather than causative, role in systemic metabolic dysfunction.


Assuntos
Ácidos Graxos/imunologia , Interleucina-4/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Doenças Metabólicas/imunologia , Obesidade/imunologia , Animais , Células Cultivadas , Dieta Hiperlipídica , Masculino , Doenças Metabólicas/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Oxirredução
13.
Respir Med ; 123: 98-104, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28137504

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive disease of dysregulated wound healing leading to unremitting scarring and loss of lung function. The predominant symptoms are dyspnea on exertion and a persistent dry cough. For patients with IPF, cough is more than just bothersome; it has a significant negative impact on quality of life and is a marker of disease severity and progression. The etiology of cough in IPF is unclear but may be due to architectural distortion of the lungs, increased sensitivity of the cough reflex, airway inflammation, or changes in mucus production and clearance. There also may be an overlap between IPF cough and cough due to other common etiologies such as asthma, gastroesophageal reflux disease, upper airway cough syndrome, and medications. There are no approved therapies to specifically treat IPF cough, and recently approved medications for IPF have not been evaluated in cough. Few clinical trials have focused on treatments for IPF cough. To date, there is only one randomized, placebo control therapeutic study for IPF cough with thalidomide, which significantly reduced IPF cough and improved quality of life. Two additional cohort studies report that interferon-α and prednisolone also decrease IPF cough. However, no medication is approved to treat IPF cough. Currently, the mainstay of therapy for IPF cough is standard cough suppressants, which have limited efficacy and often intolerable side effects. Future studies are needed to determine an effective therapy to alleviate this particularly debilitating symptom and improve overall quality of life for patients suffering with IPF.


Assuntos
Tosse/etiologia , Fibrose Pulmonar Idiopática/complicações , Antitussígenos/uso terapêutico , Comorbidade , Tosse/tratamento farmacológico , Refluxo Gastroesofágico/etiologia , Humanos , Interferon-alfa/uso terapêutico , Qualidade de Vida , Talidomida/uso terapêutico
14.
Laryngoscope ; 127(3): E107-E113, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27585358

RESUMO

OBJECTIVES/HYPOTHESIS: Laryngotracheal stenosis (LTS) is a chronic fibrotic disease characterized by fibroblast proliferation, collagen deposition, and matrix remodeling in the lamina propria of the larynx and/or trachea. Current medical therapies are limited by a poor understanding of the effector cell's (fibroblasts) cellular biology and metabolism. The purpose of this study was to compare cellular proliferation, function, and metabolism between normal and LTS-derived fibroblasts in vitro. We hypothesize that LTS-derived fibroblasts will demonstrate aberrant behavior with faster proliferation, increased collagen production, and altered metabolic allocation compared with normal fibroblasts. STUDY DESIGN: In vitro comparative analysis. METHODS: Human biopsies of normal and iatrogenic LTS tissue (n = 7) were obtained, and fibroblasts were isolated and cultured in vitro. Cellular proliferation, cellular histology, gene expression, and metabolic analyses were performed. Statistical analyses comparing normal and scar-derived fibroblasts were performed. RESULTS: LTS fibroblast proliferation rate, cellular surface area, and collagen-1 expression were increased compared to normal fibroblasts. Cellular metabolic analysis of LTS-derived fibroblasts demonstrated reduced oxidative phosphorylation and increased glycolysis/oxidative phosphorylation ratio compared with normal fibroblasts. CONCLUSIONS: Human iatrogenic LTS-derived fibroblasts demonstrated aberrant behavior when compared with normal fibroblasts. A Warburg-like effect was revealed, suggesting human iatrogenic LTS fibroblasts drive their proliferation with aerobic glycolysis. The distinct metabolism suggests metabolic inhibitors could reduce fibroblast hyperplasia and hypertrophy in LTS and fibrosis in general. LEVEL OF EVIDENCE: NA Laryngoscope, 127:E107-E113, 2017.


Assuntos
Proliferação de Células/fisiologia , Fibroblastos/metabolismo , Laringoestenose/patologia , Consumo de Oxigênio , Estenose Traqueal/patologia , Biópsia por Agulha , Técnicas de Cultura de Células , Células Cultivadas , Colágeno/metabolismo , Humanos , Imuno-Histoquímica , Laringoestenose/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Valores de Referência , Estudos de Amostragem , Estatísticas não Paramétricas , Estenose Traqueal/metabolismo
15.
PLoS One ; 11(9): e0163288, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27649073

RESUMO

Pulmonary fibrosis is a devastating, incurable disease in which chronic inflammation and dysregulated, excessive wound healing lead to progressive fibrosis, lung dysfunction, and ultimately death. Prior studies have implicated the cytokine IL-17A and Th17 cells in promoting the development of fibrosis. We hypothesized that loss of Th17 cells via CD4-specific deletion of mTORC1 activity would abrogate the development of bleomycin-induced pulmonary fibrosis. However, in actuality loss of Th17 cells led to increased mortality and fibrosis in response to bleomycin. We found that in the absence of Th17 cells, there was continued production of IL-17A by γδ T cells. These IL-17A+ γδ T cells were associated with increased lung neutrophils and M2 macrophages, accelerated development of fibrosis, and increased mortality. These data elucidate the critical role of IL-17A+ γδ T cells in promoting chronic inflammation and fibrosis, and reveal a novel therapeutic target for treatment of pulmonary fibrosis.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Complexos Multiproteicos/metabolismo , Fibrose Pulmonar/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Subpopulações de Linfócitos T/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Bleomicina , Linfócitos T CD4-Positivos/patologia , Modelos Animais de Doenças , Interleucina-17/biossíntese , Macrófagos/metabolismo , Macrófagos/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/genética , Neutrófilos/metabolismo , Neutrófilos/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/patologia , Transdução de Sinais , Subpopulações de Linfócitos T/patologia , Serina-Treonina Quinases TOR/genética
17.
Otolaryngol Head Neck Surg ; 153(2): 244-50, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26084828

RESUMO

OBJECTIVE: To define the inflammatory cell infiltrate preceding fibrosis in a laryngotracheal stenosis (LTS) murine model. STUDY DESIGN: Prospective controlled murine study. SETTING: Laboratory. SUBJECTS AND METHODS: Chemomechanical injury mice (n = 44) sustained bleomycin-coated wire-brush injury to the laryngotracheal complex while mechanical injury controls (n = 42) underwent phosphate-buffered saline (PBS)-coated wire-brush injury. Mock surgery controls (n = 34) underwent anterior transcervical tracheal exposure only. Inflammatory and fibrosis protein and gene expression were assessed in each condition. Immunohistochemistry served as a secondary outcome. RESULTS: In chemomechanical injury mice, there was an upregulation of collagen I (P < .0001, P < .0001), Tgf-ß (P = .0023, P = .0008), and elastin (P < .0001, P < .0001) on day 7; acute inflammatory gene Il1ß (P = .0027, P = .0008) on day 1; and macrophage gene CD11b (P = .0026, P = .0033) on day 1 vs mechanical and mock controls, respectively. M1 marker inducible nitric oxide synthase (iNOS) expression decreased (P = .0014) while M2 marker Arg1 (P = .0002) increased on day 7 compared with mechanical controls. Flow cytometry demonstrated increased macrophages (P = .0058, day 4) and M1 macrophages (P = .0148, day 4; P = .0343, day 7; P = .0229, day 10) compared to mock controls. There were similarities between chemomechanical and mechanical injury mice with an increase in M2 macrophages at day 10 (P = .0196). CONCLUSIONS: The bleomycin-induced LTS mouse model demonstrated increased macrophages involved with the development of fibrosis. Macrophage immunophenotype suggested that dysregulated M2 macrophages have a role in abnormal laryngotracheal wound healing. These data delineate inflammatory cells and signaling pathways in LTS that may potentially be modulated to lessen fibroblast proliferation and collagen deposition.


Assuntos
Laringoestenose/patologia , Macrófagos/patologia , Estenose Traqueal/patologia , Animais , Bleomicina , Colágeno/análise , Modelos Animais de Doenças , Elastina/análise , Citometria de Fluxo , Expressão Gênica , Imuno-Histoquímica , Laringoestenose/induzido quimicamente , Laringe/lesões , Camundongos , Camundongos Endogâmicos C57BL , Estudos Prospectivos , Traqueia/lesões , Estenose Traqueal/induzido quimicamente , Fator de Crescimento Transformador beta/análise
18.
Cancer Cell ; 27(4): 435-6, 2015 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-25873169

RESUMO

Recent clinical trials in cancer therapy have demonstrated unprecedented responses through blockade of CTLA-4 and PD-1 immune checkpoint pathways. In a provocative recent paper in Science Translational Medicine, Hatfield and colleagues demonstrate the ability of supplemental oxygen to act as a novel immune checkpoint inhibitor by disrupting the hypoxia-adenosine-A2aR pathway.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Oxigênio/uso terapêutico , Animais , Feminino , Humanos
19.
Otolaryngol Head Neck Surg ; 152(5): 881-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25754184

RESUMO

OBJECTIVE: To determine if rapamycin inhibits the growth, function, and metabolism of human laryngotracheal stenosis (LTS)-derived fibroblasts. STUDY DESIGN: Controlled in vitro study. SETTING: Tertiary care hospital in a research university. SUBJECTS AND METHODS: Fibroblasts isolated from biopsies of 5 patients with laryngotracheal stenosis were cultured. Cell proliferation, histology, gene expression, and cellular metabolism of LTS-derived fibroblasts were assessed in 4 conditions: (1) fibroblast growth medium, (2) fibroblast growth medium with dimethylsulfoxide (DMSO), (3) fibroblast growth medium with 10(-10) M (low-dose) rapamycin dissolved in DMSO, and (4) fibroblast growth medium with 10(-9) M (high-dose) rapamycin dissolved in DMSO. RESULTS: The LTS fibroblast count and DNA concentration were reduced after treatment with high-dose rapamycin compared to DMSO (P = .0007) and normal (P = .0007) controls. Collagen I expression decreased after treatment with high-dose rapamycin versus control (P = .0051) and DMSO (P = .0093) controls. Maximal respiration decreased to 68.6 pMoles of oxygen/min/10 mg/protein from 96.9 for DMSO (P = .0002) and 97.0 for normal (P = .0022) controls. Adenosine triphosphate (ATP) production decreased to 66.8 pMoles from 88.1 for DMSO (P = .0006) and 83.3 for normal (P = .0003) controls. Basal respiration decreased to 78.6 pMoles from 108 for DMSO (P = .0002) and 101 for normal (P = .0014) controls. CONCLUSIONS: Rapamycin demonstrated an anti-fibroblast effect by significantly reducing the proliferation, metabolism, and collagen deposition of human LTS fibroblast in vitro. Rapamycin significantly decreased oxidative phosphorylation of LTS fibroblasts, suggesting at a potential mechanism for the reduced proliferation and differentiation. Furthermore, rapamycin's anti-fibroblast effects indicate a promising adjuvant therapy for the treatment of laryngotracheal stenosis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Imunossupressores/farmacologia , Laringoestenose/patologia , Sirolimo/farmacologia , Técnicas de Cultura de Células , Colágeno/metabolismo , Fibroblastos/metabolismo , Humanos , Laringoestenose/imunologia , Reação em Cadeia da Polimerase em Tempo Real
20.
JAMA Otolaryngol Head Neck Surg ; 140(10): 961-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25144860

RESUMO

IMPORTANCE: Laryngotracheal stenosis (LTS) lacks an ideal animal model to study its unique wound-healing pathophysiology and the effect of interventions. OBJECTIVE: To present an in vivo, in situ mouse model of LTS that can be used to investigate its pathophysiology, mechanisms, and interventions for treatment. DESIGN, SETTING, AND SUBJECTS: Prospective controlled animal study performed at an academic animal research facility on 87 C57BL/6 mice. INTERVENTIONS: Experimental mice (n = 40) underwent bleomycin-coated wire-brush injury to the larynx and trachea, while mechanical injury controls (n = 32) underwent phosphate-buffered saline-coated wire-brush injury. Normal controls (n = 9) underwent no intervention, and mock surgery controls (n = 6) underwent anterior transcervical tracheal exposure only. Laryngotracheal complexes were harvested at days 7, 14, and 21 after injury. At the respective time points, mice in the chemomechanical and mechanical injury groups were killed, and their laryngotracheal complexes were harvested for histologic analysis. Normal and mock surgery controls were killed and then underwent histologic analysis. MAIN OUTCOMES AND MEASURES: The primary outcome measure was lamina propria thickness. RESULTS: The chemomechanical injury group maintained a significant increase in lamina propria thickness through day 21 compared with uninjured controls at day 7 (82.7 vs 41.8 µm; P<.05), day 14 (93.5 vs 26.0 µm; P<.05), and day 21 (91.2 vs 40.8 µm; P<.05). Compared with the mechanical injury group, the chemomechanical injury group demonstrated a significantly increased thickness at 21 days (91.2 vs 33.7 µm; P<.05). CONCLUSIONS AND RELEVANCE: Chemomechanical initiation of fibrosis in situ creates a viable mouse model of LTS that incorporates the physiologic circulatory supply and airflow. This small-animal model may be used to investigate the pathogenesis and inflammatory mechanisms of iatrogenic LTS and test therapeutic interventions to reverse or reduce the development of fibrosis.


Assuntos
Modelos Animais de Doenças , Laringoestenose/fisiopatologia , Laringoestenose/cirurgia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Estudos Prospectivos , Coloração e Rotulagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA