Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Nat Commun ; 14(1): 5814, 2023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37726288

RESUMO

Epithelial plasticity has been suggested in lungs of mice following genetic depletion of stem cells but is of unknown physiological relevance. Viral infection and chronic lung disease share similar pathological features of stem cell loss in alveoli, basal cell (BC) hyperplasia in small airways, and innate immune activation, that contribute to epithelial remodeling and loss of lung function. We show that a subset of distal airway secretory cells, intralobar serous (IS) cells, are activated to assume BC fates following influenza virus infection. Injury-induced hyperplastic BC (hBC) differ from pre-existing BC by high expression of IL-22Ra1 and undergo IL-22-dependent expansion for colonization of injured alveoli. Resolution of virus-elicited inflammation results in BC to IS re-differentiation in repopulated alveoli, and increased local expression of protective antimicrobial factors, but fails to restore normal alveolar epithelium responsible for gas exchange.


Assuntos
Células Epiteliais , Alvéolos Pulmonares , Animais , Camundongos , Diferenciação Celular , Hiperplasia , Imunidade Inata
2.
Am J Physiol Cell Physiol ; 325(2): C483-C495, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37458437

RESUMO

Pulmonary fibrosis comprises a range of chronic interstitial lung diseases (ILDs) that impose a significant burden on patients and public health. Among these, idiopathic pulmonary fibrosis (IPF), a disease of aging, is the most common and most severe form of ILD and is treated largely by lung transplantation. The lack of effective treatments to stop or reverse lung fibrosis-in fact, fibrosis in most organs-has sparked the need to understand causative mechanisms with the goal of identifying critical points for potential therapeutic intervention. Findings from many groups have indicated that repeated injury to the alveolar epithelium-where gas exchange occurs-leads to stem cell exhaustion and impaired alveolar repair that, in turn, triggers the onset and progression of fibrosis. Cellular senescence of alveolar epithelial progenitors is a critical cause of stemness failure. Hence, senescence impairs repair and thus contributes significantly to fibrosis. In this review, we discuss recent evidence indicating that senescence of epithelial progenitor cells impairs alveolar homeostasis and repair creating a profibrotic environment. Moreover, we discuss the impact of senescent alveolar epithelial progenitors, alveolar type 2 (AT2) cells, and AT2-derived transitional epithelial cells in fibrosis. Emerging evidence indicates that transitional epithelial cells are prone to senescence and, hence, are a new player involved in senescence-associated lung fibrosis. Understanding the complex interplay of cell types and cellular regulatory factors contributing to alveolar epithelial progenitor senescence will be crucial to developing targeted therapies to mitigate their downstream profibrotic sequelae and to promote normal alveolar repair.NEW & NOTEWORTHY With an aging population, lung fibrotic diseases are becoming a global health burden. Dysfunctional repair of the alveolar epithelium is a key causative process that initiates lung fibrosis. Normal alveolar regeneration relies on functional progenitor cells; however, the senescence of these cells, which increases with age, hinders their ability to contribute to repair. Here, we discuss studies on the control and consequence of progenitor cell senescence in fibrosis and opportunities for research.


Assuntos
Células Epiteliais Alveolares , Fibrose Pulmonar Idiopática , Humanos , Idoso , Células Epiteliais Alveolares/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Senescência Celular , Envelhecimento , Células-Tronco/metabolismo , Células Epiteliais/metabolismo , Pulmão/metabolismo
3.
Elife ; 122023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-37314162

RESUMO

Aging is a critical risk factor in idiopathic pulmonary fibrosis (IPF). Dysfunction and loss of type 2 alveolar epithelial cells (AEC2s) with failed regeneration is a seminal causal event in the pathogenesis of IPF, although the precise mechanisms for their regenerative failure and demise remain unclear. To systematically examine the genomic program changes of AEC2s in aging and after lung injury, we performed unbiased single-cell RNA-seq analyses of lung epithelial cells from uninjured or bleomycin-injured young and old mice, as well as from lungs of IPF patients and healthy donors. We identified three AEC2 subsets based on their gene signatures. Subset AEC2-1 mainly exist in uninjured lungs, while subsets AEC2-2 and AEC2-3 emerged in injured lungs and increased with aging. Functionally, AEC2 subsets are correlated with progenitor cell renewal. Aging enhanced the expression of the genes related to inflammation, stress responses, senescence, and apoptosis. Interestingly, lung injury increased aging-related gene expression in AEC2s even in young mice. The synergistic effects of aging and injury contributed to impaired AEC2 recovery in aged mouse lungs after injury. In addition, we also identified three subsets of AEC2s from human lungs that formed three similar subsets to mouse AEC2s. IPF AEC2s showed a similar genomic signature to AEC2 subsets from bleomycin-injured old mouse lungs. Taken together, we identified synergistic effects of aging and AEC2 injury in transcriptomic and functional analyses that promoted fibrosis. This study provides new insights into the interactions between aging and lung injury with interesting overlap with diseased IPF AEC2 cells.


Assuntos
Lesão Pulmonar , Fibrose Pulmonar , Humanos , Camundongos , Animais , Fibrose Pulmonar/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Pulmão/patologia , Envelhecimento , Bleomicina/toxicidade
4.
PLoS Pathog ; 19(4): e1010942, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37027441

RESUMO

During chronic cystic fibrosis (CF) infections, evolved Pseudomonas aeruginosa antibiotic resistance is linked to increased pulmonary exacerbations, decreased lung function, and hospitalizations. However, the virulence mechanisms underlying worse outcomes caused by antibiotic resistant infections are poorly understood. Here, we investigated evolved aztreonam resistant P. aeruginosa virulence mechanisms. Using a macrophage infection model combined with genomic and transcriptomic analyses, we show that a compensatory mutation in the rne gene, encoding RNase E, increased pyoverdine and pyochelin siderophore gene expression, causing macrophage ferroptosis and lysis. We show that iron-bound pyochelin was sufficient to cause macrophage ferroptosis and lysis, however, apo-pyochelin, iron-bound pyoverdine, or apo-pyoverdine were insufficient to kill macrophages. Macrophage killing could be eliminated by treatment with the iron mimetic gallium. RNase E variants were abundant in clinical isolates, and CF sputum gene expression data show that clinical isolates phenocopied RNase E variant functions during macrophage infection. Together these data show how P. aeruginosa RNase E variants can cause host damage via increased siderophore production and host cell ferroptosis but may also be targets for gallium precision therapy.


Assuntos
Ferro , Infecções por Pseudomonas , Humanos , Ferro/metabolismo , Sideróforos/farmacologia , Sideróforos/metabolismo , Pseudomonas aeruginosa/metabolismo , Virulência , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/metabolismo
5.
Nat Rev Urol ; 19(9): 515-533, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35764795

RESUMO

Bladder cancer is a prevalent but currently understudied cancer type and patient outcomes are poor when it progresses to the muscle-invasive stage. Current research in bladder cancer focuses on the genetic and epigenetic alterations occurring within the urothelial cell compartment; however, the stromal compartment receives less attention. Dynamic changes and intercellular communications occur in the tumour microenvironment (TME) of the bladder - a new concept and niche that we designate as the bladder TME (bTME) - during tumour evolution, metastatic progression and in the context of therapeutic response. Collagens and their cognate receptors, the discoidin domain receptors, have a role in various steps of the metastatic cascade and in immune checkpoint resistance. Furthermore, the presence of another TME niche, the metastatic TME (met-TME), is a novel concept that could support divergent progression of metastatic colonization in different organs, resulting in distant metastases with distinct characteristics and genetics from the primary tumour. The stroma has divergent roles in mediating therapeutic response to BCG immunotherapy and immune checkpoint inhibitors, as well as conventional chemotherapy or trimodality therapy (that is, maximal transurethral resection of bladder tumour, chemotherapy and radiotherapy). The local bTME and distant met-TME are currently conceptually and therapeutically unexploited niches that should be actively investigated. New biological insights from these TMEs will enable rational design of strategies that co-target the tumour and stroma, which are expected to improve the outcomes of patients with advanced bladder cancer.


Assuntos
Microambiente Tumoral , Neoplasias da Bexiga Urinária , Humanos , Imunoterapia/métodos , Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/patologia
6.
J Clin Invest ; 132(11)2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35389887

RESUMO

Type 2 alveolar epithelial cells (AEC2s) function as progenitor cells in the lung. We have shown previously that failure of AEC2 regeneration results in progressive lung fibrosis in mice and is a cardinal feature of idiopathic pulmonary fibrosis (IPF). In this study, we identified deficiency of a specific zinc transporter, SLC39A8 (ZIP8), in AEC2s from both IPF lungs and lungs of old mice. Loss of ZIP8 expression was associated with impaired renewal capacity of AEC2s and enhanced lung fibrosis. ZIP8 regulation of AEC2 progenitor function was dependent on SIRT1. Replenishment with exogenous zinc and SIRT1 activation promoted self-renewal and differentiation of AEC2s from lung tissues of IPF patients and old mice. Deletion of Zip8 in AEC2s in mice resulted in impaired AEC2 renewal, increased susceptibility to bleomycin injury, and development of spontaneous lung fibrosis. Therapeutic strategies to restore zinc metabolism and appropriate SIRT1 signaling could improve AEC2 progenitor function and mitigate ongoing fibrogenesis.


Assuntos
Proteínas de Transporte de Cátions , Fibrose Pulmonar Idiopática , Envelhecimento , Células Epiteliais Alveolares , Animais , Bleomicina , Proteínas de Transporte de Cátions/metabolismo , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/metabolismo , Camundongos , Sirtuína 1/genética , Sirtuína 1/metabolismo , Células-Tronco/metabolismo , Zinco/metabolismo
7.
Stem Cell Res Ther ; 13(1): 64, 2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-35130980

RESUMO

Recent advances in single-cell RNA sequencing (scRNA-seq) and epithelium lineage labeling have yielded identification of multiple abnormal epithelial progenitor populations during alveolar type 2 (ATII) cell differentiation into alveolar type 1 (ATI) cells during regenerative lung post-fibrotic injury. These abnormal cells include basaloid/basal-like cells, ATII transition cells, and persistent epithelial progenitors (PEPs). These cells occurred and accumulated during the regeneration of distal airway and alveoli in response to both chronic and acute pulmonary injury. Among the alveolar epithelial progenitors, PEPs express a distinct Krt8+ phenotype that is rarely found in intact alveoli. However, post-injury, the Krt8+ phenotype is seen in dysplastic epithelial cells. Fully understanding the characteristics and functions of these newly found, injury-induced abnormal behavioral epithelial progenitors and the signaling pathways regulating their phenotype could potentially point the way to unique therapeutic targets for fibrosing lung diseases. This review summarizes recent advances in understanding these epithelial progenitors as they relate to uncovering regenerative mechanisms.


Assuntos
Lesão Pulmonar , Células Epiteliais Alveolares , Células Epiteliais , Humanos , Pulmão , Alvéolos Pulmonares
8.
iScience ; 24(6): 102551, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34151224

RESUMO

Pulmonary mesenchymal cells are critical players in both the mouse and human during lung development and disease states. They are increasingly recognized as highly heterogeneous, but there is no consensus on subpopulations or discriminative markers for each subtype. We completed scRNA-seq analysis of mesenchymal cells from the embryonic, postnatal, adult and aged fibrotic lungs of mice and humans. We consistently identified and delineated the transcriptome of lipofibroblasts, myofibroblasts, smooth muscle cells, pericytes, mesothelial cells, and a novel population characterized by Ebf1 expression. Subtype selective transcription factors and putative divergence of the clusters during development were described. Comparative analysis revealed orthologous subpopulations with conserved transcriptomic signatures in murine and human lung mesenchymal cells. All mesenchymal subpopulations contributed to matrix gene expression in fibrosis. This analysis would enhance our understanding of mesenchymal cell heterogeneity in lung development, homeostasis and fibrotic disease conditions.

9.
Am J Respir Crit Care Med ; 203(4): 458-470, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33052708

RESUMO

Rationale: Aberrant lung remodeling in idiopathic pulmonary fibrosis (IPF) is characterized by elevated MMP9 (matrix metalloproteinase 9) expression, but the precise role of this matrix metalloproteinase in this disease has yet to be fully elucidated.Objectives: To evaluate antifibrotic effects of MMP9 inhibition on IPF.Methods: Quantitative genomic, proteomic, and functional analyses both in vitro and in vivo were used to determine MMP9 expression in IPF cells and the effects of MMP9 inhibition on profibrotic mechanisms.Measurements and Main Results: In the present study, we demonstrate that MMP9 expression was increased in airway basal cell (ABC)-like cells from IPF lungs compared with ABC cells from normal lungs. The inhibition of MMP9 activity with an anti-MMP9 antibody, andecaliximab, blocked TGF-ß1 (transforming growth factor ß1)-induced Smad2 phosphorylation. However, in a subset of cells from patients with IPF, TGF-ß1 activation in their ABC-like cells was unaffected or enhanced by MMP9 blockade (i.e., nonresponders). Further analysis of nonresponder ABC-like cells treated with andecaliximab revealed an association with type 1 IFN expression, and the addition of IFNα to these cells modulated both MMP9 expression and TGF-ß1 activation. Finally, the inhibition of MMP9 ameliorated pulmonary fibrosis induced by responder lung cells but not a nonresponder in a humanized immunodeficient mouse model of IPF.Conclusions: Together, these data demonstrate that MMP9 regulates the activation of ABC-like cells in IPF and that targeting this MMP might be beneficial to a subset of patients with IPF who show sufficient expression of type 1 IFNs.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Células Epiteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/fisiopatologia , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Metaloproteinase 9 da Matriz/metabolismo , Animais , Anticorpos Monoclonais Humanizados/metabolismo , California/epidemiologia , Feminino , Humanos , Fibrose Pulmonar Idiopática/epidemiologia , Fibrose Pulmonar Idiopática/genética , Metaloproteinase 9 da Matriz/genética , Camundongos , Michigan/epidemiologia , Modelos Animais , Proteômica , Estados Unidos
11.
J Allergy Clin Immunol ; 143(4): 1536-1548, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30445062

RESUMO

BACKGROUND: Macrophage plasticity allows cells to adopt different phenotypes, a property with important implications in disorders such as cystic fibrosis (CF) and asthma. OBJECTIVE: We sought to examine the transcriptional and functional significance of macrophage repolarization from an M1 to an M2 phenotype and assess the role of a common human genetic disorder (CF) and a prototypical allergic disease (asthma) in this transformation. METHODS: Monocyte-derived macrophages were collected from healthy subjects and patients with CF and polarized to an M2 state by using IL-4, IL-10, glucocorticoids, apoptotic PMNs, or azithromycin. We performed transcriptional profiling and pathway analysis for each stimulus. We assessed the ability of M2-repolarized macrophages to respond to LPS rechallenge and clear apoptotic neutrophils and used murine models to determine conserved functional responses to IL-4 and IL-10. We investigated whether M2 signatures were associated with alveolar macrophage phenotypes in asthmatic patients. RESULTS: We found that macrophages exhibit highly diverse responses to distinct M2-polarizing stimuli. Specifically, IL-10 activated proinflammatory pathways and abrogated LPS tolerance, allowing rapid restoration of LPS responsiveness. In contrast, IL-4 enhanced LPS tolerance, dampening proinflammatory responses after repeat LPS challenge. A common theme observed across all M2 stimuli was suppression of interferon-associated pathways. We found that CF macrophages had intact reparative and transcriptional responses, suggesting that macrophage contributions to CF-related lung disease are primarily shaped by their environment. Finally, we leveraged in vitro-derived signatures to show that allergen provocation induces distinct M2 state transcriptional patterns in alveolar macrophages. CONCLUSION: Our findings highlight the diversity of macrophage polarization, attribute functional consequences to different M2 stimuli, and provide a framework to phenotype macrophages in disease states.


Assuntos
Asma/imunologia , Fibrose Cística/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Adulto , Animais , Citocinas/imunologia , Feminino , Humanos , Masculino , Camundongos , Fenótipo , Transcrição Gênica , Transcriptoma
12.
Front Immunol ; 9: 2817, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30564235

RESUMO

Using an in vivo model of tolerance to TLR7-induced skin inflammation, we found a critical role for macrophage-derived MMP10 in mediating immune hypo-responsiveness. Cutaneous exposure to Imiquimod (IMQ), a TLR7 agonist, induced acute expression of pro-inflammatory factors (IL1ß, IL6, CXCL1) and neutrophil influx equally in both wildtype and Mmp10-/- mice. However, whereas subsequent exposure (11 and 12 days later) to IMQ led to marked abrogation of pro-inflammatory factor expression in wildtype mice, Mmp10-/- mice responded similarly as they did to the first application. In addition, the second exposure led to increased expression of negative regulators of TLR signaling (TNFAIP3, IRAK3) and immunosuppressive cytokines (IL10, TGFß1) in wildtype mice but not in Mmp10-/- mice. In vitro studies demonstrated that prior exposure of IMQ to bone marrow-derived macrophages (BMDM) made wildtype cells refractory to subsequent stimulation but did not for Mmp10-/- macrophages. These findings expand the critical roles MMP10 plays in controlling macrophage activation to indicate that the development of immune tolerance to TLR7 ligand is dependent on this macrophage-derived proteinase.


Assuntos
Tolerância Imunológica/imunologia , Macrófagos/imunologia , Metaloproteinase 10 da Matriz/imunologia , Glicoproteínas de Membrana/imunologia , Receptor 7 Toll-Like/imunologia , Animais , Citocinas/imunologia , Feminino , Imiquimode/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Masculino , Glicoproteínas de Membrana/agonistas , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Receptor 7 Toll-Like/agonistas
13.
Atherosclerosis ; 278: 124-134, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30268068

RESUMO

BACKGROUND AND AIMS: Matrix metalloproteinases (MMPs) have been implicated in atherosclerosis and vascular calcification. Among them, we reported that MMP10 is present in human atheroma, associated with atherosclerosis. However, it remains unclear whether MMP10 is involved in atherogenesis and vascular calcification. METHODS: MMP10 was measured in serum from patients with subclinical atherosclerosis and analyzed in carotid endarterectomies by immunostaining. ApoE-deficient mice (Apoe-/-) were crossed to MMP10-deficient (Mmp10-/-) mice and followed up to 20 months. Plaque area and composition were assessed by histology and immunohistochemistry. Inflammatory markers were measured in atherosclerotic plaques by RT-qPCR, and leukocyte subpopulations were analyzed by flow cytometry. In vitro calcification assays were performed in aortic vascular smooth muscle cells (VSMC). RESULTS: MMP10 serum levels were associated with coronary calcification in subjects with subclinical atherosclerosis. Immunostaining revealed MMP10 expression in human atheromas, spatially associated with calcification areas, and complicated plaques released higher amounts of MMP10 than non-diseased segments. Interestingly, vascular MMP10 expression was confined to the atherosclerotic lesion in Apoe-/- mice, and Apoe-/-Mmp10-/- showed a substantial reduction in atherosclerotic lesion size, macrophage content and plaque calcification. Reduced local and systemic inflammatory markers could be demonstrated in Apoe-/-Mmp10-/- by gene expression and flow cytometry analysis. Calcium phosphate deposition and vascular calcification markers were downregulated in VSMC from Apoe-/-Mmp10-/- mice. CONCLUSIONS: Delayed plaque progression and altered cellular composition in the absence of MMP10 suggests that MMP10 plays a role in atherosclerosis, favoring inflammation, development and complication of the plaque.


Assuntos
Metaloproteinase 10 da Matriz/deficiência , Metaloproteinase 10 da Matriz/fisiologia , Placa Aterosclerótica/metabolismo , Idoso , Animais , Antígeno CD11b/metabolismo , Progressão da Doença , Endarterectomia das Carótidas , Feminino , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Humanos , Inflamação , Masculino , Metaloproteinase 10 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Calcificação Vascular/patologia
14.
Matrix Biol ; 73: 77-104, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29524630

RESUMO

The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.


Assuntos
Matriz Extracelular/fisiologia , Pneumopatias/metabolismo , Pulmão/metabolismo , Fenômenos Biomecânicos , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Homeostase , Humanos , Fenótipo
15.
Curr Rheumatol Rep ; 20(4): 17, 2018 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-29550962

RESUMO

PURPOSE OF REVIEW: Macrophages are central players in the immune response following tissue injury. These cells perform many functions, and the changing tissue microenvironment during injury shapes macrophage phenotype down a variety of polarized pathways. This review summarizes the current knowledge on the roles of macrophages during different stages of tissue injury, repair, and-if repair is not achieved-fibrosis. RECENT FINDINGS: Macrophages present early in inflammation are functionally distinct from those at later stages. The predominant macrophage phenotype must transition from pro-inflammatory to pro-reparative to facilitate wound healing and scar resolution. If macrophages fail to acquire a tissue-healing phenotype, dysregulated signals can be drivers of disease processes, such as sustained, exuberant inflammation-as occurs in arthropathies-and fibrosis. Comprehensive understanding of the roles of specific macrophage populations at different stages of the repair process will support the development of immune-targeted therapies for diseases such as fibrosis.


Assuntos
Fibrose/metabolismo , Macrófagos/metabolismo , Cicatrização/fisiologia , Fibrose/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Ativação de Macrófagos/fisiologia , Macrófagos/patologia
16.
Matrix Biol ; 73: 34-51, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29406250

RESUMO

Several studies have implicated a causative role for specific matrix metalloproteinases (MMPs) in the development and progression of cigarette smoke-induced chronic obstructive pulmonary disease (COPD) and its severe sequela, emphysema. However, the precise function of any given MMP in emphysema remains an unanswered question. Emphysema results from the degradation of alveolar elastin - among other possible mechanisms - a process that is often thought to be caused by elastolytic proteinases made by macrophages. In this article, we discuss the data suggesting, supporting, or refuting causative roles of macrophage-derived MMPs, with a focus on MMPs-7, -9, -10, -12, and 28, in both the human disease and mouse models of emphysema. Findings from experimental models suggest that some MMPs, such as MMP-12, may directly breakdown elastin, whereas others, particularly MMP-10 and MMP-28, promote the development of emphysema by influencing the proteolytic and inflammatory activities of macrophages.


Assuntos
Elastina/metabolismo , Metaloproteinases da Matriz/genética , Enfisema Pulmonar/genética , Animais , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Macrófagos/imunologia , Metaloproteinases da Matriz/metabolismo , Camundongos , Polimorfismo de Nucleotídeo Único , Proteólise , Enfisema Pulmonar/imunologia , Enfisema Pulmonar/metabolismo
17.
Am J Pathol ; 188(4): 1094-1103, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29355516

RESUMO

Syndecan-1 is a transmembrane proteoglycan expressed prominently by lung epithelium and has pleiotropic functions such as regulating cell migration, proliferation, and survival. Loss of syndecan-1 expression by lung cancer cells is associated with higher-grade cancers and worse clinical prognosis. We evaluated the effects of syndecan-1 in various cell-based and animal models of lung cancer and found that lung tumorigenesis was moderated by syndecan-1. We also demonstrate that syndecan-1 (or lack thereof) alters the miRNA cargo carried within exosomes exported from lung cancer cells. Analysis of the changes in miRNA expression identified a distinct shift toward augmented procancer signaling consistent with the changes found in lung adenocarcinoma. Collectively, our work identifies syndecan-1 as an important factor in lung cancer cells that shapes the tumor microenvironment through alterations in miRNA packaging within exosomes.


Assuntos
Carcinogênese/metabolismo , Exossomos/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , MicroRNAs/genética , Sindecana-1/metabolismo , Células A549 , Adenocarcinoma de Pulmão/metabolismo , Adenocarcinoma de Pulmão/patologia , Animais , Proliferação de Células , Regulação para Baixo/genética , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/patologia , Camundongos , MicroRNAs/metabolismo , Análise de Sobrevida , Regulação para Cima/genética
18.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1069-L1086, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28912382

RESUMO

Growing evidence suggests that versican is important in the innate immune response to lung infection. Our goal was to understand the regulation of macrophage-derived versican and the role it plays in innate immunity. We first defined the signaling events that regulate versican expression, using bone marrow-derived macrophages (BMDMs) from mice lacking specific Toll-like receptors (TLRs), TLR adaptor molecules, or the type I interferon receptor (IFNAR1). We show that LPS and polyinosinic-polycytidylic acid [poly(I:C)] trigger a signaling cascade involving TLR3 or TLR4, the Trif adaptor, type I interferons, and IFNAR1, leading to increased expression of versican by macrophages and implicating versican as an interferon-stimulated gene. The signaling events regulating versican are distinct from those for hyaluronan synthase 1 (HAS1) and syndecan-4 in macrophages. HAS1 expression requires TLR2 and MyD88. Syndecan-4 requires TLR2, TLR3, or TLR4 and both MyD88 and Trif. Neither HAS1 nor syndecan-4 is dependent on type I interferons. The importance of macrophage-derived versican in lungs was determined with LysM/Vcan-/- mice. These studies show increased recovery of inflammatory cells in the bronchoalveolar lavage fluid of poly(I:C)-treated LysM/Vcan-/- mice compared with control mice. IFN-ß and IL-10, two important anti-inflammatory molecules, are significantly decreased in both poly(I:C)-treated BMDMs from LysM/Vcan-/- mice and bronchoalveolar lavage fluid from poly(I:C)-treated LysM/Vcan-/- mice compared with control mice. In short, type I interferon signaling regulates versican expression, and versican is necessary for type I interferon production. These findings suggest that macrophage-derived versican is an immunomodulatory molecule with anti-inflammatory properties in acute pulmonary inflammation.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Imunidade Inata , Interferon beta/imunologia , Pulmão/imunologia , Macrófagos Alveolares/imunologia , Versicanas/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Hialuronan Sintases/genética , Hialuronan Sintases/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Sindecana-4/genética , Sindecana-4/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Versicanas/genética
19.
J Immunol ; 198(12): 4813-4822, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28500076

RESUMO

We tested the role of Stat5 in dendritic cell and alveolar macrophage (AM) homeostasis in the lung using CD11c-cre mediated deletion (Cre+5f/f). We show that Stat5 is required for CD103+ dendritic cell and AM development. We found that fetal monocyte maturation into AMs was impaired in Cre+5f/f mice, and we also confirmed impaired AM development of progenitor cells using mixed chimera experiments. In the absence of Stat5 signaling in AMs, mice developed alveolar proteinosis with altered lipid homeostasis. In addition, loss of Stat5 in CD11c+ cells was associated with exaggerated LPS-induced inflammatory responses and vascular leak. In Cre+5f/f mice, there was loss of immune-dampening effects on epithelial cells, a key source of CCL2 that serves to recruit monocytes and macrophages. These findings demonstrate the critical importance of Stat5 signaling in maintaining lung homeostasis, and underscore the importance of resident macrophages in moderating tissue damage and excess inflammation.


Assuntos
Antígenos CD/imunologia , Células Dendríticas/fisiologia , Cadeias alfa de Integrinas/imunologia , Lesão Pulmonar/imunologia , Macrófagos Alveolares/fisiologia , Fator de Transcrição STAT5/metabolismo , Animais , Antígenos CD/genética , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Células Dendríticas/imunologia , Células Epiteliais/imunologia , Inflamação/imunologia , Cadeias alfa de Integrinas/genética , Macrófagos Alveolares/imunologia , Camundongos , Monócitos/imunologia , Proteinose Alveolar Pulmonar/imunologia
20.
Am J Pathol ; 187(6): 1288-1300, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28399390

RESUMO

Chronic obstructive pulmonary disease (COPD) comprises chronic bronchitis and emphysema, and is a leading cause of morbidity and mortality. Because tissue destruction is the prominent characteristic of emphysema, extracellular proteinases, particularly those with elastolytic ability, are often considered to be key drivers in this disease. Several human and mouse studies have implicated roles for matrix metalloproteinases (MMPs), particularly macrophage-derived proteinases, in COPD pathogenesis. MMP-28 is expressed by the pulmonary epithelium and macrophage, and we have found that it regulates macrophage recruitment and polarization. We hypothesized that MMP-28 has contributory roles in emphysema via alteration of macrophage numbers and activation. Because of the established association of emphysema pathogenesis to macrophage influx, we evaluated the inflammatory changes and lung histology of Mmp28-/- mice exposed to 3 and 6 months of cigarette smoke. At earlier time points, we found altered macrophage polarization in the smoke-exposed Mmp28-/- lung consistent with other published findings that MMP-28 regulates macrophage activation. At both 3 and 6 months, Mmp28-/- mice had blunted inflammatory responses more closely resembling nonsmoked mice, with a reduction in neutrophil recruitment and CXCL1 chemokine expression. By 6 months, Mmp28-/- mice were protected from emphysema. These results highlight a previously unrecognized role for MMP-28 in promoting chronic lung inflammation and tissue remodeling induced by cigarette smoke and highlight another potential target to modulate COPD.


Assuntos
Metaloproteinases da Matriz Secretadas/fisiologia , Enfisema Pulmonar/enzimologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Quimiocinas/metabolismo , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Enzimológica da Expressão Gênica/fisiologia , Pulmão/enzimologia , Macrófagos Alveolares/enzimologia , Masculino , Metaloproteinases da Matriz Secretadas/deficiência , Metaloproteinases da Matriz Secretadas/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/fisiologia , Pneumonia/enzimologia , Pneumonia/etiologia , Pneumonia/genética , Pneumonia/patologia , Doença Pulmonar Obstrutiva Crônica/enzimologia , Enfisema Pulmonar/etiologia , Enfisema Pulmonar/genética , Enfisema Pulmonar/patologia , Poluição por Fumaça de Tabaco/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...