Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Exp Biol Med (Maywood) ; 248(19): 1635-1644, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37837390

RESUMO

Respiratory viral infections with influenza A virus (IAV) or respiratory syncytial virus (RSV) pose a significant threat to public health due to excess morbidity and mortality. Dysregulated and excessive inflammatory responses are major underlying causes of viral pneumonia severity and morbidity, including aberrant host immune responses and increased risk for secondary bacterial infections. Currently available antiviral therapies have not substantially reduced the risk of severe viral pneumonia for these pathogens. Thus, new therapeutic approaches that can promote resolution of the pathogen-initiated inflammation without impairing host defense would represent a significant advance. Recent research has uncovered the potential for specialized pro-resolving mediators (SPMs) to transduce multipronged actions for the resolution of serious respiratory viral infection without increased risk for subsequent host susceptibility to bacterial infection. Here, we review recent advances in our understanding of SPM production and SPM receptor signaling in respiratory virus infections and the intriguing potential of harnessing SPM pathways to control excess morbidity and mortality from IAV and RSV pneumonia.


Assuntos
Pneumonia Viral , Infecções por Vírus Respiratório Sincicial , Humanos , Inflamação/metabolismo , Infecções por Vírus Respiratório Sincicial/complicações , Infecções por Vírus Respiratório Sincicial/metabolismo , Mediadores da Inflamação/metabolismo
3.
Am J Respir Cell Mol Biol ; 69(6): 666-677, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37552821

RESUMO

Eosinophils (Eos) reside in multiple organs during homeostasis and respond rapidly to an inflammatory challenge. Although Eos share chemical staining properties, they also demonstrate phenotypic and functional plasticity that is not fully understood. Here, we used a murine model of allergic lung inflammation to characterize Eos subsets and determine their spatiotemporal and functional regulation during inflammation and its resolution in response to resolvin D2 (RvD2), a potent specialized proresolving mediator. Two Eos subsets were identified by CD101 expression with distinct anatomic localization and transcriptional signatures at baseline and during inflammation. CD101low Eos were predominantly located in a lung vascular niche and responded to allergen challenge by moving into the lung interstitium. CD101high Eos were predominantly located in bronchoalveolar lavage (BAL) and extravascular lung, only present during inflammation, and had transcriptional evidence for cell activation. RvD2 reduced total Eos numbers and changed their phenotype and activation by at least two distinct mechanisms: decreasing interleukin 5-dependent recruitment of CD101low Eos and decreasing conversion of CD101low Eos to CD101high Eos. Collectively, these findings indicate that Eos are a heterogeneous pool of cells with distinct activation states and spatiotemporal regulation during resolution of inflammation and that RvD2 is a potent proresolving mediator for Eos recruitment and activation.


Assuntos
Alveolite Alérgica Extrínseca , Pneumonia , Eosinofilia Pulmonar , Camundongos , Animais , Eosinófilos/metabolismo , Líquido da Lavagem Broncoalveolar , Eosinofilia Pulmonar/metabolismo , Inflamação/metabolismo , Pneumonia/metabolismo , Fenótipo
4.
Proc Natl Acad Sci U S A ; 120(2): e2206480120, 2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-36595677

RESUMO

The resolution of infection is an active process with specific molecular and cellular mechanisms that temper inflammation and enhance pathogen clearance. Here, the specialized pro-resolving mediator (SPM) Maresin 1 (MaR1) inhibited respiratory syncytial virus (RSV)-induced inflammation. inlerleukin-13 production from type 2 innate lymphoid cells (ILC) and CD4 T helper type 2 cells was decreased by exogenous MaR1. In addition, MaR1 increased amphiregulin production and decreased RSV viral transcripts to promote resolution. MaR1 also promoted interferon-ß production in mouse lung tissues and also in pediatric lung slices. MaR1 significantly inhibited the RSV-triggered aberrant inflammatory phenotype in FoxP3-expressing Tregs. The receptor for MaR1, leucine-rich repeat-containing G protein-coupled receptor 6 (LGR6), was constitutively expressed on Tregs. Following RSV infection, mice lacking Lgr6 had exacerbated type 2 immune responses with an increased viral burden and blunted responses to MaR1. Together, these findings have uncovered a multi-pronged protective signaling axis for MaR1-Lgr6, improving Tregs's suppressive function and upregulating host antiviral genes resulting in decreased viral burden and pathogen-mediated inflammation, ultimately promoting restoration of airway mucosal homeostasis.


Assuntos
Pneumonia Viral , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Camundongos , Animais , Imunidade Inata , Linfócitos , Inflamação , Ácidos Docosa-Hexaenoicos/farmacologia , Receptores Acoplados a Proteínas G
5.
iScience ; 25(10): 105185, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36238896

RESUMO

Lipid phosphate phosphatases are a family of enzymes with diverse cellular metabolic functions. Phospholipid phosphatase 6 (PLPP6) is a regulator of cellular polyisoprenyl phosphates; however, its in vivo functions remain to be determined. Here, mouse PLPP6 was characterized to possess similar catalytic properties as the human enzyme. Plpp6 knockout mice (Plpp6 -/- ) were generated and displayed decreased airway allergen sensitization, pointing to a role for PLPP6 in the early events of lung allergic responses. Dendritic cell (DC) responses were investigated and endocytosis of allergen via macropinocytosis was decreased in Plpp6 -/- DCs that had lower cholesterol content. When reversed by cholesterol loading, the DC macropinocytosis defect is corrected. Adoptive transfer of Plpp6 -/- DCs to wild-type mice during sensitization was sufficient to decrease allergen-induced responses. Together, our findings have identified PLPP6 as a pivotal regulator of DC cholesterol content and macropinocytosis, cellular mechanisms that are important for pathologic responses in allergen-induced lung inflammation.

6.
mBio ; 13(4): e0126722, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35913160

RESUMO

Influenza A virus (IAV) infections are a leading cause of mortality worldwide. Excess mortality during IAV epidemics and pandemics is attributable to secondary bacterial infections, particularly pneumonia caused by Streptococcus pneumoniae. Resident alveolar macrophages (rAMs) are early responders to respiratory infections that coordinate initial host defense responses. Maresin conjugates in tissue regeneration (MCTRs) are recently elucidated cysteinyl maresins that are produced by and act on macrophages. Roles for MCTRs in responses to respiratory infections remain to be determined. Here, IAV infection led to transient decreases in rAM numbers. Repopulated lung macrophages displayed transcriptional alterations 21 days post-IAV with prolonged susceptibility to secondary pneumococcal infection. Administration of a mix of MCTR1 to 3 or MCTR3 alone post-IAV decreased lung inflammation and bacterial load 48 and 72 h after secondary pneumococcal infection. MCTR-exposed rAMs had increased migration and phagocytosis of Streptococcus pneumoniae, reduced secretion of CXCL1, and a reversion toward baseline levels of several IAV-induced pneumonia susceptibility genes. Together, MCTRs counter regulated post-IAV changes in rAMs to promote a rapid return of bacteria host defense. IMPORTANCE Secondary bacterial pneumonia is a serious and common complication of IAV infection, leading to excess morbidity and mortality. New host-directed approaches are needed to complement antibiotics to better address this important global infectious disease. Here, we show that harnessing endogenous resolution mechanisms for inflammation by exogenous administration of a family of specialized proresolving mediators (i.e., cys-MCTRs) increased macrophage resilience mechanisms after IAV to protect against secondary infection from Streptococcus pneumoniae.


Assuntos
Coinfecção , Vírus da Influenza A , Influenza Humana , Infecções por Orthomyxoviridae , Infecções Pneumocócicas , Pneumonia Bacteriana , Infecções Respiratórias , Animais , Coinfecção/microbiologia , Humanos , Influenza Humana/complicações , Pulmão/microbiologia , Macrófagos , Masculino , Camundongos , Infecções Pneumocócicas/complicações , Infecções Respiratórias/complicações , Ovinos , Streptococcus pneumoniae
7.
Curr Opin Clin Nutr Metab Care ; 25(2): 67-74, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34954726

RESUMO

PURPOSE OF REVIEW: Persistent unresolved inflammation results in a number of pathologic respiratory diseases including asthma, cystic fibrosis, acute respiratory distress syndrome (ARDS) and coronavirus disease 2019 (COVID-19)-associated ARDS. Inflammation resolution is an active series of biologic processes orchestrated by a family of bioactive specialized pro-resolving mediators (SPMs) derived from essential omega-3 and omega-6 polyunsaturated fatty acids (PUFAs). In this review, we highlight recent findings on dysregulated inflammation resolution in common respiratory diseases and recent literature on SPM generation with PUFA dietary supplementation with relevance to diseases of respiratory inflammation. RECENT FINDINGS: Human studies and preclinical models of diseases of lung inflammation have revealed disequilibrium in the levels of pro-inflammatory versus pro-resolving mediators. Recent studies identified actions for SPMs on regulating prophlogistic host responses and stimulating inflammation resolution pathways in inflammatory respiratory diseases. SUMMARY: Dietary marine oils are enriched in PUFAs and contain parent omega-3 and omega-6 fatty acids and precursors for conversion to SPMs. Nutritional supplementation with fish oils can boost SPM levels and offer a therapeutic approach targeting inflammation resolution pathways for diseases of lung inflammation.


Assuntos
COVID-19 , Ácidos Graxos Ômega-3 , Ácidos Graxos Ômega-6 , Óleos de Peixe , Humanos , Inflamação , SARS-CoV-2
8.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34544857

RESUMO

Tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM) are caused by aberrant mechanistic Target of Rapamycin Complex 1 (mTORC1) activation due to loss of either TSC1 or TSC2 Cytokine profiling of TSC2-deficient LAM patient-derived cells revealed striking up-regulation of Interleukin-6 (IL-6). LAM patient plasma contained increased circulating IL-6 compared with healthy controls, and TSC2-deficient cells showed up-regulation of IL-6 transcription and secretion compared to wild-type cells. IL-6 blockade repressed the proliferation and migration of TSC2-deficient cells and reduced oxygen consumption and extracellular acidification. U-13C glucose tracing revealed that IL-6 knockout reduced 3-phosphoserine and serine production in TSC2-deficient cells, implicating IL-6 in de novo serine metabolism. IL-6 knockout reduced expression of phosphoserine aminotransferase 1 (PSAT1), an essential enzyme in serine biosynthesis. Importantly, recombinant IL-6 treatment rescued PSAT1 expression in the TSC2-deficient, IL-6 knockout clones selectively and had no effect on wild-type cells. Treatment with anti-IL-6 (αIL-6) antibody similarly reduced cell proliferation and migration and reduced renal tumors in Tsc2+/- mice while reducing PSAT1 expression. These data reveal a mechanism through which IL-6 regulates serine biosynthesis, with potential relevance to the therapy of tumors with mTORC1 hyperactivity.


Assuntos
Interleucina-6/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Serina/metabolismo , Transaminases/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo , Animais , Interleucina-6/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transaminases/genética , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/fisiologia
9.
Cell Biochem Biophys ; 79(3): 449-459, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34176102

RESUMO

Acute inflammation in the lung is essential for host defense against pathogens and other injuries but chronic or excessive inflammation can contribute to several common respiratory diseases. In health, the inflammatory response is controlled by several cellular and molecular mechanisms. In addition to anti-inflammatory processes, there are non-phlogistic pro-resolving mechanisms that are engaged to promote the resolution of inflammation and a return to homeostasis. Defects in the production or actions of specialized pro-resolving mediators are associated with diseases characterized by excess or chronic inflammation. In this article, we review cellular and biochemical mechanisms for specialized pro-resolving mediators in health and in sepsis and the acute respiratory distress syndrome as examples of unrestrained inflammatory responses that result in life-threatening pathology. We are honored to contribute to this special edition of the Journal to help celebrate Professor Viswanathan Natarajan's contributions to our understanding of lipid-derived mediators and metabolism in lung cell responses to inflammatory, infectious, or mechanical insults; his foundational discoveries in cell biochemistry and biophysics are continuing to catalyze further advances by the field to uncover the mechanistic underpinnings of important human diseases.


Assuntos
Síndrome do Desconforto Respiratório
10.
JCI Insight ; 6(15)2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34166226

RESUMO

Sepsis is a critical illness characterized by dysregulated inflammatory responses lacking counter-regulation. Specialized proresolving mediators are agonists for antiinflammation and for promoting resolution, and they are protective in preclinical sepsis models. Here, in human sepsis, we mapped resolution circuits for the specialized proresolving mediators resolvin D1 and resolvin D2 in peripheral blood neutrophils and monocytes, their regulation of leukocyte activation and function ex vivo, and their relationships to measures of clinical severity. Neutrophils and monocytes were isolated from healthy subjects and patients with sepsis by inertial microfluidics and resolvin D1 and resolvin D2 receptor expression determined by flow cytometry. The impact of these resolvins on leukocyte activation was determined by isodielectric separation and leukocyte function by stimulated phagolysosome formation. Leukocyte proresolving receptor expression was significantly higher in sepsis. In nanomolar concentrations, resolvin D1 and resolvin D2 partially reversed sepsis-induced changes in leukocyte activation and function. Principal component analyses of leukocyte resolvin receptor expression and responses differentiated sepsis from health and were associated with measures of sepsis severity. These findings indicate that resolvin D1 and resolvin D2 signaling for antiinflammation and resolution are uncoupled from leukocyte activation in early sepsis and suggest that indicators of diminished resolution signaling correlate with clinical disease severity.


Assuntos
Ácidos Docosa-Hexaenoicos/imunologia , Monócitos/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Sepse , Feminino , Humanos , Imunidade Celular/imunologia , Testes Imunológicos/métodos , Técnicas In Vitro/métodos , Mediadores da Inflamação/imunologia , Masculino , Pessoa de Meia-Idade , Análise de Componente Principal , Sepse/sangue , Sepse/imunologia , Transdução de Sinais/imunologia
12.
Oncogene ; 38(49): 7367-7383, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31420607

RESUMO

miR-29b has been identified as a rapamycin-induced microRNA (miRNA) in Tsc2-deficient, mTORC1-hyperactive cells. The biological significance of this induction of miR-29b is unknown. We have found that miR-29b acts as an oncogenic miRNA in Tsc2-deficient cells: inhibition of miR-29b suppressed cell proliferation, anchorage-independent cell growth, cell migration, invasion, and the growth of Tsc2-deficient tumors in vivo. Importantly, the combination of miR-29b inhibition with rapamycin treatment further inhibited these tumor-associated cellular processes. To gain insight into the molecular mechanisms by which miR-29b promotes tumorigenesis, we used RNA sequencing to identify the tumor suppressor retinoid receptor beta (RARß) as a target gene of miR-29b. We found that miR-29b directly targeted the 3'UTR of RARß. Forced expression of RARß reversed the effects of miR-29b overexpression in proliferation, migration, and invasion, indicating that it is a critical target. miR-29b expression correlated with low RARß expression in renal clear cell carcinomas and bladder urothelial carcinomas, tumors associated with TSC gene mutations. We further identified growth family member 4 (ING4) as a novel interacting partner of RARß. Overexpression of ING4 inhibited the migration and invasion of Tsc2-deficient cells while silencing of ING4 reversed the RARß-mediated suppression of cell migration and invasion. Taken together, our findings reveal a novel miR-29b/RARß/ING4 pathway that regulates tumorigenic properties of Tsc2-deficient cells, and that may serve as a potential therapeutic target for TSC, lymphangioleiomyomatosis (LAM), and other mTORC1-hyperactive tumors.


Assuntos
Embrião de Mamíferos/citologia , Fibroblastos/citologia , Regulação da Expressão Gênica , MicroRNAs/genética , Receptores do Ácido Retinoico/metabolismo , Sirolimo/farmacologia , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Movimento Celular , Proliferação de Células , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Receptores do Ácido Retinoico/genética , Proteína 2 do Complexo Esclerose Tuberosa/fisiologia , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Hum Mol Genet ; 28(19): 3270-3281, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31272105

RESUMO

Lower lobe predominant pulmonary cysts occur in up to 90% of patients with Birt-Hogg-Dubé (BHD) syndrome, but the key pathologic cell type and signaling events driving this distinct phenotype remain elusive. Through examination of the LungMAP database, we found that folliculin (FLCN) is highly expressed in neonatal lung mesenchymal cells. Using RNA-Seq, we found that inactivation of Flcn in mouse embryonic fibroblasts leads to changes in multiple Wnt ligands, including a 2.8-fold decrease in Wnt2. This was associated with decreased TCF/LEF activity, a readout of canonical WNT activity, after treatment with a GSK3-α/ß inhibitor. Similarly, FLCN deficiency in HEK293T cells decreased WNT pathway activity by 76% post-GSK3-α/ß inhibition. Inactivation of FLCN in human fetal lung fibroblasts (MRC-5) led to ~ 100-fold decrease in Wnt2 expression and a 33-fold decrease in Wnt7b expression-two ligands known to be necessary for lung development. Furthermore, canonical WNT activity was decreased by 60%. Classic WNT targets such as AXIN2 and BMP4, and WNT enhanceosome members including TCF4, LEF1 and BCL9 were also decreased after GSK3-α/ß inhibition. FLCN-deficient MRC-5 cells failed to upregulate LEF1 in response to GSK3-α/ß inhibition. Finally, we found that a constitutively active ß-catenin could only partially rescue the decreased WNT activity phenotype seen in FLCN-deficient cells, whereas silencing the transcription factor TFE3 completely reversed this phenotype. In summary, our data establish FLCN as a critical regulator of the WNT pathway via TFE3 and suggest that FLCN-dependent defects in WNT pathway developmental cues may contribute to lung cyst pathogenesis in BHD.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Síndrome de Birt-Hogg-Dubé/genética , Perfilação da Expressão Gênica/métodos , Proteínas Proto-Oncogênicas/genética , Análise de Sequência de RNA/métodos , Proteínas Supressoras de Tumor/genética , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Síndrome de Birt-Hogg-Dubé/metabolismo , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Proteína Wnt2/genética , Proteína Wnt2/metabolismo
14.
Sci Rep ; 8(1): 14161, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30242175

RESUMO

Tuberous Sclerosis Complex (TSC), a rare genetic disorder with mechanistic target of rapamycin complex 1 (mTORC1) hyperactivation, is characterized by multi-organ hamartomatous benign tumors including brain, skin, kidney, and lung (Lymphangioleiomyomatosis). mTORC1 hyperactivation drives metabolic reprogramming including glucose and glutamine utilization, protein, nucleic acid and lipid synthesis. To investigate the mechanisms of exogenous nutrients uptake in Tsc2-deficient cells, we measured dextran uptake, a polysaccharide internalized via macropinocytosis. Tsc2-deficient cells showed a striking increase in dextran uptake (3-fold, p < 0.0001) relative to Tsc2-expressing cells, which was decreased (3-fold, p < 0.0001) with mTOR inhibitor, Torin1. Pharmacologic and genetic inhibition of the lipid kinase Vps34 markedly abrogated uptake of Dextran in Tsc2-deficient cells. Macropinocytosis was further increased in Tsc2-deficient cells that lack autophagic mechanisms, suggesting that autophagy inhibition leads to dependence on exogenous nutrient uptake in Tsc2-deficient cells. Treatment with a macropinocytosis inhibitor, ethylisopropylamiloride (EIPA), resulted in selective growth inhibition of Atg5-deficient, Tsc2-deficient cells (50%, p < 0.0001). Genetic inhibition of autophagy (Atg5-/- MEFs) sensitized cells with Tsc2 downregulation to the Vps34 inhibitor, SAR405, resulting in growth inhibition (75%, p < 0.0001). Finally, genetic downregulation of Vps34 inhibited tumor growth and increased tumor latency in an in vivo xenograft model of TSC. Our findings show that macropinocytosis is upregulated with Tsc2-deficiency via a Vps34-dependent mechanism to support their anabolic state. The dependence of Tsc2-deficient cells on exogenous nutrients may provide novel approaches for the treatment of TSC.


Assuntos
Carcinogênese/metabolismo , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Pinocitose/fisiologia , Esclerose Tuberosa/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Proteína 5 Relacionada à Autofagia/metabolismo , Carcinogênese/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Dextranos/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pinocitose/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteína 2 do Complexo Esclerose Tuberosa/deficiência , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
15.
Proc Natl Acad Sci U S A ; 115(27): E6274-E6282, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29891694

RESUMO

Chromophobe renal cell carcinoma (ChRCC) accounts for 5% of all sporadic renal cancers and can also occur in genetic syndromes including Birt-Hogg-Dube (BHD) and tuberous sclerosis complex (TSC). ChRCC has a distinct accumulation of abnormal mitochondria, accompanied by characteristic chromosomal imbalances and relatively few "driver" mutations. Metabolomic profiling of ChRCC and oncocytomas (benign renal tumors that share pathological features with ChRCC) revealed both similarities and differences between these tumor types, with principal component analysis (PCA) showing a distinct separation. ChRCC have a striking decrease in intermediates of the glutathione salvage pathway (also known as the gamma-glutamyl cycle) compared with adjacent normal kidney, as well as significant changes in glycolytic and pentose phosphate pathway intermediates. We also found that gamma glutamyl transferase 1 (GGT1), the key enzyme of the gamma-glutamyl cycle, is expressed at ∼100-fold lower levels in ChRCC compared with normal kidney, while no change in GGT1 expression was found in clear cell RCC (ccRCC). Significant differences in specific metabolite abundance were found in ChRCC vs. ccRCC, including the oxidative stress marker ophthalmate. Down-regulation of GGT1 enhanced the sensitivity to oxidative stress and treatment with buthionine sulfoximine (BSO), which was associated with changes in glutathione-pathway metabolites. These data indicate that impairment of the glutathione salvage pathway, associated with enhanced oxidative stress, may have key therapeutic implications for this rare tumor type for which there are currently no specific targeted therapies.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Renais/enzimologia , Neoplasias Renais/enzimologia , Proteínas de Neoplasias/metabolismo , Oligopeptídeos/metabolismo , gama-Glutamiltransferase/metabolismo , Biomarcadores Tumorais/genética , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Feminino , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Masculino , Proteínas de Neoplasias/genética , Oligopeptídeos/genética , Estresse Oxidativo/genética , Transdução de Sinais/genética , gama-Glutamiltransferase/genética
16.
Hum Mol Genet ; 27(9): 1654-1663, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29509898

RESUMO

Tuberous sclerosis complex (TSC) is an autosomal dominant disease caused by germline inactivating mutations of TSC1 or TSC2. In TSC-associated tumors of the brain, heart, skin, kidney and lung, inactivation of both alleles of TSC1 or TSC2 leads to hyperactivation of the mTORC1 pathway. The TSC/mTORC1 pathway is a key regulator of cellular processes related to growth, proliferation and autophagy. We and others have previously found that mTORC1 regulates microRNA biogenesis, but the mechanisms are not fully understood. Microprocessor, a multi-protein complex including the nuclease Drosha, processes the primary miR transcript. Using a dual-luciferase reporter, we found that inhibition of mTORC1 or downregulation of Raptor decreased Microprocessor activity, while loss of TSC2 led to a striking increase (∼5-fold) in Microprocessor activity. To determine the global impact of TSC2 on microRNAs we quantitatively analyzed 752 microRNAs in Tsc2-expressing and Tsc2-deficient cells. Out of 259 microRNAs expressed in both cell lines, 137 were significantly upregulated and 24 were significantly downregulated in Tsc2-deficient cells, consistent with the increased Microprocessor activity. Microprocessor activity is known to be regulated in part by GSK3ß. We found that total GSK3ß levels were higher in Tsc2-deficient cells, and the increase in Microprocessor activity associated with Tsc2 loss was reversed by three different GSK3ß inhibitors. Furthermore, mTOR inhibition increased the levels of phospho-GSK3ß (S9), which negatively affects Microprocessor activity. Taken together these data reveal that TSC2 regulates microRNA biogenesis and Microprocessor activity via GSK3ß.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , MicroRNAs/genética , Animais , Proliferação de Células/genética , Proliferação de Células/fisiologia , Glicogênio Sintase Quinase 3 beta/genética , Células HeLa , Humanos , Immunoblotting , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , RNA Interferente Pequeno/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
17.
Expert Rev Respir Med ; 12(2): 95-102, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29171770

RESUMO

INTRODUCTION: Lymphangioleiomyomatosis (LAM) is a destructive lung disease affecting primarily women. LAM is caused by inactivating mutations in the tuberous sclerosis complex (TSC) genes, resulting in hyperactivation of mechanistic/mammalian target of rapamycin complex 1 (mTORC1). Over the past five years, there have been remarkable advances in the diagnosis and therapy of LAM, including the identification of vascular endothelial growth factor D (VEGF-D) as a diagnostic biomarker and the US Food and Drug Administration approval of sirolimus as therapy for LAM. In appropriate clinical situations VEGF-D testing can make lung biopsy unnecessary to diagnose LAM. However, there remains an urgent unmet need for additional biomarkers of disease activity and/or response to therapy. Areas covered: This work reviews VEGF-D, an established LAM biomarker, and discusses emerging biomarkers, including circulating LAM cells, imaging, lipid, and metabolite biomarkers, focusing on those with the highest potential impact for LAM patients. Expert commentary: Ongoing research priorities include the development of validated biomarkers to 1) noninvasively diagnose LAM in women whose VEGF-D levels are not diagnostic, 2) accurately predict the likelihood of disease progression and 3) quantitatively measure disease activity and LAM cell burden. These biomarkers would enable personalized, precision clinical care and fast-track clinical trial implementation, with high clinical impact.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Pulmonares/diagnóstico , Linfangioleiomiomatose/diagnóstico , Humanos , Neoplasias Pulmonares/metabolismo , Linfangioleiomiomatose/metabolismo , Reprodutibilidade dos Testes
18.
Oncotarget ; 8(39): 64714-64727, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-29029388

RESUMO

mTORC1 hyperactivation drives the multi-organ hamartomatous disease tuberous sclerosis complex (TSC). Rapamycin inhibits mTORC1, inducing partial tumor responses; however, the tumors regrow following treatment cessation. We discovered that the oncogenic miRNA, miR-21, is increased in Tsc2-deficient cells and, surprisingly, further increased by rapamycin. To determine the impact of miR-21 in TSC, we inhibited miR-21 in vitro. miR-21 inhibition significantly repressed the tumorigenic potential of Tsc2-deficient cells and increased apoptosis sensitivity. Tsc2-deficient cells' clonogenic and anchorage independent growth were reduced by ∼50% (p<0.01) and ∼75% (p<0.0001), respectively, and combined rapamycin treatment decreased soft agar growth by ∼90% (p<0.0001). miR-21 inhibition also increased sensitivity to apoptosis. Through a network biology-driven integration of RNAseq data, we discovered that miR-21 promotes mitochondrial adaptation and homeostasis in Tsc2-deficient cells. miR-21 inhibition reduced mitochondrial polarization and function in Tsc2-deficient cells, with and without co-treatment with rapamycin. Importantly, miR-21 inhibition limited Tsc2-deficient tumor growth in vivo, reducing tumor size by approximately 3-fold (p<0.0001). When combined with rapamcyin, miR-21 inhibition showed even more striking efficacy, both during treatment and after treatment cessation, with a 4-fold increase in median survival following rapamycin cessation (p=0.0008). We conclude that miR-21 promotes mTORC1-driven tumorigenesis via a mechanism that involves the mitochondria, and that miR-21 is a potential therapeutic target for TSC-associated hamartomas and other mTORC1-driven tumors, with the potential for synergistic efficacy when combined with rapalogs.

19.
Cancer Res ; 77(12): 3255-3267, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28512249

RESUMO

p62/sequestosome-1 (SQSTM1) is a multifunctional adaptor protein and autophagic substrate that accumulates in cells with hyperactive mTORC1, such as kidney cells with mutations in the tumor suppressor genes tuberous sclerosis complex (TSC)1 or TSC2. Here we report that p62 is a critical mediator of TSC2-driven tumorigenesis, as Tsc2+/- and Tsc2f/f Ksp-CreERT2+ mice crossed to p62-/- mice were protected from renal tumor development. Metabolic profiling revealed that depletion of p62 in Tsc2-null cells decreased intracellular glutamine, glutamate, and glutathione (GSH). p62 positively regulated the glutamine transporter Slc1a5 and increased glutamine uptake in Tsc2-null cells. We also observed p62-dependent changes in Gcl, Gsr, Nqo1, and Srxn1, which were decreased by p62 attenuation and implicated in GSH production and utilization. p62 attenuation altered mitochondrial morphology, reduced mitochondrial membrane polarization and maximal respiration, and increased mitochondrial reactive oxygen species and mitophagy marker PINK1. These mitochondrial phenotypes were rescued by addition of exogenous GSH and overexpression of Sod2, which suppressed indices of mitochondrial damage and promoted growth of Tsc2-null cells. Finally, p62 depletion sensitized Tsc2-null cells to both oxidative stress and direct inhibition of GSH biosynthesis by buthionine sulfoximine. Our findings show how p62 helps maintain intracellular pools of GSH needed to limit mitochondrial dysfunction in tumor cells with elevated mTORC1, highlighting p62 and redox homeostasis as nodal vulnerabilities for therapeutic targeting in these tumors. Cancer Res; 77(12); 3255-67. ©2017 AACR.


Assuntos
Carcinogênese/metabolismo , Mitocôndrias/patologia , Complexos Multiproteicos/metabolismo , Proteína Sequestossoma-1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/metabolismo , Animais , Carcinogênese/patologia , Modelos Animais de Doenças , Imunofluorescência , Glutationa/biossíntese , Imuno-Histoquímica , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Esclerose Tuberosa/patologia , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
20.
Oncotarget ; 8(24): 38099-38112, 2017 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-28498820

RESUMO

Tuberous sclerosis complex (TSC) is a multisystem disease associated with hyperactive mTORC1. The impact of TSC1/2 deficiency on lysosome-mediated processes is not fully understood. We report here that inhibition of lysosomal function using chloroquine (CQ) upregulates cholesterol homeostasis genes in TSC2-deficient cells. This TSC2-dependent transcriptional signature is associated with increased accumulation and intracellular levels of both total cholesterol and cholesterol esters. Unexpectedly, engaging this CQ-induced cholesterol uptake pathway together with inhibition of de novo cholesterol synthesis allows survival of TSC2-deficient, but not TSC2-expressing cells. The underlying mechanism of TSC2-deficient cell survival is dependent on exogenous cholesterol uptake via LDL-R, and endosomal trafficking mediated by Vps34. Simultaneous inhibition of lysosomal and endosomal trafficking inhibits uptake of esterified cholesterol and cell growth in TSC2-deficient, but not TSC2-expressing cells, highlighting the TSC-dependent lysosome-mediated regulation of cholesterol homeostasis and pointing toward the translational potential of these pathways for the therapy of TSC.


Assuntos
Proteínas de Transporte/metabolismo , Colesterol/metabolismo , Lisossomos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de LDL/metabolismo , Esclerose Tuberosa/metabolismo , Linhagem Celular , Homeostase/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteína C1 de Niemann-Pick
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...