Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 44(1): 271-286, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37823267

RESUMEN

BACKGROUND: Prostacyclin is a fundamental signaling pathway traditionally associated with the cardiovascular system and protection against thrombosis but which also has regulatory functions in fibrosis, proliferation, and immunity. Prevailing dogma states that prostacyclin is principally derived from vascular endothelium, although it is known that other cells can also synthesize it. However, the role of nonendothelial sources in prostacyclin production has not been systematically evaluated resulting in an underappreciation of their importance relative to better characterized endothelial sources. METHODS: To address this, we have used novel endothelial cell-specific and fibroblast-specific COX (cyclo-oxygenase) and prostacyclin synthase knockout mice and cells freshly isolated from mouse and human lung tissue. We have assessed prostacyclin release by immunoassay and thrombosis in vivo using an FeCl3-induced carotid artery injury model. RESULTS: We found that in arteries, endothelial cells are the main source of prostacyclin but that in the lung, and other tissues, prostacyclin production occurs largely independently of endothelial and vascular smooth muscle cells. Instead, in mouse and human lung, prostacyclin production was strongly associated with fibroblasts. By comparison, microvascular endothelial cells from the lung showed weak prostacyclin synthetic capacity compared with those isolated from large arteries. Prostacyclin derived from fibroblasts and other nonendothelial sources was seen to contribute to antithrombotic protection. CONCLUSIONS: These observations define a new paradigm in prostacyclin biology in which fibroblast/nonendothelial-derived prostacyclin works in parallel with endothelium-derived prostanoids to control thrombotic risk and potentially a broad range of other biology. Although generation of prostacyclin by fibroblasts has been shown previously, the scale and systemic activity was unappreciated. As such, this represents a basic change in our understanding and may provide new insight into how diseases of the lung result in cardiovascular risk.


Asunto(s)
Epoprostenol , Trombosis , Ratones , Humanos , Animales , Fibrinolíticos , Células Endoteliales/metabolismo , Prostaglandinas I/metabolismo , Prostaglandinas I/farmacología , Endotelio Vascular/metabolismo , Ratones Noqueados , Fibroblastos/metabolismo , Trombosis/genética , Trombosis/prevención & control , Trombosis/metabolismo
2.
Cell Mol Gastroenterol Hepatol ; 13(4): 1095-1120, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35017061

RESUMEN

BACKGROUND AND AIMS: Phagocytosis (efferocytosis) of apoptotic neutrophils by macrophages anchors the resolution of intestinal inflammation. Efferocytosis prevents secondary necrosis and inhibits further inflammation, and also reprograms macrophages to facilitate tissue repair and promote resolution function. Macrophage efferocytosis and efferocytosis-dependent reprogramming are implicated in the pathogenesis of inflammatory bowel disease. We previously reported that absence of macrophage cyclooxygenase 2 (COX2) exacerbates inflammatory bowel disease-like intestinal inflammation. To elucidate the underlying pathogenic mechanism, we investigated here whether COX2 mediates macrophage efferocytosis and efferocytosis-dependent reprogramming, including intestinal epithelial repair capacity. METHODS: Using apoptotic neutrophils and synthetic apoptotic targets, we determined the effects of macrophage specific Cox2 knockout and pharmacological COX2 inhibition on the efferocytosis capacity of mouse primary macrophages. COX2-mediated efferocytosis-dependent eicosanoid lipidomics was determined by liquid chromatography tandem mass spectrometry. Small intestinal epithelial organoids were employed to assay the effects of COX2 on efferocytosis-dependent intestinal epithelial repair. RESULTS: Loss of COX2 impaired efferocytosis in mouse primary macrophages, in part, by affecting the binding capacity of macrophages for apoptotic cells. This effect was comparable to that of high-dose lipopolysaccharide and was accompanied by both dysregulation of macrophage polarization and the inhibited expression of genes involved in apoptotic cell binding. COX2 modulated the production of efferocytosis-dependent lipid inflammatory mediators that include the eicosanoids prostaglandin I2, prostaglandin E2, lipoxin A4, and 15d-PGJ2; and further affected secondary efferocytosis. Finally, macrophage efferocytosis induced, in a macrophage COX2-dependent manner, a tissue restitution and repair phenotype in intestinal epithelial organoids. CONCLUSIONS: Macrophage COX2 potentiates efferocytosis capacity and efferocytosis-dependent reprogramming, facilitating macrophage intestinal epithelial repair capacity.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Enfermedades Inflamatorias del Intestino , Fagocitosis , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/farmacología , Inflamación/patología , Enfermedades Inflamatorias del Intestino/patología , Macrófagos/metabolismo , Ratones , Fagocitosis/genética
3.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836566

RESUMEN

Unlike other epithelial cancer types, circulating tumor cells (CTCs) are less frequently detected in the peripheral blood of non-small cell lung cancer (NSCLC) patients using epithelial marker-based detection approaches despite the aggressive nature of NSCLC. Here, we demonstrate hexokinase-2 (HK2) as a metabolic function-associated marker for the detection of CTCs. In 59 NSCLC patients bearing cytokeratin-positive (CKpos) primary tumors, HK2 enables resolving cytokeratin-negative (HK2high/CKneg) CTCs as a prevalent population in about half of the peripheral blood samples with positive CTC counts. However, HK2high/CKneg tumor cells are a minority population in pleural effusions and cerebrospinal fluids. Single-cell analysis shows that HK2high/CKneg CTCs exhibit smaller sizes but consistent copy number variation profiles compared with CKpos counterparts. Single-cell transcriptome profiling reveals that CK expression levels of CTCs are independent of their epithelial-to-mesenchymal transition (EMT) status, challenging the long-standing association between CK expression and EMT. HK2high/CKneg CTCs display metastasis and EGFR inhibitor resistance-related molecular signatures and are selectively enriched in patients with EGFRL858R driver oncogene mutation as opposed to EGFR19Del , which is more frequently found in patients with prevalent CKpos CTCs in the blood. Consistently, treatment-naïve patients with a larger number or proportion of HK2high/CKneg CTCs in the blood exhibit poor therapy response and shorter progression-free survival. Collectively, our approach resolves a more complete spectrum of CTCs in NSCLC that can potentially be exploited to identify patient prognosis before therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Hexoquinasa/sangre , Neoplasias Pulmonares/patología , Células Neoplásicas Circulantes/patología , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Transición Epitelial-Mesenquimal , Receptores ErbB/genética , Genotipo , Humanos , Queratinas/sangre , Biopsia Líquida , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/enzimología , Pronóstico
4.
Sci Adv ; 7(12)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33741600

RESUMEN

Endothelial cyclooxygenase-1-derived prostanoids, including prostacyclin, have clear cardioprotective roles associated with their anti-thrombotic potential but have also been suggested to have paradoxical pathological activities within arteries. To date it has not been possible to test the importance of this because no models have been available that separate vascular cyclooxygenase-1 products from those generated elsewhere. Here, we have used unique endothelial-specific cyclooxygenase-1 knockout mice to show that endothelial cyclooxygenase-1 produces both protective and pathological products. Functionally, however, the overall effect of these was to drive pathological responses in the context of both vasoconstriction in vitro and the development of atherosclerosis and vascular inflammation in vivo. These data provide the first demonstration of a pathological role for the vascular cyclooxygenase-1 pathway, highlighting its potential as a therapeutic target. They also emphasize that, across biology, the role of prostanoids is not always predictable due to unique balances of context, products, and receptors.


Asunto(s)
Aterosclerosis , Ciclooxigenasa 1/metabolismo , Epoprostenol , Proteínas de la Membrana/metabolismo , Animales , Aterosclerosis/etiología , Ciclooxigenasa 1/genética , Epoprostenol/metabolismo , Ratones , Prostaglandinas , Vasoconstricción
6.
FASEB J ; 34(8): 10027-10040, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32592197

RESUMEN

Aspirin prevents thrombosis by inhibiting platelet cyclooxygenase (COX)-1 activity and the production of thromboxane (Tx)A2 , a pro-thrombotic eicosanoid. However, the non-platelet actions of aspirin limit its antithrombotic effects. Here, we used platelet-COX-1-ko mice to define the platelet and non-platelet eicosanoids affected by aspirin. Mass-spectrometry analysis demonstrated blood from platelet-COX-1-ko and global-COX-1-ko mice produced similar eicosanoid profiles in vitro: for example, formation of TxA2 , prostaglandin (PG) F2α , 11-hydroxyeicosatraenoic acid (HETE), and 15-HETE was absent in both platelet- and global-COX-1-ko mice. Conversely, in vivo, platelet-COX-1-ko mice had a distinctly different profile from global-COX-1-ko or aspirin-treated control mice, notably significantly higher levels of PGI2 metabolite. Ingenuity Pathway Analysis (IPA) predicted that platelet-COX-1-ko mice would be protected from thrombosis, forming less pro-thrombotic TxA2 and PGE2 . Conversely, aspirin or lack of systemic COX-1 activity decreased the synthesis of anti-aggregatory PGI2 and PGD2 at non-platelet sites leading to predicted thrombosis increase. In vitro and in vivo thrombosis studies proved these predictions. Overall, we have established the eicosanoid profiles linked to inhibition of COX-1 in platelets and in the remainder of the cardiovascular system and linked them to anti- and pro-thrombotic effects of aspirin. These results explain why increasing aspirin dosage or aspirin addition to other drugs may lessen antithrombotic protection.


Asunto(s)
Aspirina/farmacología , Plaquetas/metabolismo , Ciclooxigenasa 1/fisiología , Inhibidores de la Ciclooxigenasa/farmacología , Eicosanoides/metabolismo , Proteínas de la Membrana/fisiología , Trombosis/metabolismo , Animales , Ácido Araquidónico/administración & dosificación , Plaquetas/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombosis/tratamiento farmacológico , Trombosis/patología
7.
Nature ; 580(7804): 524-529, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32322056

RESUMEN

The initiation of an intestinal tumour is a probabilistic process that depends on the competition between mutant and normal epithelial stem cells in crypts1. Intestinal stem cells are closely associated with a diverse but poorly characterized network of mesenchymal cell types2,3. However, whether the physiological mesenchymal microenvironment of mutant stem cells affects tumour initiation remains unknown. Here we provide in vivo evidence that the mesenchymal niche controls tumour initiation in trans. By characterizing the heterogeneity of the intestinal mesenchyme using single-cell RNA-sequencing analysis, we identified a population of rare pericryptal Ptgs2-expressing fibroblasts that constitutively process arachidonic acid into highly labile prostaglandin E2 (PGE2). Specific ablation of Ptgs2 in fibroblasts was sufficient to prevent tumour initiation in two different models of sporadic, autochthonous tumorigenesis. Mechanistically, single-cell RNA-sequencing analyses of a mesenchymal niche model showed that fibroblast-derived PGE2 drives the expansion οf a population of Sca-1+ reserve-like stem cells. These express a strong regenerative/tumorigenic program, driven by the Hippo pathway effector Yap. In vivo, Yap is indispensable for Sca-1+ cell expansion and early tumour initiation and displays a nuclear localization in both mouse and human adenomas. Using organoid experiments, we identified a molecular mechanism whereby PGE2 promotes Yap dephosphorylation, nuclear translocation and transcriptional activity by signalling through the receptor Ptger4. Epithelial-specific ablation of Ptger4 misdirected the regenerative reprogramming of stem cells and prevented Sca-1+ cell expansion and sporadic tumour initiation in mutant mice, thereby demonstrating the robust paracrine control of tumour-initiating stem cells by PGE2-Ptger4. Analyses of patient-derived organoids established that PGE2-PTGER4 also regulates stem-cell function in humans. Our study demonstrates that initiation of colorectal cancer is orchestrated by the mesenchymal niche and reveals a mechanism by which rare pericryptal Ptgs2-expressing fibroblasts exert paracrine control over tumour-initiating stem cells via the druggable PGE2-Ptger4-Yap signalling axis.


Asunto(s)
Carcinogénesis , Neoplasias Colorrectales/patología , Intestinos/patología , Mesodermo/patología , Células Madre Neoplásicas/patología , Comunicación Paracrina , Nicho de Células Madre , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos Ly/metabolismo , Ácido Araquidónico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Proteínas de la Membrana/metabolismo , Mesodermo/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Organoides/metabolismo , Organoides/patología , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Análisis de la Célula Individual , Proteínas Señalizadoras YAP
9.
Methods Mol Biol ; 2126: 177-190, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32112389

RESUMEN

Positron emission tomography (PET) reporter genes (PRGs), when coupled with positron-emitting PET reporter probes (PRPs), are useful for tracking specific cell populations in cell-based therapies, in transgenic animal models, and in xenograft tumor progression experiments. The activities of incorporated PRGs in targeted cells can be monitored noninvasively by PET imaging in preclinical in vivo studies and clinical applications following systemic administration of the appropriate PRG. Here we describe a method that minimizes both design and variability of vector delivery vehicles for alternative PRGs and biological variability of the in vivo target when comparing the efficacy, sensitivity, and specificity of alternative PRG/PRP combinations for in vivo PRG imaging. The principles described for comparing alternative PRG/PRP reporter gene systems can be applied to comparisons of alternative fluorescence, bioluminescence, single-photon emission computerized tomography (SPECT), and magnetic resonance imaging (MRI) reporter genes.


Asunto(s)
Adenoviridae/genética , Genes Reporteros , Imagen Molecular/métodos , Tomografía de Emisión de Positrones/métodos , Timidina Quinasa/análisis , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID
10.
Sci Rep ; 10(1): 377, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941995

RESUMEN

Chronic inflammation facilitates tumor progression. We discovered that a subset of non-small cell lung cancer cells underwent a gradually progressing epithelial-to-mesenchymal (EMT) phenotype following a 21-day exposure to IL-1ß, an abundant proinflammatory cytokine in the at-risk for lung cancer pulmonary and the lung tumor microenvironments. Pathway analysis of the gene expression profile and in vitro functional studies revealed that the EMT and EMT-associated phenotypes, including enhanced cell invasion, PD-L1 upregulation, and chemoresistance, were sustained in the absence of continuous IL-1ß exposure. We referred to this phenomenon as EMT memory. Utilizing a doxycycline-controlled SLUG expression system, we found that high expression of the transcription factor SLUG was indispensable for the establishment of EMT memory. High SLUG expression in tumors of lung cancer patients was associated with poor survival. Chemical or genetic inhibition of SLUG upregulation prevented EMT following the acute IL-1ß exposure but did not reverse EMT memory. Chromatin immunoprecipitation and methylation-specific PCR further revealed a SLUG-mediated temporal regulation of epigenetic modifications, including accumulation of H3K27, H3K9, and DNA methylation, in the CDH1 (E-cadherin) promoter following the chronic IL-1ß exposure. Chemical inhibition of DNA methylation not only restored E-cadherin expression in EMT memory, but also primed cells for chemotherapy-induced apoptosis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Epigénesis Genética , Transición Epitelial-Mesenquimal , Memoria Inmunológica/inmunología , Inflamación/inmunología , Interleucina-1beta/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Memoria Inmunológica/genética , Inflamación/genética , Interleucina-1beta/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Fenotipo , Células Tumorales Cultivadas
11.
Stem Cells Transl Med ; 9(4): 499-517, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31943851

RESUMEN

Mesenchymal stem cells (MSCs) have been used in clinical studies to treat neurological diseases and damage. However, implanted MSCs do not achieve their regenerative effects by differentiating into and replacing neural cells. Instead, MSC secretome components mediate the regenerative effects of MSCs. MSC-derived extracellular vesicles (EVs)/exosomes carry cargo responsible for rescuing brain damage. We previously showed that EP4 antagonist-induced MSC EVs/exosomes have enhanced regenerative potential to rescue hippocampal damage, compared with EVs/exosomes from untreated MSCs. Here we show that EP4 antagonist-induced MSC EVs/exosomes promote neurosphere formation in vitro and increase neurogenesis and neuritogenesis in damaged hippocampi; basal MSC EVs/exosomes do not contribute to these regenerative effects. 2',3'-Cyclic nucleotide 3'-phosphodiesterase (CNP) levels in EP4 antagonist-induced MSC EVs/exosomes are 20-fold higher than CNP levels in basal MSC EVs/exosomes. Decreasing elevated exosomal CNP levels in EP4 antagonist-induced MSC EVs/exosomes reduced the efficacy of these EVs/exosomes in promoting ß3-tubulin polymerization and in converting toxic 2',3'-cAMP into neuroprotective adenosine. CNP-depleted EP4 antagonist-induced MSC EVs/exosomes lost the ability to promote neurogenesis and neuritogenesis in damaged hippocampi. Systemic administration of EV/exosomes from EP4 -antagonist derived MSC EVs/exosomes repaired cognition, learning, and memory deficiencies in mice caused by hippocampal damage. In contrast, CNP-depleted EP4 antagonist-induced MSC EVs/exosomes failed to repair this damage. Exosomal CNP contributes to the ability of EP4 antagonist-elicited MSC EVs/exosomes to promote neurogenesis and neuritogenesis in damaged hippocampi and recovery of cognition, memory, and learning. This experimental approach should be generally applicable to identifying the role of EV/exosomal components in eliciting a variety of biological responses.


Asunto(s)
2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Lesiones Encefálicas/terapia , Región CA1 Hipocampal/metabolismo , Cognición , Exosomas/enzimología , Aprendizaje , Células Madre Mesenquimatosas/enzimología , Neuritas/metabolismo , Neurogénesis , Animales , Lesiones Encefálicas/patología , Cognición/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas de Dominio Doblecortina , Exosomas/efectos de los fármacos , Humanos , Isoindoles/farmacología , Aprendizaje/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuritas/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuropéptidos/metabolismo , Polimerizacion , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Sulfonamidas/farmacología , Tubulina (Proteína)/metabolismo
12.
Circ Res ; 125(9): 847-854, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31510878

RESUMEN

RATIONALE: Endothelial cells (ECs) and platelets, which respectively produce antithrombotic prostacyclin and prothrombotic thromboxane A2, both express COX1 (cyclooxygenase1). Consequently, there has been no way to delineate any antithrombotic role for COX1-derived prostacyclin from the prothrombotic effects of platelet COX1. By contrast, an antithrombotic role for COX2, which is absent in platelets, is straightforward to demonstrate. This has resulted in an incomplete understanding of the relative importance of COX1 versus COX2 in prostacyclin production and antithrombotic protection in vivo. OBJECTIVE: We sought to identify the role, if any, of COX1-derived prostacyclin in antithrombotic protection in vivo and compare this to the established protective role of COX2. METHODS AND RESULTS: We developed vascular-specific COX1 knockout mice and studied them alongside endothelial-specific COX2 knockout mice. COX1 immunoreactivity and prostacyclin production were primarily associated with the endothelial layer of aortae; freshly isolated aortic ECs released >10-fold more prostacyclin than smooth muscle cells. Moreover, aortic prostacyclin production, the ability of aortic rings to inhibit platelet aggregation and plasma prostacyclin levels were reduced when COX1 was knocked out in ECs but not in smooth muscle cells. When thrombosis was measured in vivo after FeCl3 carotid artery injury, endothelial COX1 deletion accelerated thrombosis to a similar extent as prostacyclin receptor blockade. However, this effect was lost when COX1 was deleted from both ECs and platelets. Deletion of COX2 from ECs also resulted in a prothrombotic phenotype that was independent of local vascular prostacyclin production. CONCLUSIONS: These data demonstrate for the first time that, in healthy animals, endothelial COX1 provides an essential antithrombotic tone, which is masked when COX1 activity is lost in both ECs and platelets. These results help us define a new 2-component paradigm wherein thrombotic tone is regulated by both COX1 and COX2 through complementary but mechanistically distinct pathways.


Asunto(s)
Ciclooxigenasa 1/deficiencia , Endotelio Vascular/metabolismo , Epoprostenol/metabolismo , Fibrinolíticos/metabolismo , Eliminación de Gen , Proteínas de la Membrana/deficiencia , Agregación Plaquetaria/fisiología , Animales , Aorta/metabolismo , Ciclooxigenasa 1/genética , Epoprostenol/genética , Femenino , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos
13.
Cancer Res ; 79(23): 5907-5914, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31434645

RESUMEN

Since Warburg's observation that most cancers exhibit elevated glycolysis, decades of research have attempted to reduce tumor glucose utilization as a therapeutic approach. Hexokinase (HK) activity is the first glycolytic enzymatic step; despite many attempts to inhibit HK activity, none has reached clinical application. Identification of HK isoforms, and recognition that most tissues express only HK1 while most tumors express HK1 and HK2, stimulated reducing HK2 activity as a therapeutic option. However, studies using HK2 shRNA and isogenic HK1+HK2- and HK1+HK2+ tumor cell pairs demonstrated that tumors expressing only HK1, while exhibiting reduced glucose consumption, progressed in vivo as well as tumors expressing both HK1 and HK2. However, HK1-HK2+ tumor subpopulations exist among many cancers. shRNA HK2 suppression in HK1-HK2+ liver cancer cells reduced xenograft tumor progression, in contrast to HK1+HK2+ cells. HK2 inhibition, and partial inhibition of both oxidative phosphorylation and fatty acid oxidation using HK2 shRNA and small-molecule drugs, prevented human liver HK1-HK2+ cancer xenograft progression. Using human multiple myeloma xenografts and mouse allogeneic models to identify potential clinical translational agents, triple therapies that include antisense HK2 oligonucleotides, metformin, and perhexiline prevent progression. These results suggest an agnostic approach for HK1-HK2+ cancers, regardless of tissue origin.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Glucólisis/efectos de los fármacos , Hexoquinasa/antagonistas & inhibidores , Hexoquinasa/genética , Neoplasias/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Glucólisis/genética , Hexoquinasa/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Metformina/farmacología , Metformina/uso terapéutico , Ratones , Neoplasias/genética , Neoplasias/patología , Compuestos Onio/farmacología , Compuestos Onio/uso terapéutico , Fosforilación Oxidativa/efectos de los fármacos , Perhexilina/farmacología , Perhexilina/uso terapéutico , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Rep ; 27(12): 3413-3421.e3, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31216464

RESUMEN

Hair plays important roles, ranging from the conservation of body heat to the preservation of psychological well-being. Hair loss or alopecia affects millions worldwide, but methods that can be used to regrow hair are lacking. We report that quiescent (telogen) hair follicles can be stimulated to initiate anagen and hair growth by small molecules that activate autophagy, including the metabolites α-ketoglutarate (α-KG) and α-ketobutyrate (α-KB), and the prescription drugs rapamycin and metformin, which impinge on mTOR and AMPK signaling. Stimulation of hair growth by these agents is blocked by specific autophagy inhibitors, suggesting a mechanistic link between autophagy and hair regeneration. Consistently, increased autophagy is detected upon anagen entry during the natural hair follicle cycle, and oral α-KB prevents hair loss in aged mice. Our finding that anagen can be pharmacologically activated in telogen skin when natural anagen-inducing signal(s) are absent has implications for the treatment of hair loss patients.


Asunto(s)
Alopecia/tratamiento farmacológico , Autofagia/efectos de los fármacos , Folículo Piloso/efectos de los fármacos , Cabello/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Envejecimiento/fisiología , Compuestos Alílicos/farmacología , Alopecia/genética , Alopecia/metabolismo , Animales , Autofagia/genética , Butiratos/farmacología , División Celular/efectos de los fármacos , División Celular/genética , Femenino , Cabello/crecimiento & desarrollo , Folículo Piloso/metabolismo , Ácidos Cetoglutáricos/farmacología , Masculino , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Oligomicinas/farmacología , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética
15.
J Clin Invest ; 129(9): 3670-3685, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31184596

RESUMEN

Cyclooxygenase 2 (Cox2) total knockout and myeloid knockout (MKO) mice develop Crohn's-like intestinal inflammation when fed cholate-containing high fat diet (CCHF). We demonstrated that CCHF impaired intestinal barrier function and increased translocation of endotoxin, initiating TLR/MyD88-dependent inflammation in Cox2 KO but not WT mice. Cox2 MKO increased pro-inflammatory mediators in LPS-activated macrophages, and in the intestinal tissue and plasma upon CCHF challenge. Cox2 MKO also reduced inflammation resolving lipoxin A4 (LXA4) in intestinal tissue, while administration of an LXA4 analog rescued disease in Cox2 MKO mice fed CCHF. The apolipoprotein A-I (APOA1) mimetic 4F mitigated disease in both the Cox2 MKO/CCHF and piroxicam-accelerated Il10-/- models of inflammatory bowel disease (IBD) and reduced elevated levels of pro-inflammatory mediators in tissue and plasma. APOA1 mimetic Tg6F therapy was also effective in reducing intestinal inflammation in the Cox2 MKO/CCHF model. We further demonstrated that APOA1 mimetic peptides: i) inhibited LPS and oxidized 1-palmitoyl-2-arachidonoyl-sn-phosphatidylcholine (oxPAPC) dependent pro-inflammatory responses in human macrophages and intestinal epithelium; and ii) directly cleared pro-inflammatory lipids from mouse intestinal tissue and plasma. Our results support a causal role for pro-inflammatory and inflammation resolving lipids in IBD pathology and a translational potential for APOA1 mimetic peptides for the treatment of IBD.


Asunto(s)
Apolipoproteína A-I/farmacología , Ciclooxigenasa 2/genética , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Intestinos/patología , Animales , Modelos Animales de Enfermedad , Endotoxinas/metabolismo , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/metabolismo , Péptidos/química , Permeabilidad , Piroxicam/farmacología , Receptores de Formil Péptido/metabolismo , Transducción de Señal
16.
Cancer Res ; 79(10): 2748-2760, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30885978

RESUMEN

Although the majority of adult tissues express only hexokinase 1 (HK1) for glycolysis, most cancers express hexokinase 2 (HK2) and many coexpress HK1 and HK2. In contrast to HK1+HK2+ cancers, HK1-HK2+ cancer subsets are sensitive to cytostasis induced by HK2shRNA knockdown and are also sensitive to synthetic lethality in response to the combination of HK2shRNA knockdown, an oxidative phosphorylation (OXPHOS) inhibitor diphenyleneiodonium (DPI), and a fatty acid oxidation (FAO) inhibitor perhexiline (PER). The majority of human multiple myeloma cell lines are HK1-HK2+. Here we describe an antisense oligonucleotide (ASO) directed against human HK2 (HK2-ASO1), which suppressed HK2 expression in human multiple myeloma cell cultures and human multiple myeloma mouse xenograft models. The HK2-ASO1/DPI/PER triple-combination achieved synthetic lethality in multiple myeloma cells in culture and prevented HK1-HK2+ multiple myeloma tumor xenograft progression. DPI was replaceable by the FDA-approved OXPHOS inhibitor metformin (MET), both for synthetic lethality in culture and for inhibition of tumor xenograft progression. In addition, we used an ASO targeting murine HK2 (mHK2-ASO1) to validate the safety of mHK2-ASO1/MET/PER combination therapy in mice bearing murine multiple myeloma tumors. HK2-ASO1 is the first agent that shows selective HK2 inhibition and therapeutic efficacy in cell culture and in animal models, supporting clinical development of this synthetically lethal combination as a therapy for HK1-HK2+ multiple myeloma. SIGNIFICANCE: A first-in-class HK2 antisense oligonucleotide suppresses HK2 expression in cell culture and in in vivo, presenting an effective, tolerated combination therapy for preventing progression of HK1-HK2+ multiple myeloma tumors. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/10/2748/F1.large.jpg.


Asunto(s)
Hexoquinasa/genética , Mieloma Múltiple/patología , Oligonucleótidos Antisentido/farmacología , Mutaciones Letales Sintéticas , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Stem Cells Transl Med ; 8(7): 707-723, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30891948

RESUMEN

Adult brains have limited regenerative capacity. Consequently, both brain damage and neurodegenerative diseases often cause functional impairment for patients. Mesenchymal stem cells (MSCs), one type of adult stem cells, can be isolated from various adult tissues. MSCs have been used in clinical trials to treat human diseases and the therapeutic potentials of the MSC-derived secretome and extracellular vesicles (EVs) have been under investigation. We found that blocking the prostaglandin E2 /prostaglandin E2 receptor 4 (PGE2 /EP4 ) signaling pathway in MSCs with EP4 antagonists increased EV release and promoted the sorting of specific proteins, including anti-inflammatory cytokines and factors that modify astrocyte function, blood-brain barrier integrity, and microglial migration into the damaged hippocampus, into the EVs. Systemic administration of EP4 antagonist-elicited MSC EVs repaired deficiencies of cognition, learning and memory, inhibited reactive astrogliosis, attenuated extensive inflammation, reduced microglial infiltration into the damaged hippocampus, and increased blood-brain barrier integrity when administered to mice following hippocampal damage. Stem Cells Translational Medicine 2019.


Asunto(s)
Barrera Hematoencefálica , Cognición , Vesículas Extracelulares , Hipocampo , Isoindoles/farmacología , Aprendizaje , Células Madre Mesenquimatosas/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Recuperación de la Función , Sulfonamidas/farmacología , Adulto , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Hipocampo/lesiones , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Masculino , Ratones , Ratones Transgénicos , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo
18.
Cell Rep ; 25(10): 2919-2934.e8, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30517876

RESUMEN

It is well understood that fatty acids can be synthesized, imported, and modified to meet requisite demands in cells. However, following the movement of fatty acids through the multiplicity of these metabolic steps has remained difficult. To better address this problem, we developed Fatty Acid Source Analysis (FASA), a model that defines the contribution of synthesis, import, and elongation pathways to fatty acid homeostasis in saturated, monounsaturated, and polyunsaturated fatty acid pools. Application of FASA demonstrated that elongation can be a major contributor to cellular fatty acid content and showed that distinct pro-inflammatory stimuli (e.g., Toll-like receptors 2, 3, or 4) specifically reprogram homeostasis of fatty acids by differential utilization of synthetic and elongation pathways in macrophages. In sum, this modeling approach significantly advances our ability to interrogate cellular fatty acid metabolism and provides insight into how cells dynamically reshape their lipidomes in response to metabolic or inflammatory signals.


Asunto(s)
Ácidos Grasos/metabolismo , Marcaje Isotópico/métodos , Modelos Biológicos , Animales , Carbono/metabolismo , Línea Celular , Ácidos Grasos Insaturados/metabolismo , Homeostasis , Humanos , Inflamación/patología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL
19.
Cancer Metab ; 6: 7, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29988332

RESUMEN

BACKGROUND: Precision medicine therapies require identification of unique molecular cancer characteristics. Hexokinase (HK) activity has been proposed as a therapeutic target; however, different hexokinase isoforms have not been well characterized as alternative targets. While HK2 is highly expressed in the majority of cancers, cancer subtypes with differential HK1 and HK2 expression have not been characterized for their sensitivities to HK2 silencing. METHODS: HK1 and HK2 expression in the Cancer Cell Line Encyclopedia dataset was analyzed. A doxycycline-inducible shRNA silencing system was used to examine the effect of HK2 knockdown in cultured cells and in xenograft models of HK1-HK2+ and HK1+HK2+ cancers. Glucose consumption and lactate production rates were measured to monitor HK activity in cell culture, and 18F-FDG PET/CT was used to monitor HK activity in xenograft tumors. A high-throughput screen was performed to search for synthetically lethal compounds in combination with HK2 inhibition in HK1-HK2+ liver cancer cells, and a combination therapy for liver cancers with this phenotype was developed. A metabolomic analysis was performed to examine changes in cellular energy levels and key metabolites in HK1-HK2+ cells treated with this combination therapy. The CRISPR Cas9 method was used to establish isogenic HK1+HK2+ and HK1-HK2+ cell lines to evaluate HK1-HK2+ cancer cell sensitivity to the combination therapy. RESULTS: Most tumors express both HK1 and HK2, and subsets of cancers from a wide variety of tissues of origin express only HK2. Unlike HK1+HK2+ cancers, HK1-HK2+ cancers are sensitive to HK2 silencing-induced cytostasis. Synthetic lethality was achieved in HK1-HK2+ liver cancer cells, by the combination of DPI, a mitochondrial complex I inhibitor, and HK2 inhibition, in HK1-HK2+ liver cancer cells. Perhexiline, a fatty acid oxidation inhibitor, further sensitizes HK1-HK2+ liver cancer cells to the complex I/HK2-targeted therapeutic combination. Although HK1+HK2+ lung cancer H460 cells are resistant to this therapeutic combination, isogenic HK1KOHK2+ cells are sensitive to this therapy. CONCLUSIONS: The HK1-HK2+ cancer subsets exist among a wide variety of cancer types. Selective inhibition of the HK1-HK2+ cancer cell-specific energy production pathways (HK2-driven glycolysis, oxidative phosphorylation and fatty acid oxidation), due to the unique presence of only the HK2 isoform, appears promising to treat HK1-HK2+ cancers. This therapeutic strategy will likely be tolerated by most normal tissues, where only HK1 is expressed.

20.
J Nucl Med ; 2018 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880505

RESUMEN

Although absent in most adult tissues, hexokinase 2 (HK2) is expressed in a majority of tumors and contributes to increased glucose consumption and to in vivo tumor 18F-FDG PET signaling. Methods: Both HK2 knockdown and knockout approaches were used to investigate the role of HK2 in cancer cell proliferation, in vivo xenograft tumor progression and 18F-FDG tumor accumulation. BioProfiler analysis monitored cell culture glucose consumption and lactate production; 18F-FDG PET/CT monitored in vivo tumor glucose accumulation. Cancer Cell Line Encyclopedia data were analyzed for HK1 and HK2 expression. Results: Neither cell proliferation in culture nor xenograft tumor progression are inhibited by HK2 knockdown or knockout in cancer cells that express HK1 and HK2. However, cancer subsets from a variety of tissues of origin express only HK2, but not HK1. In contrast to HK1+HK2+ cancers, HK2 knockdown in HK1-HK2+ cancer cells results in inhibition of cell proliferation, colony formation and xenograft tumor progression. Moreover, HK1KOHK2+ cancer cells are susceptible to HK2 inhibition, in contrast to their isogenic HK1+HK2+ parental cells. Conclusion: HK1 and HK2 expression are redundant in tumors; either can provide sufficient aerobic glycolysis for tumor growth; despite a reduction in 18F-FDG PET signal. Therapeutic HK2 inhibition is likely to be restricted to HK1-HK2+ tumor subsets, but stratification of tumors that express HK2, but not HK1, should identify tumors treatable with emerging HK2 specific inhibitors.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA