Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(7): e2314085121, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38330013

RESUMEN

Cancer therapy, including immunotherapy, is inherently limited by chronic inflammation-induced tumorigenesis and toxicity within the tumor microenvironment. Thus, stimulating the resolution of inflammation may enhance immunotherapy and improve the toxicity of immune checkpoint inhibition (ICI). As epoxy-fatty acids (EpFAs) are degraded by the enzyme soluble epoxide hydrolase (sEH), the inhibition of sEH increases endogenous EpFA levels to promote the resolution of cancer-associated inflammation. Here, we demonstrate that systemic treatment with ICI induces sEH expression in multiple murine cancer models. Dietary omega-3 polyunsaturated fatty acid supplementation and pharmacologic sEH inhibition, both alone and in combination, significantly enhance anti-tumor activity of ICI in these models. Notably, pharmacological abrogation of the sEH pathway alone or in combination with ICI counter-regulates an ICI-induced pro-inflammatory and pro-tumorigenic cytokine storm. Thus, modulating endogenous EpFA levels through dietary supplementation or sEH inhibition may represent a unique strategy to enhance the anti-tumor activity of paradigm cancer therapies.


Asunto(s)
Epóxido Hidrolasas , Neoplasias , Ratones , Humanos , Animales , Epóxido Hidrolasas/metabolismo , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Neoplasias/terapia , Inmunoterapia , Microambiente Tumoral
3.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34607951

RESUMEN

Cancer therapy reduces tumor burden via tumor cell death ("debris"), which can accelerate tumor progression via the failure of inflammation resolution. Thus, there is an urgent need to develop treatment modalities that stimulate the clearance or resolution of inflammation-associated debris. Here, we demonstrate that chemotherapy-generated debris stimulates metastasis by up-regulating soluble epoxide hydrolase (sEH) and the prostaglandin E2 receptor 4 (EP4). Therapy-induced tumor cell debris triggers a storm of proinflammatory and proangiogenic eicosanoid-driven cytokines. Thus, targeting a single eicosanoid or cytokine is unlikely to prevent chemotherapy-induced metastasis. Pharmacological abrogation of both sEH and EP4 eicosanoid pathways prevents hepato-pancreatic tumor growth and liver metastasis by promoting macrophage phagocytosis of debris and counterregulating a protumorigenic eicosanoid and cytokine storm. Therefore, stimulating the clearance of tumor cell debris via combined sEH and EP4 inhibition is an approach to prevent debris-stimulated metastasis and tumor growth.


Asunto(s)
Eicosanoides/metabolismo , Epóxido Hidrolasas/biosíntesis , Macrófagos/inmunología , Metástasis de la Neoplasia/patología , Subtipo EP4 de Receptores de Prostaglandina E/biosíntesis , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/prevención & control , Citocinas/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/prevención & control , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Fagocitosis/inmunología , Células RAW 264.7
4.
Cancer Metastasis Rev ; 41(3): 737-747, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35624227

RESUMEN

Glioblastoma is the most malignant form of glioma, which is the most commonly occurring tumor of the central nervous system. Notch signaling in glioblastoma is considered to be a marker of an undifferentiated tumor cell state, associated with tumor stem cells. Notch is also known for facilitating tumor dormancy escape, recurrence and progression after treatment. Studies in vitro suggest that reducing, removing or blocking the expression of this gene triggers tumor cell differentiation, which shifts the phenotype away from stemness status and consequently facilitates treatment. In contrast, in the vasculature, Notch appears to also function as an important receptor that defines mature non-leaking vessels, and increasing its expression promotes tumor normalization in models of cancer in vivo. Failures in clinical trials with Notch inhibitors are potentially related to their opposing effects on the tumor versus the tumor vasculature, which points to the need for a greater understanding of this signaling pathway.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Glioblastoma/genética , Glioma/patología , Humanos , Células Madre Neoplásicas/patología , Transducción de Señal
5.
Cancer Metastasis Rev ; 41(1): 1-15, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35190921

RESUMEN

Coronavirus disease 2019 (COVID-19) due to infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been an ongoing pandemic causing significant morbidity and mortality worldwide. The "cytokine storm" is a critical driving force in severe COVID-19 cases, leading to hyperinflammation, multi-system organ failure, and death. A paradigm shift is emerging in our understanding of the resolution of inflammation from a passive course to an active biochemical process driven by endogenous specialized pro-resolving mediators (SPMs), such as resolvins, protectins, lipoxins, and maresins. SPMs stimulate macrophage-mediated debris clearance and counter pro-inflammatory cytokine production, a process collectively termed as the "resolution of inflammation." Hyperinflammation is not unique to COVID-19 and also occurs in neoplastic conditions, putting individuals with underlying health conditions such as cancer at elevated risk of severe SARS-CoV-2 infection. Despite approaches to block systemic inflammation, there are no current therapies designed to stimulate the resolution of inflammation in patients with COVID-19 or cancer. A non-immunosuppressive therapeutic approach that reduces the cytokine storm in patients with COVID-19 and cancer is urgently needed. SPMs are potent immunoresolvent and organ-protective lipid autacoids that stimulate the resolution of inflammation, facilitate clearance of infections, reduce thrombus burden, and promote a return to tissue homeostasis. Targeting endogenous lipid mediators, such as SPMs, offers an entirely novel approach to control SARS-CoV-2 infection and cancer by increasing the body's natural reserve of pro-resolving mediators without overt toxicity or immunosuppression.


Asunto(s)
COVID-19 , Neoplasias , Síndrome de Liberación de Citoquinas/etiología , Humanos , Inflamación , Pandemias , SARS-CoV-2
6.
Proc Natl Acad Sci U S A ; 117(35): 21576-21587, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32801214

RESUMEN

Toxic environmental carcinogens promote cancer via genotoxic and nongenotoxic pathways, but nongenetic mechanisms remain poorly characterized. Carcinogen-induced apoptosis may trigger escape from dormancy of microtumors by interfering with inflammation resolution and triggering an endoplasmic reticulum (ER) stress response. While eicosanoid and cytokine storms are well-characterized in infection and inflammation, they are poorly characterized in cancer. Here, we demonstrate that carcinogens, such as aflatoxin B1 (AFB1), induce apoptotic cell death and the resulting cell debris stimulates hepatocellular carcinoma (HCC) tumor growth via an "eicosanoid and cytokine storm." AFB1-generated debris up-regulates cyclooxygenase-2 (COX-2), soluble epoxide hydrolase (sEH), ER stress-response genes including BiP, CHOP, and PDI in macrophages. Thus, selective cytokine or eicosanoid blockade is unlikely to prevent carcinogen-induced cancer progression. Pharmacological abrogation of both the COX-2 and sEH pathways by PTUPB prevented the debris-stimulated eicosanoid and cytokine storm, down-regulated ER stress genes, and promoted macrophage phagocytosis of debris, resulting in suppression of HCC tumor growth. Thus, inflammation resolution via dual COX-2/sEH inhibition is an approach to prevent carcinogen-induced cancer.


Asunto(s)
Citocinas/metabolismo , Eicosanoides/metabolismo , Neoplasias Hepáticas/metabolismo , Aflatoxina B1/efectos adversos , Animales , Apoptosis , Carcinogénesis/metabolismo , Carcinógenos/metabolismo , Carcinoma Hepatocelular/metabolismo , Línea Celular , Ciclooxigenasa 2/metabolismo , Citocinas/inmunología , Progresión de la Enfermedad , Eicosanoides/inmunología , Epóxido Hidrolasas/metabolismo , Células Hep G2 , Humanos , Inflamación/metabolismo , Neoplasias Hepáticas/fisiopatología , Macrófagos/metabolismo , Ratones , Procesos Neoplásicos
7.
Ecotoxicol Environ Saf ; 249: 114417, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36525946

RESUMEN

Aflatoxin B1 (AFB1) contamination in food and feed leads to severe global health problems. Acting as the frontier immunological barrier, the intestinal mucosa is constantly challenged by exposure to foodborne toxins such as AFB1 via contaminated diets, but the detailed toxic mechanism and endogenous regulators of AFB1 toxicity are still unclear. Here, we showed that AFB1 disrupted intestinal immune function by suppressing macrophages, especially M2 macrophages, and antimicrobial peptide-secreting Paneth cells. Using an oxylipinomics approach, we identified that AFB1 immunotoxicity is associated with decreased epoxy fatty acids, notably epoxyeicosatrienoic acids, and increased soluble epoxide hydrolase (sEH) levels in the intestine. Furthermore, sEH deficiency or inhibition rescued the AFB1-compromised intestinal immunity by restoring M2 macrophages as well as Paneth cells and their-derived lysozyme and α-defensin-3 in mice. Altogether, our study demonstrates that AFB1 exposure impairs intestinal immunity, at least in part, in a sEH-mediated way. Moreover, the present study supports the potential application of pharmacological intervention by inhibiting the sEH enzyme in alleviating intestinal immunotoxicity and associated complications caused by AFB1 global contamination.


Asunto(s)
Aflatoxina B1 , Epóxido Hidrolasas , Animales , Ratones , Aflatoxina B1/toxicidad , Dieta , Inmunidad , Intestinos
8.
Am J Physiol Cell Physiol ; 323(5): C1444-C1474, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36189975

RESUMEN

Mycobacterium tuberculosis (Mtb) is the pathogen that causes tuberculosis (TB), a leading infectious disease of humans worldwide. One of the main histopathological hallmarks of TB is the formation of granulomas comprised of elaborately organized aggregates of immune cells containing the pathogen. Dissemination of Mtb from infected cells in the granulomas due to host and mycobacterial factors induces multiple cell death modalities in infected cells. Based on molecular mechanism, morphological characteristics, and signal dependency, there are two main categories of cell death: programmed and nonprogrammed. Programmed cell death (PCD), such as apoptosis and autophagy, is associated with a protective response to Mtb by keeping the bacteria encased within dead macrophages that can be readily phagocytosed by arriving in uninfected or neighboring cells. In contrast, non-PCD necrotic cell death favors the pathogen, resulting in bacterial release into the extracellular environment. Multiple types of cell death in the PCD category, including pyroptosis, necroptosis, ferroptosis, ETosis, parthanatos, and PANoptosis, may be involved in Mtb infection. Since PCD pathways are essential for host immunity to Mtb, therapeutic compounds targeting cell death signaling pathways have been experimentally tested for TB treatment. This review summarizes different modalities of Mtb-mediated host cell deaths, the molecular mechanisms underpinning host cell death during Mtb infection, and its potential implications for host immunity. In addition, targeting host cell death pathways as potential therapeutic and preventive approaches against Mtb infection is also discussed.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Tuberculosis/microbiología , Tuberculosis/prevención & control , Mycobacterium tuberculosis/metabolismo , Muerte Celular , Macrófagos/metabolismo , Granuloma/metabolismo , Granuloma/microbiología , Granuloma/patología , Interacciones Huésped-Patógeno
9.
Cancer Metastasis Rev ; 40(3): 791-801, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34665387

RESUMEN

Current cancer therapies aim at eradicating cancer cells from the body. However, killing cells generates cell "debris" which can promote tumor progression. Thus, therapy can be a double-edged sword. Specifically, injury and debris generated by cancer therapies, including chemotherapy, radiation, and surgery, may offset their benefit by promoting the secretion of pro-tumorigenic factors (e.g., eicosanoid-driven cytokines) that stimulate regrowth and metastasis of surviving cells. The debris produced by cytotoxic cancer therapy can also contribute to a tumor microenvironment that promotes tumor progression and recurrence. Although not well understood, several molecular mechanisms have been implicated in debris-stimulated tumor growth that we review here, such as the involvement of extracellular vesicles, exosomal miR-194-5p, Bax, Bak, Smac, HMGB1, cytokines, and caspase-3. We discuss the cases of pancreatic and other cancer types where debris promotes postoperative tumor recurrence and metastasis, thus offering a new opportunity to prevent cancer progression intrinsically linked to treatment by stimulating resolution of tumor-promoting debris.


Asunto(s)
Antineoplásicos , MicroARNs , Neoplasias , Línea Celular Tumoral , Citocinas , Eicosanoides , Humanos , Neoplasias/terapia , Microambiente Tumoral
10.
Proc Natl Acad Sci U S A ; 116(13): 6292-6297, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30862734

RESUMEN

Inflammation in the tumor microenvironment is a strong promoter of tumor growth. Substantial epidemiologic evidence suggests that aspirin, which suppresses inflammation, reduces the risk of cancer. The mechanism by which aspirin inhibits cancer has remained unclear, and toxicity has limited its clinical use. Aspirin not only blocks the biosynthesis of prostaglandins, but also stimulates the endogenous production of anti-inflammatory and proresolving mediators termed aspirin-triggered specialized proresolving mediators (AT-SPMs), such as aspirin-triggered resolvins (AT-RvDs) and lipoxins (AT-LXs). Using genetic and pharmacologic manipulation of a proresolving receptor, we demonstrate that AT-RvDs mediate the antitumor activity of aspirin. Moreover, treatment of mice with AT-RvDs (e.g., AT-RvD1 and AT-RvD3) or AT-LXA4 inhibited primary tumor growth by enhancing macrophage phagocytosis of tumor cell debris and counter-regulating macrophage-secreted proinflammatory cytokines, including migration inhibitory factor, plasminogen activator inhibitor-1, and C-C motif chemokine ligand 2/monocyte chemoattractant protein 1. Thus, the pro-resolution activity of AT-resolvins and AT-lipoxins may explain some of aspirin's broad anticancer activity. These AT-SPMs are active at considerably lower concentrations than aspirin, and thus may provide a nontoxic approach to harnessing aspirin's anticancer activity.


Asunto(s)
Antineoplásicos/farmacología , Aspirina/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/prevención & control , Animales , Aspirina/administración & dosificación , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/metabolismo , Eicosanoides/metabolismo , Ácidos Grasos Insaturados/metabolismo , Femenino , Inflamación/tratamiento farmacológico , Lipoxinas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Metabolómica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/prevención & control , Proteínas del Tejido Nervioso/metabolismo , Fagocitosis/efectos de los fármacos , Inactivadores Plasminogénicos/metabolismo , Prostaglandinas/metabolismo
11.
Proc Natl Acad Sci U S A ; 116(5): 1698-1703, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30647111

RESUMEN

Although chemotherapy is a conventional cancer treatment, it may induce a protumorigenic microenvironment by triggering the release of proinflammatory mediators. In this study, we demonstrate that ovarian tumor cell debris generated by first-line platinum- and taxane-based chemotherapy accelerates tumor progression by stimulating a macrophage-derived "surge" of proinflammatory cytokines and bioactive lipids. Thus, targeting a single inflammatory mediator or pathway is unlikely to prevent therapy-induced tumor progression. Here, we show that combined pharmacological abrogation of the cyclooxygenase-2 (COX-2) and soluble epoxide hydrolase (sEH) pathways prevented the debris-induced surge of both cytokines and lipid mediators by macrophages. In animal models, the dual COX-2/sEH inhibitor PTUPB delayed the onset of debris-stimulated ovarian tumor growth and ascites leading to sustained survival over 120 days postinjection. Therefore, dual inhibition of COX-2/sEH may be an approach to suppress debris-stimulated ovarian tumor growth by preventing the therapy-induced surge of cytokines and lipid mediators.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Epóxido Hidrolasas/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Animales , Hidrocarburos Aromáticos con Puentes/farmacología , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/metabolismo , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lípidos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias Ováricas/metabolismo , Platino (Metal)/farmacología , Transducción de Señal/efectos de los fármacos , Taxoides/farmacología
12.
Cancer Metastasis Rev ; 39(2): 337-340, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32385712

RESUMEN

Severe coronavirus disease (COVID-19) is characterized by pulmonary hyper-inflammation and potentially life-threatening "cytokine storms". Controlling the local and systemic inflammatory response in COVID-19 may be as important as anti-viral therapies. Endogenous lipid autacoid mediators, referred to as eicosanoids, play a critical role in the induction of inflammation and pro-inflammatory cytokine production. SARS-CoV-2 may trigger a cell death ("debris")-induced "eicosanoid storm", including prostaglandins and leukotrienes, which in turn initiates a robust inflammatory response. A paradigm shift is emerging in our understanding of the resolution of inflammation as an active biochemical process with the discovery of novel endogenous specialized pro-resolving lipid autacoid mediators (SPMs), such as resolvins. Resolvins and other SPMs stimulate macrophage-mediated clearance of debris and counter pro-inflammatory cytokine production, a process called inflammation resolution. SPMs and their lipid precursors exhibit anti-viral activity at nanogram doses in the setting of influenza without being immunosuppressive. SPMs also promote anti-viral B cell antibodies and lymphocyte activity, highlighting their potential use in the treatment of COVID-19. Soluble epoxide hydrolase (sEH) inhibitors stabilize arachidonic acid-derived epoxyeicosatrienoic acids (EETs), which also stimulate inflammation resolution by promoting the production of pro-resolution mediators, activating anti-inflammatory processes, and preventing the cytokine storm. Both resolvins and EETs also attenuate pathological thrombosis and promote clot removal, which is emerging as a key pathology of COVID-19 infection. Thus, both SPMs and sEH inhibitors may promote the resolution of inflammation in COVID-19, thereby reducing acute respiratory distress syndrome (ARDS) and other life-threatening complications associated with robust viral-induced inflammation. While most COVID-19 clinical trials focus on "anti-viral" and "anti-inflammatory" strategies, stimulating inflammation resolution is a novel host-centric therapeutic avenue. Importantly, SPMs and sEH inhibitors are currently in clinical trials for other inflammatory diseases and could be rapidly translated for the management of COVID-19 via debris clearance and inflammatory cytokine suppression. Here, we discuss using pro-resolution mediators as a potential complement to current anti-viral strategies for COVID-19.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antivirales/uso terapéutico , Betacoronavirus/inmunología , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/terapia , Antiinflamatorios no Esteroideos/farmacología , Betacoronavirus/aislamiento & purificación , COVID-19 , Ensayos Clínicos como Asunto , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Eicosanoides/inmunología , Eicosanoides/metabolismo , Epóxido Hidrolasas/antagonistas & inhibidores , Epóxido Hidrolasas/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/inmunología , Neumonía Viral/virología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/virología , Síndrome de Dificultad Respiratoria/inmunología , SARS-CoV-2
13.
Am J Pathol ; 190(9): 1782-1788, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32650004

RESUMEN

Severe coronavirus disease 2019 (COVID-19) symptoms, including systemic inflammatory response and multisystem organ failure, are now affecting thousands of infected patients and causing widespread mortality. Coronavirus infection causes tissue damage, which triggers the endoplasmic reticulum stress response and subsequent eicosanoid and cytokine storms. Although proinflammatory eicosanoids, including prostaglandins, thromboxanes, and leukotrienes, are critical mediators of physiological processes, such as inflammation, fever, allergy, and pain, their roles in COVID-19 are poorly characterized. Arachidonic acid-derived epoxyeicosatrienoic acids could alleviate the systemic hyperinflammatory response in COVID-19 infection by modulating endoplasmic reticulum stress and stimulating the resolution of inflammation. Soluble epoxide hydrolase (sEH) inhibitors, which increase endogenous epoxyeicosatrienoic acid levels, exhibit potent anti-inflammatory activity and inhibit various pathologic processes in preclinical disease models, including pulmonary fibrosis, thrombosis, and acute respiratory distress syndrome. Therefore, targeting eicosanoids and sEH could be a novel therapeutic approach in combating COVID-19. In this review, we discuss the predominant role of eicosanoids in regulating the inflammatory cascade and propose the potential application of sEH inhibitors in alleviating COVID-19 symptoms. The host-protective action of omega-3 fatty acid-derived epoxyeicosanoids and specialized proresolving mediators in regulating anti-inflammation and antiviral response is also discussed. Future studies determining the eicosanoid profile in COVID-19 patients or preclinical models are pivotal in providing novel insights into coronavirus-host interaction and inflammation modulation.


Asunto(s)
Antiinflamatorios/farmacología , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Animales , Betacoronavirus/patogenicidad , COVID-19 , Eicosanoides/farmacología , Eicosanoides/uso terapéutico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Pandemias , SARS-CoV-2
14.
Nature ; 523(7561): 468-71, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26201599

RESUMEN

Haematopoietic stem and progenitor cell (HSPC) transplant is a widely used treatment for life-threatening conditions such as leukaemia; however, the molecular mechanisms regulating HSPC engraftment of the recipient niche remain incompletely understood. Here we develop a competitive HSPC transplant method in adult zebrafish, using in vivo imaging as a non-invasive readout. We use this system to conduct a chemical screen, and identify epoxyeicosatrienoic acids (EETs) as a family of lipids that enhance HSPC engraftment. The pro-haematopoietic effects of EETs were conserved in the developing zebrafish embryo, where 11,12-EET promoted HSPC specification by activating a unique activator protein 1 (AP-1) and runx1 transcription program autonomous to the haemogenic endothelium. This effect required the activation of the phosphatidylinositol-3-OH kinase (PI(3)K) pathway, specifically PI(3)Kγ. In adult HSPCs, 11,12-EET induced transcriptional programs, including AP-1 activation, which modulate several cellular processes, such as migration, to promote engraftment. Furthermore, we demonstrate that the EET effects on enhancing HSPC homing and engraftment are conserved in mammals. Our study establishes a new method to explore the molecular mechanisms of HSPC engraftment, and discovers a previously unrecognized, evolutionarily conserved pathway regulating multiple haematopoietic generation and regeneration processes. EETs may have clinical application in marrow or cord blood transplantation.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Pez Cebra/embriología , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animales , Línea Celular , Movimiento Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Riñón/citología , Masculino , Ratones , Fosfatidilinositol 3-Quinasas , Factor de Transcripción AP-1/metabolismo , Transcripción Genética
15.
Am J Physiol Renal Physiol ; 319(4): F563-F570, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32799675

RESUMEN

Acute kidney injury (AKI) frequently complicates major surgery and can be associated with hypertension and progress to chronic kidney disease, but reports on blood pressure normalization in AKI are conflicting. In the present study, we investigated the effects of an angiotensin-converting enzyme inhibitor, enalapril, and a soluble epoxide hydrolase inhibitor, 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl)urea (TPPU), on renal inflammation, fibrosis, and glomerulosclerosis in a mouse model of ischemia-reperfusion injury (IRI)-induced AKI. Male CD1 mice underwent unilateral IRI for 35 min. Blood pressure was measured by tail cuff, and mesangial matrix expansion was quantified on methenamine silver-stained sections. Renal perfusion was assessed by functional MRI in vehicle- and TPPU-treated mice. Immunohistochemistry was performed to study the severity of AKI and inflammation. Leukocyte subsets were analyzed by flow cytometry, and proinflammatory cytokines were analyzed by quantitative PCR. Plasma and tissue levels of TPPU and lipid mediators were analyzed by liquid chromatography mass spectrometry. IRI resulted in a blood pressure increase of 20 mmHg in the vehicle-treated group. TPPU and enalapril normalized blood pressure and reduced mesangial matrix expansion. However, inflammation and progressive renal fibrosis were severe in all groups. TPPU further reduced renal perfusion on days 1 and 14. In conclusion, early antihypertensive treatment worsened renal outcome after AKI by further reducing renal perfusion despite reduced glomerulosclerosis.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glomerulonefritis/prevención & control , Hipertensión/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Piperidinas/farmacología , Daño por Reperfusión/tratamiento farmacológico , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Antihipertensivos/toxicidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Enalapril/farmacología , Inhibidores Enzimáticos/toxicidad , Epóxido Hidrolasas/antagonistas & inhibidores , Fibrosis , Mesangio Glomerular/efectos de los fármacos , Mesangio Glomerular/patología , Mesangio Glomerular/fisiopatología , Glomerulonefritis/etiología , Glomerulonefritis/patología , Glomerulonefritis/fisiopatología , Hipertensión/etiología , Hipertensión/fisiopatología , Masculino , Ratones , Compuestos de Fenilurea/toxicidad , Piperidinas/toxicidad , Daño por Reperfusión/complicaciones , Daño por Reperfusión/fisiopatología
16.
FASEB J ; 33(1): 114-125, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29957058

RESUMEN

Colon cancer recurrence after therapy, such as 5-fluorouracil (5-FU), remains a challenge in the clinical setting. Chemotherapy reduces tumor burden by inducing cell death; however, the resulting dead tumor cells, or debris, may paradoxically stimulate angiogenesis, inflammation, and tumor growth. Here, we demonstrate that 5-FU-generated colon carcinoma debris stimulates the growth of a subthreshold inoculum of living tumor cells in subcutaneous and orthotopic models. Debris triggered the release of osteopontin (OPN) by tumor cells and host macrophages. Both coinjection of debris and systemic treatment with 5-FU increased plasma OPN levels in tumor-bearing mice. RNA expression levels of secreted phosphoprotein 1, the gene that encodes OPN, correlate with poor prognosis in patients with colorectal cancer and are elevated in chemotherapy-treated patients who experience tumor recurrence vs. no recurrence. Pharmacologic and genetic ablation of OPN inhibited debris-stimulated tumor growth. Systemic treatment with a combination of a neutralizing OPN antibody and 5-FU dramatically inhibited tumor growth. These results demonstrate a novel mechanism of tumor progression mediated by OPN released in response to chemotherapy-generated tumor cell debris. Neutralization of debris-stimulated OPN represents a potential therapeutic strategy to overcome the inherent limitation of cytotoxic therapies as a result of the generation of cell debris.-Chang, J., Bhasin, S. S., Bielenberg, D. R., Sukhatme, V. P., Bhasin, M., Huang, S., Kieran, M. W., Panigrahy, D. Chemotherapy-generated cell debris stimulates colon carcinoma tumor growth via osteopontin.


Asunto(s)
Neoplasias del Colon/patología , Fluorouracilo/farmacología , Neovascularización Patológica/patología , Osteopontina/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis , Proliferación Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Proc Natl Acad Sci U S A ; 114(17): 4370-4375, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396419

RESUMEN

Arachidonic acid (ARA) is metabolized by cyclooxygenase (COX) and cytochrome P450 to produce proangiogenic metabolites. Specifically, epoxyeicosatrienoic acids (EETs) produced from the P450 pathway are angiogenic, inducing cancer tumor growth. A previous study showed that inhibiting soluble epoxide hydrolase (sEH) increased EET concentration and mildly promoted tumor growth. However, inhibiting both sEH and COX led to a dramatic decrease in tumor growth, suggesting that the contribution of EETs to angiogenesis and subsequent tumor growth may be attributed to downstream metabolites formed by COX. This study explores the fate of EETs with COX, the angiogenic activity of the primary metabolites formed, and their subsequent hydrolysis by sEH and microsomal EH. Three EET regioisomers were found to be substrates for COX, based on oxygen consumption and product formation. EET substrate preference for both COX-1 and COX-2 were estimated as 8,9-EET > 5,6-EET > 11,12-EET, whereas 14,15-EET was inactive. The structure of two major products formed from 8,9-EET in this COX pathway were confirmed by chemical synthesis: ct-8,9-epoxy-11-hydroxy-eicosatrienoic acid (ct-8,9-E-11-HET) and ct-8,9-epoxy-15-hydroxy-eicosatrienoic acid (ct-8,9-E-15-HET). ct-8,9-E-11-HET and ct-8,9-E-15-HET are further metabolized by sEH, with ct-8,9-E-11-HET being hydrolyzed much more slowly. Using an s.c. Matrigel assay, we showed that ct-8,9-E-11-HET is proangiogenic, whereas ct-8,9-E-15-HET is not active. This study identifies a functional link between EETs and COX and identifies ct-8,9-E-11-HET as an angiogenic lipid, suggesting a physiological role for COX metabolites of EETs.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Inductores de la Angiogénesis/metabolismo , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Ácido 8,11,14-Eicosatrienoico/metabolismo , Ácido Araquidónico/metabolismo , Humanos
18.
Cancer Metastasis Rev ; 37(2-3): 557-572, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30136088

RESUMEN

Bioactive lipids are essential components of human cells and tissues. As discussed in this review, the cancer lipidome is diverse and malleable, with the ability to promote or inhibit cancer pathogenesis. Targeting lipids within the tumor and surrounding microenvironment may be a novel therapeutic approach for treating cancer patients. Additionally, the emergence of a novel super-family of lipid mediators termed specialized pro-resolving mediators (SPMs) has revealed a new role for bioactive lipid mediators in the resolution of inflammation in cancer biology. The role of SPMs in cancer holds great promise in our understanding of cancer pathogenesis and can ultimately be used in future cancer diagnostics and therapy.


Asunto(s)
Antineoplásicos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos , Terapia Molecular Dirigida , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Humanos , Mediadores de Inflamación/metabolismo , Lípidos/química , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Relación Estructura-Actividad
19.
Artículo en Inglés | MEDLINE | ID: mdl-29933028

RESUMEN

Ovarian cancer is the most lethal gynecologic malignancy among women. Due to the heterogeneity and complexity of the disease, as well as the insidious onset of symptoms, timely diagnosis remains extremely challenging. Despite recent advances in chemotherapy regimens for ovarian cancer patients, many still suffer from recurrence and ultimately succumb to the disease; thus, there is an urgent need for the identification of novel therapeutic targets. Within this rapidly evolving field, the role of platelets in the ovarian cancer tumor microenvironment has garnered increased attention. It is well-established that platelets and tumor cells exhibit bidirectional communication in which platelets enhance tumor cell invasion, extravasation, and protection from host system defenses, while tumor cells serve as platelet agonists, increasing platelet adhesion, aggregation, and degranulation. This mini-review focuses on the platelet-tumor cell relationship in ovarian cancer, specifically highlighting the essential role of bioactive lipid mediators at this interface.


Asunto(s)
Plaquetas/metabolismo , Neoplasias Ováricas/metabolismo , Tromboxano A2/metabolismo , Animales , Plaquetas/patología , Femenino , Humanos , Invasividad Neoplásica , Neoplasias Ováricas/patología
20.
Am J Pathol ; 186(4): 1055-64, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26877262

RESUMEN

Neuropilins are receptors for disparate ligands, including proangiogenic factors such as vascular endothelial growth factor and inhibitory class 3 semaphorin (SEMA3) family members. Differentiated cells in skin epithelium and cutaneous squamous cell carcinoma highly express the neuropilin-1 (NRP1) receptor. We examined the expression of NRP1 in human and mouse oral mucosa. NRP1 was significantly up-regulated in oral epithelial dysplasia and oral squamous cell carcinoma (OSCC). NRP1 receptor localized to the outer suprabasal epithelial layers in normal tongue, an expression pattern similar to the normal skin epidermis. However, dysplastic tongue epithelium and OSCC up-regulated NRP1 in basal and proliferating epithelial layers, a profile unseen in cutaneous squamous cell carcinoma. NRP1 up-regulation is observed in a mouse carcinogen-induced OSCC model and in human tongue OSCC biopsies. Human OSCC cell lines express NRP1 protein in vitro and in mouse tongue xenografts. Sites of capillary infiltration into orthotopic OSCC tumors correlate with high NRP1 expression. HSC3 xenografts, which express the highest NRP1 levels of the cell lines examined, showed massive intratumoral lymphangiogenesis. SEMA3A inhibited OSCC cell migration, suggesting that the NRP1 receptor was bioactive in OSCC. In conclusion, NRP1 is regulated in the oral epithelium and is selectively up-regulated during epithelial dysplasia. NRP1 may function as a reservoir to sequester proangiogenic ligands within the neoplastic compartment, thereby recruiting neovessels toward tumor cells.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Epitelio/metabolismo , Neoplasias de la Boca/metabolismo , Receptores de Neuroquinina-1/metabolismo , Neoplasias Cutáneas/metabolismo , Carcinoma in Situ/metabolismo , Carcinoma de Células Escamosas/patología , Diferenciación Celular , Epitelio/patología , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/patología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA