Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 364
Filtrar
1.
Cancer Sci ; 114(5): 1929-1942, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36644823

RESUMEN

Ovarian cancer (OC) is characterized by frequent widespread peritoneal metastasis. Cancer-associated fibroblasts (CAFs) represent a critical stromal component of metastatic niche and promote omentum metastasis in OC patients. However, the role of exosomes derived from omental CAFs in metastasis remains unclear. We isolated exosomes from primary omental normal fibroblasts (NFs) and CAFs from OC patients (NF-Exo and CAF-Exo, respectively) and assessed their effect on metastasis. In mice bearing orthotopic OC xenografts, CAF-Exo treatment led to more rapid intraperitoneal tumor dissemination and shorter animal survival. Similar results were observed in mice undergoing intraperitoneal injection of tumor cells. Among the miRNAs downregulated in CAF-Exo, miR-29c-3p in OC tissues was associated with metastasis and survival in patients. Moreover, increasing miR-29c-3p in CAF-Exo significantly weakened the metastasis-promoting effect of CAF-Exo. Based on RNA sequencing, expression assays, and luciferase assays, matrix metalloproteinase 2 (MMP2) was identified as a direct target of miR-29c-3p. These results verify the significant contribution of exosomes from omental CAFs to OC peritoneal metastasis, which could be partially due to the relief of MMP2 expression inhibition mediated by low exosomal miR-29c-3p.


Asunto(s)
Fibroblastos Asociados al Cáncer , Exosomas , MicroARNs , Neoplasias Ováricas , Neoplasias Peritoneales , Femenino , Humanos , Animales , Ratones , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Exosomas/metabolismo , Neoplasias Peritoneales/patología , Epiplón/metabolismo , Epiplón/patología , Proliferación Celular , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Ováricas/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
2.
Biomaterials ; 294: 121996, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36689832

RESUMEN

Reliable and predictive experimental models are urgently needed to study metastatic mechanisms of ovarian cancer cells in the omentum. Although models for ovarian cancer cell adhesion and invasion were previously investigated, the lack of certain omental cell types, which influence the metastatic behavior of cancer cells, limits the application of these tissue models. Here, we describe a 3D multi-cellular human omentum tissue model, which considers the spatial arrangement of five omental cell types. Reproducible tissue models were fabricated combining permeable cell culture inserts and bioprinting technology to mimic metastatic processes of immortalized and patient-derived ovarian cancer cells. The implementation of an endothelial barrier further allowed studying the interaction between cancer and endothelial cells during hematogenous dissemination and the impact of chemotherapeutic drugs. This proof-of-concept study may serve as a platform for patient-specific investigations in personalized oncology in the future.


Asunto(s)
Epiplón , Neoplasias Ováricas , Humanos , Femenino , Epiplón/metabolismo , Epiplón/patología , Células Endoteliales/metabolismo , Neoplasias Ováricas/patología , Células Cultivadas , Técnicas de Cultivo de Célula
3.
Tissue Eng Part C Methods ; 28(12): 672-682, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36326206

RESUMEN

Recent advances in the field of tissue engineering and regenerative medicine have contributed to the repair of damaged tissues and organs. Renal dysfunctions such as chronic kidney disease (CKD) are considered intractable owing to its cellular heterogeneity. In addition, the absence of definitive treatment options other than dialysis or kidney transplantation in advanced CKD. In this study, we investigated therapeutic effects of a three-dimensional (3D) bio-printed omentum patch as treatment source. Because omentum contains a lot of biological sources for immune regulation and tissue regeneration, it has been used in clinic for >100 years. By using autologous tissue as a bio-ink, the patch could minimize the immune response. The mechanically micronized omentum without any additives became small enough to print, but the original components could be preserved. Then, the 3D printed omentum patch was transplanted under renal subcapsular layer in unilateral ureteral obstruction (UUO) rat model. After 14 days of patch transplantation, the kidneys were analyzed through bulk RNA sequencing and histopathological staining. From the results, decreased tubular injury was observed in the omentum patch group. In addition, the omentum patch significantly altered biological process of gene ontology such as fibrosis-related gene and growth factors. RNA sequencing confirmed the antifibrotic effect by inhibiting fibrosis-inducing mechanisms within PI3K-AKT and JAK-STAT pathways. In conclusion, the omentum patch showed the effect of antitubular injury and antifibrosis on UUO kidneys. In particular, the omentum patch is expected to protect the organ from further degeneration and loss of function by inhibiting the progression of fibrosis. The omentum patch can be a novel therapeutic option for renal dysfunction. Impact statement Many studies and clinical trials are being conducted to develop new treatments for kidney disease. However, there are no newly developed renal replacement therapies. In this study, we developed a new treatment that can ameliorate renal interstitial fibrosis using three-dimensional (3D) bio-printed autologous omentum patch. The 3D printer enables precise patch printing, and the bio-ink made of autologous tissue minimizes the immune response after transplantation. The whole kidneys were analyzed by RNA sequencing and histopathological staining 14 days after transplantation. From the results, the omentum patch had the effect of relieving tubular injury in the injured state. Also, the omentum patch significantly altered biological process of gene ontology. In particular, genes related to fibrosis were observed to be downregulated by the omentum patch. RNA sequencing confirmed that the antifibrotic effect was owing to inducing mechanisms of PI3K-AKT and JAK-STAT pathways. The findings reported in this study represent a significant advancement in the application of 3D bio-printer to damaged organ treatments, especially fibrosis-related diseases.


Asunto(s)
Insuficiencia Renal Crónica , Obstrucción Ureteral , Ratas , Animales , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/farmacología , Fosfatidilinositol 3-Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/farmacología , Proteínas Proto-Oncogénicas c-akt/uso terapéutico , Epiplón/metabolismo , Fibrosis , Riñón , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Modelos Animales de Enfermedad
4.
Cells ; 11(21)2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36359787

RESUMEN

Epithelial ovarian cancer (EOC) patients frequently develop peritoneal metastasis, especially in the human omentum. However, the mechanism underlying this propensity remains unknown. A previous study found that human omental adipose-derived mesenchymal stem cells are potentially involved in ovarian cancer growth and metastasis, but the results were inconsistent and even contradictory. In addition, the underlying mechanisms of visceral adipose metastasis remain poorly understood. Here, our goal is to clarify the role and mechanism of human omental adipose-derived mesenchymal stem cells (HO-ADSCs) in EOC cancer growth and metastasis. We first found that human omental tissue conditioned medium (HO-CM) enhances EOC cell function. Subsequent coculture studies indicated that HO-ADSCs increase the growth, migratory and invasive capabilities of ovarian cancer cells. Then, we demonstrated that exosomes secreted by HO-ADSCs (HO-ADSC exosomes) enhanced ovarian cancer cell function, and further mechanistic studies showed that the FOXM1, Cyclin F, KIF20A, and MAPK signaling pathways were involved in this process. In addition, subcutaneous tumorigenesis and peritoneal metastatic xenograft experiments provided evidence that HO-ADSC exosomes promote ovarian cancer growth and metastasis in vivo. Finally, our clinical studies provided evidence that ascites from ovarian cancer patients enhance EOC cell line proliferation, migration, and invasion in vitro. The present study indicated that HO-ADSC exosomes are secreted into ascites and exert a tumor-promoting effect on EOC growth and metastasis, providing a new perspective and method to develop future novel therapeutic strategies for the treatment of ovarian cancer.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Neoplasias Ováricas , Neoplasias Peritoneales , Humanos , Femenino , Carcinoma Epitelial de Ovario/patología , Exosomas/metabolismo , Epiplón/metabolismo , Epiplón/patología , Ascitis/patología , Tejido Adiposo/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neoplasias Ováricas/patología , Procesos Neoplásicos
5.
Cell Rep ; 39(13): 111012, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35767962

RESUMEN

Ovarian cancer (OC) is the most lethal gynecological malignancy, with aggressive metastatic disease responsible for the majority of OC-related deaths. In particular, OC tumors preferentially metastasize to and proliferate rapidly in the omentum. Here, we show that metastatic OC cells experience increased oxidative stress in the omental microenvironment. Metabolic reprogramming, including upregulation of the pentose phosphate pathway (PPP), a key cellular redox homeostasis mechanism, allows OC cells to compensate for this challenge. Inhibition of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the PPP, reduces tumor burden in pre-clinical models of OC, suggesting that this adaptive metabolic dependency is important for OC omental metastasis.


Asunto(s)
Glucosafosfato Deshidrogenasa , Neoplasias Ováricas , Carcinoma Epitelial de Ovario , Femenino , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Epiplón/metabolismo , Estrés Oxidativo , Vía de Pentosa Fosfato , Microambiente Tumoral
6.
Immunology ; 166(4): 458-474, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35437746

RESUMEN

The relationship between macrophages of the peritoneal cavity and the adjacent omentum remains poorly understood. Here, we describe two populations of omental macrophages distinguished by CD102 expression and use an adoptive cell transfer approach to investigate whether these arise from peritoneal macrophages, and whether this depends upon inflammatory status, the origin of peritoneal macrophages and availability of the omental niches. We show that whereas established resident peritoneal macrophages largely fail to migrate to the omentum, monocyte-derived resident cells readily migrate and form a substantial component of omental CD102+ macrophages in the months following resolution of peritoneal inflammation. In contrast, both populations had the capacity to migrate to the omentum in the absence of endogenous peritoneal and omental macrophages. However, inflammatory macrophages expanded more effectively and more efficiently repopulated both CD102+ and CD102- omental populations, whereas established resident macrophages partially reconstituted the omental niche via recruitment of monocytes. Hence, cell origin determines the migration of peritoneal macrophages to the omentum and predisposes established resident macrophages to drive infiltration of monocyte-derived cells.


Asunto(s)
Macrófagos Peritoneales , Epiplón , Macrófagos , Epiplón/metabolismo , Cavidad Peritoneal
7.
Circ Res ; 130(3): 366-383, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986653

RESUMEN

BACKGROUND: The chromatin-remodeling enzyme BRG1 (brahma-related gene 1) regulates gene expression in a variety of rapidly differentiating cells during embryonic development. However, the critical genes that BRG1 regulates during lymphatic vascular development are unknown. METHODS: We used genetic and imaging techniques to define the role of BRG1 in murine embryonic lymphatic development, although this approach inadvertently expanded our study to multiple interacting cell types. RESULTS: We found that omental macrophages fine-tune an unexpected developmental process by which erythrocytes escaping from naturally discontinuous omental blood vessels are collected by nearby lymphatic vessels. Our data indicate that circulating fibrin(ogen) leaking from gaps in omental blood vessels can trigger inflammasome-mediated IL-1ß (interleukin-1ß) production and secretion from nearby macrophages. IL-1ß destabilizes adherens junctions in omental blood and lymphatic vessels, contributing to both extravasation of erythrocytes and their uptake by lymphatics. BRG1 regulates IL-1ß production in omental macrophages by transcriptionally suppressing the inflammasome trigger RIPK3 (receptor interacting protein kinase 3). CONCLUSIONS: Genetic deletion of Brg1 in embryonic macrophages leads to excessive IL-1ß production, erythrocyte leakage from blood vessels, and blood-filled lymphatics in the developing omentum. Altogether, these results highlight a novel context for epigenetically regulated crosstalk between macrophages, blood vessels, and lymphatics.


Asunto(s)
Vasos Sanguíneos/metabolismo , ADN Helicasas/metabolismo , Interleucina-1beta/metabolismo , Vasos Linfáticos/metabolismo , Proteínas Nucleares/metabolismo , Epiplón/metabolismo , Factores de Transcripción/metabolismo , Uniones Adherentes/metabolismo , Animales , Vasos Sanguíneos/embriología , ADN Helicasas/genética , Eritrocitos/metabolismo , Inflamasomas/metabolismo , Vasos Linfáticos/embriología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Epiplón/irrigación sanguínea , Epiplón/embriología , Factores de Transcripción/genética
8.
Int J Mol Sci ; 23(1)2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-35008946

RESUMEN

Obesity is associated with extensive expansion and remodeling of the adipose tissue architecture, including its microenvironment and extracellular matrix (ECM). Although obesity has been reported to induce adipose tissue fibrosis, the composition of the ECM under healthy physiological conditions has remained underexplored and debated. Here, we used a combination of three established techniques (picrosirius red staining, a colorimetric hydroxyproline assay, and sensitive gene expression measurements) to evaluate the status of the ECM in metabolically healthy lean (MHL) and metabolically unhealthy obese (MUO) subjects. We investigated ECM deposition in the two major human adipose tissues, namely the omental and subcutaneous depots. Biopsies were obtained from the same anatomic region of respective individuals. We found robust ECM deposition in MHL subjects, which correlated with high expression of collagens and enzymes involved in ECM remodeling. In contrast, MUO individuals showed lower expression of ECM components but elevated levels of ECM cross-linking and adhesion proteins, e.g., lysyl oxidase and thrombospondin. Our data suggests that subcutaneous fat is more prone to express proteins involved in ECM remodeling than omental adipose tissues. We conclude that a more dynamic ability to deposit and remodel ECM may be a key signature of healthy adipose tissue, and that subcutaneous fat may adapt more readily to changing metabolic conditions than omental fat.


Asunto(s)
Tejido Adiposo/metabolismo , Matriz Extracelular/metabolismo , Expresión Génica , Epiplón/metabolismo , Grasa Subcutánea/metabolismo , Adulto , Biomarcadores , Colágeno/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Especificidad de Órganos/genética , ARN Mensajero/genética , Sensibilidad y Especificidad
9.
J Clin Endocrinol Metab ; 107(3): 755-775, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34669916

RESUMEN

CONTEXT: Adipose tissue distribution is a key factor influencing metabolic health and risk in obesity-associated comorbidities. OBJECTIVE: Here we aim to compare the proteomic profiles of mature adipocytes from different depots. METHODS: Abdominal subcutaneous (SA) and omental visceral adipocytes (VA) were isolated from paired adipose tissue biopsies obtained during bariatric surgery on 19 severely obese women (body mass index > 30 kg/m2) and analyzed using state-of-the-art mass spectrometry. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were performed to investigate proteome signature properties and to examine a possible association of the protein expression with the clinical data. RESULTS: We identified 3686 protein groups and found 1140 differentially expressed proteins (adj. P value < 0.05), of which 576 proteins were upregulated in SA and 564 in VA samples. We provide a global protein profile of abdominal SA and omental VA, present the most differentially expressed pathways and processes distinguishing SA from VA, and correlate them with clinical and body composition data. We show that SA are significantly more active in processes linked to vesicular transport and secretion, and to increased lipid metabolism activity. Conversely, the expression of proteins involved in the mitochondrial energy metabolism and translational or biosynthetic activity is higher in VA. CONCLUSION: Our analysis represents a valuable resource of protein expression profiles in abdominal SA and omental VA, highlighting key differences in their role in obesity.


Asunto(s)
Adipocitos/metabolismo , Grasa Intraabdominal/metabolismo , Obesidad Mórbida/metabolismo , Grasa Subcutánea Abdominal/metabolismo , Adulto , Cirugía Bariátrica , Femenino , Redes Reguladoras de Genes , Humanos , Grasa Intraabdominal/citología , Grasa Intraabdominal/patología , Persona de Mediana Edad , Obesidad Mórbida/patología , Obesidad Mórbida/cirugía , Epiplón/citología , Epiplón/metabolismo , Epiplón/patología , Epiplón/cirugía , Proteómica , Grasa Subcutánea Abdominal/citología , Grasa Subcutánea Abdominal/patología
10.
Int J Cancer ; 149(11): 1961-1972, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34469585

RESUMEN

Adipocyte-rich omentum offers "good soil" for disseminating ovarian cancer (OvCa), contributing to therapeutic difficulty. However, little is understood about the association between adipocytes and tumor growth at peritoneal dissemination site. Herein, we report the induction of adipocyte dedifferentiation by OvCa cells and pro-tumorigenic effects of resulted adipocyte-derived fibroblasts. We confirmed that malignant ascites promoted the dedifferentiation of the primary human adipocytes obtained from surgical omental specimen into omental adipocyte-derived fibroblast (O-ADF) that possess both mesenchymal stem cell and myofibroblast-like features. This promotion of dedifferentiation by malignant ascites was blocked by addition of Wnt signaling inhibitor. The effects of dedifferentiated adipocytes in proliferation and migration of OvCa cells were analyzed with in vitro coculturing experimental models and in vivo mice model, and we demonstrated that OvCa cell lines showed enhanced proliferative characteristics, as well as increased migratory abilities upon coculturing with O-ADF. Additionally, exogenous transforming growth factor-ß1 augmented desmoplastic morphological change of O-ADF, leading to higher proliferative ability. Our results suggest that OvCa cells promote dedifferentiation of peritoneal adipocytes by activating Wnt/ß-catenin signaling, and generated O-ADFs exhibit pro-tumoral hallmarks.


Asunto(s)
Adipocitos/patología , Fibroblastos Asociados al Cáncer/patología , Epiplón/patología , Neoplasias Ováricas/patología , Microambiente Tumoral , Células 3T3-L1 , Actinas/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Ascitis/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Desdiferenciación Celular/efectos de los fármacos , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Imidas/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/patología , Epiplón/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/secundario , Quinolinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt3A/metabolismo
11.
Obesity (Silver Spring) ; 29(6): 976-984, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33943025

RESUMEN

OBJECTIVE: Morphological alterations including adipocyte hypertrophy and fibrosis deposition are important surrogate markers of visceral adipose tissue function, but the relationships between these morphological changes and type 2 diabetes mellitus (T2DM) and impaired insulin sensitivity are poorly defined. METHODS: Omental adipose tissue was obtained from 66 individuals with obesity but without T2DM (OB group), 93 individuals with both obesity and T2DM (T2DM group), and 15 individuals with normal BMI and normal glucose tolerance (NGT group). Adipocyte diameter and volume were measured through pathological section analysis. Pericellular and perilobular fibrosis was determined through picrosirius red staining and immunochemistry, while fibrosis-related genes were tested through gene expression and hydroxyproline content. RESULTS: Compared with the NGT and OB groups, individuals from the T2DM group displayed increased adipocyte diameter and volume levels. Increased adipocyte size (diameter and volume) was positively associated with hyperglycemia and insulin resistance and inversely correlated with insulin sensitivity (using the Matsuda whole-body insulin sensitivity index assessment of insulin sensitivity) and ß-cell function (disposition index 30 and disposition index 120). The fibrosis levels of the OB group were the highest out of the three groups, whereas the fibrosis levels of T2DM individuals were lower than the OB group but higher than the NGT group. Although fibrosis was negatively correlated with T2DM, fibrosis deposition was not remarkably associated with impaired systemic insulin sensitivity and glucose metabolism. CONCLUSIONS: Compared with fibrosis deposition, adipocyte hypertrophy is more closely associated with T2DM and impaired systemic insulin sensitivity.


Asunto(s)
Diabetes Mellitus Tipo 2/epidemiología , Grasa Intraabdominal/patología , Obesidad/epidemiología , Epiplón/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Adulto , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Fibrosis/complicaciones , Fibrosis/epidemiología , Fibrosis/metabolismo , Humanos , Hipertrofia/complicaciones , Hipertrofia/epidemiología , Hipertrofia/metabolismo , Resistencia a la Insulina/fisiología , Grasa Intraabdominal/metabolismo , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología , Epiplón/patología
12.
BMC Cancer ; 21(1): 461, 2021 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-33902518

RESUMEN

BACKGROUND: Pressurized Intra-Peritoneal Aerosol Chemotherapy (PIPAC) is an innovative treatment against peritoneal carcinomatosis. Doxorubicin is a common intra-venous chemotherapy used for peritoneal carcinomatosis and for PIPAC. This study evaluated the impact of increased PIPAC intraperitoneal pressure on the distribution and cell penetration of doxorubicin in a sheep model. METHODS: Doxorubicin was aerosolized using PIPAC into the peritoneal cavity of 6 ewes (pre-alpes breed): N = 3 with 12 mmHg intraperitoneal pressure ("group 12") and N = 3 with 20 mmHg ("group 20"). Samples from peritoneum (N = 6), ovarian (N = 1), omentum (N = 1) and caecum (N = 1) were collected for each ewe. The number of doxorubicin positive cells was determined using the ratio between doxorubicine fluorescence-positive cell nuclei (DOXO+) over total number of DAPI positive cell nuclei (DAPI+). Penetration depth (µm) was defined as the distance between the luminal surface and the location of the deepest DOXO+ nuclei over the total number of cell nuclei that were stained with DAPI. Penetration depth (µm) was defined as the distance between the luminal surface and the location of the deepest DOXO+ nuclei. RESULTS: DOXO+ nuclei were identified in 87% of samples. All omental samples, directly localized in front of the nebulizer head, had 100% DOXO+ nuclei whereas very few nuclei were DOXO+ for caecum. Distribution patterns were not different between the two groups but penetration depth in ovary and caecum samples was significantly deeper in group 20. CONCLUSIONS: This study showed that applying a higher intra-peritoneal pressure during PIPAC treatment leads to a deeper penetration of doxorubicin in ovarian and caecum but does not affect distribution patterns.


Asunto(s)
Antibióticos Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Peritoneales/metabolismo , Aerosoles , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/análisis , Ciego/química , Ciego/metabolismo , Núcleo Celular/química , Doxorrubicina/administración & dosificación , Doxorrubicina/análisis , Femenino , Epiplón/química , Epiplón/metabolismo , Ovario/química , Ovario/metabolismo , Neoplasias Peritoneales/tratamiento farmacológico , Peritoneo/química , Peritoneo/metabolismo , Presión , Ovinos , Distribución Tisular
13.
FASEB J ; 35(5): e21534, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33817830

RESUMEN

The adipocyte precursors (APs) located in white adipose tissue (WAT) are functionally significant in adipose plasticity and browning. Modifying adipogenesis or WAT browning targeted on APs is a promising mechanism for anti-obesity drug. We herein explored the in vitro actions and mechanisms of glucose-dependent insulinotropic polypeptide (GIP), a gut-derived peptide, in human adipose-derived mesenchymal stem cells (hADSCs) isolated from omentum. The hADSCs were cotreated with 100 nM GIP with or without equimolar concentration of GIP3-42 (a GIP receptor antagonist), and subsequently examined in vitro. CCK-8, EdU incorporation, and flow cytometry assays were used to assess cellular proliferation. Annexin V FTIC/PI double stain, TUNEL staining, and Western blot were applied for apoptosis evaluation. Adipogenesis was reflected by Western blot, real-time PCR, Oil Red O staining, mitochondrial staining, and mitochondrial DNA analysis. Results showed that GIP promoted proliferation and inhibited apoptosis of hADSCs via pleiotropic effects. Besides, GIP facilitated de novo beige adipogenesis, by accelerating mitotic clonal expansion (MCE), upregulating core adipogenic regulators (C/EBPα and PPARγ), augmenting beige-related genes (UCP1, PGC1α, and PRDM16), increasing mitochondrial content and improving beige adipocyte functionalities. Above all, our study expands knowledge on the mechanisms of GIP modifying adipogenesis especially in inducing beige adipogenesis, and thus provides a theoretical support for clinical usage of GIP on obesity treatment.


Asunto(s)
Adipocitos Beige/citología , Adipocitos/citología , Adipogénesis , Polipéptido Inhibidor Gástrico/farmacología , Fármacos Gastrointestinales/farmacología , Células Madre Mesenquimatosas/citología , Epiplón/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos Beige/efectos de los fármacos , Adipocitos Beige/metabolismo , Diferenciación Celular , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Epiplón/efectos de los fármacos , Epiplón/metabolismo , Transducción de Señal
14.
Cell Rep ; 31(13): 107818, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32610121

RESUMEN

Obesity is characterized by adipose tissue inflammation. Because proteoglycans regulate inflammation, here we investigate their role in adipose tissue inflammation in obesity. We find that adipose tissue versican and biglycan increase in obesity. Versican is produced mainly by adipocytes and biglycan by adipose tissue macrophages. Both proteoglycans are also present in adipose tissue from obese human subjects undergoing gastric bypass surgery. Deletion of adipocyte-specific versican or macrophage-specific biglycan in mice reduces macrophage accumulation and chemokine and cytokine expression, although only adipocyte-specific versican deletion leads to sustained improvement in glucose tolerance. Macrophage-derived biglycan activates inflammatory genes in adipocytes. Versican expression increases in cultured adipocytes exposed to excess glucose, and adipocyte-conditioned medium stimulates inflammation in resident peritoneal macrophages, in part because of a versican breakdown product, versikine. These findings provide insights into the role of adipocyte- and macrophage-derived proteoglycans in adipose tissue inflammation in obesity.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/patología , Biglicano/metabolismo , Inflamación/patología , Macrófagos/metabolismo , Obesidad/patología , Versicanos/metabolismo , Células 3T3-L1 , Animales , Médula Ósea/metabolismo , Dieta Alta en Grasa , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Hipertrofia , Resistencia a la Insulina , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Persona de Mediana Edad , Epiplón/metabolismo , Especificidad de Órganos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Grasa Subcutánea/patología , Versicanos/genética
15.
Br J Cancer ; 123(3): 459-470, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32439934

RESUMEN

BACKGROUND: Gastric cancer (GC) patients frequently develop peritoneal metastasis; however, the underlying mechanism remains unknown. We hypothesised that omental adipocytes (OmAd) trigger GC cells towards malignant activity to induce peritoneal metastasis. METHODS: We analysed interactions among human GC cells, endothelial cells and OmAd using a 3D co-culture system. We also employed a multipronged animal study, including subcutaneous and orthotopic tumours, and humanised omental adipose tissue models. Urinary levels of CXCL2 were analysed in human GC patients with and without peritoneal metastasis. RESULTS: Conditioned media derived from OmAd (OmAd-CM) promoted the proliferation, migration and capacity to induce angiogenesis of GC cells through AKT phosphorylation and VEGFA overexpression, whereas silencing CXCL2 in OmAd cancelled OmAd-induced effects. In an orthotopic tumour model using SCID mice, omentectomy suppressed GC growth and peritoneal dissemination, and reduced serum levels of CXCL2. OmAd promoted GC growth in a humanised omental adipose tissue model using NSG mice, but silencing CXCL2 in OmAd cancelled OmAd-induced tumour growth. Finally, urinary levels of CXCL2 were significantly higher in GC patients with peritoneal metastasis than in those without. CONCLUSION: Omental adipocytes trigger GC cells to an aggressive phenotype through CXCL2 secretion, which induces angiogenesis followed by cell growth and peritoneal metastasis.


Asunto(s)
Quimiocina CXCL2/orina , Técnicas de Cocultivo/métodos , Epiplón/citología , Neoplasias Peritoneales/secundario , Neoplasias Gástricas/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Línea Celular Tumoral , Quimiocina CXCL2/genética , Medios de Cultivo Condicionados/química , Femenino , Humanos , Ratones , Ratones SCID , Epiplón/metabolismo , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Regulación hacia Arriba
16.
JCI Insight ; 5(6)2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32125283

RESUMEN

Severe obesity (SO) affects about 6% of youth in the United States, augmenting the risks for cardiovascular disease and type 2 diabetes. Herein, we obtained paired omental adipose tissue (omVAT) and abdominal subcutaneous adipose tissue (SAT) biopsies from girls with SO undergoing sleeve gastrectomy (SG), to test whether differences in cellular and transcriptomic profiles between omVAT and SAT depots affect insulin sensitivity differently. Following weight loss, these analyses were repeated in a subgroup of subjects having a second SAT biopsy. We found that omVAT displayed smaller adipocytes compared with SAT, increased lipolysis through adipose triglyceride lipase phosphorylation, reduced inflammation, and increased expression of browning/beiging markers. Contrary to omVAT, SAT adipocyte diameter correlated with insulin resistance. Following SG, both weight and insulin sensitivity improved markedly in all subjects. SAT adipocytes' size became smaller, showing increased lipolysis through perilipin 1 phosphorylation, decreased inflammation, and increased expression in browning/beiging markers. In summary, in adolescent girls with SO, both omVAT and SAT depots showed distinct cellular and transcriptomic profiles. Following weight loss, the SAT depot changed its cellular morphology and transcriptomic profiles into more favorable ones. These changes in the SAT depot may play a fundamental role in the resolution of insulin resistance.


Asunto(s)
Lipólisis/fisiología , Obesidad Mórbida/metabolismo , Obesidad Mórbida/cirugía , Epiplón/metabolismo , Grasa Subcutánea Abdominal/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/metabolismo , Adolescente , Femenino , Gastrectomía , Humanos , Transcriptoma , Adulto Joven
17.
J Exp Med ; 217(4)2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-31951251

RESUMEN

Experimental and clinical evidence suggests that tumor-associated macrophages (TAMs) play important roles in cancer progression. Here, we have characterized the ontogeny and function of TAM subsets in a mouse model of metastatic ovarian cancer that is representative for visceral peritoneal metastasis. We show that the omentum is a critical premetastatic niche for development of invasive disease in this model and define a unique subset of CD163+ Tim4+ resident omental macrophages responsible for metastatic spread of ovarian cancer cells. Transcriptomic analysis showed that resident CD163+ Tim4+ omental macrophages were phenotypically distinct and maintained their resident identity during tumor growth. Selective depletion of CD163+ Tim4+ macrophages in omentum using genetic and pharmacological tools prevented tumor progression and metastatic spread of disease. These studies describe a specific role for tissue-resident macrophages in the invasive progression of metastatic ovarian cancer. The molecular pathways of cross-talk between tissue-resident macrophages and disseminated cancer cells may represent new targets to prevent metastasis and disease recurrence.


Asunto(s)
Macrófagos/metabolismo , Epiplón/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/secundario , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transcriptoma
18.
Nat Commun ; 10(1): 5070, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31699980

RESUMEN

ß-Adrenergic receptor (ß-AR) signaling is a pathway controlling adaptive thermogenesis in brown or beige adipocytes. Here we investigate the biological roles of the transcription factor Foxp1 in brown/beige adipocyte differentiation and thermogenesis. Adipose-specific deletion of Foxp1 leads to an increase of brown adipose activity and browning program of white adipose tissues. The Foxp1-deficient mice show an augmented energy expenditure and are protected from diet-induced obesity and insulin resistance. Consistently, overexpression of Foxp1 in adipocytes impairs adaptive thermogenesis and promotes diet-induced obesity. A robust change in abundance of the ß3-adrenergic receptor (ß3-AR) is observed in brown/beige adipocytes from both lines of mice. Molecularly, Foxp1 directly represses ß3-AR transcription and regulates its desensitization behavior. Taken together, our findings reveal Foxp1 as a master transcriptional repressor of brown/beige adipocyte differentiation and thermogenesis, and provide an important clue for its targeting and treatment of obesity.


Asunto(s)
Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Adipogénesis/genética , Metabolismo Energético/genética , Factores de Transcripción Forkhead/genética , Receptores Adrenérgicos beta 3/genética , Proteínas Represoras/genética , Termogénesis/genética , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Ratones , Obesidad/genética , Obesidad/metabolismo , Epiplón/metabolismo , Feocromocitoma/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Proteínas Represoras/metabolismo
19.
Life Sci ; 239: 117039, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31704447

RESUMEN

AIMS: Obesity is a risk factor for endothelial dysfunction, the severity of which is likely to vary depending on extent and impact of adiposity on the vasculature. This study investigates the roles of cyclooxygenase isoforms and thromboxane receptor activities in the differential endothelial dilatory capacities of arteries derived from omental and subcutaneous adipose tissues in obesity. MAIN METHODS: Small arteries were isolated from omental and subcutaneous adipose tissues obtained from consented morbidly obese patients (n = 65, BMI 45 ±â€¯6 kg m-2 [Mean ±â€¯SD]) undergoing bariatric surgery. Relaxation to acetylcholine was studied by wire myography in the absence or presence of indomethacin (10 µM, cyclooxygenase inhibitor), FR122047 (1 µM, cyclooxygenase-1 inhibitor), Celecoxib (4 µM, cyclooxygenase-2 inhibitor), Nω-Nitro-L-arginine methyl ester (L-NAME, 100 µM, nitric oxide synthase inhibitor) or combination of apamin (0.5 µM) and charybdotoxin (0.1 µM) that together inhibit endothelium-derived hyperpolarizing factor (EDHF). Contractions to U46619 (thromboxane A2 mimetic) were also studied. KEY FINDINGS: Acetylcholine relaxation was significantly attenuated in omental compared with subcutaneous arteries from same patients (p < 0.01). Indomethacin (p < 0.01) and FR122047 (p < 0.001) but not Celecoxib significantly improved the omental arteriolar relaxation. Cyclooxygenase-1 mRNA and U46619 contractions were both increased in omental compared with subcutaneous arteries (p < 0.05). L-NAME comparably inhibited acetylcholine relaxation in both arteries, while apamin+charybdotoxin were less effective in omental compared with subcutaneous arteries. SIGNIFICANCE: The results show that the depot-specific reduction in endothelial dilatory capacity of omental compared with subcutaneous arteries in obesity is in large part due to altered cyclooxygenase-1 and enhanced thromboxane receptor activities, which cause EDHF deficiency.


Asunto(s)
Ciclooxigenasa 1/metabolismo , Arteria Gastroepiploica/efectos de los fármacos , Receptores de Tromboxanos/metabolismo , Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/metabolismo , Adulto , Apamina/farmacología , Arterias/efectos de los fármacos , Celecoxib/farmacología , Caribdotoxina/farmacología , Ciclooxigenasa 1/fisiología , Inhibidores de la Ciclooxigenasa/farmacología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Endotelio Vascular/efectos de los fármacos , Femenino , Arteria Gastroepiploica/metabolismo , Humanos , Indometacina/farmacología , Masculino , Persona de Mediana Edad , Relajación Muscular/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Obesidad Mórbida/metabolismo , Epiplón/irrigación sanguínea , Epiplón/metabolismo , Receptores de Tromboxanos/fisiología , Vasodilatación/efectos de los fármacos
20.
Indian J Gastroenterol ; 38(4): 303-309, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31643029

RESUMEN

BACKGROUND: Human ß defensins (hBD1 and hBD2) are cationic, cysteine-rich peptides and form an integral part of the mammalian innate immune system. hBD1 is constitutively expressed in epithelial cells, whereas hBD2 increases in response to bacterial infection. Human omentum is known for its anti-inflammatory properties and also possesses an antibacterial activity of its own. We hypothesized that antimicrobial peptides, ß defensins, may govern host defense mechanism in the microbe-rich environment of the peritoneal cavity. Therefore, we analyzed the expression of hBD1 and hBD2 in omentum tissue in vivo and also studied the antibacterial activity of omentum against common pathogens. METHODOLOGY: Omentum tissues were obtained from 30 patients (15 cases and 15 controls). Real-time polymerase chain reaction (PCR) was used to evaluate the mRNA expression of hBD1 and hBD2. Protein quantification was done using Western blotting technique. Antibacterial susceptibility was performed to check the antibacterial activity of omentum. RESULT: Significantly higher expression of hBD2 was observed in cases compared to controls at both the transcriptional and translational levels. In comparison with an array of antibiotics, activated omentum also showed antibacterial property even at lower concentration of its extract. CONCLUSION: Omentum directly responds to bacterial infection, which may be due to differential expression of hBD1 and hBD2 in human omental tissue. These peptides (hBD1 and hBD2) may be an ideal candidate for novel antibiotic class with a broad-spectrum activity.


Asunto(s)
Antibacterianos/metabolismo , Infecciones Bacterianas/metabolismo , Epiplón/metabolismo , beta-Defensinas/metabolismo , Estudios de Casos y Controles , Humanos , Epiplón/microbiología , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA