Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 258
Filtrar
1.
Cells ; 11(3)2022 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-35159303

RESUMO

B1 cells constitute a specialized subset of B cells, best characterized in mice, which is abundant in body cavities, including the peritoneal cavity. Through natural and antigen-induced antibody production, B1 cells participate in the early defense against bacteria. The G protein-coupled receptor 183 (GPR183), also known as Epstein-Barr virus-induced gene 2 (EBI2), is an oxysterol-activated chemotactic receptor that regulates migration of B cells. We investigated the role of GPR183 in B1 cells in the peritoneal cavity and omentum. B1 cells expressed GPR183 at the mRNA level and migrated towards the GPR183 ligand 7α,25-dihydroxycholesterol (7α,25-OHC). GPR183 knock-out (KO) mice had smaller omenta, but with normal numbers of B1 cells, whereas they had fewer B2 cells in the omentum and peritoneal cavity than wildtype (WT) mice. GPR183 was not responsible for B1 cell accumulation in the omentum in response to i.p. lipopolysaccharide (LPS)-injection, in spite of a massive increase in 7α,25-OHC levels. Lack of GPR183 also did not affect B1a- or B1b cell-specific antibody responses after vaccination. In conclusion, we found that GPR183 is non-essential for the accumulation and function of B1 cells in the omentum and peritoneal cavity, but that it influences the abundance of B2 cells in these compartments.


Assuntos
Subpopulações de Linfócitos B , Infecções por Vírus Epstein-Barr , Omento , Cavidade Peritoneal , Receptores Acoplados a Proteínas G , Animais , Subpopulações de Linfócitos B/citologia , Herpesvirus Humano 4 , Hidroxicolesteróis , Camundongos , Camundongos Knockout , Omento/citologia , Cavidade Peritoneal/citologia , Receptores Acoplados a Proteínas G/genética
2.
J Clin Endocrinol Metab ; 107(3): 755-775, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34669916

RESUMO

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.


Assuntos
Adipócitos/metabolismo , Gordura Intra-Abdominal/metabolismo , Obesidade Mórbida/metabolismo , Gordura Subcutânea Abdominal/metabolismo , Adulto , Cirurgia Bariátrica , Feminino , Redes Reguladoras de Genes , Humanos , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/patologia , Pessoa de Meia-Idade , Obesidade Mórbida/patologia , Obesidade Mórbida/cirurgia , Omento/citologia , Omento/metabolismo , Omento/patologia , Omento/cirurgia , Proteômica , Gordura Subcutânea Abdominal/citologia , Gordura Subcutânea Abdominal/patologia
3.
Surgery ; 171(2): 384-392, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34392978

RESUMO

BACKGROUND: Tissue engineering is an attractive alternative to conventional esophageal replacement techniques using intra-abdominal organs which are associated with a substantial morbidity. The objective was to evaluate the feasibility of esophageal replacement by an allogenic decellularized esophagus in a porcine model. Secondary objectives were to evaluate the benefit of decellularized esophagus recellularization with autologous bone marrow mesenchymal stromal cells and omental maturation of the decellularized esophagus. METHODS: Eighteen pigs divided into 4 experimental groups according to mesenchymal stromal cells recellularization and omental maturation underwent a 5-cm long circumferential replacement of the thoracic esophagus. Turbo green florescent protein labelling was used for in vivo mesenchymal stromal cells tracking. The graft area was covered by a stent for 3 months. Clinical and histologic outcomes were analyzed over a 6-month period. RESULTS: The median follow-up was 112 days [5; 205]. Two animals died during the first postoperative month, 2 experienced an anastomotic leakage, 13 experienced a graft area stenosis following stent migration of which 3 were sacrificed as initially planned after successful endoscopic treatment. The stent could be removed in 2 animals: the graft area showed a continuous mucosa without stenosis. After 3 months, the graft area showed a tissue specific regeneration with a mature epithelium and muscular cells. Clinical and histologic results were similar across experimental groups. CONCLUSION: Circumferential esophageal replacement by a decellularized esophagus was feasible and allowed tissue remodeling toward an esophageal phenotype. We could not demonstrate any benefit provided by the omental maturation of the decellularized esophagus nor its recellularization with mesenchymal stromal cells.


Assuntos
Esôfago/anatomia & histologia , Esôfago/cirurgia , Engenharia Tecidual/métodos , Tecidos Suporte , Animais , Estudos de Viabilidade , Feminino , Transplante de Células-Tronco Mesenquimais , Modelos Animais , Omento/citologia , Stents , Suínos , Transplante Autólogo
4.
J Exp Med ; 218(12)2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34714329

RESUMO

Two resident macrophage subsets reside in peritoneal fluid. Macrophages also reside within mesothelial membranes lining the peritoneal cavity, but they remain poorly characterized. Here, we identified two macrophage populations (LYVE1hi MHC IIlo-hi CX3CR1gfplo/- and LYVE1lo/- MHC IIhi CX3CR1gfphi subsets) in the mesenteric and parietal mesothelial linings of the peritoneum. These macrophages resembled LYVE1+ macrophages within surface membranes of numerous organs. Fate-mapping approaches and analysis of newborn mice showed that LYVE1hi macrophages predominantly originated from embryonic-derived progenitors and were controlled by CSF1 made by Wt1+ stromal cells. Their gene expression profile closely overlapped with ovarian tumor-associated macrophages previously described in the omentum. Indeed, syngeneic epithelial ovarian tumor growth was strongly reduced following in vivo ablation of LYVE1hi macrophages, including in mice that received omentectomy to dissociate the role from omental macrophages. These data reveal that the peritoneal compartment contains at least four resident macrophage populations and that LYVE1hi mesothelial macrophages drive tumor growth independently of the omentum.


Assuntos
Macrófagos Peritoneais/patologia , Omento/citologia , Neoplasias Ovarianas/patologia , Proteínas de Transporte Vesicular/metabolismo , Animais , Células Epiteliais/patologia , Feminino , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Omento/patologia , Omento/cirurgia , Peritônio/patologia , Células Estromais/metabolismo , Transcriptoma , Proteínas de Transporte Vesicular/genética , Proteínas WT1/genética , Proteínas WT1/metabolismo
5.
FASEB J ; 35(5): e21534, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33817830

RESUMO

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.


Assuntos
Adipócitos Bege/citologia , Adipócitos/citologia , Adipogenia , Polipeptídeo Inibidor Gástrico/farmacologia , Fármacos Gastrointestinais/farmacologia , Células-Tronco Mesenquimais/citologia , Omento/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipócitos Bege/efeitos dos fármacos , Adipócitos Bege/metabolismo , Diferenciação Celular , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Omento/efeitos dos fármacos , Omento/metabolismo , Transdução de Sinais
6.
Transplant Proc ; 52(10): 2977-2979, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32532558

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) may provide a novel clinical approach for acute kidney injury (AKI), which represents a severe health care condition. The human omentum is an important source of MSCs. We investigated the effects of human omental mesenchymal stem cells (HO-MSCs) after induction of ischemic AKI in a rat model. METHODS: The ischemic-reperfusion injury (IRI) was induced at reperfusion following a 45-minute clamping of renal vessels. Twenty animals were used in this study. Each rat was randomly assigned to 1 of 2 groups: G1 (control, n = 10; IRI infusion of phosphate buffer solution) or G2 (HO-MSCs, n = 10; IRI infusion of HO-MSCs). The infusions were performed in the parenchyma at reperfusion. Renal function at 1, 3, 5, and 7 days was assessed. At sacrifice, histologic samples were analyzed by light, and renal injury was graded. RESULTS: HO-MSCs induced an accelerated renal exocrine functional recovery, demonstrated by biochemical parameters and confirmed by histology showing that histopathological alterations associated with ischemic injury were less severe in cell-treated kidneys as compared with control groups (P < .05). The renal damage degree was significantly less in the animals of the HO-MSC group (P < .0001). CONCLUSIONS: These results suggest that HO-MSCs could be useful in the treatment of AKI in a rat model with possible potential implication in clinical setting.


Assuntos
Injúria Renal Aguda/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Traumatismo por Reperfusão/patologia , Animais , Modelos Animais de Doenças , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Omento/citologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica
7.
Br J Cancer ; 123(3): 459-470, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32439934

RESUMO

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.


Assuntos
Quimiocina CXCL2/urina , Técnicas de Cocultura/métodos , Omento/citologia , Neoplasias Peritoneais/secundário , Neoplasias Gástricas/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Linhagem Celular Tumoral , Quimiocina CXCL2/genética , Meios de Cultivo Condicionados/química , Feminino , Humanos , Camundongos , Camundongos SCID , Omento/metabolismo , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Regulação para Cima
8.
Cancer Res ; 80(8): 1748-1761, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32054768

RESUMO

Adipocytes are critical for ovarian cancer cells to home to the omentum, but the metabolic changes initiated by this interaction are unknown. To this end, we carried out unbiased mass spectrometry-based metabolomic and proteomic profiling of cancer cells cocultured with primary human omental adipocytes. Cancer cells underwent significant proteo-metabolomic alteration(s), typified by changes in the lipidome with corresponding upregulation of lipid metabolism proteins. FABP4, a lipid chaperone protein, was identified as the critical regulator of lipid responses in ovarian cancer cells cocultured with adipocytes. Subsequently, knockdown of FABP4 resulted in increased 5-hydroxymethylcytosine levels in the DNA, downregulation of gene signatures associated with ovarian cancer metastasis, and reduced clonogenic cancer cell survival. In addition, clustered regularly interspaced short palindromic repeats (CRISPR)-mediated knockout of FABP4 in high-grade serous ovarian cancer cells reduced metastatic tumor burden in mice. Consequently, a small-molecule inhibitor of FABP4 (BMS309403) not only significantly reduced tumor burden in a syngeneic orthotopic mouse model but also increased the sensitivity of cancer cells toward carboplatin both in vitro and in vivo. Taken together, these results show that targeting FABP4 in ovarian cancer cells can inhibit their ability to adapt and colonize lipid-rich tumor microenvironments, providing an opportunity for specific metabolic targeting of ovarian cancer metastasis. SIGNIFICANCE: Ovarian cancer metastatic progression can be restricted by targeting a critical regulator of lipid responses, FABP4.


Assuntos
Adipócitos/metabolismo , Resistencia a Medicamentos Antineoplásicos , Proteínas de Ligação a Ácido Graxo/metabolismo , Neoplasias Ovarianas/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/análise , Animais , Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacologia , Carboplatina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Técnicas de Cocultura , Metilação de DNA , Regulação para Baixo , Proteínas de Ligação a Ácido Graxo/antagonistas & inibidores , Proteínas de Ligação a Ácido Graxo/genética , Feminino , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Metabolismo dos Lipídeos , Lipidômica , Espectrometria de Massas , Metabolômica/métodos , Camundongos , Camundongos Nus , Metástase Neoplásica/prevenção & controle , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Omento/citologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Análise Serial de Proteínas , Proteômica/métodos , Pirazóis/farmacologia , Carga Tumoral/efeitos dos fármacos , Regulação para Cima
9.
Cell Signal ; 69: 109549, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31987780

RESUMO

BACKGROUND: Our previous study showed that human omental adipose-derived stem cells (ADSCs) promote ovarian cancer growth and metastasis. In this study, the role of autophagy in the ovarian cancer-promoting effects of omental ADSCs was further determined. METHODS: The growth and invasion of ovarian cancer cells were detected by CCK-8 and Transwell assays, respectively. The autophagy of ovarian cancer cells transfected with MRFP-GFP-LC3 adenoviral vectors was evaluated by confocal microscopy and western blot assay. Transfection of STAT3 siRNA was used to inhibit the expression of STAT3. RESULTS: Our results show that autophagy plays a vital role in ovarian cancer and is promoted by ADSCs. Specifically, we show that proliferation and invasion are correlated with autophagy induction by ADSCs in two ovarian cancer cell lines under hypoxic conditions. Mechanistically, ADSCs activate the STAT3 signalling pathway, thereby promoting autophagy. Knockdown of STAT3 expression using siRNA decreased hypoxia-induced autophagy and decreased the proliferation and metastasis of ovarian cancer cells. CONCLUSION: Taken together, our data indicate that STAT3-mediated autophagy induced by ADSCs promotes ovarian cancer growth and metastasis.


Assuntos
Autofagia , Células-Tronco Mesenquimais , Neoplasias Ovarianas , Fator de Transcrição STAT3/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Omento/citologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transdução de Sinais
10.
Adipocyte ; 8(1): 304-317, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31334678

RESUMO

Accumulating evidence highlights the importance of interactions between tumour cells and stromal cells for tumour initiation, progression, and metastasis. In tumours that contain adipocyte in their stroma, adipocytes contribute to modification of tumour microenvironment and affect metabolism of tumour and tumour progression by production of cytokines and adipokines from the lipids. The omentum and bone marrow (BM) are highly adipocyte-rich and are also common metastatic and primary tumour developmental sites. Omental adipocytes exhibit metabolic cross-talk, immune modulation, and angiogenesis. BM adipocytes secrete adipokines, and participate in solid tumour metastasis through regulation of the CCL2/CCR2 axis and metabolic interactions. BM adipocytes also contribute to the progression of hematopoietic neoplasms. Here, we here provide an overview of research progress on the cross-talks between omental/BM adipocytes and tumour cells, which may be pivotal modulators of tumour biology, thus highlighting novel therapeutic targets. Abbreviations: MCP-1, monocyte chemoattractant protein 1IL, interleukinSTAT3, signal transducer and activator of transcription 3FABP4, fatty acid binding protein 4PI3K/AKT, phosphoinositide 3-kinase/protein kinase BPPAR, peroxisome proliferator-activated receptorPUFA, polyunsaturated fatty acidTAM, tumour-associated macrophagesVEGF, vascular endothelial growth factorVEGFR, vascular endothelial growth factor receptorBM, bone marrowBMA, bone marrow adipocytesrBMA, regulated BMAcBMA, constitutive BMAUCP-1, uncoupling protein-1TNF-α, tumour necrosis factor-alphaRANKL, receptor activator of nuclear factor kappa-Β ligandVCAM-1, vascular cell adhesion molecule 1JAK2, Janus kinase 2CXCL (C-X-C motif) ligandPGE2, prostaglandin E2COX-2, cyclooxygenase-2CCL2, C-C motif chemokine ligand 2NF-κB, nuclear factor-kappa BMM, multiple myelomaALL, acute lymphoblastic leukemiaAML, acute myeloid leukemiaGDF15, growth differentiation factor 15AMPK, AMP-activated protein kinaseMAPK, mitogen-activated protein kinaseAPL, acute promyelocytic leukemiaCCR2, C-C motif chemokine receptor 2SDF-1α, stromal cell-derived factor-1 alphaFFA, free fatty acidsLPrA, leptin peptide receptor antagonistMCD, malonyl-CoA decarboxylase.


Assuntos
Adipócitos/metabolismo , Células da Medula Óssea/metabolismo , Carcinogênese/patologia , Metabolismo dos Lipídeos , Omento/metabolismo , Adipócitos/patologia , Adipocinas/metabolismo , Animais , Células da Medula Óssea/patologia , Carcinogênese/metabolismo , Humanos , Omento/citologia , Omento/patologia , Microambiente Tumoral
11.
Artif Organs ; 43(10): 1022-1027, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31013358

RESUMO

Permanent hypoparathyroidism is a severe clinical condition accompanied by low parathyroid hormone level. Conventional treatment requires lifelong medication, and daily drug usage has some side effects. To avoid this circumstance, transplantation is an alternative and curative option. Microencapsulation may be used as a transplantation approach particularly to evade immune response. In order to define treatment of permanent hypoparathyroidism, a 37-year-old female recipient who has permanent hypoparathyroidism was evaluated for 3 years. Routine tests, viral markers, and T and B lymphocyte cross-match tests were analyzed. In addition intradermal skin test was performed for ultrapure alginate. Microencapsulation of cultured parathyroid cells was performed with ultrapure alginate. Cell suspension was prepared and spheroids were generated with calcium chloride. Afterward, transplantation was performed with a laparoscopic approach in the omental tissue. The recipient was discharged from the hospital without complications. Serum calcium, parathyroid hormone (PTH), and phosphorus levels were observed throughout 1 year. During the follow-up period, no complications were observed. Serum calcium levels were increased significantly on day 10 and PTH levels were increased on day 25 as well. According to our knowledge, this is the first study where ultrapure alginate-based microencapsulated parathyroid cells were transplanted in the omental tissue. A significant increment of PTH levels was detected. Microencapsulated parathyroid cells showed the functionality of this technique for more than 1 year. This study showed that using ultrapure alginate-based microencapsulation without immunosuppression appears to be a promising technique.


Assuntos
Encapsulamento de Células/métodos , Células Imobilizadas/transplante , Hipoparatireoidismo/terapia , Glândulas Paratireoides/citologia , Glândulas Paratireoides/transplante , Adulto , Alginatos/química , Separação Celular , Células Cultivadas , Células Imobilizadas/citologia , Feminino , Humanos , Hipoparatireoidismo/cirurgia , Masculino , Omento/citologia , Omento/cirurgia
12.
Sci Rep ; 9(1): 3087, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30816281

RESUMO

Identification of adipose-specific genes has contributed to an understanding of mechanisms underlying adipocyte development and obesity. Herein, our analyses of the recent Genotype-Tissue Expression (GTEx) database revealed 38 adipose-specific/enhanced protein coding genes, among which 3 genes were novel adipose-specific, and 414 highly differentially expressed genes (DEGs) between subcutaneous and omental adipose depots. By integrative analyses of genome-wide association studies (GWASs), 14 adipose-specific/enhanced genes and 60 DEGs were found to be associated with obesity-related traits and diseases, consolidating evidence for contribution of these genes to the regional fat distribution and obesity phenotypes. In addition, expression of HOXC cluster was up-regulated in subcutaneous adipose tissue, and the majority of the HOXB cluster was expressed highly in omental adipose tissue, indicating differential expression patterns of HOX clusters in adipose depots. Our findings on the distinct gene expression profiles in adipose tissue and their relation to obesity provide an important foundation for future functional biological studies and therapeutic targets in obesity and associated diseases.


Assuntos
Gordura Abdominal/metabolismo , Adipócitos/metabolismo , Estudo de Associação Genômica Ampla , Obesidade/genética , Omento/metabolismo , Gordura Subcutânea/metabolismo , Gordura Abdominal/citologia , Adipócitos/citologia , Conjuntos de Dados como Assunto , Genes Homeobox , Loci Gênicos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Omento/citologia , Gordura Subcutânea/citologia , Transcriptoma
13.
Cell Cycle ; 18(3): 320-332, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30636489

RESUMO

Recent reports indicate that mesenchymal stem cells (MSCs) can fuse with cancer cells to promote cancer progression. Omental adipose-derived stromal cells (O-ASCs) are similar to MSCs, which could be recruited to the stroma in endometrial cancer. The aim of our study was to investigate whether O-ASCs can fuse with endometrial cancer cells to influence cancer cells biological characteristics. We isolated O-ASCs from patients with endometrial cancer. O-ASCs and endometrial cancer cells were labeled with different fluorescent tags and directly co-cultured in an Opera high-throughput spinning-disk confocal microscopy system to observe the processes involved in the fusion, division and migration of hybrid cells. Immunofluorescence and high-throughput imaging analyzes were performed to evaluate proteins related to epithelial-mesenchymal transition (EMT).We found O-ASCs could spontaneously fuse with endometrial cancer cells, including cytomembrane and nuclear fusion. After fusion, endometrial cancer cells assume an elongated and fibroblast-like appearance that exhibit mesenchymal phenotypes. The hybrid cells proliferated through bipolar and multipolar divisions and exhibited more rapid migratory speeds than were observed in the parental cells (P < 0.01), potentially because of their EMT-associated changes, including the down-regulation of E-cadherin and up-regulation of Vimentin. Our results collectively suggest that tumorigenic hybrids spontaneously formed between human O-ASCs and endometrial cancer cells, and that the resulting cells enhanced cancer mobility and heterogeneity by accelerated migration and undergoing multipolar divisions. These data provide a new avenue for investigating the roles of O-ASCs in endometrial cancer.


Assuntos
Tecido Adiposo/citologia , Movimento Celular , Neoplasias do Endométrio/fisiopatologia , Aneuploidia , Carcinogênese , Fusão Celular , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Transição Epitelial-Mesenquimal , Feminino , Humanos , Células Híbridas , Omento/citologia , Células Estromais/fisiologia
14.
Tissue Eng Part A ; 25(17-18): 1202-1212, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30648470

RESUMO

IMPACT STATEMENT: The methods developed in this study to manipulate pig tooth germ cells in vitro and in vivo provide a reference for studying whole-tooth regeneration and tooth development in large animals. Of importance, compared with conventional ectopic tooth regeneration, conducted in the omentum, subcutaneous tissues, or kidney capsule (among other locations) with low with immune reactivity in rodent models, this study achieved orthotopic regeneration and development of whole teeth in a large mammal, representing a large stride toward the realization of tooth regenerative therapy for humans with missing teeth.


Assuntos
Células Alógenas/citologia , Arcada Osseodentária/citologia , Regeneração/fisiologia , Dente/citologia , Células Alógenas/fisiologia , Animais , Técnicas de Cultura de Células/métodos , Células Cultivadas , Células Germinativas/citologia , Células Germinativas/fisiologia , Odontogênese/fisiologia , Omento/citologia , Omento/fisiologia , Tela Subcutânea/fisiologia , Suínos , Porco Miniatura , Técnicas de Cultura de Tecidos/métodos , Engenharia Tecidual , Dente/fisiologia
15.
Adv Mater ; 31(1): e1803895, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30406960

RESUMO

Despite incremental improvements in the field of tissue engineering, no technology is currently available for producing completely autologous implants where both the cells and the scaffolding material are generated from the patient, and thus do not provoke an immune response that may lead to implant rejection. Here, a new approach is introduced to efficiently engineer any tissue type, which its differentiation cues are known, from one small tissue biopsy. Pieces of omental tissues are extracted from patients and, while the cells are reprogrammed to become induced pluripotent stem cells, the extracellular matrix is processed into an immunologically matching, thermoresponsive hydrogel. Efficient cell differentiation within a large 3D hydrogel is reported, and, as a proof of concept, the generation of functional cardiac, cortical, spinal cord, and adipogenic tissue implants is demonstrated. This versatile bioengineering approach may assist to regenerate any tissue and organ with a minimal risk for immune rejection.


Assuntos
Hidrogéis/química , Próteses e Implantes , Animais , Diferenciação Celular , Reprogramação Celular , Células Endoteliais/citologia , Células Endoteliais/imunologia , Células Endoteliais/transplante , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/transplante , Omento/citologia , Omento/imunologia , Omento/metabolismo , Suínos , Engenharia Tecidual , Tecidos Suporte , Transplante Autólogo
16.
Artif Cells Nanomed Biotechnol ; 46(sup3): S1131-S1140, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30451550

RESUMO

The purpose of this study was to evaluate whether the prior implantation of a 3D-printed polycaprolactone (PCL) artificial trachea in the omentum is beneficial for revascularization of the scaffold and reduces associated complications in the reconstruction of a circumferential tracheal defect. Ten New Zealand rabbits were divided into 2 groups: (1) PCL-OC group (PCL scaffold cultured in omentum for 2 weeks before transplantation) and (2) PCL group. In the PCL-OC group, newly formed connective tissue completely covered the luminal surface of the scaffold with mild inflammation at 2 weeks postoperatively; a minor degree of stenosis was noted at 8 weeks postoperatively. The PCL group showed scaffold exposure without any tissue regeneration at 2 weeks postoperatively, and a moderate degree of luminal stenosis 6 weeks after implantation. Histology revealed highly organized regenerated tissue composed of ciliated respiratory epithelium, and submucosal layer in the PCL-OC group. Neo-cartilage regeneration was noted in part of the regenerated tissue. The PCL group demonstrated severe inflammation and an unorganized structure compared to that of the PCL-OC group. In vivo omentum culture of the tracheal scaffold before transplantation is beneficial for rapid re-epithelialization and revascularization of the scaffold. It also prevents postoperative luminal stenosis.


Assuntos
Órgãos Artificiais , Omento/metabolismo , Poliésteres/química , Impressão Tridimensional , Engenharia Tecidual , Tecidos Suporte/química , Traqueia/química , Animais , Cartilagem/citologia , Cartilagem/metabolismo , Omento/citologia , Coelhos
17.
Head Face Med ; 14(1): 19, 2018 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-30257689

RESUMO

BACKGROUND: Adult stem cells appear to be a promising subject for tissue engineering, representing an individual material for regeneration of aged and damaged cells. Especially adipose derived stromal cells (ADSC), which are easily to achieve, allow an encouraging perspective due to their capability of differentiating into miscellaneous cell types. Here we describe the in vitro formation of human subcutaneous, visceral and omental ADSC micromasses and compare their histological attributes while being cultivated on collagen membranes. METHODS: Subcutaneous, visceral and omental fat tissue derived cells were isolated and processed according to standard protocols. Positively stained cells for CD13, CD44 and CD90 were cultivated on agarose in order to study micromass formation using a special method of cell tracking. Stained paraffin-embedded micromasses were analysed morphologically before and after being plated on collagen membranes. RESULTS: The micromass formation process was similar in all three tissue types. Subcutaneous fat tissue derived micromasses turned out to develop a more homogeneous and compact shape than visceral and omental tissue. Nevertheless all micromasses adhered to collagen membranes with visible spreading of cells. The immune histochemical (IHC) staining of subcutaneous, visceral and omental ADSC micromasses shows a constant expression of CD13 and a decrease of CD44 and CD 90 expression within 28 days. After that period, omental fat cells don't show any expression of CD44. CONCLUSION: In conclusion micromass formation and cultivation of all analysed fat tissues can be achieved, subcutaneous cells appearing to be the best material for regenerative concepts.


Assuntos
Células-Tronco/citologia , Células Estromais/citologia , Gordura Subcutânea/citologia , Engenharia Tecidual/métodos , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Adulto , Células Cultivadas , Feminino , Humanos , Masculino , Omento/citologia , Medicina Regenerativa/métodos , Sensibilidade e Especificidade , Células-Tronco/metabolismo , Células Estromais/metabolismo , Gordura Subcutânea/metabolismo , Vísceras/citologia
18.
PLoS One ; 13(5): e0196844, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29723250

RESUMO

Transforming growth factor-ß1 (TGF-ß1) is a major mediator of peritoneal fibrosis and reportedly affects expression of the H3K4 methyltransferase, SET7/9. SET7/9-induced H3K4 mono-methylation (H3K4me1) critically activates transcription of fibrosis-related genes. In this study, we examined the effect of SET7/9 inhibition on peritoneal fibrosis in mice and in human peritoneal mesothelial cells (HPMCs). We also examined SET7/9 expression in nonadherent cells isolated from the effluent of peritoneal dialysis (PD) patients. Murine peritoneal fibrosis was induced by intraperitoneal injection of methylglyoxal (MGO) into male C57/BL6 mice over 21 days. Sinefungin, a SET7/9 inhibitor, was administered subcutaneously just before MGO injection (10 mg/kg). SET7/9 expression was elevated in both MGO-injected mice and nonadherent cells isolated from the effluent of PD patients. SET7/9 expression was positively correlated with dialysate/plasma ratio of creatinine in PD patients. Sinefungin was shown immunohistochemically to suppress expression of mesenchymal cells and collagen deposition, accompanied by decreased H3K4me1 levels. Peritoneal equilibration tests showed that sinefungin attenuated the urea nitrogen transport rate from plasma and the glucose absorption rate from the dialysate. In vitro, sinefungin suppressed TGF-ß1-induced expression of fibrotic markers and inhibited H3K4me1. These findings suggest that inhibiting the H3K4 methyltransferase SET7/9 ameliorates peritoneal fibrosis.


Assuntos
Adenosina/análogos & derivados , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Fibrose Peritoneal/prevenção & controle , Adenosina/farmacologia , Adenosina/uso terapêutico , Animais , Células Cultivadas , Colágeno Tipo I/biossíntese , Colágeno Tipo I/genética , Soluções para Diálise/farmacocinética , Epitélio , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacocinética , Código das Histonas/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/fisiologia , Humanos , Masculino , Metilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Nitrogênio/farmacocinética , Omento/citologia , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/induzido quimicamente , Fibrose Peritoneal/etiologia , Regiões Promotoras Genéticas/genética , Aldeído Pirúvico/toxicidade , Fator de Crescimento Transformador beta1/fisiologia
19.
Int J Obes (Lond) ; 42(5): 1051-1061, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29515208

RESUMO

BACKGROUND: Abdominal obesity is considered a major factor in the development of metabolic disorders. Glucagon-like peptide-1 (GLP-1) has been reported to have positive effects on improving body metabolism and to reducing insulin resistance. However, it remains less clear whether GLP-1 plays a role in the adipogenesis process of visceral fat. METHODS: Here, we analyzed the in vitro actions and probable mechanisms of Exendin-4, a GLP-1 receptor agonist, on human adipose-derived stromal cells (hADSCs) isolated from omentum. RESULTS: Our results demonstrated that Exendin-4 improved cell viability via promoting proliferation and inhibiting apoptosis in hADSCs isolated from omentum. Mechanistically, the activation of MAPK/ ERK1/2, Akt/GSK-3ß, and PKA/CREB pathways and downstream consequences induced are involved in the proliferative and anti-apoptotic roles of Exendin-4. More intriguingly, Exendin-4 could promote the differentiation of omental hADSCs. Underlying mechanisms of the differentiation of hADSCs are associated with the upregulation of the expression of pro-adipogenic genes and downregulation of the expression of anti-adipogenic genes. CONCLUSION: Our data demonstrate that Exendin-4 modifies adipogenesis of hADSCs isolated from omentum through multiple mechanisms, these effects could contribute to the protective actions of GLP-1 receptor agonist body metabolism and insulin sensitivity.


Assuntos
Adipogenia/efeitos dos fármacos , Tecido Adiposo/citologia , Fenômenos Fisiológicos Celulares/efeitos dos fármacos , Exenatida/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Omento/citologia , Adolescente , Adulto , Idoso , Células Cultivadas , Feminino , Peptídeo 1 Semelhante ao Glucagon/agonistas , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade , Adulto Jovem
20.
Artif Cells Nanomed Biotechnol ; 46(sup1): 885-895, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29446982

RESUMO

Current treatments of oesophageal diseases, such as carcinoma, congenital abnormality or trauma, require surgical intervention and oesophageal reconstruction with the stomach, jejunum or colon. However, serious side effects are possible with each treatment option. Despite tissue engineering promising to be an effective regenerative strategy, no functional solution currently exists for oesophageal reconstruction. Here, we developed an omentum-cultured oesophageal scaffold reinforced by a 3D-printed ring. The nano-structured scaffolds were wrapped into the omentum of rats and orthotopically transplanted for the repair of circumferential oesophageal defects two weeks later. The artificial oesophagus exhibited complete healing of the surgically created circumferential defects by the second week. The integration of the omentum-cultured oesophageal scaffold and the regenerative tissue remained intact. Macroscopically, there was no evidence of a fistula, perforation, abscess formation or surrounding soft-tissue necrosis. The omentum-cultured nano-structure scaffold reinforced by a 3D-printed ring is a more practical model with better vascularization for artificial neo-oesophagus reconstruction in a rat model.


Assuntos
Esôfago/citologia , Omento/citologia , Impressão Tridimensional , Engenharia Tecidual/métodos , Tecidos Suporte , Animais , Fenômenos Biomecânicos , Técnicas de Cultura de Células , Estudos de Viabilidade , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...