Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
Tipo de documento
País/Região como assunto
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(6)2022 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-35328799

RESUMO

BACKGROUND: Autism spectrum disorder (ASD) is a complex neurodevelopmental disease. To date, more than 1000 genes have been shown to be associated with ASD, and only a few of these genes account for more than 1% of autism cases. Klf7 is an important transcription factor of cell proliferation and differentiation in the nervous system, but whether klf7 is involved in autism is unclear. METHODS: We first performed ChIP-seq analysis of klf7 in N2A cells, then performed behavioral tests and RNA-seq in klf7+/- mice, and finally restored mice with adeno-associated virus (AAV)-mediated overexpression of klf7 in klf7+/- mice. RESULTS: Klf7 targeted genes are enriched with ASD genes, and 631 ASD risk genes are also differentially expressed in klf7+/- mice which exhibited the core symptoms of ASD. When klf7 levels were increased in the central nervous system (CNS) in klf7+/- adult mice, deficits in social interaction, repetitive behavior and majority of dysregulated ASD genes were rescued in the adults, suggesting transcriptional regulation. Moreover, knockdown of klf7 in human brain organoids caused dysregulation of 517 ASD risk genes, 344 of which were shared with klf7+/- mice, including some high-confidence ASD genes. CONCLUSIONS: Our findings highlight a klf7 regulation of ASD genes and provide new insights into the pathogenesis of ASD and promising targets for further research on mechanisms and treatments.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Animais , Transtorno do Espectro Autista/genética , Transtorno Autístico/complicações , Transtorno Autístico/genética , Diferenciação Celular , Regulação da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos
2.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L741-L747, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28213468

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive disease that causes unremitting deposition of extracellular matrix proteins, thus resulting in distortion of the pulmonary architecture and impaired gas exchange. Associated with high morbidity and mortality, IPF is generally refractory to current pharmacological therapies. Lefty A, a potent inhibitor of transforming growth factor-ß signaling, has been shown to have promising antifibrotic ability in vitro for the treatment of renal fibrosis and other potential organ fibroses. Here, we determined whether Lefty A can attenuate bleomycin (BLM)-induced pulmonary fibrosis in vivo based on a novel therapeutic strategy where human embryonic kidney 293 (HEK293) cells are genetically engineered with the Lefty A-associated GFP gene. The engineered HEK293 cells were encapsulated in alginate microcapsules and then subcutaneously implanted in ICR mice that had 1 wk earlier been intratracheally administered BLM to induce pulmonary fibrosis. The severity of fibrosis in lung tissue was assessed using pathological morphology and collagen expression to examine the effect of Lefty A released from the microencapsulated cells. The engineered HEK293 cells with Lefty A significantly reduced the expression of connective tissue growth factor and collagen type I mRNA, lessened the morphological fibrotic effects induced by BLM, and increased the expression of matrix metalloproteinase-9. This illustrates that engineered HEK293 cells with Lefty A can attenuate pulmonary fibrosis in vivo, thus providing a novel method to treat human pulmonary fibrotic disease and other organ fibroses.


Assuntos
Engenharia Celular , Composição de Medicamentos , Fatores de Determinação Direita-Esquerda/metabolismo , Fibrose Pulmonar/patologia , Fibrose Pulmonar/terapia , Animais , Colágeno/metabolismo , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Vetores Genéticos/metabolismo , Células HEK293 , Humanos , Imuno-Histoquímica , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos ICR , Microesferas , Retroviridae/metabolismo
3.
FEBS J ; 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38924469

RESUMO

The arrest of neural crest-derived sympathoadrenal neuroblast differentiation contributes to neuroblastoma formation, and overriding this blocked differentiation is a clear strategy for treating high-risk neuroblastoma. A better understanding of neuroblast or neuroblastoma differentiation is essential for developing new therapeutic approaches. It has been proposed that Krueppel-like factor 7 (KLF7) is a neuroblastoma super-enhancer-associated transcription factor gene. Moreover, KLF7 was found to be intensely active in postmitotic neuroblasts of the developing nervous system during embryogenesis. However, the role of KLF7 in the differentiation of neuroblast or neuroblastoma is unknown. Here, we find a strong association between high KLF7 expression and favorable clinical outcomes in neuroblastoma. KLF7 induces differentiation of neuroblastoma cells independently of the retinoic acid (RA) pathway and acts cooperatively with RA to induce neuroblastoma differentiation. KLF7 alters the GTPase activity and multiple differentiation-related genes by binding directly to the promoters of neuroblast differentiation-associated protein (AHNAK and AHNAK2) and glycerophosphodiester phosphodiesterase domain-containing protein 5 (GDPD5) and regulating their expression. Furthermore, we also observe that silencing KLF7 in neuroblastoma cells promotes the adrenergic-to-mesenchymal transition accompanied by changes in enhancer-mediated gene expression. Our results reveal that KLF7 is an inducer of neuroblast or neuroblastoma differentiation with prognostic significance and potential therapeutic value.

4.
Int J Biol Sci ; 20(1): 387-402, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38164176

RESUMO

Colon adenocarcinoma (COAD) is the most common malignancy of the digestive tract, which is characterized by a dismal prognosis. No effective treatment has been established presently, thus there is an urgent need to understand the mechanisms driving COAD progression in order to develop effective therapeutic approaches and enhance clinical outcomes. In this study, we found that KLF7 is overexpressed in COAD tissues and correlated with clinicopathological features of COAD. Both gain-of-function and loss-of-function experiments have unequivocally demonstrated that overexpression of KLF7 promotes the growth and metastasis of COAD in vitro and in vivo, while KLF7 knockdown attenuated these effects. Mechanistically, our findings reveal that KLF7 can specifically bind to the promoter region of PDGFB (TGGGTGGAG), thus promoting the transcription of PDGFB and increasing its secretion. Subsequently, secreted PDGFB facilitates the progression of COAD by activating MAPK/ERK, PI3K/AKT, and JAK/STAT3 signaling pathways through PDGFRß. Additionally, we found that sunitinib can block PDGFB signaling and inhibit COAD progression, offering a promising therapeutic strategy for COAD treatment.


Assuntos
Adenocarcinoma , Neoplasias do Colo , Humanos , Neoplasias do Colo/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Adenocarcinoma/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/genética , Becaplermina , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo
5.
Front Mol Neurosci ; 16: 1073723, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36741925

RESUMO

Primary familial brain calcification (PFBC) is a rare neurodegenerative and neuropsychiatric disorder characterized by bilateral symmetric intracranial calcification along the microvessels or inside neuronal cells in the basal ganglia, thalamus, and cerebellum. Slc20a2 homozygous (HO) knockout mice are the most commonly used model to simulate the brain calcification phenotype observed in human patients. However, the cellular and molecular mechanisms related to brain calcification, particularly at the early stage much prior to the emergence of brain calcification, remain largely unknown. In this study, we quantified the central nervous system (CNS)-infiltrating T-cells of different age groups of Slc20a2-HO and matched wild type mice and found CD45+CD3+ T-cells to be significantly increased in the brain parenchyma, even in the pre-calcification stage of 1-month-old -HO mice. The accumulation of the CD3+ T-cells appeared to be associated with the severity of brain calcification. Further immunophenotyping revealed that the two main subtypes that had increased in the brain were CD3+ CD4- CD8- and CD3+ CD4+ T-cells. The expression of endothelial cell (EC) adhesion molecules increased, while that of tight and adherents junction proteins decreased, providing the molecular precondition for T-cell recruitment to ECs and paracellular migration into the brain. The fusion of lymphocytes and EC membranes and transcellular migration of CD3-related gold particles were captured, suggesting enhancement of transcytosis in the brain ECs. Exogenous fluorescent tracers and endogenous IgG and albumin leakage also revealed an impairment of transcellular pathway in the ECs. FTY720 significantly alleviated brain calcification, probably by reducing T-cell infiltration, modulating neuroinflammation and ossification process, and enhancing the autophagy and phagocytosis of CNS-resident immune cells. This study clearly demonstrated CNS-infiltrating T-cells to be associated with the progression of brain calcification. Impairment of blood-brain barrier (BBB) permeability, which was closely related to T-cell invasion into the CNS, could be explained by the BBB alterations of an increase in the paracellular and transcellular pathways of brain ECs. FTY720 was found to be a potential drug to protect patients from PFBC-related lesions in the future.

6.
Nat Commun ; 14(1): 959, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36810848

RESUMO

The main hallmark of myocardial substrate metabolism in cardiac hypertrophy or heart failure is a shift from fatty acid oxidation to greater reliance on glycolysis. However, the close correlation between glycolysis and fatty acid oxidation and underlying mechanism by which causes cardiac pathological remodelling remain unclear. We confirm that KLF7 simultaneously targets the rate-limiting enzyme of glycolysis, phosphofructokinase-1, liver, and long-chain acyl-CoA dehydrogenase, a key enzyme for fatty acid oxidation. Cardiac-specific knockout and overexpression KLF7 induce adult concentric hypertrophy and infant eccentric hypertrophy by regulating glycolysis and fatty acid oxidation fluxes in male mice, respectively. Furthermore, cardiac-specific knockdown phosphofructokinase-1, liver or overexpression long-chain acyl-CoA dehydrogenase partially rescues the cardiac hypertrophy in adult male KLF7 deficient mice. Here we show that the KLF7/PFKL/ACADL axis is a critical regulatory mechanism and may provide insight into viable therapeutic concepts aimed at the modulation of cardiac metabolic balance in hypertrophied and failing heart.


Assuntos
Acil-CoA Desidrogenase de Cadeia Longa , Miocárdio , Animais , Masculino , Camundongos , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Cardiomegalia/patologia , Ácidos Graxos/metabolismo , Coração , Fatores de Transcrição Kruppel-Like/metabolismo , Miocárdio/metabolismo , Oxirredução , Acil-CoA Desidrogenase/metabolismo , Fosfofrutoquinases/metabolismo
7.
Cell Rep ; 41(8): 111644, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36417852

RESUMO

Oct4 is exclusively expressed in rodent inner cell mass (ICM) but silenced in its trophectoderm (TE). However, for many non-rodent animals, including pig, cattle, rabbit, goat, and human, OCT4 has a remarkable expression in early TE. This study, applying pig as the main research model, proves that OCT4 expression in TE is supported by a unique GATA motif in the OCT4 upstream conserved regulatory region, and GATA4 is responsible for its activation. Moreover, OCT4 acts as a specific regulator of a narrow range of genes (including BCL2A1 and HNRNP2AB1) that are essential for the first wave of rapid proliferation in early TE. This study describes the regulatory mechanism to direct the OCT4 expression and its significance in TE of porcine preimplantation embryo.


Assuntos
Blastocisto , Roedores , Humanos , Suínos , Animais , Bovinos , Coelhos
8.
ACS Appl Bio Mater ; 4(1): 441-450, 2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35014295

RESUMO

Cancer stem cells (CSCs) are self-renewing and constitute the primary cause of cancer relapse post-cancer therapy. The CSC niche is composed of various nonmalignant stromal cells that support CSCs' survival during cancer chemoradiotherapy. Understanding the cross-talk between CSCs and stromal cells could pave the way for developing therapeutic strategies to eradicate CSCs. Traditionally, CSC research has been relying on animal models, which can give rise to complications and poor translation in clinical practice. An efficient model to co-culture CSCs and stromal cells is urgently needed. Hence, we leveraged our expertise in enriching CSCs from in vitro cell lines with a 3D alginate-based platform, as reported previously. We established a 3D co-culture system that allowed us to study the interactions between stromal cells and CSCs over an extended period. We showed that the self-renewal capacity and stemness of CSCs were significantly enhanced when co-cultured with 3D cultured human umbilical vein endothelial cells (HUVECs) or a human monocyte cell line (THP1). Strikingly, the expression of MDR1 in 3D co-cultured CSCs was upregulated, leading to enhanced chemotoxic drug tolerance. We suggest that our in vitro co-culture model can impact CSC research and clinical practice when the goal is to develop therapeutics that target and eradicate CSCs by targeting stromal cells.


Assuntos
Alginatos/química , Técnicas de Cocultura/métodos , Células-Tronco Neoplásicas/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Técnicas de Cultura de Células em Três Dimensões , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrogéis/química , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Células-Tronco Neoplásicas/citologia , Comunicação Parácrina , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Regulação para Cima
9.
Biofabrication ; 13(3)2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33836517

RESUMO

In vitrocancer models that can largely mimic thein vivomicroenvironment are crucial for conducting more accurate research. Models of three-dimensional (3D) culture that can mimic some aspects of cancer microenvironment or cancer biopsies that can adequately represent tumor heterogeneity are intensely used currently. Those models still lack the dynamic stress stimuli in gastric carcinoma exposed to stomach peristalsisin vivo. This study leveraged a lab-developed four-dimensional (4D) culture model by a magnetic responsive alginate-based hydrogel to rotating magnets that can mimic stress stimuli in gastric cancer (GC). We used the 4D model to culture human GC cell line AGS and SGC7901, cells at the primary and metastasis stage. We revealed the 4D model altered the cancer cell growth kinetics mechanistically by alteringPCNAandp53expression compared to the 3D culture that lacks stress stimuli. We found the 4D model altered the cancer spheroids stemness as evidenced by enhanced cancer stem cells (CD44) marker expression in AGS spheroids but the expression was dampened in SGC7901 cells. We examined the multi-drug resistance (MDR1) marker expression and found the 4D model dampened the MDR1 expression in SGC7901 cell spheroids, but not in spheroids of AGS cells. Such a model provides the stomach peristalsis mimic and is promising for conducting basic or translational GC-associated research, drug screening, and culturing patient gastric biopsies to tailor the therapeutic strategies in precision medicine.


Assuntos
Técnicas de Cultura de Células , Esferoides Celulares , Neoplasias Gástricas , Linhagem Celular Tumoral , Humanos , Peristaltismo , Microambiente Tumoral
10.
Theranostics ; 11(7): 3196-3212, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33537082

RESUMO

Resistance to chemotherapy is a long-standing problem in the management of cancer, and cancer stem cells are regarded as the main source of this resistance. This study aimed to investigate metallothionein (MT)-1G involvement in the regulation of cancer stemness and provide a strategy to overcome chemoresistance in pancreatic ductal adenocarcinoma (PDAC). Methods: MT1G was identified as a critical factor related with gemcitabine resistance in PDAC cells by mRNA microarray. Its effects on PDAC stemness were evaluated through sphere formation and tumorigenicity. LC-MS/MS analysis of conditional medium revealed that activin A, a NF-κB target, was a major protein secreted from gemcitabine resistant PDAC cells. Both loss-of-function and gain-of-function approaches were used to validate that MT1G inhibited NF-κB-activin A pathway. Orthotopic pancreatic tumor model was employed to explore the effects on gemcitabine resistance with recombinant follistatin to block activin A. Results: Downregulation of MT1G due to hypermethylation of its promoter is related with pancreatic cancer stemness. Secretome analysis revealed that activin A, a NF-κB target, was highly secreted by drug resistant cells. It promotes pancreatic cancer stemness in Smad4-dependent or independent manners. Mechanistically, MT1G negatively regulates NF-κB signaling and promotes the degradation of NF-κB p65 subunit by enhancing the expression of E3 ligase TRAF7. Blockade of activin A signaling with follistatin could overcome gemcitabine resistance. Conclusions: MT1G suppresses PDAC stemness by limiting activin A secretion via NF-κB inhibition. The blockade of the activin A signaling with follistatin may provide a promising therapeutic strategy for overcoming gemcitabine resistance in PDAC.


Assuntos
Ativinas/metabolismo , Metalotioneína/metabolismo , Células-Tronco Neoplásicas/metabolismo , Animais , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , China , Cromatografia Líquida , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Metalotioneína/genética , Camundongos Endogâmicos C57BL , Camundongos Nus , NF-kappa B/metabolismo , Células-Tronco Neoplásicas/fisiologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Transdução de Sinais/efeitos dos fármacos , Espectrometria de Massas em Tandem , Fator de Transcrição RelA/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
11.
Oncogene ; 40(35): 5427-5440, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34282273

RESUMO

Hepatocellular carcinoma (HCC) is an extremely metastatic tumor. Sialic acids (SAs) are associated with cancer development and metastasis. NEU4 is a sialidase that removes SAs from glycoconjugates, while the function of the NEU4 in HCC has not been clearly explored. In our research, we found the NEU4 expression was significantly down-regulated in HCC tissues, which was correlated with high grades and poor outcomes of HCC. The NEU4 expression could be regulated by histone acetylation. In the functional analysis of NEU4, the cell motility was inhibited when NEU4 was overexpressed, and restored when NEU4 expression was down-regulated. Similarly, NEU4 over-expressed HCC cells showed less metastasis in athymic nude mice. Further study revealed that NEU4 could inhibit cell migration by enzymatic decomposition of SAs. Our results verified a NEU4 active site (NEU4E235) and overexpressing inactivates NEU4E235A that weakens the inhibition ability to cell migration. Further, 70 kinds of specific interacting proteins of NEU4 including CD44 were identified through mass spectrum. Moreover, the α2,3-linked SAs on CD44 were decreased and the hyaluronic acid (HA) binding ability was increased when NEU4 over-expressed or activated. Additionally, the mutation of CD44 with six N-glycosylation sites showed less sensibility to NEU4 on cell migration compared with wild-type CD44. In summary, our results revealed the mechanism of low expression of NEU4 in HCC and its inhibitory effect on cell migration by removal of SAs on CD44, which may provide new treatment strategies to control the motility and metastasis of HCC.


Assuntos
Carcinoma Hepatocelular , Ácidos Siálicos , Animais , Receptores de Hialuronatos , Neoplasias Hepáticas , Camundongos , Mutação , Processamento de Proteína Pós-Traducional
12.
Cell Prolif ; 53(9): e12875, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32761833

RESUMO

OBJECTIVES: R-loop is a three-stranded nucleic acid structure of RNA/DNA hybrid, which occurs naturally during transcription, and more R-loop accumulation can trigger serious DNA damage. There has been increasing attention to the issue of R-loop accumulation acted as a target for cancer therapy. However, the regulation of R-loop-associated proteins is poorly explored. MATERIAL AND METHOD: Quantitative real-time PCR and Western blot were used to measure the expression of C1orf109 in cell lines. In addition, C1orf109L (C1orf109 longest isoform) protein binding partner was identified and validated using immunoprecipitation-mass spectrometric (IP-MS) and immunoprecipitation assays. DNA-RNA immunoprecipitation (DR-IP) and immunofluorescence determined the C1orf109L location on R-loop. R-loop accumulation was determined by immunofluorescence. Cell cycle was determined by flow cytometry. Finally, time-lapse assay and cell counting were conducted to determined cell survival in response to camptothecin (CPT). RESULTS: We found that C1orf109L could mediate cell cycle arrest in the G2/M phase and DNA damage depended on R-loop accumulation. Meanwhile, C1orf109L could bind with DHX9 to trigger R-loop accumulation. And C1orf109L was competitive with PARP1 binding to DHX9, which would block the function of DHX9-PARP1 to prevent the R-loop accumulation. Furthermore, C1orf109L could enhance the chemosensitivity of CPT, a chemotherapeutic drug capable of promoting R-loop formation. CONCLUSIONS: Our data demonstrate that C1orf109L triggers R-loop accumulation and DNA damage to arrest cell cycle.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , RNA Helicases DEAD-box/metabolismo , Dano ao DNA/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Ligação Proteica/efeitos dos fármacos , Estruturas R-Loop/efeitos dos fármacos
13.
Aging (Albany NY) ; 12(7): 6415-6435, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32289749

RESUMO

The incidence of atherosclerosis (AS), a major contributor to cardiovascular disease, is steadily rising along with an increasingly older population worldwide. Pyroptosis, a form of inflammatory programmed cell death, determines the release of pro-inflammatory mediators by endothelial cells, smooth muscle cells, and atheroma-associated macrophages and foam cells, thereby playing a critical role in AS progression. Canonical pyroptosis is mediated by inflammasome formation, activation of caspase-1, and maturation and release of proinflammatory cytokines. Electrical stimulation (ES) is a noninvasive, safe therapy that has been shown to alleviate symptoms in several health conditions. Here, we investigated the anti-inflammatory and anti-pyroptotic effects of ES in human THP-1 macrophages treated with the dipeptidyl peptidase inhibitor Val-boroPro (VbP). We found that ES downregulated NOD-like receptor family protein 3 (NLRP3) inflammasome, ASC, and caspase-1 expression and abrogated the release of Interleukin-1ß (IL-1ß) and Interleukin-18 (IL-18), indicating effective pyroptosis inhibition. These changes were paralleled by a reduction in reactive oxygen species (ROS) production, reversal of VbP-induced sirtuin3 (Sirt3) downregulation, deacetylation of ATG5, and induction of autophagy. These findings suggest that ES may be a viable strategy to counteract pyroptosis-mediated inflammation in AS by raising Sirt3 to promote autophagy and inhibit ROS generation.


Assuntos
Aterosclerose , Dipeptidil Peptidases e Tripeptidil Peptidases/antagonistas & inibidores , Estimulação Elétrica/métodos , Inflamassomos/metabolismo , Macrófagos , Sirtuína 3/metabolismo , Aterosclerose/imunologia , Aterosclerose/metabolismo , Ácidos Borônicos/farmacologia , Caspase 1/metabolismo , Dipeptídeos/farmacologia , Humanos , Interleucina-1beta/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/efeitos dos fármacos , Piroptose/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Células THP-1
14.
J Mater Chem B ; 7(14): 2247-2251, 2019 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32254673

RESUMO

Photothermal therapy (PTT) has emerged as one of the promising methodologies for the treatment of cancer, and ideal photothermal agents need to be biodegradable and have strong optical absorbance in the near-infrared (NIR) optical window. Here, we report a new phthalocyanine molecule, 4OCSPC, which expands the absorbance edge to 850 nm. Under 808 nm NIR laser irradiation, 4OCSPC polymeric micelles showed robust photostability and a high photothermal conversion of 47.0%. Also, the 4OCSPC polymeric micelles exhibit a high in vivo PTT efficacy against 4T1 tumors in mice.


Assuntos
Hipertermia Induzida/métodos , Indóis/uso terapêutico , Neoplasias/terapia , Fototerapia/métodos , Polímeros/uso terapêutico , Radiossensibilizantes/uso terapêutico , Animais , Células HeLa , Humanos , Raios Infravermelhos/uso terapêutico , Isoindóis , Camundongos , Micelas , Técnicas Fotoacústicas/métodos
15.
Oncogene ; 38(22): 4297-4309, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30700830

RESUMO

The naked mole rat (nmr) is cancer resistant due to the abundant production of extremely high-molecular-weight hyaluronan (EHMW-HA). However, whether EHMW-HA has similar anti-cancer effects in mice and humans remains to be determined. The present study used breast cancer cells to clarify the effect of EHMW-HA on breast cancer. First, the overexpression of nmrHas2 in 4T1 and BT549 cell lines in both two-dimensional (2D) and three-dimensional (3D) models to mimic tumor microenvironment was established. The 4T1/BT549-nmrHas2 cells could secrete EHMW-HA (with a molecular weight of up to 6 MDa), which was similar to that found in the naked mole rat. Second, EHMW-HA altering tumor microenvironment in both 2D monolayers and 3D spheroids significantly enhanced apoptosis, inhibiting the proliferation of 4T1 and BT549 cells. The prominent anticancer effects of EHMW-HA on the cancer-cell apoptosis phenotype were further confirmed by inhibiting tumor formation in nude mice. Finally, EHMW-HA significantly induced higher p53 protein expression, which enhanced pro-apoptotic proteins p21 and Bax in breast cancer cells; this is in contrast with the triggering of hypersensitivity of the naked mole rat cells to early contact inhibition (ECI). These results have important implications for the design of therapeutic approaches based on the application of EHMW-HA.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Ácido Hialurônico/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Nus , Ratos-Toupeira , Ratos , Proteína Supressora de Tumor p53/metabolismo
16.
Biosens Bioelectron ; 141: 111481, 2019 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-31260904

RESUMO

Three-dimensional (3D) cell cultures developed with living cells and scaffolds have demonstrated outstanding potential for tissue engineering and regenerative medicine applications. However, no suitable tools are available to monitor dynamically variable cell behavior in such a complex microenvironment. In particular, simultaneously assessing cell behavior, cell secretion, and the general state of a 3D culture system is of a really challenging task. This paper presents our development of a dual-transduction-integrated biosensing system that assesses electrical impedance in conjunction with imaging techniques to simultaneously investigate the 3D cell-culture for bone regeneration. First, we created models to mimic the dynamic deposition of the extracellular matrix (ECM) in 3D culture, which underwent osteogenesis by incorporating different amounts of bone-ECM components (collagen, hydroxyapatite [HAp], and hyaluronic acid [HA]) into alginate-based hydrogels. The formed models were investigated by means of electrical impedance spectroscopy (EIS), with the results showing that the impedances increased linearly with collagen and hyaluronan, but changed in a more complex manner with HAp. Thereafter, we created two models that consisted of primary osteoblast cells (OBs), which expressed the enhanced green fluorescent protein (EGFP), and 4T1 cells, which secreted the EGFP-HA, in the alginate hydrogel. We found the capacitance (associated with impedance and measured by EIS) increased with the increases in initial embedded OBs, and also confirmed the cell proliferation over 3 days with the EGFP signal as monitored by the fluorescent imaging component in our system. Interestingly, the change in capacitance is found to be associated with OB migration following stimulation. Also, we show higher capacitance in 4T1 cells that secret HA when compared to control 4T1 cells after a 3-day culture. Taken together, we demonstrate that our biosensing system is able to investigate the dynamic process of 3D culture in a non-invasive and real-time manner.


Assuntos
Técnicas Biossensoriais/instrumentação , Regeneração Óssea , Técnicas de Cultura de Células/instrumentação , Animais , Linhagem Celular , Células Cultivadas , Colágeno/metabolismo , Durapatita/metabolismo , Impedância Elétrica , Desenho de Equipamento , Matriz Extracelular/metabolismo , Ácido Hialurônico/metabolismo , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Alicerces Teciduais/química , Transdutores
17.
Adv Healthc Mater ; 7(12): e1800080, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29719134

RESUMO

Transplanted stem cells constitute a new therapeutic strategy for the treatment of neurological disorders. Emerging evidence indicates that a negative microenvironment, particularly one characterized by the acute inflammation/immune response caused by physical injuries or transplanted stem cells, severely impacts the survival of transplanted stem cells. In this study, to avoid the influence of the increased inflammation following physical injuries, an intelligent, double-layer, alginate hydrogel system is designed. This system fosters the matrix metalloproeinases (MMP) secreted by transplanted stem cell reactions with MMP peptide grafted on the inner layer and destroys the structure of the inner hydrogel layer during the inflammatory storm. Meanwhile, the optimum concentration of the arginine-glycine-aspartate (RGD) peptide is also immobilized to the inner hydrogels to obtain more stem cells before arriving to the outer hydrogel layer. It is found that blocking Cripto-1, which promotes embryonic stem cell differentiation to dopamine neurons, also accelerates this process in neural stem cells. More interesting is the fact that neural stem cell differentiation can be conducted in astrocyte-differentiation medium without other treatments. In addition, the system can be adjusted according to the different parameters of transplanted stem cells and can expand on the clinical application of stem cells in the treatment of this neurological disorder.


Assuntos
Células Imobilizadas/transplante , Hidrogéis/química , Células-Tronco Neurais/transplante , Doenças Neurodegenerativas/terapia , Oligopeptídeos/química , Transplante de Células-Tronco/métodos , Células Alógenas/metabolismo , Animais , Células Imobilizadas/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Doenças Neurodegenerativas/metabolismo , Transplante de Células-Tronco/instrumentação
18.
Poult Sci ; 96(1): 200-210, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27587730

RESUMO

Gga-miR-21 is abundantly expressed in chicken pre-adipocytes, but its role is unclear. The present study investigated the role of gga-miR-21 in chicken pre-adipocyte proliferation. Cell proliferation assay and gene expression analysis of proliferating cell nuclear antigen (PCNA) showed that the gga-miR-21 mimic inhibited pre-adipocyte proliferation. In contrast, the gga-miR-21 inhibitor enhanced pre-adipocyte proliferation. The subsequent investigation identified Kruppel-like factor 5 (KLF5) mRNA as a target of gga-miR-21. The gga-miR-21 mimic inhibited KLF5 3'UTR reporter activity and decreased endogenous KLF5 expression in primary pre-adipocytes. KLF5 knockdown using RNAi had a similar effect to that of the gga-miR-21 mimic on cell proliferation. The promoting effect of the gga-miR-21 inhibitor on pre-adipocyte proliferation was partially attenuated by KLF5 knockdown. Taken together, our results demonstrated that miR-21 inhibits chicken pre-adipocyte proliferation, at least in part, by targeting KLF5.


Assuntos
Adipócitos/citologia , Proteínas Aviárias/genética , Galinhas/fisiologia , Fatores de Transcrição Kruppel-Like/genética , MicroRNAs/metabolismo , Animais , Proteínas Aviárias/metabolismo , Sequência de Bases , Proliferação de Células , Galinhas/genética , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , MicroRNAs/genética
19.
Sci Rep ; 7(1): 15852, 2017 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-29158522

RESUMO

The miR-17-92 cluster has been well studied in mammals but less extensively studied in birds. Here, we demonstrated that miR-17-92 cluster overexpression promoted the proliferation of DF1 cells and immortalized chicken preadipocytes (ICPA-1), and miR-17-5p and miR-20a, members of the miR-17-92 cluster, targeted MAP3K2. Further analysis showed that MAP3K2 overexpression reduced the proliferation of DF1 and ICPA-1 cells and attenuated the promotive effect of the miR-17-92 cluster on cell proliferation. Downstream gene expression analysis of the MAPK signalling pathway showed that MAP3K2 overexpression decreased c-Myc expression; in contrast, MAP3K2 knockdown using RNA interference and miR-17-92 cluster overexpression increased c-Myc expression. Furthermore, c-Myc overexpression promoted miR-17-92 cluster expression and DF1 cell proliferation. Taken together, these data indicated that miR-17-92 promotes chicken cell proliferation at least in part by the upregulation of c-Myc via targeting MAP3K2, and the miR-17-92 cluster, c-Myc and E2F1 form a complex regulatory network in chicken cell proliferation.


Assuntos
Proliferação de Células/genética , MAP Quinase Quinase Quinase 2/genética , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Apoptose/genética , Movimento Celular/genética , Galinhas/genética , Fator de Transcrição E2F1/genética , Regulação da Expressão Gênica/genética , Humanos , Interferência de RNA , Transdução de Sinais/genética
20.
ACS Appl Mater Interfaces ; 9(11): 9327-9338, 2017 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-28240531

RESUMO

Hyaluronan (HA), a polymer with various molecular weights (MW) found in tumor microenvironments, is associated with malignant progression of breast cancer. Reducing the amount of high-MW HA in the microenvironment by hyaluronidase is a promising approach for breast cancer treatment. However, whether the generation of HA fragments negatively affects breast cancer cells remains to be determined. Furthermore, HA forms three-dimensional (3D) networks by cross-linking with other extracellular molecules to function. Therefore, a model mimicking the cross-linked HA network is required to determine the effect of HA fragments on breast cancer cells. To clarify the differential roles of low (HA35) versus high (HA117) MW HA on cancer cell phenotype, a 3D culture system was set up by covalently cross-linking HA with alginate and investigating the behavior of 4T-1 and SKBR3 breast cancer cells alongside a two-dimensional (2D) control. The results show the invasion and migration abilities of 4T-1 and SKBR3 cells are significantly enhanced by the presence of HA35 but inhibited by HA117 in both 2D monolayers and 3D spheroids. The differential effects of HA35 and HA117 on cancer cell epithelial-mesenchymal transition (EMT) phenotype were further confirmed in terms of differential regulation of E-cadherin and vimentin as important EMT markers at both the cellular and mRNA levels. Additional experiments show the CD44-Twist signaling pathway might be involved in the differential effects of HA35 and HA117. These results have important implications with respect to understanding the role of HA in breast cancer development and for the design of therapeutic approaches based on the eradication of HA with hyaluronidase.


Assuntos
Ácido Hialurônico/química , Neoplasias da Mama , Linhagem Celular Tumoral , Movimento Celular , Humanos , Receptores de Hialuronatos , Hialuronoglucosaminidase , Peso Molecular , Proteínas de Neoplasias , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA