Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
J Mol Histol ; 52(4): 635-642, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34076833

RESUMO

Endothelial dysfunction is considered as the main hallmark of Preeclampsia (PE). Despite the unknown pathogenesis of PE, different possible causes have been suggested in various studies. In this review, we first studied the Leukemia inhibitory factor (LIF) role in the related pathways to the PE pathogenesis, such as inflammation, endothelial dysfunction and hypertension. LIF can increase the expression of ICAM-1 and VCAM-1 via the JAK/STAT3 pathway, thereby inducing inflammatory responses and endothelial dysfunction. It can also be involved in the vascular vasoconstriction and hypertension by reducing the nitric oxide (NO) synthesis. Identifying the link between LIF and pathways associated with PE pathogenesis could be effective to achieve an effective PE treatment in the future.


Assuntos
Fator Inibidor de Leucemia/fisiologia , Pré-Eclâmpsia/etiologia , Transdução de Sinais/fisiologia , Feminino , Humanos , Biologia Molecular , Pré-Eclâmpsia/fisiopatologia , Gravidez
2.
Gynecol Endocrinol ; 37(8): 725-729, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33974468

RESUMO

AIM: Study the effect of low-dose aspirin on the endometrial receptivity in endometriosis rat models. MATERIALS AND METHODS: This study is to explore the expressions of progesterone receptor and LIF among three groups of endometriosis rat models: control group (n = 12), EMs group (n = 15), and aspirin group (n = 17). The expressions of progesterone receptor (PR), PRA, PRB, and leukemia inhibitory factor receptor (LIFR) in eutopic endometrium were determined using immunohistochemistry technology, western blot, and qRT-PCR. The levels of LIF in eutopic endometrium and serum were detected by western blot, qRT-PCR, and ELISA. RESULTS: The expressions of PR, PRA, and PRB protein were significantly increased in the eutopic endometrium after low-dose aspirin treatment, and the level of PRB mRNA was also increased while the ratio of PRA/PRB mRNA was decreased in the eutopic endometrium. The levels of LIF in eutopic endometrium and serum were increased compared with the untreated endometriosis rats. However, the expression of LIFR was not statistically different among the three groups. CONCLUSIONS: The results suggest that the low-dose aspirin treatment could downregulate progesterone resistance and increase the expression of LIF of endometriosis rats during the implantation window, which could improve endometrial receptivity and enhance the pregnant rate of endometriosis. It may provide a potential treatment method for endometriosis-related infertility.


Assuntos
Aspirina/administração & dosagem , Implantação do Embrião/fisiologia , Endometriose/tratamento farmacológico , Fator Inibidor de Leucemia/efeitos dos fármacos , Progesterona/fisiologia , Receptores de Progesterona/análise , Animais , Endometriose/complicações , Endometriose/metabolismo , Endométrio/química , Feminino , Infertilidade Feminina/tratamento farmacológico , Infertilidade Feminina/etiologia , Fator Inibidor de Leucemia/análise , Fator Inibidor de Leucemia/fisiologia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/análise , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/fisiologia , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Progesterona/efeitos dos fármacos
3.
Arch Immunol Ther Exp (Warsz) ; 69(1): 2, 2021 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-33630157

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive, treatment-resistant cancer. Five-year survival rate is about 9%, one of the lowest among all solid tumors. Such a poor outcome is partly due to the limited knowledge of tumor biology, and the resulting lack of effective treatment options and robust predictive biomarkers. The leukemia inhibitory factor (LIF) has recently emerged as a potential biomarker and therapeutic target for PDAC. Accumulating evidence has suggested that LIF plays a role in supporting cancer evolution as a regulator of cell differentiation, renewal and survival. Interestingly, it can be detected in the serum of PDAC patients at higher concentrations than healthy individuals, this supporting its potential value as diagnostic biomarker. Furthermore, preliminary data indicate that testing for LIF serum concentration or tissue expression may help with treatment response monitoring and prognostication. Finally, studies in PDAC mouse models have also shown that LIF may be a valuable therapeutic target, and first-in-human clinical trial is currently ongoing. This article aims to review the available data on the role of LIF in PDAC promotion, and to discuss the evidence supporting its potential role as a biomarker and target of effective anti-cancer therapy in this setting.


Assuntos
Carcinoma Ductal Pancreático/patologia , Fator Inibidor de Leucemia/fisiologia , Neoplasias Pancreáticas/patologia , Biomarcadores Tumorais/análise , Fibroblastos Associados a Câncer/fisiologia , Carcinoma Ductal Pancreático/etiologia , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/terapia , Resistencia a Medicamentos Antineoplásicos , Humanos , Tolerância Imunológica , Fator Inibidor de Leucemia/análise , Invasividade Neoplásica , Células-Tronco Neoplásicas/fisiologia , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/terapia , Microambiente Tumoral
4.
Stem Cell Reports ; 16(3): 582-596, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33636115

RESUMO

Naive pluripotency can be maintained in medium with two inhibitors plus leukemia inhibitory factor (2i/LIF) supplementation, which primarily affects canonical WNT, FGF/ERK, and JAK/STAT3 signaling. However, whether one of these three supplements alone is sufficient to maintain naive self-renewal remains unclear. Here we show that LIF alone in medium is sufficient for adaptation of 2i/L-ESCs to embryonic stem cells (ESCs) in a hypermethylated state (L-ESCs). Global transcriptomic analysis shows that L-ESCs are close to 2i/L-ESCs and in a stable state between naive and primed pluripotency. Notably, our results demonstrate that DNA methyltransferases (DNMTs) play an important role in LIF-dependent mouse ESC adaptation and self-renewal. LIF-dependent ESC adaptation efficiency is significantly increased in serum treatment and reduced in Dnmt3a or Dnmt3l knockout ESCs. Importantly, unlike epiblast stem cells, L-ESCs contribute to somatic tissues and germ cells in chimeras. L-ESCs cultured under such simple conditions as in this study would provide a more conducive platform to clarify the molecular mechanism of ESCs in in vitro culture.


Assuntos
DNA (Citosina-5-)-Metiltransferases/fisiologia , DNA Metiltransferase 3A/metabolismo , Fator Inibidor de Leucemia/fisiologia , Células-Tronco Embrionárias Murinas/fisiologia , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Autorrenovação Celular , Células Cultivadas , Meios de Cultura/química , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Impressão Genômica , Camadas Germinativas/metabolismo , Janus Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Transcriptoma
5.
Sheng Li Xue Bao ; 72(1): 105-114, 2020 Feb 25.
Artigo em Chinês | MEDLINE | ID: mdl-32099988

RESUMO

Embryo implantation is crucial for the establishment and maintenance of successful pregnancy and requires the synchronization between implantation-competent blastocyst and receptive uterus. In assisted reproductive technologies, recognition of uterine receptivity is the limiting factor for improving pregnancy rate. It has been previously reported that embryo implantation involves the activation and inactivation of numerous signaling molecules which may influence the proliferation and differentiation of uterine epithelial cells, epithelial polarity, luminal closure, embryo orientation, epithelial-stromal interactions, gland development, etc. Here we summarize the function of estrogen, progesterone, leukemia inhibitory factor (LIF), microRNA (miRNA), channel protein and signaling pathways in embryo implantation and explore their regulatory network to provide theoretical basis for the treatment of infertility and development of safe and efficient contraceptives.


Assuntos
Implantação do Embrião , Útero/fisiologia , Blastocisto/fisiologia , Estrogênios/fisiologia , Feminino , Humanos , Fator Inibidor de Leucemia/fisiologia , MicroRNAs/genética , Gravidez , Progesterona/fisiologia , Transdução de Sinais
6.
Cell Metab ; 31(2): 327-338.e6, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31928884

RESUMO

The beta (ß)-cell mass formed during embryogenesis is amplified by cell replication during fetal and early postnatal development. Thereafter, ß cells become functionally mature, and their mass is maintained by a low rate of replication. For those few ß cells that replicate in adult life, it is not known how replication is initiated nor whether this occurs in a specialized subset of ß cells. We capitalized on a YAP overexpression system to induce replication of stem-cell-derived ß cells and, by single-cell RNA sequencing, identified an upregulation of the leukemia inhibitory factor (LIF) pathway. Activation of the LIF pathway induces replication of human ß cells in vitro and in vivo. The expression of the LIF receptor is restricted to a subset of transcriptionally distinct human ß cells with increased proliferative capacity. This study delineates novel genetic networks that control the replication of LIF-responsive, replication-competent human ß cells.


Assuntos
Linfócitos B/citologia , Proliferação de Células , Fator Inibidor de Leucemia/fisiologia , Adulto , Idoso , Animais , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Células-Tronco Pluripotentes , Fator de Transcrição STAT3/metabolismo , Análise de Célula Única
7.
Development ; 146(24)2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31740534

RESUMO

Embryonic stem cells (ESCs) exist in at least two states that transcriptionally resemble different stages of embryonic development. Naïve ESCs resemble peri-implantation stages and primed ESCs the pre-gastrulation epiblast. In mouse, primed ESCs give rise to definitive endoderm in response to the pathways downstream of Nodal and Wnt signalling. However, when these pathways are activated in naïve ESCs, they differentiate to a cell type resembling early primitive endoderm (PrE), the blastocyst-stage progenitor of the extra-embryonic endoderm. Here, we apply this context dependency to human ESCs, showing that activation of Nodal and Wnt signalling drives the differentiation of naïve pluripotent cells toward extra-embryonic PrE, or hypoblast, and these can be expanded as an in vitro model for naïve extra-embryonic endoderm (nEnd). Consistent with observations made in mouse, human PrE differentiation is dependent on FGF signalling in vitro, and we show that, by inhibiting FGF receptor signalling, we can simplify naïve pluripotent culture conditions, such that the inhibitor requirements closer resemble those used in mouse. The expandable nEnd cultures reported here represent stable extra-embryonic endoderm, or human hypoblast, cell lines.This article has an associated 'The people behind the papers' interview.


Assuntos
Endoderma/embriologia , Fator Inibidor de Leucemia/fisiologia , Ligantes da Sinalização Nodal/fisiologia , Células-Tronco Pluripotentes/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Células Cultivadas , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Endoderma/citologia , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camadas Germinativas/fisiologia , Humanos , Fator Inibidor de Leucemia/metabolismo , Camundongos , Ligantes da Sinalização Nodal/metabolismo , Transdução de Sinais/fisiologia
8.
Placenta ; 88: 20-27, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31586768

RESUMO

INTRODUCTION: Leukemia Inhibitory Factor (LIF) regulates behavior of trophoblast cells and their interaction with immune and endothelial cells. In vitro, trophoblast cell response to LIF may vary depending on the cell model. Reported differences in the miRNA profile of trophoblastic cells may be responsible for these observations. Therefore, miRNA expression was investigated in four trophoblastic cell lines under LIF stimulation followed by in silico analysis of altered miRNAs and their associated pathways. METHODS: Low density TaqMan miRNA assays were used to quantify levels of 762 mature miRNAs under LIF stimulation in three choriocarcinoma-derived (JEG-3, ACH-3P and AC1-M59) and a trophoblast immortalized (HTR-8/SVneo) cell lines. Expression of selected miRNAs was confirmed in primary trophoblast cells and cell lines by qPCR. Targets and associated pathways of the differentially expressed miRNAs were inferred from the miRTarBase followed by a KEGG Pathway Enrichment Analysis. HTR-8/SVneo and JEG-3 cells were transfected with miR-21-mimics and expression of miR-21 targets was assessed by qPCR. RESULTS: A similar number of miRNAs changed in each tested cell line upon LIF stimulation, however, low coincidence of individual miRNA species was observed and occurred more often among choriocarcinoma-derived cells (complete data set at http://www.ncbi.nlm.nih.gov/geo/ under GEO accession number GSE130489). Altered miRNAs were categorized into pathways involved in human diseases, cellular processes and signal transduction. Six cascades were identified as significantly enriched, including JAK/STAT and TGFB-SMAD. Upregulation of miR-21-3p was validated in all cell lines and primary cells and STAT3 was confirmed as its target. DISCUSSION: Dissimilar miRNA responses may be involved in differences of LIF effects on trophoblastic cell lines.


Assuntos
Fator Inibidor de Leucemia/fisiologia , MicroRNAs/metabolismo , Trofoblastos/fisiologia , Linhagem Celular , Humanos , Fator de Transcrição STAT3/metabolismo
9.
Dev Biol ; 445(2): 145-155, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30448440

RESUMO

The mammary gland (MG) is a unique organ responsible for milk synthesis, secretion, and involution to prepare the gland for subsequent lactation. The mammary epithelial cells (MECs), which are the milk synthesizing units of the MG, proliferate, differentiate, undergo apoptosis and regenerate following a cyclic pathway of lactation - involution - lactation, fine-tuning these molecular events through hormones, growth factors and other regulatory molecules. The developmental stages of the MG are embryonic, prepubertal, pubertal, pregnancy, lactation and involution, with major developmental processes occurring after puberty. The involution stage includes interesting physiological processes such as MEC apoptosis, matrix remodeling, and the generation of cells regaining the shape of a virgin MG. Signal transducer and activator of transcription 3 (STAT3) is the established master regulator of this process and aberrant expression of STAT3 leads to subnormal involution and may induce neoplasia. Several studies have reported on the molecular mechanism of MG involution with substantial knowledge being gained about this process; however, a deep understanding of this phenomenon has yet to be attained. This review focuses deeply on the molecular details of post-lactational regression, the signaling pathways involved in the lactation-involution cycle, and the latest developments in STAT3-associated MG neoplasia. Deep insight into the involution process will pave the way towards understanding the biology, apoptosis, and oncogenesis of the MG.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/fisiologia , Animais , Apoptose/genética , Neoplasias da Mama/etiologia , Citocinas/genética , Citocinas/fisiologia , Progressão da Doença , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Matriz Extracelular/fisiologia , Feminino , Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Humanos , Lactação/genética , Lactação/fisiologia , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/fisiologia , Gotículas Lipídicas , Glândulas Mamárias Animais/anatomia & histologia , Camundongos , MicroRNAs/genética , Modelos Biológicos , Gravidez , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais , Fator de Crescimento Transformador beta/fisiologia
10.
Adv Exp Med Biol ; 1099: 125-139, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30306520

RESUMO

Opioid analgesics remain the most effective and widely used analgesics for the management of moderate to severe pain, including cancer pain and chronic non-cancer pain. However, the efficacy of long-term opioid analgesics is attenuated by tolerance and/or hyperalgesia after long-term use, preventing adequate pain relief under stable opioid dosages for chronic pain patients. Classical neuron-centered concepts about tolerance, such as internalization of opioid receptors, upregulation of N-methyl-D-aspartate receptor function, or downregulation of glutamate transporter activity, can only partially explain the phenomenon of tolerance. Recent evidence revealing glial activation and upregulation of inflammatory mediators in the rodent central nervous system has confirmed the pivotal role of neuroinflammation in neuropathic pain or opioid tolerance, or both. However, human evidence is still sparse.Based on our clinical practice, we conducted translational research by investigating the cerebrospinal fluid (CSF) cytokine and chemokine profiles of opioid-tolerant patients after research ethic committee approval. CSF samples from opioid-tolerant patients and opioid-naive subjects were compared. We found CXCL1, CXCL12, and leukemia inhibitory factor (LIF) were significantly upregulated among the opioid-tolerant patients and positively correlated with the opioid dosage.We translated these findings back to lab animal experiment; after induction of tolerance by morphine infusion, the spinal cord expression of CXCL1, CXCL12, and LIF were all upregulated. Although CXCL1 and CXCL12 infusion alone did not affect baseline tail-flick latency, morphine analgesic efficacy dropped significantly after intrathecal infusion of CXCL1 and CXCL12. After establishing tolerance by intrathecal continuous infusion of morphine, tolerance development was accelerated by co-administration of CXCL1 and CXCL12. In parallel, the effect was attenuated by co-administration of CXCL1- or CXCL12-neutralizing antibody or concordant receptor antagonists.On the contrary, although chronic morphine administration still induced LIF upregulation in rat spinal cords, intrathecal injection of LIF potentiated the analgesic action of morphine and delayed the development of morphine tolerance. Upregulation of endogenously released LIF by long-term use of opioids might counterbalance the tolerance induction effects of other pro-inflammatory cytokines.CXCL1, CXCL12, and LIF are upregulated in both opioid-tolerant patients and rodents. The onset and extent of opioid tolerance were affected by modulating the intrathecal CXCL1/CXCR2, CXCL12/CXCR4, and LIF signaling and could be novel drug targets for the treatment of opioid tolerance.


Assuntos
Analgésicos Opioides/farmacologia , Quimiocina CXCL12/fisiologia , Quimiocina CXCL1/fisiologia , Tolerância a Medicamentos , Inflamação/fisiopatologia , Fator Inibidor de Leucemia/fisiologia , Animais , Humanos , Ratos , Medula Espinal/efeitos dos fármacos
12.
PLoS One ; 13(2): e0191865, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29444110

RESUMO

The interaction between cancer cells and the cancer stroma plays a crucial role in tumor progression and metastasis in diverse malignancies, including oral cancer. However, the mechanism underlying this interaction remains incompletely elucidated. Here, to investigate the interaction between oral cancer cells and fibroblasts, which are major cellular components of the tumor stroma, we conducted an in vitro study by using human oral squamous cell carcinoma (OSCC) cell lines and normal human dermal fibroblasts (NHDFs). The results of transwell assays revealed that the migration and invasion of 2 OSCC cell lines, HO1-N-1 and HSC3, were markedly stimulated upon coculturing with NHDFs. To investigate the factors that promote tumor invasion, we isolated NHDFs from cocultures prepared with HO1-N-1 cells and performed microarray analysis. Among the various genes that were upregulated, we identified the gene encoding leukemia inhibitory factor (LIF), and we focused on LIF in further analyses. We confirmed that all OSCC-derived conditioned media potently upregulated LIF expression in NHDFs, and the results of our transwell analysis demonstrated that NHDF-induced OSCC migration and invasion were inhibited by LIF-neutralizing antibodies. Furthermore, immunohistochemical analysis of patient samples revealed that in 44 out of 112 OSCC cases, LIF was expressed in the tumor stroma, particularly in cancer-associated fibroblasts (CAFs), and, notably, clinicopathological analyses confirmed that LIF expression in CAFs was significantly correlated with increased depth of tumor invasion. Collectively, our results suggest that OSCC stimulates fibroblasts to produce LIF, which, in turn, participates in cancer-cell invasion. Our finding offers a potential therapeutic strategy targeting the cancer stroma for the treatment of OSCC patients.


Assuntos
Carcinoma de Células Escamosas/patologia , Fator Inibidor de Leucemia/fisiologia , Neoplasias Bucais/patologia , Invasividade Neoplásica , Linhagem Celular Tumoral , Técnicas de Cocultura , Meios de Cultivo Condicionados , Fibroblastos/metabolismo , Humanos
13.
Am J Physiol Cell Physiol ; 313(5): C487-C500, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28768641

RESUMO

IL-6 and leukemia inhibitory factor (LIF), members of the IL-6 family of cytokines, play recognized paradoxical roles in skeletal muscle mass regulation, being associated with both growth and atrophy. Overload or muscle contractions can induce a transient increase in muscle IL-6 and LIF expression, which has a regulatory role in muscle hypertrophy. However, the cellular mechanisms involved in this regulation have not been completely identified. The induction of mammalian target of rapamycin complex 1 (mTORC1)-dependent myofiber protein synthesis is an established regulator of muscle hypertrophy, but the involvement of the IL-6 family of cytokines in this process is poorly understood. Therefore, we investigated the acute effects of IL-6 and LIF administration on mTORC1 signaling and protein synthesis in C2C12 myotubes. The role of glycoprotein 130 (gp130) receptor and downstream signaling pathways, including phosphoinositide 3-kinase (PI3K)-Akt-mTORC1 and signal transducer and activator of transcription 3 (STAT3)-suppressor of cytokine signaling 3 (SOCS3), was investigated by administration of specific siRNA or pharmaceutical inhibitors. Acute administration of IL-6 and LIF induced protein synthesis, which was accompanied by STAT3 activation, Akt-mTORC1 activation, and increased SOCS3 expression. This induction of protein synthesis was blocked by both gp130 siRNA knockdown and Akt inhibition. Interestingly, STAT3 inhibition or Akt downstream mTORC1 signaling inhibition did not fully block the IL-6 or LIF induction of protein synthesis. SOCS3 siRNA knockdown increased basal protein synthesis and extended the duration of the protein synthesis induction by IL-6 and LIF. These results demonstrate that either IL-6 or LIF can activate gp130-Akt signaling axis, which induces protein synthesis via mTORC1-independent mechanisms in cultured myotubes. However, IL-6- or LIF-induced SOCS3 negatively regulates the activation of myotube protein synthesis.


Assuntos
Interleucina-6/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Biossíntese de Proteínas/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Citocinas/farmacologia , Citocinas/fisiologia , Interleucina-6/farmacologia , Fator Inibidor de Leucemia/farmacologia , Fator Inibidor de Leucemia/fisiologia , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Mioblastos/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos
14.
Obesity (Silver Spring) ; 24(11): 2361-2367, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27663886

RESUMO

OBJECTIVE: High-fat diet (HFD) is known to induce low-grade hypothalamic inflammation. Whether inflammation occurs in other brain areas remains unknown. This study tested the effect of short-term HFD on cytokine gene expression and identified leukemia inhibitory factor (LIF) as a responsive cytokine in the brain stem. Thus, functional and cellular effects of LIF in the brain stem were investigated. METHODS: Male rats were fed chow or HFD for 3 days, and then gene expression was analyzed in different brain regions for IL-1ß, IL-6, TNF-α, and LIF. The effect of intracerebroventricular injection of LIF on chow intake and body weight was also tested. Patch clamp recording was performed in the nucleus tractus solitarius (NTS). RESULTS: HFD increased pontine TNF-α mRNA while downregulating LIF in all major parts of the brain stem, but not in the hypothalamus or hippocampus. LIF injection into the cerebral aqueduct suppressed food intake without conditioned taste aversion, suggesting that LIF can induce anorexia via lower brain regions without causing malaise. In the NTS, a key brain stem nucleus for food intake regulation, LIF induced acute changes in neuronal excitability. CONCLUSIONS: HFD-induced downregulation of anorexic LIF in the brain stem may provide a permissive condition for HFD overconsumption. This may be at least partially mediated by the NTS.


Assuntos
Anorexia/fisiopatologia , Tronco Encefálico/metabolismo , Dieta Hiperlipídica/efeitos adversos , Regulação para Baixo , Fator Inibidor de Leucemia/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/metabolismo , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Fator Inibidor de Leucemia/administração & dosagem , Masculino , RNA Mensageiro/metabolismo , Ratos , Núcleo Solitário/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
15.
Hepatology ; 64(5): 1606-1622, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27533020

RESUMO

Increasing evidence supports a role for N-myc downstream-regulated gene 2 (NDRG2) deregulation in tumorigenesis. We investigated the roles and mechanisms of NDRG2 in human cholangiocarcinoma (CCA) progression. In the present study, expression of NDRG2, microRNA (miR)-181c and leukemia inhibitory factor (LIF) in human CCA and adjacent nontumor tissues were examined. The effects of NDRG2 on CCA tumor growth and metastasis were determined both in vivo and in vitro. The role of the NDRG2/LIF/miR-181c signaling pathway in cholangiocarcinogenesis and metastasis were investigated both in vivo and in vitro. The results showed that human CCA tissues exhibited decreased levels of NDRG2 and increased levels of miR-181c and LIF compared with nontumor tissues. NDRG2 could inhibit CCA cell proliferation, chemoresistance, and metastasis both in vitro and in vivo. We found that NDRG2 is a target gene of miR-181c, and the down-regulation of NDRG2 was attributed to miR-181c overexpression in CCA. Furthermore, miR-181c can be activated by LIF treatment, whereas NDRG2 could inhibit LIF transcription through disrupting the binding between Smad, small mothers against decapentaplegic complex and LIF promoter. Down-regulation of NDRG2 and overexpression of miR-181c or LIF are significantly associated with a poorer overall survival (OS) in CCA patients. Finally, we found that a combination of NDRG2, miR-181c, and LIF expression is a strong predictor of prognosis in CCA patients. CONCLUSION: These results establish the counteraction between NDRG2 and LIF/miR-181c as a key mechanism that regulates cholangiocarcinogenesis and metastasis. Our results elucidated a novel pathway in NDRG2-mediated inhibition of cholangiocarcinogenesis and metastasis and suggest new therapeutic targets, including NDRG2, LIF, miR-181c, and transforming growth factor beta, in CCA prevention and treatment. (Hepatology 2016;64:1606-1622).


Assuntos
Neoplasias dos Ductos Biliares/etiologia , Colangiocarcinoma/etiologia , Retroalimentação Fisiológica , Fator Inibidor de Leucemia/fisiologia , MicroRNAs/fisiologia , Proteínas/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Progressão da Doença , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Transdução de Sinais
16.
Br J Anaesth ; 117(4): 512-520, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28077540

RESUMO

BACKGROUND: The efficacy of opioids typically decreases after long-term use owing to the development of tolerance. Glial activation and the upregulation of proinflammatory cytokines are related to the induction of tolerance. We investigated the effect of leukemia inhibitory factor (LIF) on morphine analgesia and tolerance. METHODS: LIF concentrations in rat spinal cords were measured by polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA) after morphine administration. LIF distribution was examined using confocal microscopy. To evaluate the effects of LIF on morphine analgesia and tolerance, LIF was intrathecally administered 30 min before morphine injection. The analgesic effect of morphine was evaluated by measuring tail-flick latency. Human LIF concentrations from the cerebrospinal fluid (CSF) of opioid tolerant patients were also determined by specific ELISA. RESULTS: Chronic morphine administration upregulated LIF concentrations in rat spinal cords. Intrathecal injection of LIF potentiated the analgesic action of morphine. Patch clamp recording of spinal cord slices showed that LIF enhanced DAMGO ([D-Ala2, N-MePhe4, Gly-ol]-enkephalin)-induced outward potassium current. The development of tolerance was markedly suppressed by exogenous LIF, whereas neutralizing the endogenously released LIF with anti-LIF antibodies accelerated the tolerance induction. Moreover, LIF concentrations in the CSF of opioid-tolerant patients were higher than those in the opioid-naive controls. CONCLUSIONS: Intrathecal administration of LIF potentiated morphine antinociceptive activity and attenuated the development of morphine tolerance. Upregulation of endogenously released LIF by long-term use of opioids might counterbalance the tolerance induction effects of other proinflammatory cytokines. LIF might be a novel drug candidate for inhibiting opioid tolerance induction.


Assuntos
Analgésicos Opioides/farmacologia , Fator Inibidor de Leucemia/fisiologia , Morfina/farmacologia , Animais , Citocinas/análise , Tolerância a Medicamentos , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
17.
J Neurotrauma ; 33(16): 1522-34, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-26541248

RESUMO

Reactions of both astrocytes and microglia to central nervous system injury can be beneficial or detrimental to recovery. To gain insights into the functional importance of gliosis, we developed a new model of adolescent closed-head injury (CHI) and interrogated the behavioral, physiological, and cellular outcomes after a concussive CHI in leukemia inhibitory factor (LIF) haplodeficient mice. These mice were chosen because LIF is important for astrocyte and microglial activation. Behaviorally, the LIF haplodeficient animals were equally impaired 4 h after the injury, but in the subsequent 2 weeks, the LIF haplodeficient mice acquired more severe motor and sensory deficits, compared with wild type mice. The prolonged accumulation of neurological impairment was accompanied by desynchronization of the gliotic response, increased cell death, axonal degeneration, diminished callosal compound action potential, and hypomyelination. Our results clearly show that LIF is an essential injury-induced cytokine that is required to prevent the propagation of secondary neurodegeneration.


Assuntos
Citocinas/fisiologia , Traumatismos Cranianos Fechados/metabolismo , Traumatismos Cranianos Fechados/patologia , Traumatismos Cranianos Fechados/fisiopatologia , Fator Inibidor de Leucemia/fisiologia , Animais , Citocinas/deficiência , Modelos Animais de Doenças , Feminino , Fator Inibidor de Leucemia/deficiência , Masculino , Camundongos
18.
J. physiol. biochem ; 71(4): 807-821, dic. 2015.
Artigo em Inglês | IBECS | ID: ibc-145732

RESUMO

Myeloid leukemia factor 1 (MLF1) was involved in t(3;5) chromosomal rearrangement and aberrantly expressed in myelodysplastic syndromes/acute myeloid leukemia patients. Ex vivo experiments showed that the lymphocytes from the Mlf1-deficient mice were more resistant to apoptotic stimulations than the wild-type cells. Furthermore, the ectopically expressed MLF1 induced apoptosis in the cell models. These findings revealed that MLF1 was required for the cells to respond to the apoptotic stimulations. Ex vivo experiments also demonstrated that cytokine withdrawal significantly up-regulated Mlf1’s expression and promoted its association with B cell lymphoma-extra large (Bcl-XL) in the lymphocytes, at the same time reduced the association of Bax with Bcl-XL The same effects were also observed in the cells that over-expressed MLF1. However, these effects were observed in Mlf1 null lymphocytes as well as the cells over-expressing Bcl-XL. In addition, MLF1’s proapoptosis could be completely prevented by co-expression of Bcl-XL and significantly attenuated in Bax/Bak double null cells. These data, taken together, strongly suggested that in response to the stresses, up-regulated Mlf1 promoted its association with Bcl-XL and reduced the available Bcl-XL for associating with Bax, which resulted in releasing Bax from the Bcl-XL and apoptosis in turn. Lastly, we showed that MLF1 was negatively regulated by 14-3-3 and revealed that 14-3-3 bound to MLF1 and physically blocked MLF1’s Bcl-2 homology domain 3 (BH3) as well as Bcl-XL from associating with MLF1. Our findings suggested that ectopically expressed MLF1 could be responsible for the pathological apoptosis in early myelodysplastic syndrome (MDS) patients


Assuntos
Leucemia Mieloide/fisiopatologia , Fator Inibidor de Leucemia/fisiologia , Proteína bcl-X/fisiologia , Síndromes Mielodisplásicas/fisiopatologia , Modelos Animais de Doenças , Apoptose/fisiologia
19.
Development ; 142(20): 3488-99, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26395492

RESUMO

Embryonic stem cells (ESCs) are pluripotent cell lines that can be maintained indefinitely in an early developmental state. ESC culture conditions almost always require the cytokine LIF to maintain self-renewal. As ESCs are not homogeneous but contain multiple populations reminiscent of the blastocyst, identifying the target cells of LIF is necessary to understand the propagation of pluripotency. We recently found that LIF acts under self-renewing conditions to stimulate the fraction of ESCs that express extraembryonic markers, but has little impact on pluripotent gene expression. Here, we report that LIF has two distinct roles: it blocks early epiblast (Epi) differentiation, and it supports the expansion of primitive endoderm (PrE)-primed ESCs and PrE in vivo. We find that activation of JAK/STAT signalling downstream of LIF occurs initially throughout the pre-implantation embryo, but later marks the PrE. Moreover, the addition of LIF to cultured embryos increases the GATA6(+) PrE population, whereas inhibition of JAK/STAT signalling reduces both NANOG(+) epiblast and GATA6(+) PrE. The reduction of the NANOG(+) Epi might be explained by its precocious differentiation to later Epi derivatives, whereas the increase in PrE is mediated both by an increase in proliferation and inhibition of PrE apoptosis that is normally triggered in embryos with an excess of GATA6(+) cells. Thus, it appears that the relative size of the PrE is determined by the number of LIF-producing cells in the embryo. This suggests a mechanism by which the embryo adjusts the relative ratio of the primary lineages in response to experimental manipulation.


Assuntos
Blastocisto/citologia , Endoderma/citologia , Regulação da Expressão Gênica no Desenvolvimento , Fator Inibidor de Leucemia/fisiologia , Animais , Apoptose , Diferenciação Celular , Linhagem da Célula , Citocinas/metabolismo , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Feminino , Citometria de Fluxo , Fator de Transcrição GATA6/metabolismo , Perfilação da Expressão Gênica , Interleucina-6/metabolismo , Janus Quinases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Fenótipo , Células-Tronco Pluripotentes/citologia , Fator de Transcrição STAT3/metabolismo , Fatores de Tempo
20.
Mol Cell Biol ; 35(19): 3284-300, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26169829

RESUMO

The transcription factor STAT3 is constitutively active in many cancers, where it mediates important biological effects, including cell proliferation, differentiation, survival, and angiogenesis. The N-terminal domain (NTD) of STAT3 performs multiple functions, such as cooperative DNA binding, nuclear translocation, and protein-protein interactions. However, it is unclear which subsets of STAT3 target genes depend on the NTD for transcriptional regulation. To identify such genes, we compared gene expression in STAT3-null mouse embryonic fibroblasts (MEFs) stably expressing wild-type STAT3 or STAT3 from which NTD was deleted. NTD deletion reduced the cytokine-induced expression of specific STAT3 target genes by decreasing STAT3 binding to their regulatory regions. To better understand the potential mechanisms of this effect, we determined the crystal structure of the STAT3 NTD and identified a dimer interface responsible for cooperative DNA binding in vitro. We also observed an Ni(2+)-mediated oligomer with an as yet unknown biological function. Mutations on both dimer and Ni(2+)-mediated interfaces affected the cytokine induction of STAT3 target genes. These studies shed light on the role of the NTD in transcriptional regulation by STAT3 and provide a structural template with which to design STAT3 NTD inhibitors with potential therapeutic value.


Assuntos
Fator de Transcrição STAT3/química , Transcrição Gênica , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Cristalografia por Raios X , Fator Inibidor de Leucemia/fisiologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Fator de Transcrição STAT3/fisiologia , Ativação Transcricional , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...