Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
J Immunol ; 204(4): 879-891, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31924647

RESUMO

Hematopoiesis is tightly regulated by the bone marrow (BM) niche. The niche is robust, allowing for the return of hematopoietic homeostasis after insults such as infection. Hematopoiesis is partly regulated by soluble factors, such as neuropeptides, substance P (SP), and neurokinin A (NK-A), which mediate hematopoietic stimulation and inhibition, respectively. SP and NK-A are derived from the Tac1 gene that is alternately spliced into four variants. The hematopoietic effects of SP and NK-A are mostly mediated via BM stroma. Array analyses with 2400 genes indicated distinct changes in SP-stimulated BM stroma. Computational analyses indicated networks of genes with hematopoietic regulation. Included among these networks is the high-mobility group box 1 gene (HMGB1), a nonhistone chromatin-associated protein. Validation studies indicated that NK-A could reverse SP-mediated HMGB1 decrease. Long-term culture-initiating cell assay, with or without NK-A receptor antagonist (NK2), showed a suppressive effect of HMGB1 on hematopoietic progenitors and increase in long-term culture-initiating cell assay cells (primitive hematopoietic cells). These effects occurred partly through NK-A. NSG mice with human hematopoietic system injected with the HMGB1 antagonist glycyrrhizin verified the in vitro effects of HMGB1. Although the effects on myeloid lineage were suppressed, the results suggested a more complex effect on the lymphoid lineage. Clonogenic assay for CFU- granulocyte-monocyte suggested that HMGB1 may be required to prevent hematopoietic stem cell exhaustion to ensure immune homeostasis. In summary, this study showed how HMGB1 is linked to SP and NK-A to protect the most primitive hematopoietic cell and also to maintain immune/hematopoietic homeostasis.


Assuntos
Proteína HMGB1/metabolismo , Hematopoese/genética , Neuroimunomodulação/genética , Neurocinina A/metabolismo , Substância P/metabolismo , Adolescente , Adulto , Processamento Alternativo , Animais , Benzamidas/farmacologia , Biópsia , Medula Óssea/metabolismo , Medula Óssea/patologia , Transplante de Medula Óssea , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/imunologia , Células HEK293 , Hematopoese/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Neuroimunomodulação/imunologia , Neurocinina A/antagonistas & inibidores , Análise de Sequência com Séries de Oligonucleotídeos , Piperidinas/farmacologia , Cultura Primária de Células , Taquicininas/genética , Quimeras de Transplante , Adulto Jovem
2.
FASEB J ; 30(1): 149-59, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26373800

RESUMO

Substance P and its truncated receptor exert oncogenic effects. The high production of substance P in breast cancer cells (BCCs) is caused by the enhancement of tachykinin (TAC)1 translation by cytosolic factor. In vitro translational studies and mRNA stabilization analyses indicate that BCCs contain the factor needed to increase TAC1 translation and to stabilize the mRNA. Prediction of protein folding, RNA-shift analysis, and proteomic analysis identified a 40 kDa molecule that interacts with the noncoding exon 7. Western blot analysis and RNA supershift identified Musashi 1 (Msi1) as the binding protein. Ectopic expression of TAC1 in nontumorigenic breast cells (BCs) indicates that TAC1 regulates its stability by increasing Msi1. Using a reporter gene system, we showed that Msi1 competes with microRNA (miR)130a and -206 for the 3' UTR of exon 7/TAC1. In the absence of Msi1 and miR130a and -206, reporter gene activity decreased, indicating that Msi1 expression limits TAC1 expression. Tumor growth was significantly decreased when nude BALB/c mice were injected with Msi1-knockdown BCCs. In summary, the RNA-binding protein Msi1 competes with miR130a and -206 for interaction with TAC1 mRNA, to stabilize and increase its translation. Consequently, these interactions increase tumor growth.


Assuntos
Neoplasias da Mama/genética , Ciclo Celular/genética , Morte Celular/genética , Proliferação de Células/genética , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral/metabolismo , Feminino , Humanos , Camundongos Endogâmicos BALB C , Proteômica/métodos
3.
Exp Cell Res ; 318(20): 2604-15, 2012 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-22981979

RESUMO

Neurokinin 1 (NK1) encodes full-length (NK1-FL) and truncated (NK1-Tr) receptors, with distinct 3' UTR. NK1-Tr exerts oncogenic functions and is increased in breast cancer (BC). Enhanced transcription of NK1 resulted in higher level of NK1-Tr. The 3' UTR of these two transcripts are distinct with NK1-Tr terminating at a premature stop codon. NK1-Tr mRNA gained an advantage over NK1-FL with regards to translation. This is due to the ability of miR519B to interact with sequences within the 3' UTR of NK1-FL, but not NK1-Tr since the corresponding region is omitted. MiR519b suppressed the translation of NK1-FL in T47D and MDA-MB-231 resulting in increased NK1-Tr protein. Cytokines can induce the transcription of NK1. However, our studies indicated that translation appeared to be independent of cytokine production by the BC cells (BCCs). This suggested that transcription and translation of NK1 might be independent. The findings were validated in vivo. MiR-519b suppressed the growth of MDA-MB-231 in 7/10 nude BALB/c. In total, increased NK1-Tr in BCCs is due to enhanced transcription and suppressed translation of NK1-FL by miR-519b to reduced tumor growth. In summary, we report on miRNA as a method to further regulate the expression of a spiced variant to promote oncogenesis. In addition, the findings have implications for therapy with NK1 antagonists. The oncogenic effect of NK1-Tr must be considered to improve the efficacy of current drugs to NK1.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , Neurocinina A/genética , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , MicroRNAs/biossíntese , Neurocinina A/biossíntese , Regulação para Cima/genética
4.
Differentiation ; 84(2): 214-22, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22824626

RESUMO

Mesenchymal stromal cells (MSCs) show promise for treatment of a variety of neurological and other disorders. Cat has a high degree of linkage with the human genome and has been used as a model for analysis of neurological disorders such as stroke, Alzheimer's disease and motor disorders. The present study was designed to characterize bone marrow-derived MSCs from cats and to investigate the capacity to generate functional peptidergic neurons. MSCs were expanded with cells from the femurs of cats and then characterized by phenotype and function. Phenotypically, feline and human MSCs shared surface markers, and lacked hematopoietic markers, with similar morphology. As compared to a subset of human MSCs, feline MSCs showed no evidence of the major histocompatibility class II. Since the literature suggested Stro-1 as an indicator of pluripotency, we compared early and late passages feline MSCs and found its expression in >90% of the cells. However, the early passage cells showed two distinct populations of Stro-1-expressing cells. At passage 5, the MSCs were more homogeneous with regards to Stro-1 expression. The passage 5 MSCs differentiated to osteogenic and adipogenic cells, and generated neurons with electrophysiological properties. This correlated with the expression of mature neuronal markers with concomitant decrease in stem cell-associated genes. At day 12 induction, the cells were positive for MAP2, Neuronal Nuclei, tubulin ßIII, Tau and synaptophysin. This correlated with electrophysiological maturity as presented by excitatory postsynaptic potentials (EPSPs). The findings indicate that the cat may constitute a promising biomedical model for evaluation of novel therapies such as stem cell therapy in such neurological disorders as Alzheimer's disease and stroke.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Mesenquimais/citologia , Neurogênese , Neurônios/metabolismo , Fenótipo , Adipogenia , Animais , Antígenos de Superfície/metabolismo , Gatos , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores , Humanos , Neurônios/citologia , Osteogênese , Especificidade da Espécie
5.
J Immunol ; 184(10): 5885-94, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20382885

RESUMO

Mesenchymal stem cells (MSCs) have been shown to support breast cancer growth. Because MSCs also increase the frequency of regulatory T cells (T(regs)), this study tested the hypothesis that human MSCs, via Tregs, protect breast cancer cells (BCCs) from immune clearance MSCs suppressed the proliferation of PBMCs when the latter were exposed to gamma-irradiated BCCs. Similarly, MSCs showed significant inhibition of PBMC migration toward BCCs and a corresponding decrease in CXCL12. MSCs also inhibited NK cell and CTL functions, which correlated with reduced numbers of CD8(+) and CD56(+) cells compared with parallel cultures without MSCs. The reduced NK and CTL activities correlated with a decrease in intracellular and secreted granzyme B. To explain these immunosuppressive findings, we compared T(reg) levels after coculture with MSCs and found an approximately 2-fold increase in T(regs), with associated decreases in antitumor Th1 cytokines and increases in Th2 cytokines. MSC-derived TGF-beta1 was largely responsible for the increase in T(regs) based on knockdown studies. In the presence of T(reg) depletion, PBMC proliferation and effector functions were partially restored. Together, these studies show an MSC-mediated increase in T(regs) in cocultures of PBMCs and BCCs. The results could be explained, in part, by the increase in Th2-type cytokines and MSC-generated TGF-beta1. These findings demonstrate immune protection by MSCs to BCCs. The reduction in immune cell proliferation and recruitment mediated by MSCs has implications for treatment of breast cancer with chemotherapy.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/fisiologia , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Feminino , Humanos , Células K562 , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Linfopenia/imunologia , Linfopenia/metabolismo , Linfopenia/patologia , Células-Tronco Mesenquimais/patologia , Camundongos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/patologia , Linfócitos T Reguladores/metabolismo
6.
Proc Natl Acad Sci U S A ; 106(11): 4408-13, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19246391

RESUMO

Breast cancer remains the most prevalent cancer among women in the United States. Substance P, a peptide derived from the TAC1 gene, mediates oncogenic properties in breast and other cancers. TAC1 expression facilitates the entry of breast cancer cells into bone marrow. The transcriptional repressor element 1-silencing transcription factor (REST) has been implicated in both oncogenic and tumor-suppressor functions. REST binds to the 5' untranslated region of the TAC1 promoter and suppresses its expression. This study investigated a role for REST in TAC1 induction in breast cancer. Western blots and real-time PCR indicated that REST expression in breast cancer cells was inversely proportional to the cells' aggressiveness, for both cell lines and primary breast cancer cells. REST knockdown in low-metastatic T47D cells and nontumorigenic MCF12A cells resulted in increases in TAC1 induction, proliferation, and migration. These parameters were negatively affected by ectopic expression of REST in highly aggressive MDA-MB-231 cells. Together, these findings show a central role for REST in the oncogenic function of TAC1 and suggest a tumor-suppressor role for REST in breast cancer.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Repressoras/fisiologia , Substância P/genética , Proteínas Supressoras de Tumor/fisiologia , Sítios de Ligação , Neoplasias da Mama/patologia , Movimento Celular , Proliferação de Células , Feminino , Humanos , Regiões Promotoras Genéticas , Índice de Gravidade de Doença , Células Tumorais Cultivadas
7.
Biochem Biophys Res Commun ; 414(1): 170-4, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21945934

RESUMO

Leptin is a pleiotropic hormone primarily secreted by adipocytes. A high density of functional Leptin receptors has been reported to be expressed in the hippocampus and other cortical regions of the brain, the physiological significance of which has not been explored extensively. Alzheimer's disease (AD) is marked by impaired brain metabolism with decreased glucose utilization in those regions which often precede pathological changes. Recent epidemiological studies suggest that plasma Leptin is protective against AD. Specifically, elderly with plasma Leptin levels in the lowest quartile were found to be four times more likely to develop AD than those in the highest quartile. We have previously reported that Leptin modulates AD pathological pathways in vitro through a mechanism involving the energy sensor, AMP-activated protein kinase (AMPK). To this end, we investigated the extent to which activation of AMPK as well as another class of sensors linking energy availability to cellular metabolism, the sirtuins (SIRT), mediate Leptin's biological activity. Leptin directly activated neuronal AMPK and SIRT in cell lines. Additionally, the ability of Leptin to reduce tau phosphorylation and ß-amyloid production was sensitive to the AMPK and sirtuin inhibitors, compound C and nicotinamide, respectively. These findings implicate that Leptin normally acts as a signal for energy homeostasis in neurons. Perhaps Leptin deficiency in AD contributes to a neuronal imbalance in handling energy requirements, leading to higher Aß and phospho-tau, which can be restored by replenishing low Leptin levels. This may also be a legitimate strategy for therapy.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Amiloide/antagonistas & inibidores , Leptina/farmacologia , Neurônios/efeitos dos fármacos , Proteínas Quinases/biossíntese , Sirtuínas/biossíntese , Proteínas tau/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos
8.
Therapy ; 8(5): 481-490, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22121334

RESUMO

The data from the initial clinical trials utilizing recombinant human leptin as an obesity therapy were published in 1998. Since then, numerous studies have been described which address dosage, safety and efficacy of leptin replacement for a variety of disorders with diverse patient groups, including pediatric and adult subjects. We review the current clinical trial data, demonstrate that leptin administration is safe for long term use in humans, and summarize reported cognitive benefits. The functions of leptin in neuroprotection and cognition have been largely overlooked. Accumulating data suggest a very significant application of leptin may be a therapy for Alzheimer's disease.

9.
Methods Mol Biol ; 2224: 195-202, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33606216

RESUMO

Mice with human hematopoietic system have become critical for research and preclinical studies. Mice with patient-derived xenografts of different tumors exist without human immune system. Answers can be addressed with the same immunodeficient mice that are chimeric for the human hemato-lymphoid system (humanized mice). The growing field of immune-oncology could benefit from preclinical studies with the humanized mice. Other fields will also benefit such as studies of infectious disease, regenerative medicine, organ transplant, and allergies. Here, we describe the method to humanize immune-deficient mice with human CD34+ hematopoietic cells.


Assuntos
Sistema Imunitário/imunologia , Síndromes de Imunodeficiência/imunologia , Adulto , Animais , Antígenos CD34/imunologia , Modelos Animais de Doenças , Feminino , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade
10.
Aging (Albany NY) ; 13(21): 23981-24016, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34762598

RESUMO

This study addresses the potential to reverse age-associated morbidity by establishing methods to restore the aged hematopoietic system. Parabiotic animal models indicated that young secretome could restore aged tissues, leading us to establish a heterochronic transwell system with aged mobilized peripheral blood (MPB), co-cultured with young MPB or umbilical cord blood (UCB) cells. Functional studies and omics approaches indicate that the miRNA cargo of microvesicles (MVs) restores the aged hematopoietic system. The in vitro findings were validated in immune deficient (NSG) mice carrying an aged hematopoietic system, improving aged hallmarks such as increased lymphoid:myeloid ratio, decreased inflammation and cellular senescence. Elevated MYC and E2F pathways, and decreased p53 were key to hematopoietic restoration. These processes require four restorative miRs that target the genes for transcription/differentiation, namely PAX and phosphatase PPMIF. These miRs when introduced in aged cells were sufficient to restore the aged hematopoietic system in NSG mice. The aged MPBs were the drivers of their own restoration, as evidenced by the changes from distinct baseline miR profiles in MPBs and UCB to comparable expressions after exposure to aged MPBs. Restorative natural killer cells eliminated dormant breast cancer cells in vivo, indicating the broad relevance of this cellular paradigm - preventing and reversing age-associated disorders such as clearance of early malignancies and enhanced responses to vaccine and infection.


Assuntos
Células da Medula Óssea , Micropartículas Derivadas de Células , Senescência Celular/fisiologia , Hematopoese/fisiologia , Adulto , Idoso , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/fisiologia , Feminino , Sangue Fetal/citologia , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Secretoma , Adulto Jovem
11.
Life Sci Alliance ; 4(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34078741

RESUMO

The challenge for treating breast cancer (BC) is partly due to long-term dormancy driven by cancer stem cells (CSCs) capable of evading immune response and resist chemotherapy. BC cells show preference for the BM, resulting in poor prognosis. CSCs use connexin 43 (Cx43) to form gap junctional intercellular communication with BM niche cells, fibroblasts, and mesenchymal stem cells (MSCs). However, Cx43 is an unlikely target to reverse BC dormancy because of its role as a hematopoietic regulator. We found N-cadherin (CDH2) and its associated pathways as potential drug targets. CDH2, highly expressed in CSCs, interacts intracellularly with Cx43, colocalizes with Cx43 in BC cells within BM biopsies of patients, and is required for Cx43-mediated gap junctional intercellular communication with BM niche cells. Notably, CDH2 and anti-apoptotic pathways maintained BC dormancy. We thereby propose these pathways as potential pharmacological targets to prevent dormancy and chemosensitize resistant CSCs.


Assuntos
Antígenos CD/metabolismo , Neoplasias da Mama/metabolismo , Caderinas/metabolismo , Conexina 43/metabolismo , Antígenos CD/genética , Medula Óssea/metabolismo , Caderinas/genética , Caderinas/fisiologia , Conexina 43/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Junções Comunicantes/metabolismo , Junções Comunicantes/patologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Metástase Neoplásica/patologia , Células-Tronco Neoplásicas/metabolismo , Evasão Tumoral/fisiologia
12.
Biochem Biophys Res Commun ; 380(1): 98-104, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19166821

RESUMO

Leptin, which serves as a lipid-modulating hormone to control metabolic energy availability, is decreased in Alzheimer's disease (AD) patients, and serum levels are inversely correlated to severity of dementia. We have previously described the effects of leptin in reducing amyloid beta protein both in vitro and in vivo, and tau phosphorylation in vitro. Herein, we systematically investigated the signaling pathways activated by leptin, leading to these molecular endpoints, to better understand its mechanism of action. Inhibition of amyloid beta production and tau phosphorylation in leptin-treated human and/or rat neuronal cultures were both dependent on activation of AMP-activated protein kinase (AMPK). Direct stimulation of AMPK with the cell-permeable activator, 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR), replicated leptin's effects and conversely, Compound C, an inhibitor of AMPK, blocked leptin's action. The data implicate that AMPK is a key regulator of both AD-related pathways.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Proteínas Quinases/metabolismo , Proteínas tau/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Células Cultivadas , Humanos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Ribonucleotídeos/farmacologia
13.
J Alzheimers Dis ; 16(4): 731-40, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19387109

RESUMO

Adipocyte-derived leptin appears to regulate a number of features defining Alzheimer's disease (AD) at the molecular and physiological level. Leptin has been shown to reduce the amount of extracellular amyloid beta, both in cell culture and animal models, as well as to reduce tau phosphorylation in neuronal cells. Importantly, chronic administration of leptin resulted in a significant improvement in the cognitive performance of transgenic animal models. In AD, weight loss often precedes the onset of dementia and the level of circulating leptin is inversely proportional to the severity of cognitive decline. It is speculated that a deficiency in leptin levels or function may contribute to systemic and CNS abnormalities leading to disease progression. Furthermore, a leptin deficiency may aggravate insulin-controlled pathways, known to be aberrant in AD. These observations suggest that a leptin replacement therapy may be beneficial for these patients.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Leptina/metabolismo , Leptina/uso terapêutico , Animais , Humanos
14.
Front Pharmacol ; 10: 134, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30853911

RESUMO

Glioblastoma multiforme (GBM) is a fatal malignancy of the central nervous system, commonly associated with chemoresistance. The alkylating agent Temozolomide (TMZ) is the front-line chemotherapeutic agent and has undergone intense studies on resistance. These studies reported on mismatch repair gene upregulation, ABC-targeted drug efflux, and cell cycle alterations. The mechanism by which TMZ induces cell cycle arrest has not been well-established. TMZ-resistant GBM cells have been linked to microRNA (miRNA) and exosomes. A cell cycle miRNA array identified distinct miRNAs only in exosomes from TMZ-resistant GBM cell lines and primary spheres. We narrowed the miRs to miR-93 and -193 and showed in computational analyses that they could target Cyclin D1. Since Cyclin D1 is a major regulator of cell cycle progression, we performed cause-effect studies and showed a blunting effects of miR-93 and -193 in Cyclin D1 expression. These two miRs also decreased cell cycling quiescence and induced resistance to TMZ. Taken together, our data provide a mechanism by which GBM cells can exhibit TMZ-induced resistance through miRNA targeting of Cyclin D1. The data provide a number of therapeutic approaches to reverse chemoresistance at the miRNA, exosomal and cell cycle points.

15.
Cell Death Dis ; 10(2): 59, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30683851

RESUMO

Breast cancer (BC) cells (BCCs) can retain cellular quiescence for decades, a phenomenon referred to as dormancy. BCCs show preference for the bone marrow (BM) where they can remain dormant for decades. Targeting BCCs within the BM is a challenge since the dormant BCCs reside within BM stroma, also residence for hematopoietic stem cells (HSCs). Dormant BCCs could behave as cancer stem cells (CSCs). The CSCs and HSCs are similar by function and also, by commonly expressed genes. The method by which dormant BCCs transition into clinically metastatic cells remains unclear. This study tested the hypothesis that macrophages (MΦs) within BM stroma, facilitates dormancy or reverse this state into metastatic cells. MΦs exhibiting an M2 phenotype constitute ~10% of cultured BM stroma. The M2 MΦs form gap junctional intercellular communication (GJIC) with CSCs, resulting in cycling quiescence, reduced proliferation and carboplatin resistance. In contrast, MΦs expressing the M1 phenotype reversed BC dormancy. Activation of M2a MΦs via the toll-like receptor 4 (TLR4) switched to M1 phenotype. The switch can occur by direct activation of M2a MΦs, or indirectly through activation of mesenchymal stem cells. M1 MΦ-derived exosomes activated NFкB to reverse quiescent BCCs to cycling cells. Using an in vivo model of BC dormancy, injected Mi MOs sensitized BCCs to carboplatin and increased host survival. In summary, we have shown how BM stromal MΦs, through exosomes, regulate the behavior of BCCs, by either inducing or reversing dormancy.


Assuntos
Medula Óssea/patologia , Neoplasias da Mama/patologia , Comunicação Celular , Exossomos/metabolismo , Macrófagos/metabolismo , Células-Tronco Neoplásicas/metabolismo , Adolescente , Adulto , Animais , Neoplasias da Mama/tratamento farmacológico , Carboplatina/uso terapêutico , Células Cultivadas , Técnicas de Cocultura , Resistencia a Medicamentos Antineoplásicos , Feminino , Junções Comunicantes , Xenoenxertos , Humanos , Macrófagos/classificação , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptor 4 Toll-Like/metabolismo , Adulto Jovem
16.
Biochem Biophys Res Commun ; 376(3): 536-41, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18801339

RESUMO

Leptin is a centrally acting hormone controlling metabolic pathways. Recently, it was shown that leptin can reduce amyloid beta levels both in vitro and in vivo. Herein, phosphorylation of tau was investigated following treatment of neuronal cells with leptin and insulin. Specifically, phosphorylation of tau at amino acid residues Ser(202), Ser(396) and Ser(404) was monitored in retinoic acid induced, human cell lines: SH-SY5Y and NTera-2. Both hormones induced a concentration- and time-dependent reduction of tau phosphorylation, and were synergistic at suboptimum concentrations. Importantly, leptin was 300-fold more potent than insulin (IC(50)L=46.9 nM vs. IC(50)I=13.8 microM). A central role for AMP-dependent kinase as a mediator of leptin's action is demonstrated by the ability of 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR) to decrease tau phosphorylation, and by blocking leptin in the presence of Compound C. Thus, leptin, which ameliorates both amyloid beta and tau-related pathological pathways, holds promise as a novel therapeutic for Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Leptina/farmacologia , Neurônios/efeitos dos fármacos , Proteínas tau/metabolismo , Proteínas Quinases Ativadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Linhagem Celular Tumoral , Humanos , Insulina/farmacologia , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/metabolismo , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ribonucleotídeos/farmacologia , Transdução de Sinais
17.
Biol Proced Online ; 10: 90-101, 2008 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-19461957

RESUMO

The present study describes a protocol to generate heterogenous populations of neurotransmitter-producing neurons from human mesenchymal stem cells (MSCs). MSCs are bone marrow (BM)-derived cells which undergo lineage- specific differentiation to generate bone, fat, cartilage and muscle, but are also capable of transdifferentiating into defined ectodermal and endodermal tissues. The purpose of this study is to evaluate the potential of MSCs as an alternative source of customized neurons for experimental neurobiology or other regenerative approaches. Our neuronal protocol utilizes freshly harvested human MSCs cultured on specific surfaces and exposed to an induction cocktail consisting of low serum concentration, retinoic acid (RA), growth factors and supplements. Here we report on the types of neurotransmitters produced by the neurons, and demonstrate that the cells are electrically responsive to exogenous neurotransmitter administration.

18.
Stem Cells ; 25(12): 3143-54, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17761754

RESUMO

OCT4 is a master transcriptional regulator, which mediates pluripotency in ESCs through inhibition of tissue-specific and promotion of stem cell-specific genes. Suppression of OCT4, along with other regulators of pluripotency, such as SOX2 and NANOG, has been correlated with cell-fate specification and lineage-specific differentiation. Recent reports have shown the expression of OCT4 in adult MSCs but have not ascribed functional homology with ESCs. MSCs are mesoderm-derived cells, primarily resident in adult bone marrow, that undergo lineage-specific differentiation to generate specialized cells such as stroma, fat, bone, and cartilage. We have previously demonstrated the plasticity of MSCs through their ability to generate neuronal cells. Here, we show that OCT4 provides similar regulatory circuitries in human MSCs and ESCs, using chromatin immunoprecipitation-DNA selection and ligation technology and loss-of-function studies. MSCs were found to express the embryonic transcription factors OCT4, NANOG, and SOX2. In addition, OCT4 was found to (a) target similar genes in MSCs and ESCs, (b) promote the expression of MSC-specific genes, and (c) regulate MSC cell cycle progression. The results suggest similar regulatory mechanisms for OCT4 in MSCs and ESCs and have implications regarding MSC plasticity. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células-Tronco Mesenquimais/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Fator 3 de Transcrição de Octâmero/genética
19.
Brain Behav Immun ; 22(4): 442-50, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18061399

RESUMO

Hematopoiesis is the process by which immune and blood cells are produced from a finite number of relatively few hematopoietic stem cells (HSCs). In adults, hematopoiesis occurs in the adult bone marrow (BM), with the support of stromal cells. This support partly occurs through the production of hematopoietic regulators belonging to the families of cytokines and neuropeptides/neurotransmitters, which mediate their actions through specific receptors. Thus, stromal cells could be central to the neural-hematopoietic-immune axis. This study focuses on Tac1, which encodes hematopoietic regulators belonging to the tachykinin family of neuropeptides. We examined post-transcriptional regulation of Tac1 in BM stroma. Since this gene is inducible in stroma, we selected cytokines with varying hematopoietic effects: stimulator Stem Cell Factor (SCF), broad-acting IL-11 and suppressive TGF-beta1. RNA shift with Tac1 mRNA and cytoplasmic extracts from IL-11 and SCF-stimulated stroma showed RNA shift after 15min at 37 degrees C, whereas a shift was detected with extracts from TGF-beta1-stimulated stroma after 5min at room temperature. Another level of post-transcriptional regulation was observed by the detection of miRNAs that interact with the 3' untranslated region of Tac1 mRNA. In summary, this study showed that cytokine induced miRNA downregulation and RNA-binding protein(s) are involved in post-transcriptional regulation of Tac1 in BM stroma. The broad categories of cytokines as hematopoietic stimulators or inhibitors might depend on the avidity of RNA-binding protein(s) for Tac1 mRNA, as well as the ability to degrade or stabilize the specific miRNAs.


Assuntos
Medula Óssea/fisiologia , Hematopoese/genética , MicroRNAs/genética , Proteínas de Ligação a RNA/metabolismo , Taquicininas/genética , Regiões 3' não Traduzidas/fisiologia , Adolescente , Adulto , Citosol/metabolismo , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Genes Reporter , Hematopoese/efeitos dos fármacos , Hematopoese/imunologia , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Interleucina-11/farmacologia , MicroRNAs/imunologia , Neuroimunomodulação/genética , Neuroimunomodulação/fisiologia , Neurocinina A/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Células Estromais/efeitos dos fármacos , Células Estromais/imunologia , Substância P/metabolismo , Taquicininas/imunologia , Fator de Crescimento Transformador beta1/farmacologia
20.
Sci Rep ; 8(1): 367, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29321622

RESUMO

This study proposes that a novel developmental hierarchy of breast cancer (BC) cells (BCCs) could predict treatment response and outcome. The continued challenge to treat BC requires stratification of BCCs into distinct subsets. This would provide insights on how BCCs evade treatment and adapt dormancy for decades. We selected three subsets, based on the relative expression of octamer-binding transcription factor 4 A (Oct4A) and then analysed each with Affymetrix gene chip. Oct4A is a stem cell gene and would separate subsets based on maturation. Data analyses and gene validation identified three membrane proteins, TMEM98, GPR64 and FAT4. BCCs from cell lines and blood from BC patients were analysed for these three membrane proteins by flow cytometry, along with known markers of cancer stem cells (CSCs), CD44, CD24 and Oct4, aldehyde dehydrogenase 1 (ALDH1) activity and telomere length. A novel working hierarchy of BCCs was established with the most immature subset as CSCs. This group was further subdivided into long- and short-term CSCs. Analyses of 20 post-treatment blood indicated that circulating CSCs and early BC progenitors may be associated with recurrence or early death. These results suggest that the novel hierarchy may predict treatment response and prognosis.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama/genética , Biologia Computacional , Perfilação da Expressão Gênica , Transcriptoma , Adulto , Idoso , Idoso de 80 Anos ou mais , Família Aldeído Desidrogenase 1 , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Biologia Computacional/métodos , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Imunofenotipagem , Isoenzimas/metabolismo , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Estadiamento de Neoplasias , Retinal Desidrogenase/metabolismo , Homeostase do Telômero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA