Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Clin Med ; 11(16)2022 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-36013165

RESUMO

Plasmablastic lymphoma (PBL) is a rare and aggressive form of large B-cell lymphoma (LBCL) most commonly seen in the setting of chronic immunosuppression or autoimmune disease. The prognosis is poor and CHOP-like regimens often fail to produce durable remission; therefore, there is no established standard of care treatment. However, PBL demonstrates substantial morphologic and immunophenotypic overlap with multiple myeloma (MM), suggesting that MM therapeutics might prove useful in treating PBL. We studied the effects of treatment using the first-in-class monoclonal antibody directed against CD38, daratumumab, in combination with chemotherapy in seven patients with advanced-stage LBCL with plasmablastic features. Treatment was safe and well-tolerated. Among six evaluable patients, six patients had complete response after treatment, and four patients who met strict WHO criteria for PBL had durable response (12-31 months and ongoing).

3.
Semin Oncol ; 48(1): 57-68, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33965249

RESUMO

Pancreas ductal adenocarcinoma (PDAC) has a dismal prognosis with a 5-year survival rate of 10%. Currently, chemotherapy remains the standard of care for systemic treatment. Immunotherapy with checkpoint inhibitors unfortunately has not been found to be effective in the treatment of PDAC to date, likely due to the highly desmoplastic and immunosuppressive tumor microenvironment (TME). Treatment targeting pathways against the immunosuppressive mechanisms of PDAC are of mounting interest to improve outcomes in PDAC. In this review, we discuss prior efforts and the current state of immunotherapy in PDAC. We will also review the emerging targets and treatments with significant clinical potential for the treatment of PDAC such as: CD40 pathway, the adenosine pathway, the CXCR4/CXCL12 axis, the CCR2/CCL2 axis, IDO pathway, and others.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/tratamento farmacológico , Humanos , Imunoterapia , Neoplasias Pancreáticas/tratamento farmacológico , Microambiente Tumoral , Neoplasias Pancreáticas
4.
Infect Immun ; 89(4)2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33468581

RESUMO

Thymosin beta-4 (Tß4) is an actin-sequestering peptide that plays important roles in regeneration and remodeling of injured tissues. However, its function in a naturally occurring pathogenic bacterial infection model has remained elusive. We adopted Tß4-overexpressing transgenic (Tg) mice to investigate the role of Tß4 in acute pulmonary infection and systemic sepsis caused by Legionella pneumophila Upon infection, Tß4-Tg mice demonstrated significantly lower bacterial loads in the lung, less hyaline membranes and necrotic abscess, with lower interstitial infiltration of neutrophils, CD4+, and CD8+ T cells. Bronchoalveolar lavage fluid of Tß4-Tg mice possessed higher bactericidal activity against exogenously added L. pneumophila, suggesting that constitutive expression of Tß4 could efficiently control L. pneumophila Furthermore, qPCR analysis of lung homogenates demonstrated significant reduction of interleukin 1 beta (IL-1ß) and tumor necrosis factor alpha (TNF-α), which primarily originate from lung macrophages, in Tß4-Tg mice after pulmonary infection. Upon L. pneumophila challenge of bone marrow-derived macrophages (BMDM) in vitro, secretion of IL-1ß and TNF-α proteins was also reduced in Tß4-Tg macrophages, without affecting their survival. The anti-inflammatory effects of BMDM in Tß4-Tg mice on each cytokine were affected when triggering with tlr2, tlr4, tlr5, or tlr9 ligands, suggesting that anti-inflammatory effects of Tß4 are likely mediated by the reduced activation of Toll-like receptors (TLR). Finally, Tß4-Tg mice in a systemic sepsis model were protected from L. pneumophila-induced lethality compared to wild-type controls. Therefore, Tß4 confers effective resistance against L. pneumophila via two pathways, a bactericidal and an anti-inflammatory pathway, which can be harnessed to treat acute pneumonia and septic conditions caused by L. pneumophila in humans.


Assuntos
Resistência à Doença/genética , Expressão Ectópica do Gene , Legionella pneumophila/fisiologia , Doença dos Legionários/genética , Doença dos Legionários/microbiologia , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/microbiologia , Timosina/genética , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno/genética , Humanos , Imuno-Histoquímica , Imunofenotipagem , Doença dos Legionários/patologia , Ligantes , Masculino , Camundongos , Camundongos Transgênicos , Pneumonia Bacteriana/patologia , Sepse/genética , Sepse/microbiologia , Sepse/patologia , Receptores Toll-Like/metabolismo
5.
Mult Scler J Exp Transl Clin ; 6(3): 2055217320942931, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32850133

RESUMO

We report a fatal case of COVID-19 in a 51-year-old African American woman with multiple sclerosis on natalizumab. She had multiple risk factors for severe COVID-19 disease including race, obesity, hypertension, and elevated inflammatory markers, but the contribution of natalizumab to her poor outcome remains unknown. We consider whether altered dynamics of peripheral immune cells in the context of natalizumab treatment could worsen the cytokine storm syndrome associated with severe COVID-19. We discuss extended interval dosing as a risk-reduction strategy for multiple sclerosis patients on natalizumab, and the use of interleukin-6 inhibitors in such patients who contract COVID-19.

6.
Curr Probl Diagn Radiol ; 48(2): 117-120, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29402490

RESUMO

PURPOSE: To evaluate whether in the setting of negative diagnostic mammogram for breast pain additional ultrasound is necessary. METHODS: Retrospective IRB-approved review of our database identified 8085 women who underwent ultrasound evaluation for breast pain from 1/1/2013-12/31/2013. Of 8085 women, 559 women had mammogram evaluation preceding the ultrasound and these women comprise the basis of this study. The patient's age, type of mammogram examination (screening or diagnostic), Breast Imaging-Reporting and Data System (BI-RADS) breast density (BD), type of breast pain (focal, diffuse, cyclical, unilateral, bilateral), additional breast symptoms (palpable concern, nipple discharge, skin changes, others), mammogram or ultrasound findings and final BI-RADS assessment, follow-up imaging, and follow-up biopsy results were reviewed and recorded. RESULTS: The median age of patients was 46 years old (range: 27-97). Patients recalled from negative screening mammogram were 29.8% (167/559). Patients with preceding negative diagnostic mammogram were 70.2% (392/559). The BI-RADS BD distribution was BD1: 5.5%, BD2: 39.9%, BD3: 46.0%, BD4: 8.6%. Final BI-RADS assessments were BI-RADS 1/2 (79%), BI-RADS 3 (12.9%), BI-RADS 4 (8.1%), BI-RADS 5 (0%). Majority (66.9%, 374/559) of the patient had breast pain alone. Additional breast symptoms were also noted as follows: palpable concern (24%), nipple discharge (3.9%), skin changes or other (5.2%). On follow-up evaluation, 26 findings were recommended for tissue sampling yielding 2 malignancies (0.4%, 2/559) in 2 patients. In the setting of negative mammogram and clinical symptom of breast pain alone yielded no malignances (NPV, 100%, 374/374) and was not impacted by BD. In patients with additional symptoms accompanying pain, malignancies were present despite negative mammogram in 2 patients; nipple discharge (4.5%, 1/22), and palpable concern (0.7%, 1/134). CONCLUSION: In the setting of negative mammogram and breast pain alone, additional evaluation with ultrasound is likely low yield and may be unnecessary. However, with additional symptoms such as palpable concern or nipple discharge, ultrasound is likely an important adjunct modality for identifying mammographically occult tumors.


Assuntos
Mastodinia/diagnóstico por imagem , Ultrassonografia Mamária , Procedimentos Desnecessários/estatística & dados numéricos , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Mamografia , Pessoa de Meia-Idade , Estudos Retrospectivos
8.
Biomed Pharmacother ; 94: 430-438, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28778046

RESUMO

Salicornia europaea L. (SE) has been used as folk medicine for the treatment of various diseases such as obesity, diabetes, and cancer. However, its effects on atherosclerotic events in vascular smooth muscle cells (VSMCs) remain unknown. The present study explored the effects of the ethyl acetate fraction of desalted SE hot water extract (SEWEAF) on atherosclerotic responses (especially migration and proliferation) in VSMCs and vascular neointima formation. Treatment with the SEWEAF significantly suppressed the platelet-derived growth factor (PDGF)-BB-induced VSMC migration and proliferation as well the phosphorylation of mitogen-activated protein kinases (MAPKs) such as the p38 MAPK and extracellular signal-regulated kinase (ERK) 1/2. Moreover, oral administration of the SEWEAF resulted in the attenuation of neointima formation in balloon-injured rat carotid arteries. Additionally, HPLC analysis showed that the major components in the two subfractions of the SEWEAF were five phenolic acids and four flavonols. In the SEWEAF components, for which atherosclerosis-linked responses in VSMCs have not been known, p-coumaric acid, quercetin-3-ß-d-glucoside, and isorhamnetin-3-ß-d-glucoside inhibited both PDGF-BB-induced migration and proliferation and isorhamnetin attenuated only PDGF-BB-stimulated VSMC proliferation. These results suggest that the SEWEAF may suppress PDGF-BB-induced VSMC migration by downregulating the phosphorylation of p38 MAPK and ERK1/2, thus leading to the reduction of neointimal hyperplasia during vascular remodeling. Therefore, the desalted SE extract, SEWEAF may be a potential ingredient for dietary supplements or nutraceuticals to ameliorate and/or prevent vascular remodeling-related disorders.


Assuntos
Movimento Celular/efeitos dos fármacos , Chenopodiaceae/química , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Neointima/enzimologia , Neointima/patologia , Extratos Vegetais/farmacologia , Acetatos/química , Animais , Becaplermina , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Flavonóis/farmacologia , Hidroxibenzoatos/farmacologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/farmacologia , Ratos Sprague-Dawley , Água
9.
PLoS Biol ; 15(7): e2001246, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28683067

RESUMO

Clinical and preclinical studies indicate that early postnatal exposure to anesthetics can lead to lasting deficits in learning and other cognitive processes. The mechanism underlying this phenomenon has not been clarified and there is no treatment currently available. Recent evidence suggests that anesthetics might cause persistent deficits in cognitive function by disrupting key events in brain development. The hippocampus, a brain region that is critical for learning and memory, contains a large number of neurons that develop in the early postnatal period, which are thus vulnerable to perturbation by anesthetic exposure. Using an in vivo mouse model we demonstrate abnormal development of dendrite arbors and dendritic spines in newly generated dentate gyrus granule cell neurons of the hippocampus after a clinically relevant isoflurane anesthesia exposure conducted at an early postnatal age. Furthermore, we find that isoflurane causes a sustained increase in activity in the mechanistic target of rapamycin pathway, and that inhibition of this pathway with rapamycin not only reverses the observed changes in neuronal development, but also substantially improves performance on behavioral tasks of spatial learning and memory that are impaired by isoflurane exposure. We conclude that isoflurane disrupts the development of hippocampal neurons generated in the early postnatal period by activating a well-defined neurodevelopmental disease pathway and that this phenotype can be reversed by pharmacologic inhibition.


Assuntos
Anestésicos Inalatórios/toxicidade , Disfunção Cognitiva/induzido quimicamente , Hipocampo/efeitos dos fármacos , Isoflurano/toxicidade , Serina-Treonina Quinases TOR/metabolismo , Animais , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Exposição Ambiental , Hipocampo/crescimento & desenvolvimento , Hipocampo/patologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/patologia
10.
Neurotoxicol Teratol ; 60: 33-39, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27751818

RESUMO

The mechanism by which anesthetics might act on the developing brain in order to cause long term deficits remains incompletely understood. The hippocampus has been identified as a structure that is likely to be involved, as rodent models show numerous deficits in behavioral tasks of learning that are hippocampal-dependent. The hippocampus is an unusual structure in that it is the site of large amounts of neurogenesis postnatally, particularly in the first year of life in humans, and these newly generated neurons are critical to the function of this structure. Intriguingly, neurogenesis is a major developmental event that occurs during postulated windows of vulnerability to developmental anesthetic neurotoxicity across the different species in which it has been studied. In this review, we examine the evidence for anesthetic effects on neurogenesis in the early postnatal period and ask whether neurogenesis should be studied further as a putative mechanism of injury. Multiple anesthetics are considered, and both in vivo and in vitro work is presented. While there is abundant evidence that anesthetics act to suppress neurogenesis at several different phases, evidence of a causal link between these effects and any change in learning behavior remains elusive.


Assuntos
Anestésicos/efeitos adversos , Encéfalo/crescimento & desenvolvimento , Neurogênese/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Animais , Humanos
11.
J Neurosurg Anesthesiol ; 28(4): 405-412, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27768676

RESUMO

BACKGROUND: Early postnatal exposure to general anesthetic agents causes a lasting impairment in learning and memory in animal models. One hypothesis to explain this finding is that exposure to anesthetic agents during critical points in neural development disrupts the formation of brain circuitry. Here, we explore the effects of sevoflurane on the neuronal growth cone, a specialization at the growing end of axons and dendrites that is responsible for the targeted growth that underlies connectivity between neurons. METHODS: Dissociated neuronal cultures were prepared from embryonic mouse neocortex. Time-lapse images of live growth cones exposed to anesthetics were taken using differential interference contrast microscopy, and the rate of change of the area of the lamellipodia and the speed of the filopodial tip were quantified as measures of motility. The involvement of the p75 neurotropin receptor (p75NTR) was tested using inhibitors applied to the media and by a coimmunoprecipitation assay. RESULTS: The rate of lamellipodial area change and filopodial tip velocity in both axonal and dendritic growth cones was significantly reduced with sevoflurane exposure between 2% and 6%. Motility could be substantially restored by treatment with Y27632 and TAT-peptide 5, which are inhibitors of Rho Kinase and p75NTR, respectively. Sevoflurane results in reduced coimmunoprecipitation of Rho-Guanosine-5'-diphosphate dissociation inhibitor after pulldown with p75NTR. CONCLUSIONS: Sevoflurane interferes with growth cone motility, which is a critical process in brain circuitry formation. Our data suggest that this may occur through an action on the p75NTR, which promotes growth inhibitory signaling by the Rho pathway.


Assuntos
Anestésicos Inalatórios/efeitos adversos , Cones de Crescimento/efeitos dos fármacos , Éteres Metílicos/efeitos adversos , Animais , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Sevoflurano , Transdução de Sinais/efeitos dos fármacos
12.
Nat Commun ; 6: 10119, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26658127

RESUMO

Down syndrome is the most common chromosomal disorder affecting the nervous system in humans. To date, investigations of neural anomalies in Down syndrome have focused on the central nervous system, although dysfunction of the peripheral nervous system is a common manifestation. The molecular and cellular bases underlying peripheral abnormalities have remained undefined. Here, we report the developmental loss of sympathetic innervation in human Down syndrome organs and in a mouse model. We show that excess regulator of calcineurin 1 (RCAN1), an endogenous inhibitor of the calcineurin phosphatase that is triplicated in Down syndrome, impairs neurotrophic support of sympathetic neurons by inhibiting endocytosis of the nerve growth factor (NGF) receptor, TrkA. Genetically correcting RCAN1 levels in Down syndrome mice markedly improves NGF-dependent receptor trafficking, neuronal survival and innervation. These results uncover a critical link between calcineurin signalling, impaired neurotrophin trafficking and neurodevelopmental deficits in the peripheral nervous system in Down syndrome.


Assuntos
Síndrome de Down/metabolismo , Regulação da Expressão Gênica/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Musculares/metabolismo , Fatores de Crescimento Neural/metabolismo , Transporte Proteico/fisiologia , Sistema Nervoso Simpático/crescimento & desenvolvimento , Animais , Proteínas de Ligação ao Cálcio , Dinamina I/genética , Dinamina I/metabolismo , Endocitose , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos , Proteínas Musculares/genética , Fatores de Crescimento Neural/genética , Fosforilação , Receptor trkA/genética , Receptor trkA/metabolismo , Sistema Nervoso Simpático/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
14.
Dev Neurosci ; 37(4-5): 428-39, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26087836

RESUMO

Neonatal hypoxic-ischemic injury (HI) results in widespread cerebral encephalopathy and affects structures that are essential for neurocognitive function, such as the hippocampus. The dentate gyrus contains a reservoir of neural stem and progenitor cells (NSPCs) that are critical for postnatal development and normal adult function of the hippocampus, and may also facilitate the recovery of function after injury. Using a neonatal mouse model of mild-to-moderate HI and immunohistochemical analysis of NSPC development markers, we asked whether these cells are vulnerable to HI and how they respond to both injury and hypothermic therapy. We found that cleaved caspase-3 labeling in the subgranular zone, where NSPCs are located, is increased by more than 30-fold after HI. The population of cells positive for both proliferating cell nuclear antigen and nestin (PCNA+Nes+), which represent primarily actively proliferating NSPCs, are acutely decreased by 68% after HI. The NSPC population expressing NeuroD1, a marker for NSPCs transitioning to become fate-committed neural progenitors, was decreased by 47%. One week after HI, there was a decrease in neuroblasts and immature neurons in the dentate gyrus, as measured by doublecortin (DCX) immunolabeling, and at the same time PCNA+Nes+ cell density was increased by 71%. NSPCs expressing Tbr2, which identifies a highly proliferative intermediate neural progenitor population, increased by 107%. Hypothermia treatment after HI partially rescues both the acute decrease in PCNA+Nes+ cell density at 1 day after injury and the chronic loss of DCX immunoreactivity and reduction in NeuroD1 cell density measured at 1 week after injury. Thus, we conclude that HI causes an acute loss of dentate gyrus NSPCs, and that hypothermia partially protects NSPCs from HI.


Assuntos
Giro Denteado/patologia , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/terapia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Animais Recém-Nascidos , Giro Denteado/citologia , Modelos Animais de Doenças , Proteína Duplacortina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia
15.
Oncotarget ; 6(12): 9820-33, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25888632

RESUMO

Signaling by small guanosine triphosphatases (GTPase), Rap1/Rac1, is one of the major pathways controlling cancer cell migration and tumor metastasis. Thymosin beta-4 (Tß4), an actin-sequestering protein, has been shown to increase migration of cancer cells. Episodes of hypoxia and re-oxygenation (H/R) are an important phenomenon in tumor microenvironment (TME). We investigated whether Tß4 could play as an intermediary to crosstalk between Rac1- and Rap1- GTPase activation under hypoxia/reoxygenation (H/R) conditions. Inhibition of Tß4 expression using transcription activator-like effector nucleases (TALEN) significantly decreased lung metastasis of B16F10 cells. Rac1 and Rap1 activity, as well as cancer cell migration, increased following induction of Tß4 expression in normoxia- or H/R-experienced cells, but were barely detectable in Tß4-depleted cells. Rap1-regulated Rac1 activity was decreased by a dominant negative Rap1 (Rap1N17), and increased by 8-(4-chloro-phenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate (CPT), a Rap1 activator. In contrast, a Rac1-specific inhibitor, NSC23766, and dominant negative Rac1 (Rac1N17) enhanced Tß4 expression and aberrant Rap1 activity. While NSC23766 and Rac1N17 incompletely inhibited tumor metastasis in vivo, and H/R-experienced cancer cell migration in vitro, more efficient attenuation of cancer cell migration was accomplished by simultaneous inactivation of Rap1 and Rac1 with Rap1N17 and Rac1N17, respectively. These data suggest that a combination therapy targeting both Rap1 and Rac1 activity may be an effective method of inhibiting tumor metastasis.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Regulação Neoplásica da Expressão Gênica , Oxigênio/química , Proteínas de Ligação a Telômeros/metabolismo , Timosina/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Aminoquinolinas/química , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Movimento Celular , Ativação Enzimática , Células HeLa , Humanos , Masculino , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Neuropeptídeos/metabolismo , Pirimidinas/química , Complexo Shelterina , Transdução de Sinais , Microambiente Tumoral , Técnicas do Sistema de Duplo-Híbrido
16.
Biomol Ther (Seoul) ; 23(1): 19-25, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25593639

RESUMO

Vascular endothelial growth factor (VEGF) is an important regulator of neovascularization. Hypoxia inducible nitric oxide (NO) enhanced the expression of VEGF and thymosin beta-4 (Tß4), actin sequestering protein. Here, we investigated whether NO-mediated VEGF expression could be regulated by Tß4 expression in HeLa cervical cancer cells. Hypoxia inducible NO production and VEGF expression were reduced by small interference (si) RNA of Tß4. Hypoxia response element (HRE)-luciferase activity and VEGF expression were increased by the treatment with N-(ß-D-Glucopyranosyl)-N2-acetyl-S-nitroso-D, L-penicillaminamide (SNAP-1), to generate NO, which was inhibited by the inhibition of Tß4 expression with Tß4-siRNA. In hypoxic condition, HRE-luciferase activity and VEGF expression were inhibited by the treatment with N(G)-monomethyl-L-arginine (L-NMMA), an inhibitor to nitric oxide synthase (NOS), which is accompanied with a decrease in Tß4 expression. VEGF expression inhibited by L-NMMA treatment was restored by the transfection with pCMV-Tß4 plasmids for Tß4 overexpression. Taken together, these results suggest that Tß4 could be a regulator for the expression of VEGF via the maintenance of NOS activity.

17.
J Leukoc Biol ; 97(4): 711-21, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25632046

RESUMO

RT is commonly used to treat malignant tumors. However, tumor regrowth is a major limitation to RT as an antitumor treatment. In the present study, we investigated the tumor-promoting effects of high-dose (or ablative) RT treatments on tumor-bearing mice. We focused on the role of macrophages that interact with IR-CCs in the TME, which cause tumor regrowth. We observed that CT26(H-2(d)) tumor growth was enhanced by i.v. injection of IR-CT26 cells compared with NR control CT26 cells. The levels of iNOS gene expression and NO production from RAW264.7 macrophages (H-2(d)) in response to the interaction with IR-CT26 cells were higher than with NR-CT26 cells. When CT26 tumor-bearing mice were treated i.v. with L-NMMA, a NOS inhibitor, the reduction in in vivo tumor growth was higher in the IR-CT26-injected group compared with the NR-CT26-injected control group. In vivo CT26 tumor growth was decreased after transplanting PEM extracted from L-NMMA-treated, tumor-bearing mice. Although iNOS activity was reduced by inhibiting TLR1 expression with TLR1-siRNA, it was enhanced by TLR1 overexpression. Transcriptional activation and protein expression levels of iNOS were also decreased in the presence of TLR1-siRNA but increased as a result of TLR1 overexpression. These results demonstrate that postradiotherapeutic tumor regrowth may be caused by interaction of IR-CCs with macrophages that induce TLR1-mediated iNOS expression and NO production. Our data suggest that iNOS in macrophages could be a useful target to regulate postradiotherapeutic responses in hosts and subsequently limit tumor regrowth.


Assuntos
Adenocarcinoma/radioterapia , Neoplasias do Colo/radioterapia , Raios gama , Macrófagos/metabolismo , Melanoma Experimental/radioterapia , Proteínas de Neoplasias/fisiologia , Óxido Nítrico Sintase Tipo II/fisiologia , Óxido Nítrico/fisiologia , Receptor 1 Toll-Like/fisiologia , Microambiente Tumoral/efeitos da radiação , Células 3T3 , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Células da Medula Óssea/metabolismo , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Técnicas de Cocultura , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Progressão da Doença , Indução Enzimática , Macrófagos/classificação , Macrófagos Peritoneais/metabolismo , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Recidiva , Receptor 1 Toll-Like/biossíntese , Receptor 1 Toll-Like/genética , ômega-N-Metilarginina/farmacologia
19.
Nephron Exp Nephrol ; 128(1-2): 80-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25412793

RESUMO

BACKGROUND: Wnt5a is important for the development of various organs and postnatal cellular function. Little is known, however, about the role of Wnt5a in kidney development, although WNT5A mutations were identified in patients with Robinow syndrome, a genetic disease which includes developmental defects in kidneys. Our goal in this study was to determine the role of Wnt5a in kidney development. METHODS: Whole-mount in situ hybridization was used to establish the expression pattern of Wnt5a during kidney development. Zebrafish with wnt5a knockdown and Wnt5a global knockout mice were used to identify kidney phenotypes. RESULTS: In zebrafish, wnt5a knockdown resulted in glomerular cyst formation and dilated renal tubules. In mice, Wnt5a global knockout resulted in pleiotropic, but severe, kidney phenotypes, including agenesis, fused kidney, hydronephrosis and duplex kidney/ureter. CONCLUSIONS: Our data demonstrated the important role of Wnt5a in kidney development. Disrupted Wnt5a resulted in kidney cysts in zebrafish and pleiotropic abnormal kidney development in mice.


Assuntos
Rim/embriologia , Rim/fisiologia , Proteínas Wnt/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Técnicas de Inativação de Genes , Incidência , Rim/anormalidades , Doenças Renais Císticas/epidemiologia , Doenças Renais Císticas/etiologia , Doenças Renais Císticas/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Proteínas Wnt/deficiência , Proteínas Wnt/genética , Proteína Wnt-5a , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
20.
PLoS One ; 9(10): e106532, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25271630

RESUMO

The actin-sequestering protein thymosin beta-4 (Tß4) is involved in various cellular and physiological processes such as proliferation, motility, growth and metastasis. Nitric oxide (NO) promotes tumor invasiveness and metastasis by activating various enzymes. Herein, we investigated whether hypoxia-inducible NO regulates Tß4 expression and cancer cell migration using HeLa cervical cancer cells. NO production and Tß4 expression were increased in a hypoxic condition. The treatment with N-(ß-D-Glucopyranosyl)-N2-acetyl-S-nitroso-D, L-penicillaminamide (SNAP-1), to generate NO, enhanced the transcription of Tß4 and cancer cell migration. SNAP-1-induced cell migration was decreased by the inhibition of Tß4 with small interference (si) RNA. In a hypoxic condition, treatment with N(G)-monomethyl-L-arginine (L-NMMA), nitric oxide synthase (NOS) inhibitor, reduced Tß4 transcriptional activity, and hypoxia-inducible factor (HIF)-1α. Hypoxia-induced cancer cell migration was also decreased by L-NMMA treatment. In a normoxic condition, Tß4 transcriptional activity was decreased in the cells incubated in the presence of L-NMMA after co-transfection with Tß4 promoter and GST-conjugated HIF-1α. Collectively, these results suggest that NO could regulate the expression of Tß4 by direct or indirect effect of HIF-1α on Tß4 promoter.


Assuntos
Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Timosina/genética , Timosina/metabolismo , Actinas/metabolismo , Movimento Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Hipóxia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/antagonistas & inibidores , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/genética , ômega-N-Metilarginina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...