Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(11)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38892038

RESUMO

The effects of the enzyme N-acetylgalactosamine-4-sulfatase (Arylsulfatase B, ARSB), which removes the 4-sulfate group at the non-reducing end of chondroitin 4-sulfate, on the expression of PD-L1 were determined, and the underlying mechanism of PD-L1 expression was elucidated. Initial experiments in human melanoma cells (A375) showed that PD-L1 expression increased from 357 ± 31 to 796 ± 50 pg/mg protein (p < 10-11) when ARSB was silenced in A375 cells. In subcutaneous B16F10 murine melanomas, PD-L1 declined from 1227 ± 189 to 583 ± 110 pg/mg protein (p = 1.67 × 10-7), a decline of 52%, following treatment with exogenous, bioactive recombinant ARSB. This decline occurred in association with reduced tumor growth and prolongation of survival, as previously reported. The mechanism of regulation of PD-L1 expression by ARSB is attributed to ARSB-mediated alteration in chondroitin 4-sulfation, leading to changes in free galectin-3, c-Jun nuclear localization, HDAC3 expression, and effects of acetyl-H3 on the PD-L1 promoter. These findings indicate that changes in ARSB contribute to the expression of PD-L1 in melanoma and can thereby affect the immune checkpoint response. Exogenous ARSB acted on melanoma cells and normal melanocytes through the IGF2 receptor. The decline in PD-L1 expression by exogenous ARSB may contribute to the impact of ARSB on melanoma progression.


Assuntos
Antígeno B7-H1 , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases , Melanoma Experimental , Melanoma , N-Acetilgalactosamina-4-Sulfatase , Animais , Humanos , Camundongos , N-Acetilgalactosamina-4-Sulfatase/metabolismo , N-Acetilgalactosamina-4-Sulfatase/genética , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Histona Desacetilases/metabolismo , Histona Desacetilases/genética , Linhagem Celular Tumoral , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/genética , Melanoma/metabolismo , Melanoma/genética , Melanoma/patologia , Galectina 3/metabolismo , Galectina 3/genética , Regiões Promotoras Genéticas , Proteínas Sanguíneas , Galectinas
3.
Sci Adv ; 10(7): eadi5501, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38354243

RESUMO

Osteoarthritis (OA) is characterized by cartilage damage, inflammation, and pain. Vascular endothelial growth factor receptors (VEGFRs) have been associated with OA severity, suggesting that inhibitors targeting these receptors alleviate pain (via VEGFR1) or cartilage degeneration (via VEGFR2). We have developed a nanoparticle-based formulation of pazopanib (Votrient), an FDA-approved anticancer drug that targets both VEGFR1 and VEGFR2 (Nano-PAZII). We demonstrate that a single intraarticular injection of Nano-PAZII can effectively reduce joint pain for a prolonged time without substantial side effects in two different preclinical OA rodent models involving either surgical (upon partial medial meniscectomy) or nonsurgical induction (with monoiodoacetate). The injection of Nano-PAZII blocks VEGFR1 and relieves OA pain by suppressing sensory neuronal ingrowth into the knee synovium and neuronal plasticity in the dorsal root ganglia and spinal cord. Simultaneously, the inhibition of VEGFR2 reduces cartilage degeneration. These findings provide a mechanism-based disease-modifying drug strategy that addresses both pain symptoms and cartilage loss in OA.


Assuntos
Osteoartrite , Fator A de Crescimento do Endotélio Vascular , Animais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Osteoartrite/tratamento farmacológico , Osteoartrite/etiologia , Osteoartrite/metabolismo , Dor/tratamento farmacológico , Dor/etiologia , Articulação do Joelho/metabolismo , Artralgia , Modelos Animais de Doenças
4.
Gene ; 893: 147920, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-37890601

RESUMO

Pain is the prime symptom of osteoarthritis (OA) that directly affects the quality of life. Protein kinase Cδ (PKCδ/Prkcd) plays a critical role in OA pathogenesis; however, its significance in OA-related pain is not entirely understood. The present study investigated the functional role of PKCδ in OA pain sensation. OA was surgically induced in control (Prkcdfl/fl), global- (Prkcdfl/fl; ROSACreERT2), and sensory neuron-specific conditional knockout (cKO) mice (Prkcdfl/fl; NaV1.8/Scn10aCreERT2) followed by comprehensive analysis of longitudinal behavioral pain, histopathology and immunofluorescence studies. GlobalPrkcd cKO mice prevented cartilage deterioration by inhibiting matrix metalloproteinase-13 (MMP13) in joint tissues but significantly increased OA pain. Sensory neuron-specificdeletion of Prkcd in mice did not protect cartilage from degeneration but worsened OA-associated pain. Exacerbated pain sensitivity observed in global- and sensory neuron-specific cKO of Prkcd was corroborated with markedly increased specific pain mediators in knee synovium and dorsal root ganglia (DRG). These specific pain markers include nerve growth factor (NGF) and vascular endothelial growth factor (VEGF), and their cognate receptors, including tropomyosin receptor kinase A (TrkA) and vascular endothelial growth factor receptor-1 (VEGFR1). The increased levels of NGF/TrkA and VEGF/VEGFR1 were comparable in both global- and sensory neuron-specific cKO groups. These data suggest that the absence of Prkcd gene expression in the sensory neurons is strongly associated with OA hyperalgesia independent of cartilage protection. Thus, inhibition of PKCδ may be beneficial for cartilage homeostasis but could aggravate OA-related pain symptoms.


Assuntos
Hiperalgesia , Osteoartrite , Animais , Camundongos , Modelos Animais de Doenças , Hiperalgesia/genética , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , Osteoartrite/metabolismo , Dor/complicações , Dor/genética , Qualidade de Vida , Fator A de Crescimento do Endotélio Vascular/genética
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166913, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37813168

RESUMO

In the syngeneic, subcutaneous B16F10 mouse model of malignant melanoma, treatment with exogenous ARSB markedly reduced tumor size and extended survival. In vivo experiments showed that local treatment with exogenous N-acetylgalactosamine-4-sulfatase (Arylsulfatase B; ARSB) led to reduced tumor growth over time (p < 0.0001) and improved the probability of survival up to 21 days (p = 0.0391). Tumor tissue from the treated mice had lower chondroitin 4-sulfate (C4S) content and lower sulfotransferase activity. The free galectin-3 declined, and the SHP2 activity increased, due to altered binding with chondroitin 4-sulfate. These changes induced effects on transcription, which were mediated by Sp1, phospho-ERK1/2, and phospho-p38 MAPK. Reduced mRNA expression of chondroitin sulfate proteoglycan 4 (CSPG4), carbohydrate sulfotransferase 15 (N-acetylgalactosamine 4-sulfate 6-O-sulfotransferase), and matrix metalloproteinases 2 and 9 resulted. Experiments in the human melanoma cell line A375 demonstrated similar responses to exogenous ARSB as in the tumors, and inverse effects followed ARSB siRNA. ARSB, which removes the 4-sulfate group at the non-reducing end of C4S, acts as a tumor suppressor, and treatment with exogenous ARSB impacts on vital cell signaling and reduces the expression of critical genes associated with melanoma progression.


Assuntos
Melanoma , N-Acetilgalactosamina-4-Sulfatase , Neoplasias Cutâneas , Animais , Humanos , Camundongos , Sulfatos de Condroitina/metabolismo , Melanoma/tratamento farmacológico , N-Acetilgalactosamina-4-Sulfatase/genética , N-Acetilgalactosamina-4-Sulfatase/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/tratamento farmacológico , Melanoma Maligno Cutâneo
6.
Int J Biol Sci ; 19(2): 675-690, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36632459

RESUMO

Pain is the major reason that patients suffering from osteoarthritis (OA) seek medical care. We found that vascular endothelial growth factors (VEGFs) mediate signaling in OA pain pathways. To determine the specific contributions of VEGFs and their receptors (VEGFRs) to joint pathology and pain transmission during OA progression, we studied intra-articular (IA) injections of VEGF ligands into murine knee joints. Only VEGF ligands specific for the activation of VEGFR1, but not VEGFR2, induced allodynia within 30 min. Interventions in OA by inhibitors of VEGFRs were done in vivo using a preclinical murine OA model by IA injections of selective inhibitors of VEGFR1/VEGFR2 kinase (pazopanib) or VEGFR2 kinase (vandetanib). OA phenotypes were evaluated using pain-associated murine behavioral tests and histopathologic analyses. Alterations in VEGF/VEGFR signaling by drugs were determined in knee joints, dorsal root ganglia, and spinal cord by immunofluorescence microscopy. Pazopanib immediately relieved OA pain by interfering with pain transmission pathways. Pain reduction by vandetanib was mainly due to the inhibition of cartilage degeneration by suppressing VEGFR2 expression. In conclusion, IA administration of pazopanib, which simultaneously inhibits VEGFR1 and VEGFR2, can be developed as an ideal OA disease-modifying drug that rapidly reduces joint pain and simultaneously inhibits cartilage degeneration.


Assuntos
Terapia de Alvo Molecular , Osteoartrite , Receptores de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Animais , Camundongos , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Dor/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
7.
Int J Mol Sci ; 23(20)2022 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-36292950

RESUMO

Tropomyosin receptor kinase A (TrkA/NTRK1) is a high-affinity receptor for nerve growth factor (NGF), a potent pain mediator. NGF/TrkA signaling elevates synovial sensory neuronal distributions in the joints and causes osteoarthritis (OA) pain. We investigated the mechanisms of pain transmission as to whether peripheral sensory neurons are linked to the cellular plasticity in the dorsal root ganglia (DRG) and are critical for OA hyperalgesia. Sensory neuron-specific deletion of TrkA was achieved by tamoxifen injection in 4-week-old TrkAfl/fl;NaV1.8CreERT2 (Ntrk1 fl/fl;Scn10aCreERT2) mice. OA was induced by partial medial meniscectomy (PMM) in 12-week-old mice, and OA-pain-related behavior was analyzed for 12 weeks followed by comprehensive histopathological examinations. OA-associated joint pain was markedly improved without cartilage protection in sensory-neuron-specific conditional TrkA knock-out (cKO) mice. Alleviated hyperalgesia was associated with suppression of the NGF/TrkA pathway and reduced angiogenesis in fibroblast-like synovial cells. Elevated pain transmitters in the DRG of OA-induced mice were significantly diminished in sensory-neuron-specific TrkA cKO and global TrkA cKO mice. Spinal glial activity and brain-derived neurotropic factor (BDNF) were significantly increased in OA-induced mice but were substantially eliminated by sensory-neuron-specific deletion. Our results suggest that augmentation of NGF/TrkA signaling in the joint synovium and the peripheral sensory neurons facilitate pro-nociception and centralized pain sensitization.


Assuntos
Fator de Crescimento Neural , Osteoartrite , Camundongos , Animais , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo , Tropomiosina/metabolismo , Hiperalgesia/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Receptoras Sensoriais/metabolismo , Dor/metabolismo , Gânglios Espinais/metabolismo , Osteoartrite/metabolismo , Tamoxifeno/metabolismo
8.
J Cell Physiol ; 235(6): 5305-5317, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31875985

RESUMO

Although degenerative disc disease (DDD) and related low back pain (LBP) are growing public health problems, the underlying disease mechanisms remain unclear. An increase in the vascular endothelial growth factor (VEGF) levels in DDD has been reported. This study aimed to examine the role of VEGF receptors (VEGFRs) in DDD, using a mouse model of DDD. Progressive DDD was induced by anterior stabbing of lumbar intervertebral discs in wild type (WT) and VEGFR-1 tyrosine-kinase deficient mice (vegfr-1TK-/- ). Pain assessments were performed weekly for 12 weeks. Histological and immunohistochemical assessments were made for discs, dorsal root ganglions, and spinal cord. Both vegfr-1TK-/- and WT mice presented with similar pathological changes in discs with an increased expression of inflammatory cytokines and matrix-degrading enzymes. Despite the similar pathological patterns, vegfr-1TK-/- mice showed insensitivity to pain compared with WT mice. This insensitivity to discogenic pain was related to lower levels of pain factors in the discs and peripheral sensory neurons and lower spinal glial activation in the vegfr-1TK- /- mice than in the WT mice. Exogenous stimulation of bovine disc cells with VEGF increased inflammatory and cartilage degrading enzyme. Silencing vegfr-1 by small-interfering-RNA decreased VEGF-induced expression of pain markers, while silencing vegfr-2 decreased VEGF-induced expression of inflammatory and metabolic markers without changing pain markers. This suggests the involvement of VEGFR-1 signaling specifically in pain transmission. Collectively, our results indicate that the VEGF signaling is involved in DDD. Particularly, VEGFR-1 is critical for discogenic LBP transmission independent of the degree of disc pathology.


Assuntos
Disco Intervertebral/metabolismo , Dor Lombar/genética , Vértebras Lombares/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Modelos Animais de Doenças , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Regulação da Expressão Gênica/genética , Humanos , Disco Intervertebral/lesões , Disco Intervertebral/patologia , Dor Lombar/patologia , Vértebras Lombares/lesões , Vértebras Lombares/patologia , Camundongos , Medição da Dor , Transdução de Sinais/genética
9.
J Biol Chem ; 294(3): 1059-1069, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30459233

RESUMO

FoxO proteins are major targets of insulin action, and FoxO1 mediates the effects of insulin on hepatic glucose metabolism. We reported previously that serpinB1 is a liver-secreted factor (hepatokine) that promotes adaptive ß-cell proliferation in response to insulin resistance in the liver-specific insulin receptor knockout (LIRKO) mouse. Here we report that FoxO1 plays a critical role in promoting serpinB1 expression in hepatic insulin resistance in a non-cell-autonomous manner. Mice lacking both the insulin receptor and FoxO1 (LIRFKO) exhibit reduced ß-cell mass compared with LIRKO mice because of attenuation of ß-cell proliferation. Although hepatic expression of serpinB1 mRNA and protein levels was increased in LIRKO mice, both the mRNA and protein levels returned to control levels in LIRFKO mice. Furthermore, liver-specific expression of constitutively active FoxO1 in transgenic mice induced an increase in hepatic serpinB1 mRNA and protein levels in refed mice. Conversely, serpinB1 mRNA and protein levels were reduced in mice lacking FoxO proteins in the liver. ChIP studies demonstrated that FoxO1 binds to three distinct sites located ∼9 kb upstream of the serpinb1 gene in primary mouse hepatocytes and that this binding is enhanced in hepatocytes from LIRKO mice. However, adenoviral expression of WT or constitutively active FoxO1 and insulin treatment are sufficient to regulate other FoxO1 target genes (IGFBP-1 and PEPCK) but not serpinB1 expression in mouse primary hepatocytes. These results indicate that liver FoxO1 promotes serpinB1 expression in hepatic insulin resistance and that non-cell-autonomous factors contribute to FoxO1-dependent effects on serpinB1 expression in the liver.


Assuntos
Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Fígado/metabolismo , Serpinas/biossíntese , Animais , Proteína Forkhead Box O1/genética , Hepatócitos/citologia , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fígado/citologia , Masculino , Camundongos , Camundongos Transgênicos , Fosfoenolpiruvato Carboxiquinase (ATP)/genética , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Serpinas/genética
10.
Gene ; 655: 1-12, 2018 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-29474860

RESUMO

Environmental disruption of the circadian rhythm is linked with increased pain due to osteoarthritis (OA). We aimed to characterize the role of the clock gene in OA-induced pain more systemically using both genetic and pharmacological approaches. Genetically modified mice, (bmal1f/fNav1.8CreERT mice), generated by deleting the critical clock gene, bmal1, from Nav1.8 sensory neurons, were resistant to the development of mechanical hyperalgesia associated with OA induced by partial medial meniscectomy (PMM) of the knee. In wild-type mice, induction of OA by PMM surgery led to a substantial increase in BMAL1 expression in DRG neurons. Interestingly, pharmacological activation of the REV-ERB (a negative regulator of bmal1 transcription) with SR9009 resulted in reduction of BMAL1 expression, and a significant decrease in mechanical hyperalgesia associated with OA. Cartilage degeneration was also significantly reduced in mice treated with the REV-ERB agonist SR9009. Based on these data, we also assessed the effect of pharmacological activation of REV-ERB using a model of environmental circadian disruption with its associated mechanical hyperalgesia, and noted that SR9009 was an effective analgesic in this model as well. Our data clearly demonstrate that genetic disruption of the molecular clock, via deletion of bmal1 in the sensory neurons of the DRG, decreases pain in a model of OA. Furthermore, pharmacological activation of REV-ERB leading to suppression of BMAL1 expression may be an effective method for treating OA-related pain, as well as to reduce joint damage associated with this disease.


Assuntos
Analgésicos/uso terapêutico , Artralgia/tratamento farmacológico , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Terapia de Alvo Molecular/métodos , Osteoartrite/tratamento farmacológico , Animais , Artralgia/genética , Proteínas CLOCK/genética , Feminino , Hiperalgesia/tratamento farmacológico , Hiperalgesia/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Osteoartrite/genética
11.
Cell Rep ; 22(2): 523-534, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29320745

RESUMO

Liver-specific disruption of the type 2 deiodinase gene (Alb-D2KO) results in resistance to both diet-induced obesity and liver steatosis in mice. Here, we report that this is explained by an ∼60% reduction in liver zinc-finger protein-125 (Zfp125) expression. Zfp125 is a Foxo1-inducible transcriptional repressor that causes lipid accumulation in the AML12 mouse hepatic cell line and liver steatosis in mice by reducing liver secretion of triglycerides and hepatocyte efflux of cholesterol. Zfp125 acts by repressing 18 genes involved in lipoprotein structure, lipid binding, and transport. The ApoE promoter contains a functional Zfp125-binding element that is also present in 17 other lipid-related genes repressed by Zfp125. While liver-specific knockdown of Zfp125 causes an "Alb-D2KO-like" metabolic phenotype, liver-specific normalization of Zfp125 expression in Alb-D2KO mice rescues the phenotype, restoring normal susceptibility to diet-induced obesity, liver steatosis, and hypercholesterolemia.


Assuntos
Proteínas de Ligação a DNA/genética , Fígado Gorduroso/genética , Proteína Forkhead Box O1/genética , Hipercolesterolemia/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Fígado Gorduroso/patologia , Proteína Forkhead Box O1/metabolismo , Camundongos
12.
Gene Rep ; 11: 94-100, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30873504

RESUMO

Osteoarthritis (OA) is a painful and debilitating disease. A striking feature of OA is the dramatic increase in vascular endothelial growth factor (VEGF) levels and in new blood vessel formation in the joints, both of which correlate with the severity of OA pain. Our aim was to determine whether anti-VEGF monoclonal antibodies (mAbs) - MF-1 (mAb to VEGFR1) and DC101 (mAb to VEGFR2) - can reduce OA pain and can do so by targeting VEGF signaling pathways such as Flt-1 (VEGFR1) and Flk-1 (VEGFR2). After IACUC approval, OA was induced by partial medial meniscectomy (PMM) in C57/BL6 mice (20 g). ln the first experiment, for validation of VEGFR1 in DRG, the mouse dorsal root ganglion (DRG) was stimulated with NGF for 48 hours to find the relative gene induction for VEGFR1 vs. 18S by RT-PCR. In the second experiment, Biotin-conjugated VEGFA (1 µg/knee joint) was administered in the left knee joint of mice with advanced OA in order to characterization of VEGFR1 and VEGFR2. pVEGFR1/VEGFR2 was detected by immunostaining in DRGs. Finally, MF-1 and DC101 were administered in OA mice by both intrathecal (IT) and intraarticular (IA) injections, and the change in paw withdrawal threshold (PWT) was measured. Retrograde transport of VEGF was confirmed for detection of pVEGFR1/VEGFR2 in the DRG. PMM surgery led to development of OA and mechanical allodynia, with reduced paw withdrawal thresholds (PWT) (P<0.0001). IT injection of MF-1 led to a reduction of allodynia in advanced OA, but injection of DC101 did not. IA injection of MF-1 or DC101 at one week after PMM injury did not reduce allodynia, but when injected in advanced OA mice joints at 12 weeks, both Mabs increased PWT an indicator of analgesia. Our data show that MF-1 (VEGR1 inhibition) decreases pain in advanced OA after IT or IA injection. Activation of MF-1 or DC101 may ameliorate OA-related joint pain.

13.
J Cell Physiol ; 233(10): 6589-6602, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29150945

RESUMO

Discogenic low back pain (DLBP) is extremely common and costly. Effective treatments are lacking due to DLBP's unknown pathogenesis. Currently, there are no in vivo mouse models of DLBP, which restricts research in this field. The aim of this study was to establish a reliable DLBP model in mouse that captures the pathological changes in the disc and allows longitudinal pain testing. The model was generated by puncturing the mouse lumbar discs (L4/5, L5/6, and L6/S1) and removing the nucleus pulposus using a microscalpel under the microscope. Histology, molecular pathways, and pain-related behaviors were examined. Over 12 weeks post-surgery, animals displayed the mechanical, heat, and cold hyperalgesia along with decreased burrowing and rearing. Histology showed progressive disc degeneration with loss of disc height, nucleus pulposus reduction, proteoglycan depletion, and annular fibrotic disorganization. Immunohistochemistry revealed a substantial increase in inflammatory mediators at 2 and 4 weeks. Nerve growth factor was upregulated from 2 weeks to the end of the experiment. Nerve fiber ingrowth was induced in the injured discs after 4 weeks. Disc-puncture also produced an upregulation of neuropeptides in dorsal root ganglia neurons and an activation of glial cells in the spinal cord dorsal horn. These findings indicate that the cellular and structural changes in discs, as well as peripheral and central nervous system plasticity, paralleled persistent, and robust behavioral pain responses. Therefore, this mouse DLBP model could be used to investigate mechanisms underlying discogenic pain, thereby facilitating effective drug screening and development of treatments for DLBP.


Assuntos
Degeneração do Disco Intervertebral/fisiopatologia , Dor Lombar/fisiopatologia , Corno Dorsal da Medula Espinal/fisiopatologia , Punção Espinal , Animais , Sistema Nervoso Central/fisiopatologia , Modelos Animais de Doenças , Gânglios Espinais/fisiopatologia , Humanos , Degeneração do Disco Intervertebral/genética , Degeneração do Disco Intervertebral/cirurgia , Dor Lombar/genética , Dor Lombar/cirurgia , Camundongos , Neuroglia/patologia , Neuropeptídeos/genética , Núcleo Pulposo/fisiopatologia , Corno Dorsal da Medula Espinal/cirurgia
14.
Cell Rep ; 15(2): 349-59, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27050511

RESUMO

Metabolism is a highly integrated process that is coordinately regulated between tissues and within individual cells. FoxO proteins are major targets of insulin action and contribute to the regulation of gluconeogenesis, glycolysis, and lipogenesis in the liver. However, the mechanisms by which FoxO proteins exert these diverse effects in an integrated fashion remain poorly understood. We report that FoxO proteins also exert important effects on intrahepatic lipolysis and fatty acid oxidation via the regulation of adipose triacylglycerol lipase (ATGL), which mediates the first step in lipolysis, and its inhibitor, the G0/S1 switch 2 gene (G0S2). We also find that ATGL-dependent lipolysis plays a critical role in mediating diverse effects of FoxO proteins in the liver, including effects on gluconeogenic, glycolytic, and lipogenic gene expression and metabolism. These results indicate that intrahepatic lipolysis plays a critical role in mediating and integrating the regulation of glucose and lipid metabolism downstream of FoxO proteins.


Assuntos
Tecido Adiposo/metabolismo , Proteína Forkhead Box O1/metabolismo , Glucose/metabolismo , Lipase/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Homeostase , Humanos , Lipase/genética , Metabolismo dos Lipídeos/genética , Lipogênese , Masculino , Camundongos Transgênicos , Modelos Biológicos , Oxirredução
15.
J Biol Chem ; 290(51): 30551-61, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26499800

RESUMO

The activity of the thyroid gland is stimulated by food availability via leptin-induced thyrotropin-releasing hormone/thyroid-stimulating hormone expression. Here we show that food availability also stimulates thyroid hormone activation by accelerating the conversion of thyroxine to triiodothyronine via type 2 deiodinase in mouse skeletal muscle and in a cell model transitioning from 0.1 to 10% FBS. The underlying mechanism is transcriptional derepression of DIO2 through the mTORC2 pathway as defined in rictor knockdown cells. In cells kept in 0.1% FBS, there is DIO2 inhibition via FOXO1 binding to the DIO2 promoter. Repression of DIO2 by FOXO1 was confirmed using its specific inhibitor AS1842856 or adenoviral infection of constitutively active FOXO1. ChIP studies indicate that 4 h after 10% FBS-containing medium, FOXO1 binding markedly decreases, and the DIO2 promoter is activated. Studies in the insulin receptor FOXO1 KO mouse indicate that insulin is a key signaling molecule in this process. We conclude that FOXO1 represses DIO2 during fasting and that derepression occurs via nutritional activation of the PI3K-mTORC2-Akt pathway.


Assuntos
Jejum/metabolismo , Iodeto Peroxidase/biossíntese , Músculo Esquelético/metabolismo , Tiroxina/metabolismo , Tri-Iodotironina/metabolismo , Animais , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Iodeto Peroxidase/genética , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Tiroxina/genética , Tri-Iodotironina/genética , Iodotironina Desiodinase Tipo II
16.
Nat Commun ; 6: 7079, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25963540

RESUMO

FoxO proteins are major targets of insulin action. To better define the role of FoxO1 in mediating insulin effects in the liver, we generated liver-specific insulin receptor knockout (LIRKO) and IR/FoxO1 double knockout (LIRFKO) mice. Here we show that LIRKO mice are severely insulin resistant based on glucose, insulin and C-peptide levels, and glucose and insulin tolerance tests, and genetic deletion of hepatic FoxO1 reverses these effects. (13)C-glucose and insulin clamp studies indicate that regulation of both hepatic glucose production (HGP) and glucose utilization is impaired in LIRKO mice, and these defects are also restored in LIRFKO mice corresponding to changes in gene expression. We conclude that (1) inhibition of FoxO1 is critical for both direct (hepatic) and indirect effects of insulin on HGP and utilization, and (2) extrahepatic effects of insulin are sufficient to maintain normal whole-body and hepatic glucose metabolism when liver FoxO1 activity is disrupted.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Gluconeogênese/fisiologia , Glucose/metabolismo , Insulina/metabolismo , Fígado/metabolismo , Animais , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica/fisiologia , Técnica Clamp de Glucose , Masculino , Camundongos
17.
J Biol Chem ; 287(24): 20132-43, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22511764

RESUMO

Induction of lipogenesis in response to insulin is critically dependent on the transcription factor, sterol regulatory element-binding protein-1c (SREBP-1c). FoxO1, a forkhead box class-O transcription factor, is an important mediator of insulin action, but its role in the regulation of lipid metabolism has not been clearly defined. We examined the effects of FoxO1 on srebp1 gene expression in vivo and in vitro. In vivo studies showed that constitutively active (CA) FoxO1 (CA-FoxO1) reduced basal expression of SREBP-1c mRNA in liver by ∼60% and blunted induction of SREBP-1c in response to feeding. In liver-specific FoxO knock-out mice, SREBP-1c expression was increased ∼2-fold. Similarly, in primary hepatocytes, CA-FoxO1 suppressed SREBP1-c expression and inhibited basal and insulin-induced SREBP-1c promoter activity. SREBP-1c gene expression is induced by the liver X receptor (LXR), but CA-FoxO1 did not block the activation of SREBP-1c by the LXR agonist TO9. Insulin stimulates SREBP-1c transcription through Sp1 and via "feed forward" regulation by newly synthesized SREBP-1c. CA-FoxO1 inhibited SREBP-1c by reducing the transactivational capacity of both Sp1 and SREBP-1c. In addition, chromatin immunoprecipitation assays indicate that FoxO1 can associate with the proximal promoter region of the srebp1 gene and disrupt the assembly of key components of the transcriptional complex of the SREBP-1c promoter. We conclude that FoxO1 inhibits SREBP-1c transcription via combined actions on multiple transcription factors and that this effect is exerted at least in part through reduced transcriptional activity of Sp1 and SREBP-1c and disrupted assembly of the transcriptional initiation complex on the SREBP-1c promoter.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Fígado/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição Sp1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Células Cultivadas , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Hepatócitos/metabolismo , Insulina/genética , Insulina/metabolismo , Receptores X do Fígado , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Ratos , Elementos de Resposta/fisiologia , Fator de Transcrição Sp1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Transcrição Gênica/fisiologia
18.
Vaccine ; 28(49): 7757-63, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-20887831

RESUMO

This study describes the immunotherapeutic properties of vaccines that encode tumor-associated calcium signal transducer-1 (Trop-1), a newly identified breast cancer antigen, in mice with breast cancer. Previously we found that Trop-1 was over-expressed in cellular breast cancer vaccines that were highly enriched for cells that induced therapeutic CTL-mediated immune responses in mice with breast cancer, as compared with non-enriched vaccines. In this study, to determine if the expression of Trop-1 by cells in the enriched vaccine was responsible for its therapeutic benefits, an expression plasmid that specified the Trop-1 gene was transfected into the LM fibroblast cells, which was then used as a vaccine. To augment their immunogenic properties, the fibroblasts were genetically modified before Trop-1 DNA-transfer to secrete IL-2 and to express allogeneic MHC class I H-2K(b)-determinants. Mice with established breast cancer treated solely by immunization with fibroblasts modified to express Trop-1 developed CD8(+) cell-mediated immunity to the breast cancer cells. The immunity was sufficient to prolong the survival of mice with established breast cancer. In some instances, the immunity was sufficient to result in rejection of the tumor; the mice remained tumor free more than 60 days.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Moléculas de Adesão Celular/imunologia , Neoplasias Mamárias Experimentais/imunologia , Animais , Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/genética , Linhagem Celular , Molécula de Adesão da Célula Epitelial , Feminino , Fibroblastos/imunologia , Antígenos H-2 , Imunidade Celular , Interleucina-2/imunologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos C3H , Plasmídeos , Baço/imunologia , Linfócitos T Citotóxicos/imunologia , Transfecção
19.
Immunology ; 126(1): 84-91, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18547366

RESUMO

T helper type 1 (Th1) cell-mediated immune responses contribute to host defences against intracellular pathogen infections and cancer. Previously, we found that aminoacyl tRNA synthetase-interacting multifunctional protein 1 (AIMP1) activated macrophages and dendritic cells to enhance Th1 responses. Herein, we manipulated this property to improve Th1 immune responses in vivo by constructing a mammalian expression plasmid (pAnti-CD3sFv/AIMP1) encoding AIMP1 fused to the anti-CD3 single-chain Fv (sFv), the smallest unit of the antibody that interacts with the CD3epsilon region of the T-cell receptor. Intramuscular injection of ovalbumin (OVA)-sensitized BALB/c mice with pAnti-CD3sFv/AIMP1 DNA adjuvant increased the OVA-specific, interferon-gamma production by their CD4(+) T cells and the levels of anti-OVA immunoglobulin G2a (IgG2a) isotype in their sera. Furthermore, the pAnti-CD3sFv/AIMP1 DNA adjuvant decreased interleukin-4 production and anti-OVA IgE levels in the OVA-injected mice. Importantly, the pAnti-CD3sFv/AIMP1 was more efficient than a mixture of pAnti-CD3sFv and pAIMP1 in inducing OVA-specific Th1 immune responses and also in inhibiting OVA-specific Th2 responses during antigen priming. These studies indicated that the pAnti-CD3sFv/AIMP1 fusion DNA adjuvant enhanced Th1 immune responses in antigen-sensitized mice.


Assuntos
Complexo CD3/imunologia , Citocinas/imunologia , DNA/imunologia , Células Th1/imunologia , Adjuvantes Imunológicos , Animais , Complexo CD3/genética , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Feminino , Imunoglobulina E/biossíntese , Imunoglobulina G/biossíntese , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Injeções Intramusculares , Interleucina-4/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Baço/imunologia
20.
Immunotherapy ; 1(3): 495-504, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-20352015

RESUMO

Various strategies have been used to generate cellular cancer vaccines with the expectation that they will become an effective part of the overall management of cancer patients. However, with few notable exceptions, immunization has not resulted in significant long-term therapeutic benefits. Tumor growth has continued and patient survival has been at best only modestly prolonged. One possible explanation is that as only a small proportion of the constituents of malignant cells are "tumor specific" and the vast majority are the products of nonantigenic, normal "housekeeping" genes, the immune response in patients immunized with cellular cancer vaccines is not sufficient to result in tumor rejection. Here, we review and characterize various types of cellular cancer vaccines. In addition, in a mouse breast cancer model system, we describe a unique strategy designed to enrich cellular vaccines for cells that induce tumor immunity. Numerous advantages and disadvantages of cancer immunotherapy with cellular vaccines are also presented.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer , Células Dendríticas/imunologia , Imunoterapia Adotiva , Animais , Apresentação de Antígeno , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Engenharia Genética , Biblioteca Genômica , Humanos , Ativação Linfocitária , Camundongos , Vacinas de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA