Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nat Immunol ; 18(2): 161-172, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27941786

RESUMO

Aire is a transcriptional regulator that induces promiscuous expression of thousands of genes encoding tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs). While the target genes of Aire are well characterized, the transcriptional programs that regulate its own expression have remained elusive. Here we comprehensively analyzed both cis-acting and trans-acting regulatory mechanisms and found that the Aire locus was insulated by the global chromatin organizer CTCF and was hypermethylated in cells and tissues that did not express Aire. In mTECs, however, Aire expression was facilitated by concurrent eviction of CTCF, specific demethylation of exon 2 and the proximal promoter, and the coordinated action of several transcription activators, including Irf4, Irf8, Tbx21, Tcf7 and Ctcfl, which acted on mTEC-specific accessible regions in the Aire locus.


Assuntos
Células Epiteliais/imunologia , Redes Reguladoras de Genes , Linfócitos T/fisiologia , Timo/imunologia , Fatores de Transcrição/metabolismo , Animais , Apresentação de Antígeno/genética , Autoantígenos/metabolismo , Fator de Ligação a CCCTC , Diferenciação Celular , Células Cultivadas , Seleção Clonal Mediada por Antígeno , Metilação de DNA , Regulação da Expressão Gênica , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Especificidade de Órgãos/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Timo/citologia , Fatores de Transcrição/genética , Proteína AIRE
2.
Nat Immunol ; 16(7): 737-45, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26006015

RESUMO

Aire is a transcriptional regulator that induces the promiscuous expression of thousands of tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs), a step critical for the induction of immunological self-tolerance. Studies have offered molecular insights into how Aire operates, but more comprehensive understanding of this process still remains elusive. Here we found abundant expression of the protein deacetylase Sirtuin-1 (Sirt1) in mature Aire(+) mTECs, wherein it was required for the expression of Aire-dependent TRA-encoding genes and the subsequent induction of immunological self-tolerance. Our study elucidates a previously unknown molecular mechanism for Aire-mediated transcriptional regulation and identifies a unique function for Sirt1 in preventing organ-specific autoimmunity.


Assuntos
Tolerância Central/imunologia , Sirtuína 1/imunologia , Fatores de Transcrição/imunologia , Ativação Transcricional/imunologia , Acetilação , Animais , Antígenos/imunologia , Tolerância Central/genética , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Citometria de Fluxo , Células HEK293 , Humanos , Immunoblotting , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos/imunologia , Ligação Proteica/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 1/genética , Sirtuína 1/metabolismo , Timo/citologia , Timo/imunologia , Timo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma/imunologia , Proteína AIRE
3.
Cancer Immunol Immunother ; 69(10): 2021-2031, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32405793

RESUMO

Following excision of colorectal tumors, metastatic disease is prevalent, primarily occurs in the liver, and is highly predictive of poor prognosis. The perioperative period is now recognized as critical in determining the incidence of postoperative metastases and long-term cancer outcomes. Thus, various perioperative prophylactic interventions are currently studied during this time frame. However, immune stimulation during the perioperative period has rarely been attempted due to specific contraindications to surgery and various adverse effects. Here, to prevent liver metastases, we perioperatively employed a TLR-9 agonist, CpG-C, which exhibits minimal pyrogenic and other adverse effects in patients. We found that marginating-hepatic (MH) cells in BALB/c mice contained high percentage of NK cells, but exhibited negligible NK cytotoxicity, as previously reported in humans. However, a single CpG-C administration (25-100 µg/mouse) doubled MH-NK cell numbers, increased NK cell activation and maturation markers (NKp46, CD11b), decreased the inhibitory NKG2A ligand, and dramatically increased MH-NK-cell cytotoxicity against the syngeneic CT26 colon cancer line. Moreover, in operated mice, this innocuous intervention also markedly improved resistance to CT26 and MC38 hepatic metastases in BALB/c and C57BL/6 mice, respectively. Beneficial effects of CpG-C were mediated through activation of MH-NK cells, as indicated by an in vivo NK depletion study. Last, CpG-C protected against surgery-induced suppression of MH-NK cytotoxicity and improved their activation indices. Thus, we suggest that systemic perioperative CpG-C treatment should be considered and studied as a novel therapeutic approach to improve long-term cancer outcomes in colorectal cancer patients.


Assuntos
Neoplasias do Colo/prevenção & controle , Células Matadoras Naturais/efeitos dos fármacos , Neoplasias Hepáticas/prevenção & controle , Oligodesoxirribonucleotídeos/administração & dosagem , Animais , Apoptose , Proliferação de Células , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Período Perioperatório , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
PLoS Genet ; 12(1): e1005776, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26789017

RESUMO

Thymic medullary regions are formed in neonatal mice as islet-like structures, which increase in size over time and eventually fuse a few weeks after birth into a continuous structure. The development of medullary thymic epithelial cells (TEC) is dependent on NF-κB associated signaling though other signaling pathways may contribute. Here, we demonstrate that Stat3-mediated signals determine medullary TEC cellularity, architectural organization and hence the size of the medulla. Deleting Stat3 expression selectively in thymic epithelia precludes the postnatal enlargement of the medulla retaining a neonatal architecture of small separate medullary islets. In contrast, loss of Stat3 expression in cortical TEC neither affects the cellularity or organization of the epithelia. Activation of Stat3 is mainly positioned downstream of EGF-R as its ablation in TEC phenocopies the loss of Stat3 expression in these cells. These results indicate that Stat3 meditated signal via EGF-R is required for the postnatal development of thymic medullary regions.


Assuntos
Diferenciação Celular/genética , Células Epiteliais , Receptores ErbB/genética , Fator de Transcrição STAT3/biossíntese , Animais , Desenvolvimento Embrionário , Receptores ErbB/biossíntese , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Fator de Transcrição STAT3/genética , Transdução de Sinais , Linfócitos T/metabolismo , Timócitos/metabolismo , Timo/crescimento & desenvolvimento , Timo/metabolismo
5.
Brain Behav Immun ; 37: 207-19, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24333572

RESUMO

Most in vitro and ex-vivo studies indicate a profound suppression of NK cell cytotoxicity (NKCC) by glucocorticoids; while catecholamines and prostaglandins were reported both to suppress and to enhance NKCC. However, methodological considerations hinder our ability to deduce from these findings to the impact of endogenous release of these factors on in vivo levels of NKCC and their implications to NK-dependent resistance to pathologies in living humans or animals. Here we used an in vivo approach that sensitively and specifically reflects NKCC in living F344 rats, based on lung clearance of NK-sensitive tumor cells (MADB106), and based on comparing effects between NK-intact and NK-depleted rats. To study the role of corticosterone, epinephrine, and prostaglandins, we administered these factors to rats, or antagonized their endogenous release following different stress paradigms or surgery. The results indicated that endogenous or exogenous elevated corticosterone levels can suppress in vivo NKCC levels, but only under some conditions, and mostly secondarily to the NK-suppressing impact of epinephrine. Specifically, corticosterone-induced NKCC suppression occurred (i) only under prolonged, but not short exposure to stress, and mainly in males; (ii) was smaller than the prominent impact of epinephrine; (iii) was mostly ascribed to corticosterone-induced potentiation of the effects of epinephrine or/and prostaglandins; and (iv) was completely abolished through antagonizing epinephrine or/and prostaglandins. Overall, these findings markedly limit the significance of stress/surgery-induced corticosterone release in the in vivo suppression of NKCC, and highlight the blockade of epinephrine or/and prostaglandins as effective and clinically feasible approaches to overcome such immuno-suppressive effects.


Assuntos
Catecolaminas/fisiologia , Citotoxicidade Imunológica , Glucocorticoides/fisiologia , Células Matadoras Naturais/imunologia , Prostaglandinas/fisiologia , Estresse Fisiológico , Adrenalectomia , Animais , Catecolaminas/metabolismo , Linhagem Celular Tumoral , Corticosterona/farmacocinética , Corticosterona/farmacologia , Feminino , Glucocorticoides/metabolismo , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Masculino , Prostaglandinas/metabolismo , Ratos , Ratos Endogâmicos F344
6.
Surg Today ; 44(10): 1925-34, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24190423

RESUMO

PURPOSE: Extensive oncological surgeries were previously suggested to increase cancer recurrence rates. We herein studied the impact of different surgical procedures and sex on colorectal cancer liver metastasis, employing several tumor inoculation approaches in BALB/c mice. METHODS: Experimental hepatic metastases of the syngeneic CT26 colorectal cancer line were induced either by intra-portal inoculation or intra-splenic inoculation, employing different tumor loads. Following intra-splenic inoculation, the entire spleen or an injected hemi-spleen was removed. Additionally, the magnitude of the surgical trauma accompanying the injection procedure was manipulated. RESULTS: Increasing the surgical trauma by adding laparotomy or extending the length of the surgery and hypothermia did not significantly affect the number of liver metastases or liver weight for any of the injection methods and tumor loads. The development of metastasis was significantly greater in males than in females under all conditions studied--a difference not explained by the direct effects of sex hormones on in vitro CT26 proliferation or vitality. CONCLUSION: Concurring with less controlled clinical observations, the surgical extensiveness did not significantly affect CT26 hepatic metastasis, potentially due to a ceiling effect of the surgical trauma on the metastatic process. The sexual dimorphism observed for the CT26 metastasis should be investigated in the context of surgical stress and considering anti-CT26 immunoreactivity.


Assuntos
Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/cirurgia , Caracteres Sexuais , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Feminino , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos BALB C , Procedimentos Cirúrgicos Minimamente Invasivos , Transplante de Neoplasias/métodos
7.
Neuroimmunomodulation ; 20(4): 194-204, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23635771

RESUMO

OBJECTIVE: Young adults often encounter sleep deprivation and stressful events. Both have been separately reported to modulate immunity, and occasionally they occur simultaneously. We assessed the combined effects of these conditions on immune competence in healthy students. METHODS: Twenty-three participants (mean age 24 years; SD 1.86; 14 females) were exposed to 30 h of sleep deprivation during which they conducted physiological, social and cognitive tasks. The control group consisted of 18 participants (mean age 23.67 years; SD 1.46; 11 females). All participants underwent cognitive and psychological evaluations at 10:00 AM, followed by blood and saliva collection, 3 days before sleep deprivation induction and on the morning following it. Immune/endocrine measures included blood counts of lymphocytes, granulocytes, monocytes and natural killer (NK) cells; levels of several cell surface markers; NK cytotoxicity; plasma levels of interleukin (IL)-6, IL-10, dehydroepiandrosterone and neuropeptide Y, and plasma and salivary cortisol levels. RESULTS: Although the experimental protocol significantly elevated state anxiety and psychological dissociation levels, no effects were evident in any of the immunological/endocrine indices. In contrast, expected sex differences in immune measures were found, including significantly higher NK cytotoxicity and monocyte counts in males, validating the integrity of the measurements. CONCLUSIONS: The findings suggest resilience of the immune system to a combined sleep deprivation and stressful exposure in young adults, while previous studies reported immune perturbations following either of these conditions separately. These apparent contradictions might reflect differences in the study design or in the methodology used for immunological assessments, including the time of sample collection, the combination of sleep deprivation with stress and our in vivo assessment of cytokine levels.


Assuntos
Privação do Sono/imunologia , Privação do Sono/psicologia , Estresse Psicológico/imunologia , Estresse Psicológico/psicologia , Estudantes/psicologia , Feminino , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Células K562 , Células Matadoras Naturais/imunologia , Contagem de Leucócitos/métodos , Masculino , Adulto Jovem
8.
Int Immunol ; 23(4): 287-96, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21427174

RESUMO

IFN regulatory factor-8 (IRF-8, previously known as ICSBP) is a key transcription factor driving the differentiation of granulocyte\monocyte progenitor (GMP) cells toward monocyte\macrophage lineage. The promyelocytic leukemia (PML) gene is an immediate target gene regulated by IRF-8 in response to IFN-γ activation. PML is a multifunctional protein that has many isoforms serving as the scaffold components for nuclear bodies (NBs) engaged in numerous proteins interactions. The role of PML in the retinoic acid pathway that drives GMPs to granulopoiesis is documented in the literature. Here, we show that PML is also involved in monopoiesis by mediating some of the IRF-8 activities during the differentiation of murine-derived bone marrow macrophages (BMMs). PML silencing resulted in altered expression level of key transcription factors essential for monopoiesis that was accompanied by silencing of typical myeloid-specific genes. Interestingly, this altered expression resembled that of the GMPs and that of BMMs derived from IRF-8(-/-) mice altogether supporting the role of PML in monopoiesis. Further, PML silencing led to reduced colony-forming capacity of bone marrow cells highlighting the dual function of PML in myelopoiesis. Last, PML overexpression only partially rescued the phenotype of IRF-8(-/-) BMMs. Together, our data show that PML is an important factor for monopoiesis and not solely for granulopoiesis. This suggests that PML-NBs respond to an incoming signal that affects the fate of GMP driving cell differentiation to granulocytes or monocytes.


Assuntos
Macrófagos/metabolismo , Células Progenitoras Mieloides/citologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Proteína da Leucemia Promielocítica , RNA Interferente Pequeno/genética , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
9.
J Craniofac Surg ; 23(1): 113-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22337385

RESUMO

BACKGROUND: Hypertelorbitism has been associated with a variety of congenital deformities. Appropriate timing for surgical correction remains controversial. We present our long-term experience of 33 patients with hypertelorbitism undergoing facial bipartition or orbital box osteotomy. METHODS: Patients with hypertelorbitism treated with either facial bipartition or orbital box osteotomy and repositioning who had long-term follow-up were studied (n=33). Age at the time of first surgery, preoperative interdacryon distance, and immediate postoperative interdacryon distance were recorded. Relapse was determined on postoperative follow-up, and the need for secondary correction was noted. Physician satisfaction score (range, 0-4) was also assessed. RESULTS: Patients had a mean total follow-up of 14.0 years. With regard to age at the time of initial procedure, patients younger than 6 years were all noted to have relapse, and 83% underwent revision surgery. In patients 6 years or older, only 11% had relapse and required a second operation. Yet, satisfaction scores were similar (3.2 versus 3.5). With regard to the severity of hypertelorbitism, there was no relapse noted among patients with mild hypertelorbitism (interorbital distance [IOD], 30-34 mm). Among those with moderate hypertelorbitism (IOD, 35-40 mm), 29.4% developed relapse. By contrast, all patients with severe hypertelorbitism (IOD, >40 mm) were noted to have relapse requiring repeat correction. Satisfaction scores were similar (3.4 versus 3.3 versus 3.1). CONCLUSIONS: Relapse after surgery for hypertelorbitism is related to the age of the patient at correction and the preoperative severity. When possible, surgical repositioning of the orbits should be delayed until later childhood.


Assuntos
Hipertelorismo/cirurgia , Procedimentos de Cirurgia Plástica/métodos , Fatores Etários , Placas Ósseas , Parafusos Ósseos , Cefalometria , Criança , Pré-Escolar , Osso Etmoide/cirurgia , Seguimentos , Osso Frontal/cirurgia , Humanos , Hipertelorismo/classificação , Cuidados Intraoperatórios , Estudos Longitudinais , Maxila/cirurgia , Septo Nasal/cirurgia , Órbita/cirurgia , Osteotomia/instrumentação , Osteotomia/métodos , Recidiva , Reoperação , Estudos Retrospectivos , Osso Esfenoide/cirurgia , Tomografia Computadorizada por Raios X , Resultado do Tratamento
10.
Cells ; 11(8)2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35455969

RESUMO

Vascular endothelial growth factor A (VEGF-A) is a secreted protein that stimulates angiogenesis in response to hypoxia. Under hypoxic conditions, a non-canonical long isoform called L-VEGF is concomitantly expressed with VEGF-A. Once translated, L-VEGF is proteolytically cleaved to generate N-VEGF and VEGF-A. Interestingly, while VEGF-A is secreted and affects the surrounding cells, N-VEGF is mobilized to the nucleus. This suggests that N-VEGF participates in transcriptional response to hypoxia. In this study, we performed a series of complementary experiments to examine the functional role of N-VEGF. Strikingly, we found that the mere expression of N-VEGF followed by its hypoxia-independent mobilization to the nucleus was sufficient to induce key genes associated with angiogenesis, such as Hif1α,VEGF-A isoforms, as well as genes associated with cell survival under hypoxia. Complementarily, when N-VEGF was genetically depleted, key hypoxia-induced genes were downregulated and cells were significantly susceptible to hypoxia-mediated apoptosis. This is the first report of N-VEGF serving as an autoregulatory arm of VEGF-A. Further experiments will be needed to determine the role of N-VEGF in cancer and embryogenesis.


Assuntos
Neovascularização Patológica , Fator A de Crescimento do Endotélio Vascular , Apoptose , Hipóxia Celular , Humanos , Hipóxia , Neovascularização Patológica/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Ann Surg ; 253(4): 798-810, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21475023

RESUMO

BACKGROUND: Surgical procedures, including primary tumor resection, have been suggested to suppress immune competence and to promote postoperative infections and cancer metastasis. Catecholamines and prostaglandins were recently implicated in these processes, and in directly promoting tumor angiogenesis and invasion. OBJECTIVE: To examine the integration of 2 complementary approaches to reduce postoperative immunosuppression and metastatic progression: (1) perioperative immunostimulation with CpG-C and (2) pharmacological blockade of the tumor-promoting and immunosuppressing effects of catecholamines and prostaglandins, using propranolol (P) and etodolac (E), respectively. METHODS: F344 rats were treated before surgery with CpG-C, P+E, both interventions, or vehicles, and were intravenously inoculated with syngeneic MADB106 mammary adenocarcinoma cells. Blood was withdrawn, marginating-pulmonary leukocytes were harvested, and NK activity and lung MADB106 tumor retention were assessed. In addition, C57BL/6 mice were implanted with syngeneic B16F10.9 melanoma cells. When tumors reached 100 mm, mice were treated with CpG-C/vehicle, and 24 hours later the tumor was excised along with P+E/vehicle treatment. Recurrence-free survival was monitored thereafter. RESULTS: Each of the regimens alone, CpG-C or P+E, showed improvement in most indices examined, including improved long-term recurrence-free survival rates. Most importantly, the combined treatment yielded additive or synergistic effects, further improving tumor clearance from the lungs and enhancing NK numbers and cytotoxicity via different, but complimentary, mechanisms. CONCLUSIONS: Treatment aimed at perioperative enhancement of CMI and simultaneous inhibition of excessive catecholamine and prostaglandin responses, employing CpG-C, propranolol, and etodolac, could be successful in limiting postoperative immunosuppression and metastatic progression, more so than each treatment alone.


Assuntos
Hospedeiro Imunocomprometido/efeitos dos fármacos , Imunoterapia Ativa/métodos , Metástase Neoplásica/imunologia , Metástase Neoplásica/prevenção & controle , Neoplasias/imunologia , Neoplasias/cirurgia , Neovascularização Patológica/prevenção & controle , Oligodesoxirribonucleotídeos/farmacologia , Análise de Variância , Animais , Quimioterapia Adjuvante , Modelos Animais de Doenças , Etodolac/farmacologia , Feminino , Tolerância Imunológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Assistência Perioperatória/métodos , Complicações Pós-Operatórias/prevenção & controle , Propranolol/farmacologia , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Valores de Referência , Medição de Risco , Sensibilidade e Especificidade , Estresse Psicológico/etiologia , Estresse Psicológico/prevenção & controle , Células Tumorais Cultivadas
12.
Brain Behav Immun ; 25(1): 67-76, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20656015

RESUMO

Bi-directional influences between stress hormones and immune responses have been repeatedly documented, however, in the clinical setting they are rarely considered when immunotherapeutic approaches are used or studied in patients. As some immunotherapeutic treatments have shown great potential in animal models but have had limited success in patients, we hypothesize that ongoing psychological and physiological stress responses in patients, which do not characterize the setting of animal studies, contribute to this discrepancy. In the current study we examined the interaction between ongoing water stress and CpG-C immunotherapy to determine whether stress that precedes immunotherapy can modulate the efficacy of CpG-C immunostimulation. C57BL/6 mice were exposed to water stress or served as controls. Two hours following the commencement of the stress protocol animals were injected with CpG-C, non-CpG, or PBS, and sacrificed 1, 4 or 12h thereafter. We found that in CpG-C-treated animals stress eliminated the elevation of plasma IL-12, and synergistically elevated corticosterone levels. Furthermore, stress markedly reduced the total number of myeloid (33D1(+)), plasmacytoid (mPDCA-1(+)) and plasmacytoid-derived (33D1(+)mPDCA-1(+)) dendritic cells in CpG-C-treated animals, as well as the numbers of these cell sub-types expressing CD11b, CD80 and CD69. These changes were more dramatic in the blood than in the spleen. Overall, these findings indicate that under no-stress conditions CpG-C induces a robust immune response, which is significantly diminished when immunostimulation is attempted during ongoing stress. If these findings hold in humans, potential prophylactic treatments should be found to limit the deleterious effects of ongoing stress on the efficacy of immunotherapy.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Imunoterapia , Oligodesoxirribonucleotídeos/uso terapêutico , Estresse Psicológico/imunologia , Animais , Corticosterona/sangue , Ilhas de CpG , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Citometria de Fluxo , Imersão/fisiopatologia , Interleucina-10/biossíntese , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/imunologia , Baço/metabolismo , Estresse Psicológico/complicações , Receptor Toll-Like 9/metabolismo
13.
Brain Behav Immun ; 25(4): 727-35, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21277367

RESUMO

Immune stimulation by biological response modifiers is a common approach in tumor immunotherapy. IL-12 was found effective in various animal studies, but clinical trials showed limited success. However, among other differences, animal models do not simulate psychological or physiological stress while employing IL-12, whereas cancer patients often experience distress while treated with immunostimulants. Thus, in the current study we assessed the impact of continuous stress on the efficacy of IL-12 immunostimulation. F344 rats were subjected to a pharmacological stress paradigm (continuous administration of a ß-adrenergic agonist) or to a 20 h behavioral stress paradigm (wet cage exposure) commencing 2h before IL-12 administration. Twenty-six hours after stress initiation, we studied indices known to reflect IL-12 immunostimulatory impacts, including NK cell numbers and activity in different immune compartments, and in vivo resistance to MADB106 lung tumor colonization. The results indicated that both the pharmacological and behavioral stress paradigms significantly reduced the increase in the number and activity of marginating-pulmonary NK cells evident in non-stressed IL-12 treated animals. Additionally, stressed animals exhibited a lower IL-12-induced improvement of MADB106 lung clearance, an in vivo index that markedly depends on total marginating-pulmonary NK activity. These deleterious effects of stress were more prominent in males than in females. Overall, the findings demonstrate that prolonged stress exposure can disrupt the efficacy of simultaneous immunostimulatory treatments, irrespective of stress effects on baseline immune measures. Neuroendocrine and cellular mediating mechanisms are yet unknown, but the potential clinical ramifications of these findings warrant consideration in clinical trials employing immunostimulatory agents.


Assuntos
Adenocarcinoma/imunologia , Adjuvantes Imunológicos/administração & dosagem , Interleucina-12/imunologia , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/imunologia , Estresse Psicológico/imunologia , Adenocarcinoma/complicações , Agonistas de Receptores Adrenérgicos beta 2 , Análise de Variância , Animais , Doença Crônica , Corticosterona/sangue , Epinefrina/sangue , Feminino , Interleucina-12/administração & dosagem , Células Matadoras Naturais/citologia , Neoplasias Pulmonares/complicações , Masculino , Metaproterenol , Neoplasias Experimentais/complicações , Neoplasias Experimentais/imunologia , Ratos , Ratos Endogâmicos F344 , Fatores Sexuais , Estresse Psicológico/sangue , Estresse Psicológico/induzido quimicamente , Estresse Psicológico/complicações
14.
J Mol Biol ; 432(16): 4544-4560, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32534063

RESUMO

The regulation of gene expression is a result of a complex interplay between chromatin remodeling, transcription factors, and signaling molecules. Cell differentiation is accompanied by chromatin remodeling of specific loci to permanently silence genes that are not essential for the differentiated cell activity. The molecular cues that recruit the chromatin remodeling machinery are not well characterized. IRF8 is an immune-cell specific transcription factor and its expression is augmented by interferon-γ. Therefore, it serves as a model gene to elucidate the molecular mechanisms governing its silencing in non-immune cells. Ahigh-throughput shRNA library screen in IRF8 expression-restrictive cells enabled the identification of MafK as modulator of IRF8 silencing, affecting chromatin architecture. ChIP-Seq analysis revealed three MafK binding regions (-25 kb, -20 kb, and IRF8 6th intron) within the IRF8 locus. These MafK binding sites are sufficient to repress a reporter gene when cloned in genome-integrated lentiviral reporter constructs in only expression-restrictive cells. Conversely, plasmid-based constructs do not demonstrate such repressive effect. These results highlight the role of these MafK binding sites in mediating repressed chromatin assembly. Finally, a more thorough genomic analysis was performed, using CRISPR-Cas9 to delete MafK-int6 binding region in IRF8 expression-restrictive cells. Deleted clones exhibited an accessible chromatin conformation within the IRF8 locus that was accompanied by a significant increase in basal expression of IRF8 that was further induced by interferon-γ. Taken together, we identified and characterized several MafK binding elements within the IRF8 locus that mediate repressive chromatin conformation resulting in the silencing of IRF8 expression in a celltype-specific manner.


Assuntos
Cromatina/metabolismo , Fatores Reguladores de Interferon/genética , Fator de Transcrição MafK/genética , Fator de Transcrição MafK/metabolismo , Animais , Sítios de Ligação , Sistemas CRISPR-Cas , Montagem e Desmontagem da Cromatina , Regulação da Expressão Gênica , Células HEK293 , Humanos , Fatores Reguladores de Interferon/química , Fatores Reguladores de Interferon/metabolismo , Camundongos , Células NIH 3T3 , Especificidade de Órgãos , Células RAW 264.7 , RNA Interferente Pequeno/farmacologia
15.
Brain Behav Immun ; 23(5): 611-21, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18951972

RESUMO

Inflammation is implicated in several medical conditions that are sexually dimorphic, including depression, cardiovascular diseases, autoimmunity, and presumably cancer progression. Here we studied the effects of the proinflammatory agent, LPS, on MADB106 lung tumor retention (LTR), and sought to elucidate underlying mechanisms and sexual dimorphism. F344 male and female rats were administered with LPS (0.001-1mg/kg i.v.) simultaneously with tumor cell inoculation, and treated with a beta-blocker (nadolol, 0.2-0.3mg/kg s.c.), a COX inhibitor (indomethacin, 4mg/kg s.c.) or both drugs. To study the role of NK cells, numbers and cytotoxicity of marginating-pulmonary NK cells were studied, and selective in vivo NK-depletion was employed. Serum levels of corticosterone, IL-6, and TNF-alpha were also assessed. The findings indicated that LPS increased LTR in both sexes, but 10-fold higher doses were needed in females to reach the increase evident in males. Additionally, nadolol and indomethacin reduced the effects of LPS, more so in males. In vivo NK-depletion and ex vivo NK activity studies suggested that LPS affected LTR through both NK-independent and NK-dependent mechanisms, the latter mediated through prostaglandin release in males. Corticosterone, IL-6, and TNF-alpha responses to LPS were sexually dimorphic, but were not associated with LPS or drugs' impacts on LTR. Overall, our findings demonstrate sexual dimorphism in LPS-induced elevated susceptibility to MADB106 experimental metastasis, and in potential humoral underlying mechanisms. Further studies are needed to elucidate additional immunological and non-immunological mediators of these dimorphisms, as well as to assess their involvement in other sexually dimorphic pathologies that are associated with inflammation.


Assuntos
Adenocarcinoma/secundário , Catecolaminas/fisiologia , Lipopolissacarídeos/toxicidade , Neoplasias Pulmonares/secundário , Prostaglandinas/fisiologia , Caracteres Sexuais , Adenocarcinoma/sangue , Adenocarcinoma/prevenção & controle , Antagonistas Adrenérgicos beta/farmacologia , Animais , Linhagem Celular Tumoral/transplante , Corticosterona/sangue , Inibidores de Ciclo-Oxigenase/farmacologia , Citotoxicidade Imunológica , Feminino , Indometacina/farmacologia , Interleucina-6/sangue , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/prevenção & controle , Masculino , Neoplasias Mamárias Experimentais/patologia , Nadolol/farmacologia , Ratos , Ratos Endogâmicos F344 , Fator de Necrose Tumoral alfa/análise
16.
J Mol Biol ; 431(2): 210-222, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30502383

RESUMO

Interferon regulatory factor 8 (IRF8) is a nuclear transcription factor that plays a key role in the hierarchical differentiation of hematopoietic stem cells toward monocyte/dendritic cell lineages. Therefore, its expression is mainly limited to bone marrow-derived cells. The molecular mechanisms governing this cell-type-restricted expression have been described. However, the molecular mechanisms that are responsible for its silencing in non-hematopoietic cells are elusive. Recently, we demonstrated a role for IRF8 third intron in restricting its expression in non-hematopoietic cells. Furthermore, we showed that this intron alone is sufficient to promote repressed chromatin a cell-type-specific manner. Here we demonstrate the effect of the IRF8 third intron on chromatin conformation during murine embryonal stem cell differentiation. Using genome editing, we provide data showing that the third intron plays a key role in priming the chromatin state of the IRF8 locus during cell differentiation. It mediates dual regulatory effects in a cell-type-specific mode. It acts as a repressor element governing chromatin state of the IRF8 locus during embryonal stem cell differentiation to cardiomyocytes that are expression-restrictive cells. Conversely, it functions as an activator element that is essential for open chromatin structure during the differentiation of these cells to dendritic cells that are expression-permissive cells. Together, these results point to the role of IRF8 third intron as a cell-type-specific chromatin priming element during embryonal stem cell differentiation. These data add another layer to our understanding of the molecular mechanisms governing misexpression of a cell-type-specific gene such as IRF8.


Assuntos
Diferenciação Celular/genética , Cromatina/genética , Fatores Reguladores de Interferon/genética , Íntrons/genética , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Linhagem Celular , Linhagem da Célula/genética , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/genética , Células HEK293 , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Monócitos/metabolismo , Miócitos Cardíacos/metabolismo , Células NIH 3T3 , Células RAW 264.7
17.
Ann Surg Oncol ; 15(7): 2042-52, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18398660

RESUMO

BACKGROUND: COX inhibitors and beta-blockers were recently suggested to reduce cancer progression through inhibition of tumor proliferation and growth factor secretion, induction of tumor apoptosis, and prevention of cellular immune suppression during the critical perioperative period. Here we evaluated the perioperative impact of clinically applicable drugs from these categories in the context of surgery, studying natural killer (NK) cell activity and resistance to experimental metastases. METHODS: F344 rats were treated with COX-1 inhibitors (SC560), COX-2 inhibitors (indomethacin, etodolac, or celecoxib), a beta-blocker (propranolol), or a combination of a COX-2 inhibitor and a beta-blocker (etodolac and propranolol). Rats underwent laparotomy, and were inoculated intravenously with syngeneic MADB106 tumor cells for the assessment of lung tumor retention (LTR). Additionally, the impact of these drug regimens on postoperative levels of NK cytotoxicity was studied in peripheral blood and marginating-pulmonary leukocytes. RESULTS: Surgery increased MADB106 LTR. COX-2 inhibition, but not COX-1 inhibition, reduced postoperative LTR. Etodolac and propranolol both attenuated the deleterious impact of surgery, and their combined use abolished it. Surgery decreased NK cytotoxicity per NK cell in both immune compartments, and only the combination of etodolac and propranolol significantly attenuated these effects. Lastly, the initiation of drug treatment three days prior to surgery yielded the same beneficial effects as a single pre-operative administration, but, as discussed, prolonged treatment may be more advantageous clinically. CONCLUSIONS: Excess prostaglandin and catecholamine release contributes to postoperative immune-suppression. Treatment combining perioperative COX-2 inhibition and beta-blockade is practical in operated cancer patients, and our study suggests potential immunological and clinical benefits.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Imunidade/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Metástase Neoplásica/imunologia , Metástase Neoplásica/prevenção & controle , Animais , Linhagem Celular Tumoral , Inibidores de Ciclo-Oxigenase 2/farmacologia , Quimioterapia Combinada , Masculino , Assistência Perioperatória , Ratos , Ratos Endogâmicos F344 , Fatores de Risco
18.
Mol Immunol ; 44(4): 338-46, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16597464

RESUMO

Interferon regulatory factor 1 (IRF-1) and IRF-8, also known as interferon consensus sequence binding protein (ICSBP), are important regulators of macrophage differentiation and function. These factors exert their activities through the formation of heterocomplexes. As such, they are coactivators of various interferon-inducible genes in macrophages. To gain better insights into the involvement of these two transcription factors in the onset of the innate immune response and to identify their regulatory network in activated macrophages, DNA microarray was employed. Changes in the expression profile were analyzed in peritoneal macrophages from wild type mice and compared to IRF-1 and IRF-8 null mice, before and following 4 h exposure to IFN-gamma and LPS. The expression pattern of 265 genes was significantly changed (up/down) in peritoneal macrophages extracted from wild type mice following treatment with IFN-gamma and LPS, while no changes in the expression levels of these genes were observed in samples of the same cell-type from both IRF-1 and IRF-8 null mice. Among these putative target genes, numerous genes are involved in macrophage activity during inflammation. The expression profile of 10 of them was further examined by quantitative RT-PCR. In addition, the promoter regions of three of the identified genes were analyzed by reporter gene assay for the ability to respond to IRF-1 and IRF-8. Together, our results suggest that both IRF-1 and IRF-8 are involved in the transcriptional regulation of these genes. We therefore suggest a broader role for IRF-1 and IRF-8 in macrophages differentiation and maturation, being important inflammatory mediators.


Assuntos
Fator Regulador 1 de Interferon/genética , Fatores Reguladores de Interferon/genética , Ativação de Macrófagos/genética , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Fator Regulador 1 de Interferon/metabolismo , Fatores Reguladores de Interferon/metabolismo , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Ativação Transcricional/efeitos dos fármacos
19.
PLoS One ; 11(6): e0156812, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27257682

RESUMO

Interferon Regulatory Factor-8 (IRF-8) serves as a key factor in the hierarchical differentiation towards monocyte/dendritic cell lineages. While much insight has been accumulated into the mechanisms essential for its hematopoietic specific expression, the mode of restricting IRF-8 expression in non-hematopoietic cells is still unknown. Here we show that the repression of IRF-8 expression in restrictive cells is mediated by its 3rd intron. Removal of this intron alleviates the repression of Bacterial Artificial Chromosome (BAC) IRF-8 reporter gene in these cells. Fine deletion analysis points to conserved regions within this intron mediating its restricted expression. Further, the intron alone selectively initiates gene silencing only in expression-restrictive cells. Characterization of this intron's properties points to its role as an initiator of sustainable gene silencing inducing chromatin condensation with suppressive histone modifications. This intronic element cannot silence episomal transgene expression underlining a strict chromatin-dependent silencing mechanism. We validated this chromatin-state specificity of IRF-8 intron upon in-vitro differentiation of induced pluripotent stem cells (iPSCs) into cardiomyocytes. Taken together, the IRF-8 3rd intron is sufficient and necessary to initiate gene silencing in non-hematopoietic cells, highlighting its role as a nucleation core for repressed chromatin during differentiation.


Assuntos
Cromatina/metabolismo , Fatores Reguladores de Interferon/metabolismo , Íntrons/genética , Macrófagos/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Citometria de Fluxo , Humanos , Fatores Reguladores de Interferon/genética , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Miócitos Cardíacos/metabolismo , Células NIH 3T3 , Células RAW 264.7 , Reação em Cadeia da Polimerase em Tempo Real
20.
Cell Rep ; 15(3): 651-665, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27068467

RESUMO

The thymus provides a unique microenvironment enabling development and selection of T lymphocytes. Medullary thymic epithelial cells (mTECs) play a pivotal role in this process by facilitating negative selection of self-reactive thymocytes and the generation of Foxp3(+) regulatory T cells. Although studies have highlighted the non-canonical nuclear factor κB (NF-κB) pathway as the key regulator of mTEC development, comprehensive understanding of the molecular pathways regulating this process still remains incomplete. Here, we demonstrate that the development of functionally competent mTECs is regulated by the histone deacetylase 3 (Hdac3). Although histone deacetylases are global transcriptional regulators, this effect is highly specific only to Hdac3, as neither Hdac1 nor Hdac2 inactivation caused mTEC ablation. Interestingly, Hdac3 induces an mTEC-specific transcriptional program independently of the previously recognized RANK-NFκB signaling pathway. Thus, our findings uncover yet another layer of complexity of TEC lineage divergence and highlight Hdac3 as a major and specific molecular switch crucial for mTEC differentiation.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/enzimologia , Histona Desacetilases/metabolismo , Timócitos/citologia , Timócitos/enzimologia , Animais , Núcleo Celular/metabolismo , Proteínas Correpressoras/metabolismo , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Histona Desacetilases/deficiência , Tolerância Imunológica , Camundongos , Modelos Biológicos , NF-kappa B/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA