Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 4792, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839760

RESUMO

Innate lymphoid cell precursors (ILCPs) develop into distinct subsets of innate lymphoid cells (ILCs) with specific functions. The epigenetic program underlying the differentiation of ILCPs into ILC subsets remains poorly understood. Here, we reveal the genome-wide distribution and dynamics of the DNA methylation and hydroxymethylation in ILC subsets and their respective precursors. Additionally, we find that the DNA hydroxymethyltransferase TET1 suppresses ILC1 but not ILC2 or ILC3 differentiation. TET1 deficiency promotes ILC1 differentiation by inhibiting TGF-ß signaling. Throughout ILCP differentiation at postnatal stage, gut microbiota contributes to the downregulation of TET1 level. Microbiota decreases the level of cholic acid in the gut, impairs TET1 expression and suppresses DNA hydroxymethylation, ultimately resulting in an expansion of ILC1s. In adult mice, TET1 suppresses the hyperactivation of ILC1s to maintain intestinal homeostasis. Our findings provide insights into the microbiota-mediated epigenetic programming of ILCs, which links microbiota-DNA methylation crosstalk to ILC differentiation.


Assuntos
Diferenciação Celular , Metilação de DNA , Proteínas de Ligação a DNA , Imunidade Inata , Linfócitos , Proteínas Proto-Oncogênicas , Animais , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Camundongos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Linfócitos/metabolismo , Linfócitos/imunologia , Camundongos Endogâmicos C57BL , Microbioma Gastrointestinal , Epigênese Genética , Camundongos Knockout , Fator de Crescimento Transformador beta/metabolismo , Transdução de Sinais
2.
Biofactors ; 50(2): 266-293, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38059412

RESUMO

Ferroptosis is a new form of regulated cell death caused by iron-dependent accumulation of lethal polyunsaturated phospholipids peroxidation. It has received considerable attention owing to its putative involvement in a wide range of pathophysiological processes such as organ injury, cardiac ischemia/reperfusion, degenerative disease and its prevalence in plants, invertebrates, yeasts, bacteria, and archaea. To counter ferroptosis, living organisms have evolved a myriad of intrinsic efficient defense systems, such as cyst(e)ine-glutathione-glutathione peroxidase 4 system (cyst(e)ine-GPX4 system), guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin (BH4) system (GCH1/BH4 system), ferroptosis suppressor protein 1/coenzyme Q10 system (FSP1/CoQ10 system), and so forth. Among these, GPX4 serves as the only enzymatic protection system through the reduction of lipid hydroperoxides, while other defense systems ultimately rely on small compounds to scavenge lipid radicals and prevent ferroptotic cell death. In this article, we systematically summarize the chemical biology of lipid radical trapping process by endogenous chemicals, such as coenzyme Q10 (CoQ10), BH4, hydropersulfides, vitamin K, vitamin E, 7-dehydrocholesterol, with the aim of guiding the discovery of novel ferroptosis inhibitors.


Assuntos
Cistos , Ubiquinona , Humanos , Ubiquinona/metabolismo , Peroxidação de Lipídeos , Morte Celular , Peróxidos Lipídicos/metabolismo
3.
Small ; : e2307365, 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37990372

RESUMO

In search of effective therapeutics for breast cancers, establishing physiologically relevant in vitro models is of great benefit to facilitate the clinical translation. Despite extensive progresses, it remains to develop the tumor models maximally recapturing the key pathophysiological attributes of their native counterparts. Therefore, the current study aimed to develop a microsphere-enabled modular approach toward the formation of in vitro breast tumor models with the capability of incorporating various selected cells while retaining spatial organization. Poly (lactic-co-glycolic acid) microspheres (150-200 mm) with tailorable pore size and surface topography are fabricated and used as carriers to respectively lade with breast tumor-associated cells. Culture of cell-laden microspheres assembled within a customized microfluidic chamber allowed to form 3D tumor models with spatially controlled cell distribution. The introduction of endothelial cell-laden microspheres into cancer-cell laden microspheres at different ratios would induce angiogenesis within the culture to yield vascularized tumor. Evaluation of anticancer drugs such as doxorubicin and Cediranib on the tumor models do demonstrate corresponding physiological responses. Clearly, with the ability to modulate microsphere morphology, cell composition and spatial distribution, microsphere-enabled 3D tumor tissue formation offers a high flexibility to satisfy the needs for pathophysiological study, anticancer drug screening or design of personalized treatment.

4.
J Exp Med ; 220(10)2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37549024

RESUMO

The neuro-immune regulation is associated with homeostasis of the intestine. Intestinal group 3 innate lymphoid cells (ILC3s) are tissue-resident lymphocytes whose functions are affected by the intestine niche. However, how a gut neuronal signal coordinates the immune response of ILC3s is largely unknown. Here, we found that cyclic adenosine monophosphate (cAMP) signaling exacerbated the inflammatory response and attenuated the expression level of the transcription factor forkhead box O1 (FOXO1) in ILC3s. Deficiency of FOXO1 drove the hyperactivation of ILC3s and resulted in gut inflammation independently of T cells. Mechanistically, FOXO1 promoted the transcription of neuropeptide receptor VIPR2 and inhibited the transcription of adrenoceptor ADRA2A in ILC3s. FOXO1-related regulation of VIPR2 and ADRA2A signaling balanced the activation of ILC3s under steady condition or during colitis. Moreover, chronic stress elevated cAMP level and downregulated FOXO1 level, exacerbating intestinal inflammation. Our findings reveal that FOXO1 balances the activation of ILC3s via VIP and adrenergic signaling and regulates intestinal homeostasis.


Assuntos
Imunidade Inata , Linfócitos , Humanos , Transdução de Sinais , Inflamação/metabolismo , Homeostase , Proteína Forkhead Box O1/metabolismo
5.
Cell Host Microbe ; 31(3): 418-432.e8, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36893736

RESUMO

The intestinal microbiota plays an important role in colorectal cancer (CRC) progression. However, the effect of tissue-resident commensal bacteria on CRC immune surveillance remains poorly understood. Here, we analyzed the intratissue bacteria from CRC patient colon tissues. We found that the commensal bacteria belonging to the Lachnospiraceae family, including Ruminococcus gnavus (Rg), Blautia producta (Bp), and Dorea formicigenerans (Df), were enriched in normal tissues, while Fusobacterium nucleatum (Fn) and Peptostreptococcus anaerobius (Pa) were abundant in tumor tissues. Tissue-resident Rg and Bp reduced colon tumor growth and promoted the activation of CD8+ T cells in immunocompetent mice. Mechanistically, intratissue Rg and Bp degraded lyso-glycerophospholipids that inhibited CD8+ T cell activity and maintained the immune surveillance function of CD8+ T cells. Lyso-glycerophospholipids alone promoted tumor growth that was abrogated with Rg and Bp injection. Collectively, intratissue Lachnospiraceae family bacteria facilitate the immune surveillance function of CD8+ T cells and control colorectal cancer progression.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Animais , Camundongos , Neoplasias Colorretais/microbiologia , Linfócitos T CD8-Positivos , Carcinogênese , Neoplasias do Colo/microbiologia , Fusobacterium nucleatum
6.
Cell Mol Immunol ; 20(3): 264-276, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36600053

RESUMO

The NLRP3 inflammasome plays an essential role in resistance to bacterial infection. The nervous system secretes multiple neuropeptides affecting the nervous system as well as immune cells. The precise impact of the neuropeptide CGRP on NLRP3 inflammasome activation is still unclear. Here, we show that CGRP negatively regulates the antibacterial process of host cells. CGRP prevents NLRP3 inflammasome activation and reduces mature IL-1ß secretion. Following NLRP3 inflammasome stimulation that triggers endosome leakage, CGRP internalized to endosomal compartments is released into the cell cytosol. Cytosolic CGRP binds directly to NLRP3 and dismantles the NLRP3-NEK7 complex, which is crucial for NLRP3 inflammasome activation. CGRP administration exacerbates bacterial infection, while the treatment with a CGRP antagonist has the opposite effect. Our study uncovers a unique role of CGRP in inhibiting inflammasome activation during infections, which might shed new light on antibacterial therapies in the future.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Citosol/metabolismo , Macrófagos , Interleucina-1beta/metabolismo
7.
WIREs Mech Dis ; 14(2): e1543, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35266652

RESUMO

Immune memory is well known as a signature of the adaptive immune system. Recently, enhanced responses to subsequent triggers are also observed in innate immune system, termed trained immunity (TI). Awakening of innate immune memory is required for host defense, such as anti-pathogen and anti-tumor responses. However, hyper-reactivation of trained innate immune cells also gives rise to undesirable inflammation. Mucosa immune system serves as the first defense line against pathogens. Trained immunity of mucosal immune system is tightly associated with the outcomes of mucosal diseases. In this review, we discuss the role of trained immunity in mucosal-associated diseases and the underlying mechanisms. We summarize the metabolic and epigenetic changes of trained immune cells and highlight their potential in clinical treatment. This article is categorized under: Infectious Diseases > Molecular and Cellular Physiology.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Humanos , Sistema Imunitário , Memória Imunológica , Inflamação
8.
Oncogene ; 39(27): 5068-5081, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32528131

RESUMO

Topoisomerase II poisons are one of the most common class of chemotherapeutics used in cancer. We and others had shown that a subset of glioblastomas, the most malignant of all primary brain tumors in adults, is responsive to TOP2 poisons. To identify genes that confer susceptibility to this drug in gliomas, we performed a genome-scale CRISPR knockout screen with etoposide. Genes involved in protein synthesis and DNA damage were implicated in etoposide susceptibility. To define potential biomarkers for TOP2 poisons, CRISPR hits were overlapped with genes whose expression correlates with susceptibility to this drug across glioma cell lines, revealing ribosomal protein subunit RPS11, 16, and 18 as putative biomarkers for response to TOP2 poisons. Loss of RPS11 led to resistance to etoposide and doxorubicin and impaired the induction of proapoptotic gene APAF1 following treatment. The expression of these ribosomal subunits was also associated with susceptibility to TOP2 poisons across cell lines from gliomas and multiple other cancers.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Etoposídeo/farmacologia , Glioblastoma/tratamento farmacológico , Proteínas Ribossômicas/metabolismo , Inibidores da Topoisomerase II/farmacologia , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Neoplasias Encefálicas/genética , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Doxorrubicina/farmacologia , Técnicas de Inativação de Genes , Glioblastoma/genética , Humanos
9.
Protein Expr Purif ; 167: 105544, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31759085

RESUMO

Leucine aminopeptidase (LAP), an exopeptidase that releases amino acid residues, especially leucine, from the N-terminus of polypeptides, is often applied to debitter protein hydrolysate in the food industry. However, there are no thermostable and high activity enzymes that can be used in the food industry. In this study, we obtained the highly active and thermostable leucine aminopeptidases screened from the thermophilic fungi Thermomyces lanuginosus, Talaromyces thermophilus, and Malbranchea cinnamomea. The activity of the recombinant leucine aminopeptidase Thelap was significantly increased to 2771.5 U/mL, as mediated by the CRISPR/Cas9 tool. The recombinant Thelap was easily purified from fermentation broth by Ni-affinity chromatography, and the specific activity of the purified Thelap was increased to 7449.6 U/mg. The recombinant Thelap showed optimal activity at pH 8.5 and 75 °C and remained above 70% of the maximum activity over a wide temperature range (30-80 °C). With regard to temperature stability, Thelap retained more than 90% activity when it was incubated at 65-75 °C for 2 h. K+ and Co2+ increased the enzyme activity of the recombinant Thelap, while Ba2+, Mn2+, Ni2+, Ca2+, Mg2+ and SDS inhibited its enzyme activity, and the inhibition capacity of Mg2+ was the weakest. Upon application in soy protein hydrolysis, Thelap could significantly increase the degree of hydrolysis and remove more hydrophobic amino acids from the N-terminal region of the polypeptide to decrease the bitterness.


Assuntos
Eurotiales/metabolismo , Leucil Aminopeptidase/biossíntese , Aspergillus niger/genética , Fermentação , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Expressão Gênica , Leucil Aminopeptidase/química , Leucil Aminopeptidase/metabolismo , Proteínas Recombinantes , Proteínas de Soja/metabolismo
10.
Exp Neurol ; 320: 112980, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31229638

RESUMO

Our earlier work generated a powerful platform technology of polymeric scaffolding of stem cells to investigate and treat the injured or diseased central nervous system. However, the reciprocal sequelae between biophysical properties of the polymer and responses of the stem cell have not been examined in situ in lesioned spinal cords. We postulated that implantable synthetic scaffolds, acting through physical features, might affect donor cell behavior and host tissue remodeling. To test this hypothesis, poly(d,l-lactic-co-glycolic acid) (PLGA) in either low/soft or high/hard rigidity was fabricated for carrying adult human bone marrow mesenchymal stromal stem cells (hMSCs). The construct was transplanted into the epicenter of a rat model of acute T9-10 segmental hemisection to evaluate the effect of PLGA rigidity on the therapeutic potential and fate of hMSCs for neural repair. Compared to controls, only treatment with soft PLGA-scaffolded hMSCs significantly improved sensorimotor function via activation of recovery neurobiology mechanisms. The main benefits included inhibiting neuroinflammation and enhancing tissue protection. Also detected in the treated lesion region were expressions of neurotrophic and anti-inflammatory factors together with proliferation of endogenous neural stem cells, impacts likely derived from hMSCs' functional multipotency maintained by soft PLGA-scaffolding. Conversely, hard rigidity PLGA activated mechanotransduction and mesoderm lineage differentiation of hMSCs that ectopically produced bone, cartilage and muscle markers in neural parenchyma. The findings collectively suggested that the physical texture of polymeric scaffolds should be tailored for sustaining the stemness of hMSCs to constructively interact with the spinal cord for functional restoration.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Traumatismos da Medula Espinal , Alicerces Teciduais/química , Animais , Feminino , Humanos , Ratos , Ratos Sprague-Dawley
11.
Theranostics ; 9(7): 2071-2083, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31037157

RESUMO

Glioblastoma (GBM) remains the most lethal and untreatable central nervous system malignancy. The challenges to devise novel and effective anti-tumor therapies include difficulty in locating the precise tumor border for complete surgical resection, and rapid regrowth of residual tumor tissue after standard treatment. Repeatable and non-invasive intranasal application of neural stem cells (NSCs) was recently shown to enable clinically relevant delivery of therapy to tumors. Treatment with chemotactic NSCs demonstrated significant survival benefits when coupled with radiation and oncolytic virotherapy in preclinical models of GBM. In order to further augment the clinical applicability of this novel therapeutic platform, we postulate that the FDA-approved compound, methimazole (MT), can be safely utilized to delay the nasal clearance and improve the ability of NSCs to penetrate the olfactory epithelium for robust in vivo brain tumor targeting and therapeutic actions. METHODS: To examine the role of reversible reduction of the olfactory epithelial barrier in non-invasive intranasal delivery, we explored the unique pharmacologic effect of MT at a single dosage regimen. In our proof-of-concept studies, quantitative magnetic resonance imaging (MRI), immunocytochemistry, and survival analysis were performed on glioma-bearing mice treated with a single dose of MT prior to intranasal anti-GBM therapy using an oncolytic virus (OV)-loaded NSCs. RESULTS: Based on histology and in vivo imaging, we found that disrupting the olfactory epithelium with MT effectively delays clearance and allows NSCs to persist in the nasal cavity for at least 24 h. MT pretreatment amplified the migration of NSCs to the tumor. The therapeutic advantage of this enhancement was quantitatively validated by tissue analysis and MRI tracking of NSCs loaded with superparamagnetic iron oxide nanoparticles (SPIOs) in live animals. Moreover, we observed significant survival benefits in GBM-bearing mice treated with intranasal delivery of oncolytic virus-loaded NSCs following MT injection. CONCLUSION: Our work identified a novel pharmacologic strategy to accelerate the clinical application of the non-invasive NSCs-based therapeutic platform to tackle aggressive brain tumors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Mucosa Nasal/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Adenoviridae/genética , Animais , Neoplasias Encefálicas/virologia , Linhagem Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glioblastoma/virologia , Glioma/tratamento farmacológico , Glioma/virologia , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos , Camundongos Nus , Mucosa Nasal/virologia , Células-Tronco Neurais/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
12.
Mol Neurobiol ; 56(7): 5032-5040, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30460615

RESUMO

The immunosuppressive microenvironment is one of the major factors promoting the growth of glioblastoma multiforme (GBM). Infiltration of CD4+CD25+Foxp3+ regulatory T cells (Tregs) into the tumor microenvironment plays a significant role in the suppression of the anti-tumor immunity and portends a dismal prognosis for patients. Glioma-mediated secretion of chemo-attractant C-C motif ligand 2 and 22 (CCL2/22) has previously been shown by our group to promote Treg migration in vitro. In this study, we show that a local implantation of platelet-rich fibrin patch (PRF-P) into the brain of GL261 glioma-bearing mice prolonged the survival of affected animals by 42.85% (p = 0.0011). Analysis performed on brain tumor tissue harvested from PRF-P-treated mice revealed a specific decrease in intra-tumoral lymphocytes with a preferential depletion of immunosuppressive Tregs. Importantly, co-culture of GL261 or chemo-attractants (CCL2/22) with PRF-P abrogated Treg migration. Pharmacological blockade of the CCL2/22 interaction with their receptors potentiated the inhibitory effect of PRF-P on Tregs recruitment in culture. Moreover, our findings revealed the soluble CD40 ligand (sCD40L) as a major Treg inhibitory player produced by activated platelets entrapped within the fibrin matrix of the PRF-P. Blockade of sCD40L restored the migratory capacity of Tregs, emphasizing the role of PRF-P in preventing the Treg migration to glioma tissue. Our findings highlight autologous PRF-P as a personalized, Treg-selective suppression platform that can potentially supplement and enhance the efficacy of glioma therapies.


Assuntos
Autoenxertos , Neoplasias Encefálicas/terapia , Glioma/terapia , Fibrina Rica em Plaquetas/fisiologia , Linfócitos T Reguladores/imunologia , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Craniotomia/métodos , Glioma/imunologia , Glioma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Reguladores/metabolismo , Microambiente Tumoral/imunologia
13.
Neurotherapeutics ; 15(4): 1127-1138, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30027430

RESUMO

Antitumor immunotherapeutic strategies represent an especially promising set of approaches with rapid translational potential considering the dismal clinical context of high-grade gliomas. Dendritic cells (DCs) are the body's most professional antigen-presenting cells, able to recruit and activate T cells to stimulate an adaptive immune response. In this regard, specific loading of tumor-specific antigen onto dendritic cells potentially represents one of the most advanced strategies to achieve effective antitumor immunization. In this study, we developed a DC-specific adenoviral (Ad) vector, named Ad5scFvDEC205FF, targeting the DC surface receptor, DEC205. In vitro analysis shows that 60% of DCs was infected by this vector while the infectivity of other control adenoviral vectors was less than 10%, demonstrating superior infectivity on DCs. Moreover, an average of 14% of DCs were infected by Ad5scFvDEC205FF-GFP, while less than 3% of non-DCs were infected following in vivo administration, demonstrating highly selective in vivo DC infection. Importantly, vaccination with this vehicle expressing human glioma-specific antigen, Ad5scFvDEC205FF-CMV-IE, shows a prolonged survival benefit in GL261CMV-IE-implanted murine glioma models (p < 0.0007). Furthermore, when rechallenged, cancerous cells were completely rejected. In conclusion, our novel, viral-mediated, DC-based immunization approach has the significant therapeutic potential for patients with high-grade gliomas.


Assuntos
Imunidade Adaptativa/genética , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Neoplasias Encefálicas , Células Dendríticas/imunologia , Glioma , Lectinas Tipo C/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Receptores de Superfície Celular/metabolismo , Adenoviridae/genética , Análise de Variância , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Células Dendríticas/virologia , Modelos Animais de Doenças , Vetores Genéticos/metabolismo , Glioma/imunologia , Glioma/patologia , Glioma/terapia , Células HEK293 , Humanos , Linfonodos/citologia , Camundongos , Baço/citologia , Transdução Genética , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Biomed Pharmacother ; 103: 1658-1663, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29864955

RESUMO

Platinum-based chemotherapeutic drugs, especial cisplatin, are the most common and effective anticancer drugs to treat the non-small cell lung cancer (NSCLC), but the major obstacle of this treatment is the resistance to chemotherapeutic drugs due to the anti-apoptosis of cancer cells. In our study, we found that Canopy homolog 2 (CNPY2) is increased in NSCLC tissues compared to the normal lung tissues, and the upregulation of CNPY2 is correlated with poor survival. Next, colony formation, annexin V-binding and TUNEL assays revealed that overexpression of CNPY2 inhibits the apoptosis of NSCLC cells induced by cisplatin. Further assays demonstrated that the anti-apoptosis may be aroused by the hyperactivation of NF-κB signaling pathway, and blocking the NF-κB pathway promotes the apoptosis of CNPY2-upregulating cells. The above results suggest that CNPY2 can serves as a therapeutic target to promote the effect of chemotherapy in NSCLC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose , Carcinoma Pulmonar de Células não Pequenas/patologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares/patologia , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
16.
Proc Natl Acad Sci U S A ; 114(30): E6147-E6156, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28696296

RESUMO

Brain tumor-initiating cells (BTICs) have been identified as key contributors to therapy resistance, recurrence, and progression of diffuse gliomas, particularly glioblastoma (GBM). BTICs are elusive therapeutic targets that reside across the blood-brain barrier, underscoring the urgent need to develop novel therapeutic strategies. Additionally, intratumoral heterogeneity and adaptations to therapeutic pressure by BTICs impede the discovery of effective anti-BTIC therapies and limit the efficacy of individual gene targeting. Recent discoveries in the genetic and epigenetic determinants of BTIC tumorigenesis offer novel opportunities for RNAi-mediated targeting of BTICs. Here we show that BTIC growth arrest in vitro and in vivo is accomplished via concurrent siRNA knockdown of four transcription factors (SOX2, OLIG2, SALL2, and POU3F2) that drive the proneural BTIC phenotype delivered by multiplexed siRNA encapsulation in the lipopolymeric nanoparticle 7C1. Importantly, we demonstrate that 7C1 nano-encapsulation of multiplexed RNAi is a viable BTIC-targeting strategy when delivered directly in vivo in an established mouse brain tumor. Therapeutic potential was most evident via a convection-enhanced delivery method, which shows significant extension of median survival in two patient-derived BTIC xenograft mouse models of GBM. Our study suggests that there is potential advantage in multiplexed targeting strategies for BTICs and establishes a flexible nonviral gene therapy platform with the capacity to channel multiplexed RNAi schemes to address the challenges posed by tumor heterogeneity.


Assuntos
Glioblastoma/patologia , Nanopartículas/uso terapêutico , Interferência de RNA , Animais , Carcinogênese/genética , Resistencia a Medicamentos Antineoplásicos , Feminino , Terapia Genética/métodos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Vis Exp ; (124)2017 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-28605391

RESUMO

The intrinsic tropism towards brain malignancies renders stem cells as promising carriers of therapeutic agents against malignant tumors. The delivery of therapeutic stem cells via the intranasal route is a recently discovered alternative strategy, with strong potential for clinical translation, due to its non-invasive nature compared to intracranial implantation or delivery via systemic routes. The lack of blood brain barrier further strengthens the therapeutic potential of stem cells undergoing intranasal brain entry. This article summarizes the essential techniques utilized in our studies and outlines the basic principles of intranasal strategy for stem cell delivery using a mouse model of intracranial glioma xenografts. We demonstrate the optimized procedures that generate consistent and reproducible results with specific predetermined experimental parameters and offer guidelines for streamlined work flow that ensure efficient execution and reliable experimental outcome. The article is designed to serve as a baseline for further experimental customization based on hypothesis, stem cell types, or tumor specifics.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Administração Intranasal , Animais , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Receptores CXCR4/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Proc Natl Acad Sci U S A ; 114(5): E820-E829, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28096400

RESUMO

Mesenchymal stromal stem cells (MSCs) isolated from adult tissues offer tangible potential for regenerative medicine, given their feasibility for autologous transplantation. MSC research shows encouraging results in experimental stroke, amyotrophic lateral sclerosis, and neurotrauma models. However, further translational progress has been hampered by poor MSC graft survival, jeopardizing cellular and molecular bases for neural repair in vivo. We have devised an adult human bone marrow MSC (hMSC) delivery formula by investigating molecular events involving hMSCs incorporated in a uniquely designed poly(lactic-co-glycolic) acid scaffold, a clinically safe polymer, following inflammatory exposures in a dorsal root ganglion organotypic coculture system. Also, in rat T9-T10 hemisection spinal cord injury (SCI), we demonstrated that the tailored scaffolding maintained hMSC stemness, engraftment, and led to robust motosensory improvement, neuropathic pain and tissue damage mitigation, and myelin preservation. The scaffolded nontransdifferentiated hMSCs exerted multimodal effects of neurotrophism, angiogenesis, neurogenesis, antiautoimmunity, and antiinflammation. Hindlimb locomotion was restored by reestablished integrity of submidbrain circuits of serotonergic reticulospinal innervation at lumbar levels, the propriospinal projection network, neuromuscular junction, and central pattern generator, providing a platform for investigating molecular events underlying the repair impact of nondifferentiated hMSCs. Our approach enabled investigation of recovery neurobiology components for injured adult mammalian spinal cord that are different from those involved in normal neural function. The uncovered neural circuits and their molecular and cellular targets offer a biological underpinning for development of clinical rehabilitation therapies to treat disabilities and complications of SCI.


Assuntos
Transplante de Células-Tronco Mesenquimais , Traumatismos da Medula Espinal/terapia , Adulto , Animais , Técnicas de Cocultura , Feminino , Gânglios Espinais , Membro Posterior , Humanos , Ácido Láctico , Lipopolissacarídeos , Locomoção , Masculino , Células-Tronco Mesenquimais , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/fisiopatologia , Alicerces Teciduais
19.
Stem Cell Reports ; 7(3): 471-482, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27594591

RESUMO

The challenges to effective drug delivery to brain tumors are twofold: (1) there is a lack of non-invasive methods of local delivery and (2) the blood-brain barrier limits systemic delivery. Intranasal delivery of therapeutics to the brain overcomes both challenges. In mouse model of malignant glioma, we observed that a small fraction of intranasally delivered neural stem cells (NSCs) can migrate to the brain tumor site. Here, we demonstrate that hypoxic preconditioning or overexpression of CXCR4 significantly enhances the tumor-targeting ability of NSCs, but without altering their phenotype only in genetically modified NSCs. Modified NSCs deliver oncolytic virus to glioma more efficiently and extend survival of experimental animals in the context of radiotherapy. Our findings indicate that intranasal delivery of stem cell-based therapeutics could be optimized for future clinical applications, and allow for safe and repeated administration of biological therapies to brain tumors and other CNS disorders.


Assuntos
Glioma/genética , Glioma/patologia , Células-Tronco Neurais/metabolismo , Terapia Viral Oncolítica , Receptores CXCR4/genética , Administração Intranasal , Animais , Biomarcadores , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glioma/mortalidade , Glioma/terapia , Humanos , Hipóxia/metabolismo , Camundongos , Vírus Oncolíticos/genética , Fenótipo , Receptores CXCR4/metabolismo , Transplante de Células-Tronco , Transdução Genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cancer Res ; 76(19): 5671-5682, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27530322

RESUMO

In many aggressive cancers, such as glioblastoma multiforme, progression is enabled by local immunosuppression driven by the accumulation of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC). However, the mechanistic details of how Tregs and MDSCs are recruited in various tumors are not yet well understood. Here we report that macrophages and microglia within the glioma microenvironment produce CCL2, a chemokine that is critical for recruiting both CCR4+ Treg and CCR2+Ly-6C+ monocytic MDSCs in this disease setting. In murine gliomas, we established novel roles for tumor-derived CCL20 and osteoprotegerin in inducing CCL2 production from macrophages and microglia. Tumors grown in CCL2-deficient mice failed to maximally accrue Tregs and monocytic MDSCs. In mixed-bone marrow chimera assays, we found that CCR4-deficient Treg and CCR2-deficient monocytic MDSCs were defective in glioma accumulation. Furthermore, administration of a small-molecule antagonist of CCR4 improved median survival in the model. In clinical specimens of glioblastoma multiforme, elevated levels of CCL2 expression correlated with reduced overall survival of patients. Finally, we found that CD163-positive infiltrating macrophages were a major source of CCL2 in glioblastoma multiforme patients. Collectively, our findings show how glioma cells influence the tumor microenvironment to recruit potent effectors of immunosuppression that drive progression. Cancer Res; 76(19); 5671-82. ©2016 AACR.


Assuntos
Neoplasias Encefálicas/imunologia , Quimiocina CCL2/fisiologia , Glioma/imunologia , Células Supressoras Mieloides/fisiologia , Linfócitos T Reguladores/fisiologia , Microambiente Tumoral , Animais , Neoplasias Encefálicas/mortalidade , Movimento Celular , Glioma/mortalidade , Humanos , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Receptores CCR4/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA