Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 7(74): eabl3795, 2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-35984892

RESUMO

A diet rich in saturated fat and carbohydrates causes low-grade chronic inflammation in several organs, including the liver, ultimately driving nonalcoholic steatohepatitis. In this setting, environment-driven lipotoxicity and glucotoxicity induce liver damage, which promotes dendritic cell activation and generates a major histocompatibility complex class II (MHC-II) immunopeptidome enriched with peptides derived from proteins involved in cellular metabolism, oxidative phosphorylation, and the stress responses. Here, we demonstrated that lipotoxicity and glucotoxicity, as driven by a high-fat and high-fructose (HFHF) diet, promoted MHC-II presentation of nested T and B cell epitopes from protein disulfide isomerase family A member 3 (PDIA3), which is involved in immunogenic cell death. Increased MHC-II presentation of PDIA3 peptides was associated with antigen-specific proliferation of hepatic CD4+ immune infiltrates and isotype switch of anti-PDIA3 antibodies from IgM to IgG3, indicative of cellular and humoral PDIA3 autoreactivity. Passive transfer of PDIA3-specific T cells or PDIA3-specific antibodies also exacerbated hepatocyte death, as determined by increased hepatic transaminases detected in the sera of mice subjected to an HFHF but not control diet. Increased humoral responses to PDIA3 were also observed in patients with chronic inflammatory liver conditions, including autoimmune hepatitis, primary biliary cholangitis, and type 2 diabetes. Together, our data indicated that metabolic insults caused by an HFHF diet elicited liver damage and promoted pathogenic immune autoreactivity driven by T and B cell PDIA3 epitopes.


Assuntos
Autoimunidade , Diabetes Mellitus Tipo 2 , Fígado , Isomerases de Dissulfetos de Proteínas , Animais , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Epitopos , Antígenos de Histocompatibilidade Classe II , Fígado/patologia , Camundongos , Peptídeos , Isomerases de Dissulfetos de Proteínas/imunologia , Isomerases de Dissulfetos de Proteínas/metabolismo
2.
Cancer Biother Radiopharm ; 35(6): 459-473, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32013538

RESUMO

An α particle-emitting nanodrug that is a potent and specific antitumor agent and also prompts significant remodeling of local immunity in the tumor microenvironment (TME) has been developed and may impact the treatment of melanoma. Biocompatible ultrasmall fluorescent core-shell silica nanoparticles (C' dots, diameter ∼6.0 nm) have been engineered to target the melanocortin-1 receptor expressed on melanoma through α melanocyte-stimulating hormone peptides attached to the C' dot surface. Actinium-225 is also bound to the nanoparticle to deliver a densely ionizing dose of high-energy α particles to cancer. Nanodrug pharmacokinetic properties are optimal for targeted radionuclide therapy as they exhibit rapid blood clearance, tumor-specific accumulation, minimal off-target localization, and renal elimination. Potent and specific tumor control, arising from the α particles, was observed in a syngeneic animal model of melanoma. Surprisingly, the C' dot component of this drug initiates a favorable pseudopathogenic response in the TME generating distinct changes in the fractions of naive and activated CD8 T cells, Th1 and regulatory T cells, immature dendritic cells, monocytes, MΦ and M1 macrophages, and activated natural killer cells. Concomitant upregulation of the inflammatory cytokine genome and adaptive immune pathways each describes a macrophage-initiated pseudoresponse to a viral-shaped pathogen. This study suggests that therapeutic α-particle irradiation of melanoma using ultrasmall functionalized core-shell silica nanoparticles potently kills tumor cells, and at the same time initiates a distinct immune response in the TME.


Assuntos
Partículas alfa/uso terapêutico , Portadores de Fármacos/química , Melanoma Experimental/radioterapia , Compostos Radiofarmacêuticos/administração & dosagem , Neoplasias Cutâneas/radioterapia , Microambiente Tumoral/efeitos da radiação , Actínio/administração & dosagem , Actínio/farmacocinética , Animais , Linhagem Celular Tumoral/transplante , Biologia Computacional , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Imunidade Celular/genética , Imunidade Celular/efeitos da radiação , Masculino , Dose Máxima Tolerável , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Terapia de Alvo Molecular/métodos , Nanopartículas/química , RNA-Seq , Compostos Radiofarmacêuticos/farmacocinética , Receptor Tipo 1 de Melanocortina/antagonistas & inibidores , Receptor Tipo 1 de Melanocortina/metabolismo , Dióxido de Silício/química , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Distribuição Tecidual , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
3.
ACS Biomater Sci Eng ; 5(3): 1487-1496, 2019 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33405623

RESUMO

Hypertrophic scarring is a dermal disorder resulting from collagen and other extra cellular matrix protein depositions following the deep trauma, severe burn injury, and surgery incisions. A variety of therapeutic procedures are currently available, however, achieving an ideal treatment method remains a challenge. In our recently published report, a 3D bilayered decellularized human amniotic membrane/electrospun silk fibroin membrane was fabricated and characterized for regenerative medical applications. To obtain a solid bind between two layers, the samples were immersed in 70% ethanol. In this study, the effects of amniotic membrane/electrospun silk fibroin on minimizing the postinjury hypertrophic scar formation were determined in the rabbit ear model. In vivo experiments were carried out to assess the bilayer membrane characteristics on full thickness hypertrophic scar at days 28 and 50 postimplantations. A significant decrease in collagen deposition and expression and increased expression and deposition of MMP1 in the wound bed were observed on the wounds dressed with bilayered membrane when compared to the amniotic membrane alone and controls (wound with no implant). The current study shows that our fabricated construct has potential as an efficient antiscarring wound dressing material and may also serve for the subsequent soft tissue engineering needs.

4.
Cancer Res ; 78(14): 3747-3754, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29959150

RESUMO

Despite esophageal adenocarcinoma (EAC) being the most widespread among gastrointestinal cancers, with an 11-fold increase in the risk of cancer for patients with Barrett esophagus (BE), its prognosis is still poor. There is a critical need to better perceive the biology of cancer progression and identification of specific targets that are the hallmark of BE's progression. This review explores the established animal models of BE, including genetic, surgical and nonsurgical approaches, potential chemoprevention targets, and the reasoning behind their applications to prevent Barrett-related EAC. The key methodological features in the design feasibility of relevant studies are also discussed. Cancer Res; 78(14); 3747-54. ©2018 AACR.


Assuntos
Esôfago de Barrett/patologia , Esôfago de Barrett/prevenção & controle , Adenocarcinoma/patologia , Adenocarcinoma/prevenção & controle , Animais , Progressão da Doença , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/prevenção & controle , Humanos , Lesões Pré-Cancerosas/patologia , Prognóstico
5.
Biomacromolecules ; 19(7): 2409-2422, 2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29529861

RESUMO

Severe burn injuries can lead to delays in healing and devastating scar formation. Attempts have been made to develop a suitable skin substitute for the scarless healing of such skin wounds. Currently, there is no effective strategy for completely scarless healing after the thermal injuries. In our recent work, we fabricated and evaluated a 3D protein-based artificial skin made from decellularized human amniotic membrane (AM) and electrospun nanofibrous silk fibroin (ESF) in vitro. We also characterized both biophysical and cell culture investigation to establish in vitro performance of the developed bilayer scaffolds. In this report, we evaluate the appropriate utility of this fabricated bilayered artificial skin in vivo with particular emphasis on healing and scar formation due to the biochemical and biomechanical complexity of the skin. For this work, AM and AM/ESF membranes alone or seeded with adipose-tissue-derived mesenchymal stem cells (AT-MSCs) are implanted on full-thickness burn wounds in mice. The healing efficacy and scar formation are evaluated at 7, 14, and 28 days post-implantation in vivo. Our data reveal that ESF accelerates the wound-healing process through the early recruitment of inflammatory cells such as macrophages into the defective site as well as the up-regulation of angiogenic factors from the AT-MSCs and the facilitation of the remodeling phase. In vivo application of the prepared AM/ESF membrane seeded with the AT-MSCs reduces significantly the post-burn scars. The in vivo data suggest that the potential applications of the AM/ESF bilayered artificial skin may be considered a clinical translational product with stem cells to guide the scarless healing of severe burn injuries.


Assuntos
Queimaduras/terapia , Regeneração Tecidual Guiada/métodos , Pele Artificial , Cicatrização , Âmnio/química , Animais , Fibroínas/química , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C
6.
Carbohydr Polym ; 187: 66-84, 2018 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-29486846

RESUMO

Agarose is a natural polysaccharide polymer having unique characteristics that give reason to consider it for tissue engineering applications. Special characteristics of agarose such as its excellent biocompatibility, thermo-reversible gelation behavior and physiochemical features support its use as a biomaterial for cell growth and/or controlled/localized drug delivery. The resemblance of this natural carbohydrate polymer to the extracellular matrix results in attractive features that bring about a strong interest in its usage in the field. The scope of this review is to summarize the extensive researches addressing agarose-based biomaterials in order to provide an in-depth understanding of its tissue engineering-related applications.


Assuntos
Materiais Biocompatíveis/química , Hidrogéis/química , Polímeros/química , Engenharia Tecidual/métodos , Medicina Regenerativa/métodos
7.
J Colloid Interface Sci ; 516: 57-66, 2018 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-29408144

RESUMO

An innovative drug-loaded colloidal hydrogel was synthesized for applications in neural interfaces in tissue engineering by reacting carboxyl capped aniline dimer and gelatin molecules. Dexamethasone was loaded into the gelatin-aniline dimer solution as a model drug to form an in situ drug-loaded colloidal hydrogel. The conductivity of the hydrogel samples fluctuated around 10-5 S/cm which appeared suitable for cellular activities. Cyclic voltammetry was used for electroactivity determination, in which 2 redox states were observed, suggesting that the short chain length and steric hindrance prevented the gel from achieving a fully oxidized state. Rheological data depicted the modulus decreasing with aniline dimer increment due to limited hydrogen bonds accessibility. Though the swelling ratio of pristine gelatin (600%) decreased by the introduction and increasing the concentration of aniline dimer because of its hydrophobic nature, it took the value of 300% at worst, which still seems promising for drug delivery uses. Degradation rate of hydrogel was similarly decreased by adding aniline dimer. Drug release was evaluated in passive and stimulated patterns demonstrating tendency of aniline dimer to form a vesicle that controls the drug release behavior. The optimal cell viability, proper cell attachment and neurite extension was achieved in the case of hydrogel containing 10 wt% aniline dimer. Based on tissue/organ behavior, it was promisingly possible to adjust the characteristics of the hydrogels for an optimal drug release. The outcome of this simple and effective approach can potentially offer additional tunable characteristics for recording and stimulating purposes in neural interfaces.

8.
Biomed Mater ; 13(3): 035003, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29125135

RESUMO

Burn injuries have been reported to be an important cause of morbidity and mortality and they are still considered as unmet clinical need. Although there is a myriad of effective stem cells that have been suggested for skin regeneration, there is no one ideal scaffold. The aim of this study was to develop a three-dimensional (3D) bi-layer scaffold made of biological decellularized human amniotic membrane (AM) with viscoelastic electrospun nanofibrous silk fibroin (ESF) spun on top. The fabricated 3D bi-layer AM/ESF scaffold was submerged in ethanol to induce ß-sheet transformation as well as to get a tightly coated and inseparable bi-layer. The biomechanical and biological properties of the 3D bi-layer AM/ESF scaffold were investigated. The results indicate significantly improved mechanical properties of the AM/ESF compared with the AM alone. Both the AM and AM/ESF possess a variety of suitable adhesion cells without detectable cytotoxicity against adipose tissue-derived mesenchymal stem cells (AT-MSCs). The AT-MSCs show increased expression of two main pro-angiogenesis factors, vascular endothelial growth factor and basic fibroblast growth factor, when cultured on the AM/ESF for 7 days, when comparing with AM alone. The results suggest that the AM/ESF scaffold with autologous AT-MSCs has excellent cell adhesion and proliferation along with production of growth factors which serves as a possible application in a clinical setting for skin regeneration.


Assuntos
Âmnio/metabolismo , Fibroínas/química , Células-Tronco Mesenquimais/citologia , Pele Artificial , Animais , Fenômenos Biomecânicos , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Elasticidade , Eletroquímica , Etanol/química , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Nanofibras , Neovascularização Patológica , Neovascularização Fisiológica , Regeneração , Pele/lesões , Espectroscopia de Infravermelho com Transformada de Fourier , Estresse Mecânico , Engenharia Tecidual/métodos , Alicerces Teciduais , Viscosidade
9.
Curr Pharm Des ; 23(20): 2918-2929, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28425863

RESUMO

Drug delivery systems are effective and attractive methods which allow therapeutic substances to be introduced into the body more effectively and safe by having tunable delivery rate and release target site. Gold nanoparticles (AuNPs) have a myriad of favorable physical, chemical, optical, thermal and biological properties that make them highly suitable candidates as non-toxic carriers for drug and gene delivery. The surface modifications of AuNPs profoundly improve their circulation, minimize aggregation rates, enhance attachment to therapeutic molecules and target agents due to their nano range size which further increases their ability to cross cell membranes and reduce overall cytotoxicity. This comprehensive article reviews the applications of the AuNPs in drug delivery systems along with their corresponding surface modifications. The highlighting results obtained from the preclinical trial are promising and next five years have huge possibility move to the clinical setting.


Assuntos
Sistemas de Liberação de Medicamentos , Ouro/química , Nanopartículas Metálicas/química , Preparações Farmacêuticas/administração & dosagem , Humanos , Preparações Farmacêuticas/química
10.
ACS Appl Mater Interfaces ; 9(13): 11392-11404, 2017 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-28117963

RESUMO

In this study, we present a novel chitosan-intercalated montmorillonite/poly(vinyl alcohol) (OMMT/PVA) nanofibrous mesh as a microenvironment for guiding differentiation of human dental pulp stem cells (hDPSCs) toward neuronlike cells. The OMMT was prepared through ion exchange reaction between the montmorillonite (MMT) and chitosan. The PVA solutions containing various concentrations of OMMT were electrospun to form 3D OMMT-PVA nanofibrous meshes. The biomechanical and biological characteristics of the nanofibrous meshes were evaluated by ATR-FTIR, XRD, SEM, MTT, and LDH specific activity, contact angle, and DAPI staining. They were carried out for mechanical properties, overall viability, and toxicity of the cells. The hDPSCs were seeded on the prepared scaffolds and induced with neuronal specific differentiation media at two differentiation stages (2 days at preinduction stage and 6 days at induction stage). The neural differentiation of the cells cultured on the meshes was evaluated by determining the expression of Oct-4, Nestin, NF-M, NF-H, MAP2, and ßIII-tubulin in the cells after preinduction, at induction stages by real-time PCR (RT-PCR) and immunostaining. All the synthesized nanofibers exhibited a homogeneous morphology with a favorable mechanical behavior. The population of the cells differentiated into neuronlike cells in all the experimental groups was significantly higher than that in control group. The expression level of the neuronal specific markers in the cells cultured on 5% OMMT/PVA meshes was significantly higher than the other groups. This study demonstrates the feasibility of the OMMT/PVA artificial nerve graft cultured with hDPSCs for regeneration of damaged neural tissues. These fabricated matrices may have a potential in neural tissue engineering applications.


Assuntos
Polpa Dentária , Bentonita , Diferenciação Celular , Proliferação de Células , Quitosana , Humanos , Nanofibras , Álcool de Polivinil , Células-Tronco , Engenharia Tecidual , Alicerces Teciduais
11.
Oncotarget ; 8(1): 203-214, 2017 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-27448962

RESUMO

OBJECTIVE: The incidence of esophageal adenocarcinoma (EAC) is increasing, but factors contributing to malignant progression of its precursor lesion, Barrett's esophagus (BE), have not been defined. Hypergastrinemia caused by long-term use of proton pump inhibitors (PPIs), has been suggested as one possible risk factor. The gastrin receptor, CCK2R, is expressed in the cardia and upregulated in BE, suggesting the involvement of the gastrin-CCK2R pathway in progression. In the L2-IL-1ß mouse model, Barrett's-like esophagus arises from the gastric cardia. Therefore, we aimed to analyze the effect of hypergastrinemia on CCK2R+ progenitor cells in L2-IL-1ß mice. DESIGN: L2-IL-1ß mice were mated with hypergastrinemic (INS-GAS) mice or treated with PPIs to examine the effect of hypergastrinemia in BE progression. CCK2R-CreERT crossed with L2-IL-1ß mice were used to analyze the lineage progenitor potential of CCK2R+ cells. Cardia glands were cultured in vitro, and the effect of gastrin treatment analyzed. L2-IL-1ß mice were treated with a CCK2R antagonist YF476 as a potential chemopreventive drug. RESULTS: Hypergastrinemia resulted in increased proliferation and expansion of Barrett's-like esophagus. Lineage tracing experiments revealed that CCK2R+ cells are long-lived progenitors that can give rise to such lesions under chronic inflammation. Gastrin stimulated organoid growth in cardia culture, while CCK2R inhibition prevented Barrett's-like esophagus and dysplasia. CONCLUSIONS: Our data suggest a progression model for BE to EAC in which CCK2R+ progenitor cells, stimulated by hypergastrinemia, proliferate to give rise to metaplasia and dysplasia. Hypergastrinemia can result from PPI use, and the effects of hypergastrinemia in human BE should be studied further.


Assuntos
Esôfago de Barrett/etiologia , Esôfago de Barrett/metabolismo , Gastrinas/metabolismo , Regulação da Expressão Gênica , Mioblastos Cardíacos/metabolismo , Receptor de Colecistocinina B/genética , Animais , Esôfago de Barrett/patologia , Biomarcadores , Modelos Animais de Doenças , Progressão da Doença , Gastrinas/farmacologia , Hiperglicemia , Imuno-Histoquímica , Metaplasia , Camundongos , Camundongos Transgênicos , Mioblastos Cardíacos/efeitos dos fármacos , Fenótipo , Receptor de Colecistocinina B/metabolismo
12.
Cancer Cell ; 31(1): 21-34, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-27989802

RESUMO

Within the gastrointestinal stem cell niche, nerves help to regulate both normal and neoplastic stem cell dynamics. Here, we reveal the mechanisms underlying the cancer-nerve partnership. We find that Dclk1+ tuft cells and nerves are the main sources of acetylcholine (ACh) within the gastric mucosa. Cholinergic stimulation of the gastric epithelium induced nerve growth factor (NGF) expression, and in turn NGF overexpression within gastric epithelium expanded enteric nerves and promoted carcinogenesis. Ablation of Dclk1+ cells or blockade of NGF/Trk signaling inhibited epithelial proliferation and tumorigenesis in an ACh muscarinic receptor-3 (M3R)-dependent manner, in part through suppression of yes-associated protein (YAP) function. This feedforward ACh-NGF axis activates the gastric cancer niche and offers a compelling target for tumor treatment and prevention.


Assuntos
Acetilcolina/fisiologia , Fator de Crescimento Neural/fisiologia , Transdução de Sinais/fisiologia , Neoplasias Gástricas/etiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas de Ciclo Celular , Quinases Semelhantes a Duplacortina , Mucosa Gástrica/inervação , Camundongos , Camundongos Endogâmicos C57BL , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/análise , Receptor Muscarínico M3/fisiologia , Proteínas de Sinalização YAP
13.
Int J Artif Organs ; 39(10): 524-533, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27901555

RESUMO

INTRODUCTION: Amid the plethora of methods to repair critical bone defects, there is no one perfect approach. In this study, we sought to evaluate a potent 3-dimensional (3D) bioactive SiO2-CaO-P2O5 glasses (bioglass)/gelatin (gel) scaffold for its biocompatibility by seeding cells as well as for its regenerative properties by animal implantation. METHODS: Osteoblast cells were seeded onto nanocomposite scaffolds to investigate the process of critical-size calvarial defect via new bone formation. Scanning electron microscopy (SEM) was used to validate topography of the scaffolds, its homogeneity and ideal cellular attachment. Proliferation assay and confocal microscopy were used to evaluate its biocompatibility. To validate osteogenesis of the bioactive nanocomposite scaffolds, they were first implanted into rats and later removed and analyzed at different time points post mortem using histological, immunohistochemical and histomorphometric methods. RESULTS: Based on in vitro results, we showed that our nanocomposite is highly cell-compatible material and allows for osteoblasts to adhere, spread and proliferate. In vivo results indicate that our nanocomposite provides a significant contribution to bone regeneration and is highly biodegradable and biocompatible. So, seeded scaffolds with osteoblasts enhanced repair of critical bone defects via osteogenesis. CONCLUSIONS: We demonstrate the feasibility of engineering a nanocomposite scaffold with an architecture resembling the human bone, and provide proof-of-concept validation for our scaffold using a rat animal model.


Assuntos
Substitutos Ósseos , Cerâmica , Osteoblastos , Crânio/cirurgia , Alicerces Teciduais , Animais , Regeneração Óssea , Adesão Celular , Linhagem Celular , Sobrevivência Celular , Gelatina/química , Teste de Materiais , Nanocompostos , Osteogênese , Ratos Wistar , Crânio/lesões , Cicatrização
14.
Nanomedicine ; 12(7): 2181-2200, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27247186

RESUMO

Tissue engineering aims to develop therapeutic products that utilize a combination of scaffolds with viable cell systems or responsive biomolecules derived from such cells, for the repair, restoration/regeneration of tissues. Here, the main goal is to enable the body to heal itself by the introduction of electrospun scaffolds, such that the body recognizes them as its own and in turn uses them to regenerate "neo-native" functional tissues. During the last decade, innovative nanofibrous scaffolds have attracted substantial interest in bone tissue engineering. The electrospinning process makes it possible to fabricate appropriate scaffolds for bone tissue engineering from different categories of nanobiomaterials having the ability of controlled delivery of drugs in the defective tissues. It is expected that with the progress in science and technology, better bone constructs will be proposed in the future. This review discusses the innovative approaches into electrospinning techniques for the fabrication of nanofibrous scaffolds for bone tissue engineering.


Assuntos
Osso e Ossos , Nanofibras , Engenharia Tecidual , Humanos , Regeneração , Alicerces Teciduais
15.
Cell Stem Cell ; 18(4): 441-55, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27058937

RESUMO

The existence of adult pancreatic progenitor cells has been debated. While some favor the concept of facultative progenitors involved in homeostasis and repair, neither a location nor markers for such cells have been defined. Using genetic lineage tracing, we show that Doublecortin-like kinase-1 (Dclk1) labels a rare population of long-lived, quiescent pancreatic cells. In vitro, Dclk1+ cells proliferate readily and sustain pancreatic organoid growth. In vivo, Dclk1+ cells are necessary for pancreatic regeneration following injury and chronic inflammation. Accordingly, their loss has detrimental effects after cerulein-induced pancreatitis. Expression of mutant Kras in Dclk1+ cells does not affect their quiescence or longevity. However, experimental pancreatitis converts Kras mutant Dclk1+ cells into potent cancer-initiating cells. As a potential effector of Kras, Dclk1 contributes functionally to the pathogenesis of pancreatic cancer. Taken together, these observations indicate that Dclk1 marks quiescent pancreatic progenitors that are candidates for the origin of pancreatic cancer.


Assuntos
Carcinogênese/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Pancreatite/metabolismo , Pancreatite/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Administração Oral , Animais , Carcinogênese/patologia , Carcinoma Ductal Pancreático/induzido quimicamente , Carcinoma Ductal Pancreático/etiologia , Carcinoma Ductal Pancreático/patologia , Proliferação de Células , Quinases Semelhantes a Duplacortina , Camundongos , Organoides/citologia , Organoides/crescimento & desenvolvimento , Neoplasias Pancreáticas/induzido quimicamente , Neoplasias Pancreáticas/etiologia , Pancreatite/induzido quimicamente , Pancreatite/complicações , Proteínas Serina-Treonina Quinases/genética , Tamoxifeno/administração & dosagem
16.
Nat Commun ; 7: 10517, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26841680

RESUMO

CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSCs) expand in the spleen during cancer and promote progression through suppression of cytotoxic T cells. An anti-inflammatory reflex arc involving the vagus nerve and memory T cells is necessary for resolution of acute inflammation. Failure of this neural circuit could promote procarcinogenic inflammation and altered tumour immunity. Here we show that splenic TFF2, a secreted anti-inflammatory peptide, is released by vagally modulated memory T cells to suppress the expansion of MDSCs through CXCR4. Splenic denervation interrupts the anti-inflammatory neural arc, resulting in the expansion of MDSCs and colorectal cancer. Deletion of Tff2 recapitulates splenic denervation to promote carcinogenesis. Colorectal carcinogenesis could be suppressed through transgenic overexpression of TFF2, adenoviral transfer of TFF2 or transplantation of TFF2-expressing bone marrow. TFF2 is important to the anti-inflammatory reflex arc and plays an essential role in arresting MDSC proliferation. TFF2 offers a potential approach to prevent and to treat cancer.


Assuntos
Proliferação de Células/genética , Colite/genética , Neoplasias Colorretais/genética , Mucinas/genética , Proteínas Musculares/genética , Células Mieloides/imunologia , Peptídeos/genética , Receptores CXCR4/imunologia , Baço/imunologia , Linfócitos T Citotóxicos/imunologia , Nervo Vago , Transferência Adotiva , Animais , Western Blotting , Transplante de Medula Óssea , Colite/imunologia , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/imunologia , Citocinas/imunologia , Denervação , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imuno-Histoquímica , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/imunologia , Mucinas/metabolismo , Proteínas Musculares/imunologia , Proteínas Musculares/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Peptídeos/imunologia , Peptídeos/metabolismo , Permeabilidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/inervação , Subpopulações de Linfócitos T/imunologia , Fator Trefoil-2 , Vagotomia Troncular , Estimulação do Nervo Vago
17.
Sci Rep ; 6: 19311, 2016 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-26776680

RESUMO

Protein aggregation is a common biological phenomenon, observed in different physiological and pathological conditions. Decreased protein solubility and a tendency to aggregate is also observed during physiological aging but the causes are currently unknown. Herein we performed a biophysical separation of aging-related high molecular weight aggregates, isolated from the bone marrow and splenic cells of aging mice and followed by biochemical and mass spectrometric analysis. The analysis indicated that compared to younger mice an increase in protein post-translational carbonylation was observed. The causative role of these modifications in inducing protein misfolding and aggregation was determined by inducing carbonyl stress in young mice, which recapitulated the increased protein aggregation observed in old mice. Altogether our analysis indicates that oxidative stress-related post-translational modifications accumulate in the aging proteome and are responsible for increased protein aggregation and altered cell proteostasis.


Assuntos
Envelhecimento/metabolismo , Agregados Proteicos , Carbonilação Proteica , Proteínas/metabolismo , Envelhecimento/patologia , Animais , Células da Medula Óssea/metabolismo , Feminino , Camundongos , Estresse Oxidativo , Agregação Patológica de Proteínas , Espécies Reativas de Oxigênio/metabolismo , Baço/metabolismo
18.
Exp Cell Res ; 347(1): 1-13, 2016 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-26500109

RESUMO

The mechanisms by which macrophages control the inflammatory response, wound healing, biomaterial-interactions, and tissue regeneration appear to be related to their activation/differentiation states. Studies of macrophage behavior in vitro can be useful for elucidating their mechanisms of action, but it is not clear to what extent the source of macrophages affects their apparent behavior, potentially affecting interpretation of results. Although comparative studies of macrophage behavior with respect to cell source have been conducted, there has been no direct comparison of the three most commonly used cell sources: murine bone marrow, human monocytes from peripheral blood (PB), and the human leukemic monocytic cell line THP-1, across multiple macrophage phenotypes. In this study, we used multivariate discriminant analysis to compare the in vitro expression of genes commonly chosen to assess macrophage phenotype across all three sources of macrophages, as well as those derived from induced pluripotent stem cells (iPSCs), that were polarized towards four distinct phenotypes using the same differentiation protocols: M(LPS,IFN) (aka M1), M(IL4,IL13) (aka M2a), M(IL10) (aka M2c), and M(-) (aka M0) used as control. Several differences in gene expression trends were found among the sources of macrophages, especially between murine bone marrow-derived and human blood-derived M(LPS,IFN) and M(IL4,IL13) macrophages with respect to commonly used phenotype markers like CCR7 and genes associated with angiogenesis and tissue regeneration like FGF2 and MMP9. We found that the genes with the most similar patterns of expression among all sources were CXCL-10 and CXCL-11 for M(LPS,IFN) and CCL17 and CCL22 for M(IL4,IL13). Human PB-derived macrophages and human iPSC-derived macrophages showed similar gene expression patterns among the groups and genes studied here, suggesting that iPSC-derived monocytes have the potential to be used as a reliable cell source of human macrophages for in vitro studies. These findings could help select appropriate markers when testing macrophage behavior in vitro and highlight those markers that may confuse interpretation of results from experiments employing macrophages from different sources.


Assuntos
Polaridade Celular/genética , Perfilação da Expressão Gênica , Macrófagos/citologia , Macrófagos/metabolismo , Animais , Materiais Biocompatíveis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Polaridade Celular/efeitos dos fármacos , Análise Discriminante , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon gama/farmacologia , Análise dos Mínimos Quadrados , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Fenótipo
19.
Drug Discov Today ; 21(4): 540-3, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26646254

RESUMO

Ultrasound-targeted microbubble destruction (UTMD) is a promising technique with an immense target-specific gene delivery potential deep inside the human body. The potential of this technique has recently been confirmed for diabetic patients. This technology allows the genes to transfer specifically into the inefficient pancreas using ultrasound energy without viral vector utilization. It has been speculated that this idea and the advent of modern gene therapy techniques could result in significant future advances. Undoubtedly, this strategy needs further investigation and many critical questions have to be answered before it can be successfully advanced. Herein, we introduce the salient features of this approach, the hurdles that must be overcome, the hopes associated with it and practical constraints to develop this method for diabetes treatment.


Assuntos
Diabetes Mellitus/terapia , Técnicas de Transferência de Genes , Microbolhas , Ondas Ultrassônicas , Animais , Humanos , Pâncreas/metabolismo
20.
J Control Release ; 221: 48-61, 2016 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-26646543

RESUMO

A myriad of pathologies affect the gastrointestinal tract, citing this affected area as a significant target for therapeutic intervention. One group of therapeutic agents, antisense and oligonucleotides and small interfering RNAs, offer a promising platform for treating a wide variety of diseases ranging from cancer to auto-immune diseases. Current delivery methods are carried out either systemically or locally into diseased areas, both of which involve needles. The challenge in orally administering this type of treatment lies in the complications that arise due to the vast environmental extremes found within the gastrointestinal tract, owing to the fact that, as the drug travels down the gastrointestinal tract, it is subjected to pH changes and interactions with bacteria and a variety of digestive and protective enzymes including proteases, DNAses, and RNAses. Overcoming these challenges to allow the practical application of these drugs is a priority that has invoked a multitude of research in the chemical, biological, and material sciences. In this review, we will address common gastrointestinal pathologies, the barriers to oral-based therapies and antisense-interfering technologies, the approaches that have already been applied for their delivery, and the current status of antisense drug therapy clinical trials for gastrointestinal-related disorders.


Assuntos
Sistemas de Liberação de Medicamentos , Gastroenteropatias/terapia , Técnicas de Transferência de Genes , Oligonucleotídeos Antissenso/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Animais , Gastroenteropatias/patologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Humanos , Agulhas , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/uso terapêutico , RNA Interferente Pequeno/química , RNA Interferente Pequeno/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...