Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 453
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Braz. J. Pharm. Sci. (Online) ; 59: e21461, 2023. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1429963

RESUMO

Abstract he innate immune response plays an important role in the pathophysiology of acute respiratory distress syndrome (ARDS); however, no drug has been proven to be beneficial in the management of ARDS. Therefore, the aim of this study was to investigate the effects of using combined sedatives on systemic inflammatory responses in patients with ARDS. A total of 90 patients with ARDS and an intubation time of > 120 h were randomly divided into the propofol group (group P), midazolam group (group M), and combined sedation group (group U). Patients in groups P and M were sedated with propofol and midazolam, respectively, whereas patients in group U were sedated with a combination of propofol, midazolam, and dexmedetomidine. The dosage of sedatives and vasoactive drugs, duration of mechanical ventilation, and incidence of sedative adverse reactions were documented. The dosage of sedatives and vasoactive drugs, as well as the incidence of sedative adverse reactions in group U, was significantly lower than those in groups P and M. Similarly, the duration of mechanical ventilation in group U was significantly shorter than that in groups P and M. Hence, inducing sedation through a combination of multiple drugs can significantly reduce their adverse effects, improve their sedative effect, inhibit systemic inflammatory responses, and improve oxygenation in patients with ARDS


Assuntos
Humanos , Masculino , Feminino , Adulto , Pacientes/classificação , Síndrome do Desconforto Respiratório do Recém-Nascido/diagnóstico , Preparações Farmacêuticas/análise , Sedação Consciente/efeitos adversos , Midazolam/agonistas , Propofol/agonistas , Citocinas/administração & dosagem , Dexmedetomidina/agonistas
2.
Adv Drug Deliv Rev ; 179: 114035, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34740765

RESUMO

Glioblastoma multiform (GBM) is considered as the most lethal tumor among CNS malignancies. Although immunotherapy has achieved remarkable advances in cancer treatment, it has not shown satisfactory results in GBM patients. Biomaterial science, along with nanobiotechnology, is able to optimize the efficiency of immunotherapy in these patients. They can be employed to provide the specific activation of immune cells in tumor tissue and combinational therapy as well as preventing systemic adverse effects resulting from hyperactivation of immune responses and off-targeting effect. Advance biomaterials in this field are classified into targeting nanocarriers and localized delivery systems. This review will offer an overview of immunotherapy strategies for glioblastoma and advance delivery systems for immunotherapeutics that may have a high potential in glioblastoma treatment.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Materiais Biocompatíveis/química , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Sistemas de Liberação de Fármacos por Nanopartículas/química , Antineoplásicos Imunológicos/uso terapêutico , Neoplasias Encefálicas/patologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/farmacologia , Citocinas/administração & dosagem , Citocinas/farmacologia , Preparações de Ação Retardada , Liberação Controlada de Fármacos , Glioblastoma/patologia , Humanos , Inibidores de Checkpoint Imunológico/administração & dosagem , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia Adotiva/métodos , Microambiente Tumoral/fisiologia
3.
Oxid Med Cell Longev ; 2021: 4539453, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34621464

RESUMO

Mesenchymal stem cells (MSCs) are the most exploited stem cells with multilineage differentiation potential and immunomodulatory properties. Numerous lines of findings have reported their successful applications in a multitude of inflammatory conditions and immune disorders. However, it is currently discovered that these effects are mainly mediated in a paracrine manner by MSC-exosomes. Moreover, MSC-exosomes have been implicated in a wide variety of biological responses including immunomodulation, oxidative stress, tumor progression, and tissue regeneration. Meanwhile, they are reported to actively participate in various hematological diseases by the means of transferring different types of exosomal components to the target cells. Therefore, in this review, we briefly discuss the sources and biological features of MSCs and then illustrate the biogenesis and biological processes of MSC-exosomes. Of note, this paper especially highlights the latest research progress of MSC-exosomes in hematological diseases.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Citocinas/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Exossomos/imunologia , Exossomos/metabolismo , Doenças Hematológicas/tratamento farmacológico , Imunomodulação , Células-Tronco Mesenquimais/citologia , RNA/administração & dosagem , Animais , Doenças Hematológicas/imunologia , Humanos
4.
Radiat Res ; 196(6): 668-679, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34554263

RESUMO

Treatment of accidental radiation-induced myelosuppression is primarily based on supportive care and requires specific treatment based on hematopoietic growth factors injection or hematopoietic cell transplantation for the most severe cases. The cytokines used consisted of pegylated erythropoietin (darbepoetin alfa) 500 IU once per week, pegylated G-CSF (pegfilgrastim) 6 mg × 2 once, stem cell factor 20 µg.kg-1 for five days, and romiplostim (TPO analog) 10 µg.kg -1 once per week, with different combinations depending on the accidents. As the stem cell factor did not have regulatory approval for clinical use in France, the French regulatory authorities (ANSM, formerly, AFSSAPS) approved their compassionate use as an investigational drug "on a case-by-case basis". According to the evolution and clinical characteristics, each patient's treatment was adopted on an individual basis. Daily blood count allows initiating G-CSF and SCF delivery when granulocyte <1,000/mm3, TPO delivery when platelets <50,000/mm3, and EPO when Hb<80 g/L. The length of each treatment was based on blood cell recovery criteria. The concept of "stimulation strategy" is linked to each patient's residual hematopoiesis, which varies among them, depending on the radiation exposure's characteristics and heterogeneity. This paper reports the medical management of 8 overexposed patients to ionizing radiation. The recovery of bone marrow function after myelosuppression was accelerated using growth factors, optimized by multiple-line combinations. Particularly in the event of prolonged exposure to ionizing radiation in dose ranges inducing severe myelosuppression (in the order of 5 to 8 Gy), with no indication of hematopoietic stem cell transplantation.


Assuntos
Medula Óssea/efeitos da radiação , Citocinas/uso terapêutico , Liberação Nociva de Radioativos , Medula Óssea/metabolismo , Citocinas/administração & dosagem , Humanos , Irradiação Corporal Total
5.
Int J Mol Sci ; 22(13)2021 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-34201546

RESUMO

Tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) are two cytokines involved in the perpetuation of the chronic inflammation state characterizing rheumatoid arthritis (RA). Significant advances in the treatment of this pathology have been made over the past ten years, partially through the development of anti-TNF and anti-IL-1 therapies. However, major side effects still persist and new alternative therapies should be considered. The formulation of the micro-immunotherapy medicine (MIM) 2LARTH® uses ultra-low doses (ULD) of TNF-α, IL-1ß, and IL-2, in association with other immune factors, to gently restore the body's homeostasis. The first part of this review aims at delineating the pivotal roles played by IL-1ß and TNF-α in RA physiopathology, leading to the development of anti-TNF and anti-IL-1 therapeutic agents. In a second part, an emphasis will be made on explaining the rationale of using multiple therapeutic targets, including both IL-1ß and TNF-α in 2LARTH® medicine. Particular attention will be paid to the ULD of those two main pro-inflammatory factors in order to counteract their overexpression through the lens of their molecular implication in RA pathogenesis.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Citocinas/administração & dosagem , Imunoterapia/métodos , Interleucina-1beta/administração & dosagem , Fator de Necrose Tumoral alfa/administração & dosagem , Administração Oral , Animais , Artrite Reumatoide/fisiopatologia , Relação Dose-Resposta a Droga , Humanos , Interleucina-1beta/efeitos adversos , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/fisiologia , Interleucina-2/administração & dosagem , Interleucina-2/efeitos adversos , Terapia de Alvo Molecular/métodos , Medicina de Precisão , Fator de Necrose Tumoral alfa/efeitos adversos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia
6.
Immunopharmacol Immunotoxicol ; 43(5): 554-561, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34314307

RESUMO

BACKGROUND: Indoleamine 2,3-dioxygenase (IDO) inhibition has received much attention in cancer immunotherapy due to its role in immune escape in cancer cells. Additionally, changes in the pro-inflammatory cytokine levels can affect tumor growth and metastasis as well as the effectiveness of immunotherapy. The purpose of this study was for the first time to determine the effects of indoximod as an IDO inhibitor on triple-negative breast cancer (TNBC) and to assess the link between the efficacy of indoximod and IFN-γ or TNF-α stimulation. METHODS: The cytotoxic and apoptotic effects of indoximod alone or IFN-γ or TNF-α induction to mimic an inflammatory environment were evaluated by WST-1, Annexin V, cell cycle analysis, and acridine orange (AO)/ethidium bromide (EtBr) staining. Furthermore, the expression levels of IDO1 and PD-L1 expression were analyzed by RT-PCR. RESULTS: Our results demonstrated that indoximod significantly decreased the TNBC cell viability through apoptotic cell death (p < .05). The combination of indoximod and TNF-α was more effective than indoximod and IFN-γ stimulation or indoximod alone in TNBC cells. Additionally, PD-L1 expression level was significantly up-regulated after treatment with indoximod and TNF-α or IFN-γ combinations (p < .05). CONCLUSIONS: Indoximod exhibited a therapeutic potential in TNBC cells and pro-inflammatory cytokines could affect the effectiveness of indoximod. However, further studies are required to identify the role of the IDO-associated signaling pathways, the molecular mechanisms of indoximod induced apoptotic cell death, and the relationship between IDO inhibition by IDO inhibitors and pro-inflammatory cytokine levels.


Assuntos
Citocinas/administração & dosagem , Mediadores da Inflamação/administração & dosagem , Neoplasias de Mama Triplo Negativas/metabolismo , Triptofano/análogos & derivados , Antígeno B7-H1/biossíntese , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Humanos , Interferon gama/administração & dosagem , Resultado do Tratamento , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Triptofano/administração & dosagem , Fator de Necrose Tumoral alfa/administração & dosagem
7.
Int J Mol Sci ; 22(11)2021 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-34071037

RESUMO

Knee osteoarthritis (KOA) represents a clinical challenge due to poor potential for spontaneous healing of cartilage lesions. Several treatment options are available for KOA, including oral nonsteroidal anti-inflammatory drugs, physical therapy, braces, activity modification, and finally operative treatment. Intra-articular (IA) injections are usually used when the non-operative treatment is not effective, and when the surgery is not yet indicated. More and more studies suggesting that IA injections are as or even more efficient and safe than NSAIDs. Recently, research to improve intra-articular homeostasis has focused on biologic adjuncts, such as platelet-rich plasma (PRP). The catabolic and inflammatory intra-articular processes that exists in knee osteoarthritis (KOA) may be influenced by the administration of PRP and its derivatives. PRP can induce a regenerative response and lead to the improvement of metabolic functions of damaged structures. However, the positive effect on chondrogenesis and proliferation of mesenchymal stem cells (MSC) is still highly controversial. Recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, significant progress has been made in understanding the mechanism of PRP action. In this review, we will discuss mechanisms related to inflammation and chondrogenesis in cartilage repair and regenerative processes after PRP administration in in vitro and animal studies. Furthermore, we review clinical trials of PRP efficiency in changing the OA biomarkers in knee joint.


Assuntos
Plasma Rico em Plaquetas , Animais , Células Cultivadas , Microambiente Celular , Condrócitos/efeitos dos fármacos , Condrogênese , Citocinas/administração & dosagem , Citocinas/uso terapêutico , Grânulos Citoplasmáticos/química , Cobaias , Humanos , Ácido Hialurônico/farmacologia , Ácido Hialurônico/uso terapêutico , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/uso terapêutico , Injeções Intra-Articulares , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Neurotransmissores/administração & dosagem , Neurotransmissores/uso terapêutico , Osteoartrite do Joelho , Plasma Rico em Plaquetas/química , Resultado do Tratamento
8.
Nat Rev Clin Oncol ; 18(9): 558-576, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34006998

RESUMO

Immune-checkpoint inhibitors and chimeric antigen receptor (CAR) T cells are revolutionizing oncology and haematology practice. With these and other immunotherapies, however, systemic biodistribution raises safety issues, potentially requiring the use of suboptimal doses or even precluding their clinical development. Delivering or attracting immune cells or immunomodulatory factors directly to the tumour and/or draining lymph nodes might overcome these problems. Hence, intratumoural delivery and tumour tissue-targeted compounds are attractive options to increase the in situ bioavailability and, thus, the efficacy of immunotherapies. In mouse models, intratumoural administration of immunostimulatory monoclonal antibodies, pattern recognition receptor agonists, genetically engineered viruses, bacteria, cytokines or immune cells can exert powerful effects not only against the injected tumours but also often against uninjected lesions (abscopal or anenestic effects). Alternatively, or additionally, biotechnology strategies are being used to achieve higher functional concentrations of immune mediators in tumour tissues, either by targeting locally overexpressed moieties or engineering 'unmaskable' agents to be activated by elements enriched within tumour tissues. Clinical trials evaluating these strategies are ongoing, but their development faces issues relating to the administration methodology, pharmacokinetic parameters, pharmacodynamic end points, and immunobiological and clinical response assessments. Herein, we discuss these approaches in the context of their historical development and describe the current landscape of intratumoural or tumour tissue-targeted immunotherapies.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Animais , Anticorpos Monoclonais/administração & dosagem , Citocinas/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Humanos , Fatores Imunológicos/administração & dosagem , Imunoterapia/tendências , Injeções Intralesionais , Camundongos , Terapia de Alvo Molecular/métodos , Neoplasias/patologia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Distribuição Tecidual , Microambiente Tumoral/imunologia
9.
Reprod Sci ; 28(6): 1659-1670, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33886116

RESUMO

Endometrial receptivity and thickness play an important role in achieving a pregnancy. Intrauterine autologous platelet-rich plasma (PRP) infusion has been used in infertile women with recurrent implantation failure (RIF) and thin endometrial lining thickness (EMT). Literature search was performed in PubMed for studies including in vitro, animal, and human studies as well as in abstracts presented at national conferences. Animal studies demonstrated a decrease in the expression of inflammatory markers and fibrosis, and increased endometrial proliferation rate, increased expression of proliferative genes, and increased pregnancy rates. The in vitro studies showed that PRP was associated with increased stromal and mesenchymal cell proliferation, increased expression of regenerative enzymes, and enhancement in cell migration. In infertile women undergoing assisted reproductive technology, one randomized clinical trial showed that PRP intrauterine infusion improved EMT, implantation rate, and clinical pregnancy rate (CPR) in patients with thin EMT, while 3 other trials involving subjects with RIF showed conflicting results related to CPR. Case series and cohort studies showed conflicting results pertaining to CPR. Data to date suggest that PRP may be beneficial in improving endometrial thickness and endometrial receptivity. However, further large prospective and high-quality trials are needed to assert its effect and to identify the population of patients that would benefit the most.


Assuntos
Endométrio/fisiologia , Plasma Rico em Plaquetas , Útero/fisiologia , Adulto , Animais , Quimiocinas/administração & dosagem , Citocinas/administração & dosagem , Implantação do Embrião , Transferência Embrionária , Endométrio/anatomia & histologia , Endométrio/efeitos dos fármacos , Feminino , Ginatresia/complicações , Humanos , Infertilidade Feminina/etiologia , Infertilidade Feminina/terapia , Injeções , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Plasma Rico em Plaquetas/química , Plasma Rico em Plaquetas/fisiologia , Gravidez , Técnicas de Reprodução Assistida , Útero/efeitos dos fármacos
10.
Biochem Biophys Res Commun ; 558: 22-28, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33894674

RESUMO

Autocrine motility factor (AMF) stimulates the motility of cancer cells via an autocrine route and has been implicated in tumor progression and metastasis. Overexpression of AMF is correlated with the aggressive nature of breast cancer and is negatively associated with clinical outcomes. In contrast, AMF also has the ability to suppress cancer cells. In this study, AMFs from different cancer cells were demonstrated to have suppressive activity against MCF-7 and MDA-MB-231 breast cancer cells. In a growth and colony formation assay, AMF from AsPC-1 pancreatic cancer cells (ASPC-1:AMF) was determined to be more suppressive compared to other AMFs. It was also demonstrated that AsPC-1:AMF could arrest breast cancer cells at the G0/G1 cell cycle phase. Quantified by Western blot analysis, AsPC-1:AMF lowered levels of the AMF receptor (AMFR) and G-protein-coupled estrogen receptor (GPER), concomitantly regulating the activation of the AKT and ERK signaling pathways. JAK/STAT activation was also decreased. These results were found in estrogen receptor (ER)-positive MCF-7 cells but not in triple-negative MDA-MB-231 cells, suggesting that AsPC-1:AMF could work through multiple pathways led to apoptosis. More importantly, AsPC-1:AMF and methyl jasmonate (MJ) cooperatively and synergistically acted against breast cancer cells. Thus, AMF alone or along with MJ may be a promising breast cancer treatment option.


Assuntos
Acetatos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Ciclopentanos/administração & dosagem , Glucose-6-Fosfato Isomerase/administração & dosagem , Oxilipinas/administração & dosagem , Antineoplásicos Fitogênicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Clonagem Molecular , Citocinas/administração & dosagem , Citocinas/genética , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Glucose-6-Fosfato Isomerase/genética , Humanos , Células MCF-7 , Terapia de Alvo Molecular , Receptores do Fator Autócrino de Motilidade/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Transdução de Sinais/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
11.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32824966

RESUMO

Myocardial infarction causes cardiac tissue damage and the release of damage-associated molecular patterns leads to activation of the immune system, production of inflammatory mediators, and migration of various cells to the site of infarction. This complex response further aggravates tissue damage by generating oxidative stress, but it eventually heals the infarction site with the formation of fibrotic tissue and left ventricle remodeling. However, the limited self-renewal capability of cardiomyocytes cannot support sufficient cardiac tissue regeneration after extensive myocardial injury, thus, leading to an irreversible decline in heart function. Approaches to improve cardiac tissue regeneration include transplantation of stem cells and delivery of inflammation modulatory and wound healing factors. Nevertheless, the harsh environment at the site of infarction, which consists of, but is not limited to, oxidative stress, hypoxia, and deficiency of nutrients, is detrimental to stem cell survival and the bioactivity of the delivered factors. The use of biomaterials represents a unique and innovative approach for protecting the loaded factors from degradation, decreasing side effects by reducing the used dosage, and increasing the retention and survival rate of the loaded cells. Biomaterials with loaded stem cells and immunomodulating and tissue-regenerating factors can be used to ameliorate inflammation, improve angiogenesis, reduce fibrosis, and generate functional cardiac tissue. In this review, we discuss recent findings in the utilization of biomaterials to enhance cytokine/growth factor and stem cell therapy for cardiac tissue regeneration in small animals with myocardial infarction.


Assuntos
Sistemas de Liberação de Medicamentos , Cardiopatias/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Medicina Regenerativa/métodos , Animais , Citocinas/administração & dosagem , Citocinas/uso terapêutico , Cardiopatias/tratamento farmacológico , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Nanopartículas/química
12.
Nat Commun ; 11(1): 3546, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32669559

RESUMO

Advanced ovarian cancer usually spreads to the omentum. However, the omental cell-derived molecular determinants modulating its progression have not been thoroughly characterized. Here, we show that circulating ITLN1 has prognostic significance in patients with advanced ovarian cancer. Further studies demonstrate that ITLN1 suppresses lactotransferrin's effect on ovarian cancer cell invasion potential and proliferation by decreasing MMP1 expression and inducing a metabolic shift in metastatic ovarian cancer cells. Additionally, ovarian cancer-bearing mice treated with ITLN1 demonstrate marked decrease in tumor growth rates. These data suggest that downregulation of mesothelial cell-derived ITLN1 in the omental tumor microenvironment facilitates ovarian cancer progression.


Assuntos
Carcinoma Epitelial do Ovário/secundário , Citocinas/metabolismo , Lectinas/metabolismo , Omento/patologia , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/secundário , Animais , Carcinoma Epitelial do Ovário/sangue , Carcinoma Epitelial do Ovário/mortalidade , Carcinoma Epitelial do Ovário/terapia , Linhagem Celular Tumoral/transplante , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Citocinas/administração & dosagem , Citocinas/sangue , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Proteínas Ligadas por GPI/administração & dosagem , Proteínas Ligadas por GPI/sangue , Proteínas Ligadas por GPI/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Lactoferrina/metabolismo , Lectinas/administração & dosagem , Lectinas/sangue , Metaloproteinase 1 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/terapia , Ovário , Proteínas Recombinantes/administração & dosagem , Taxa de Sobrevida , Microambiente Tumoral
13.
Science ; 369(6504): 712-717, 2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32527928

RESUMO

Excessive cytokine signaling frequently exacerbates lung tissue damage during respiratory viral infection. Type I (IFN-α and IFN-ß) and III (IFN-λ) interferons are host-produced antiviral cytokines. Prolonged IFN-α and IFN-ß responses can lead to harmful proinflammatory effects, whereas IFN-λ mainly signals in epithelia, thereby inducing localized antiviral immunity. In this work, we show that IFN signaling interferes with lung repair during influenza recovery in mice, with IFN-λ driving these effects most potently. IFN-induced protein p53 directly reduces epithelial proliferation and differentiation, which increases disease severity and susceptibility to bacterial superinfections. Thus, excessive or prolonged IFN production aggravates viral infection by impairing lung epithelial regeneration. Timing and duration are therefore critical parameters of endogenous IFN action and should be considered carefully for IFN therapeutic strategies against viral infections such as influenza and coronavirus disease 2019 (COVID-19).


Assuntos
Células Epiteliais Alveolares/patologia , Citocinas/metabolismo , Interferon Tipo I/metabolismo , Interferons/metabolismo , Pulmão/patologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Células Epiteliais Alveolares/imunologia , Animais , Apoptose , Líquido da Lavagem Broncoalveolar/imunologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Citocinas/administração & dosagem , Citocinas/imunologia , Feminino , Vírus da Influenza A Subtipo H3N2 , Interferon Tipo I/administração & dosagem , Interferon Tipo I/farmacologia , Interferon-alfa/administração & dosagem , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Interferon beta/administração & dosagem , Interferon beta/metabolismo , Interferon beta/farmacologia , Interferons/administração & dosagem , Interferons/farmacologia , Masculino , Camundongos , Infecções por Orthomyxoviridae/metabolismo , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Interferon lambda
14.
Biomater Sci ; 8(6): 1734-1747, 2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-31998886

RESUMO

Insufficient endothelialization of cardiovascular devices is a high-risk factor for implant failure. Presentation of extracellular matrix (ECM)-derived coatings is a well-known strategy to improve implant integration. However, the complexity of the system is challenging and strategies for applying multifunctionality are required. Here, we engineered mussel-derived surface-binding peptides equipped with integrin (c[RGDfK]) and proteoglycan binding sites (FHRRIKA) for enhanced endothelialization. Surface-binding properties of the platform containing l-3,4-dihydroxyphenylalanine (DOPA) residues were confirmed for hydrophilized polycaprolactone-co-lactide scaffolds as well as for glass and polystyrene. Further, heparin and the heparin-binding angiogenic factors VEGF, FGF-2 and CXCL12 were immobilized onto the peptide in a modular assembly. Presentation of bioactive peptides greatly enhanced human umbilical vein endothelial cell (HUVEC) adhesion and survival under static and fluidic conditions. In subsequent investigations, peptide-heparin-complexes loaded with CXCL12 or VEGF had an additional increasing effect on cell viability, differentiation and migration. Finally, hemocompatibility of the coatings was ensured. This study demonstrates that coatings combining adhesion peptides, glycosaminoglycans and modulators are a versatile tool to convey ECM-inspired multifunctionality to biomaterials and efficiently promote their integration.


Assuntos
Citocinas/administração & dosagem , Células Endoteliais da Veia Umbilical Humana/citologia , Levodopa/química , Peptídeos/administração & dosagem , Implantes Absorvíveis , Animais , Prótese Vascular , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Citocinas/farmacologia , Matriz Extracelular , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Integrinas/química , Camundongos , Peptídeos/química , Peptídeos/farmacologia , Proteoglicanas/química , Propriedades de Superfície
15.
Biochem Biophys Res Commun ; 523(4): 829-834, 2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-31954515

RESUMO

The cytokine-like protein FAM19A5 is highly expressed in the brain, but little is known about its functions there. Here, we found that FAM19A5 was expressed in mouse hypothalamic cells expressing proopiomelanocortin (POMC) and neuropeptide Y (NPY)/agouti-related peptide (AgRP), and in the microglia. Tumor necrosis factor-α (TNF-α), which induces inflammatory sickness responses, greatly increased hypothalamic expression of FAM19A5. Knockdown of FAM19A5 expression resulted in decreased TNF-α-induced anorexia, body weight loss and TNF-α-induced expression of inflammatory factors. In contrast, intracerebroventricular administration of FAM19A5 induced anorexia, body weight loss and hyperthermia, together with increased expression of inflammatory factors. FAM19A5 injection also induced increases in c-fos activation and POMC mRNA level in hypothalamic POMC neurons. Together, these results suggest that FAM19A5 plays an important role in hypothalamic inflammatory responses.


Assuntos
Citocinas/metabolismo , Hipotálamo/metabolismo , Hipotálamo/patologia , Inflamação/metabolismo , Animais , Citocinas/administração & dosagem , Citocinas/farmacologia , Humanos , Hipotálamo/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Especificidade de Órgãos/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fator de Necrose Tumoral alfa/administração & dosagem
16.
Front Immunol ; 11: 615240, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33679703

RESUMO

Although the discovery and characterization of multiple tumor antigens have sparked the development of many antigen/derived cancer vaccines, many are poorly immunogenic and thus, lack clinical efficacy. Adjuvants are therefore incorporated into vaccine formulations to trigger strong and long-lasting immune responses. Adjuvants have generally been classified into two categories: those that 'depot' antigens (e.g. mineral salts such as aluminum hydroxide, emulsions, liposomes) and those that act as immunostimulants (Toll Like Receptor agonists, saponins, cytokines). In addition, several novel technologies using vector-based delivery of antigens have been used. Unfortunately, the immune system declines with age, a phenomenon known as immunosenescence, and this is characterized by functional changes in both innate and adaptive cellular immunity systems as well as in lymph node architecture. While many of the immune functions decline over time, others paradoxically increase. Indeed, aging is known to be associated with a low level of chronic inflammation-inflamm-aging. Given that the median age of cancer diagnosis is 66 years and that immunotherapeutic interventions such as cancer vaccines are currently given in combination with or after other forms of treatments which themselves have immune-modulating potential such as surgery, chemotherapy and radiotherapy, the choice of adjuvants requires careful consideration in order to achieve the maximum immune response in a compromised environment. In addition, more clinical trials need to be performed to carefully assess how less conventional form of immune adjuvants, such as exercise, diet and psychological care which have all be shown to influence immune responses can be incorporated to improve the efficacy of cancer vaccines. In this review, adjuvants will be discussed with respect to the above-mentioned important elements.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/uso terapêutico , Imunoterapia Ativa/métodos , Neoplasias/terapia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/classificação , Fatores Etários , Compostos de Alúmen/administração & dosagem , Antineoplásicos/uso terapêutico , Ensaios Clínicos Fase III como Assunto/métodos , Terapia Combinada , Citocinas/administração & dosagem , Citocinas/imunologia , Sinergismo Farmacológico , Emulsões , Microbioma Gastrointestinal/imunologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia/métodos , Estilo de Vida , Lipossomos/administração & dosagem , Depleção Linfocítica , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/imunologia , Nanopartículas/administração & dosagem , Radioterapia , Saponinas/administração & dosagem , Saponinas/imunologia , Receptores Toll-Like/agonistas , Receptores Toll-Like/imunologia , Potência de Vacina , Virossomos/administração & dosagem
17.
Fertil Steril ; 112(5): 849-857.e1, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31551154

RESUMO

OBJECTIVE: To evaluate the effects of cytokine enrichment of culture medium on embryological and clinical outcomes after intracytoplasmic sperm injection (ICSI). DESIGN: A randomized clinical trial. SETTING: In vitro fertilization centers. PATIENT(S): This trial included 443 ICSI cycles randomized into two groups. INTERVENTION(S): This study evaluated the influence of integration of granulocyte-macrophage colony-stimulating factor, heparin-binding epidermal growth factor-like growth factor, and leukemia inhibitory factor into culture media on human embryo development after ICSI. MAIN OUTCOME MEASURE(S): Ongoing pregnancy rate per a randomized participant. RESULT(S): Cytokine enrichment of culture medium showed improvement in ongoing pregnancy rate compared with no cytokines (106/224 [47%] vs. 78/219 [36%]; absolute rate difference [ARD] = 12; 95% confidence interval [CI], 2.5-21). This integration of cytokines also showed better rates of live birth (101/224 [45%] vs. 71/219 [33%]; ARD = 13; 95% CI, 4-21) and cumulative live birth (132/224 [60%] vs. 97/219 [44%]; ARD = 12; 95% CI, 4-20) and lower rate of pregnancy loss (27/124 [22%] vs. 37/103 [36%]; ARD = -14; 95% CI, -26 to -2) than conventional medium. Embryos developed in the cytokine-supplemented medium showed better blastocyst formation, quality, cryopreservation, and use than control medium. CONCLUSION(S): Integration of cytokines into human embryo culture media showed improvement in embryological and clinical outcomes after ICSI. However, the long-term effect of cytokine enrichment of a medium is still unclear and warrants further studies with longitudinal follow-up. CLINICAL TRIAL REGISTRATION NUMBER: NCT02420886 at ClinicalTrials.gov.


Assuntos
Citocinas/administração & dosagem , Técnicas de Cultura Embrionária/métodos , Transferência Embrionária/métodos , Embrião de Mamíferos/efeitos dos fármacos , Injeções de Esperma Intracitoplásmicas , Adulto , Meios de Cultura/farmacologia , Técnicas de Cultura Embrionária/tendências , Embrião de Mamíferos/fisiologia , Feminino , Humanos , Gravidez , Taxa de Gravidez/tendências , Injeções de Esperma Intracitoplásmicas/tendências
18.
J Plast Surg Hand Surg ; 53(6): 347-355, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31244355

RESUMO

Background: Viability decreases at the distal parts with an increase in the length of flaps. In this study, we evaluated the effects of subcutaneously administered omentin on flap viability, where it is applied to distal one-third part of McFarlane flaps elevated from the rat's dorsal skin.Materials and methods: Twenty-four adult, female, Sprague-Dawley rats were used. Subjects were divided into three groups; group 1 is the control group, group 2 received omentin 1 week before flap elevation, and group 3 received omentin 2 d before and at the day of flap elevation. About 1 cc (300 nanogram/cc) omentin applied by subcutaneous injections to the distal one-third flap. Photos are taken daily for macroscopic evaluations. The 3-mm full thickness punch biopsies at the third day and 1-cm2 biopsies at the seventh day from the middle of the one-third distal third of the flaps were taken. Necrotic and viable areas were measured. Neutrophil counting, epidermis thickness, inflammation, edema, and vascular endothelial growth factor (VEGF) immune staining were evaluated using histopathological analyses. Endothelial Nitric Oxide Synthase (eNOS) expression was performed by ELISA.Results: Omentin increased the percentage of the viable areas of flaps, epidermal thickness, number of newly formed blood vessels, and eNOS expression levels. The results showed statistical significance.Conclusions: Omentin human increases the viable areas of flaps and may be used for enhancement of flap survival.


Assuntos
Citocinas/administração & dosagem , Sobrevivência de Enxerto , Lectinas/administração & dosagem , Retalhos Cirúrgicos , Animais , Contagem de Células , Endotélio/metabolismo , Epiderme/patologia , Injeções Subcutâneas , Modelos Animais , Neovascularização Fisiológica , Neutrófilos/metabolismo , Óxido Nítrico Sintase/metabolismo , Ratos Sprague-Dawley , Retalhos Cirúrgicos/irrigação sanguínea
19.
Sci Transl Med ; 11(498)2019 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-31243150

RESUMO

The clinical application of cytokine therapies for cancer treatment remains limited due to severe adverse reactions and insufficient therapeutic effects. Although cytokine localization by intratumoral administration could address both issues, the rapid escape of soluble cytokines from the tumor invariably subverts this effort. We find that intratumoral administration of a cytokine fused to the collagen-binding protein lumican prolongs local retention and markedly reduces systemic exposure. Combining local administration of lumican-cytokine fusions with systemic immunotherapies (tumor-targeting antibody, checkpoint blockade, cancer vaccine, or T cell therapy) improves efficacy without exacerbating toxicity in syngeneic tumor models and the BrafV600E /Ptenfl/fl genetically engineered melanoma model. Curative abscopal effects on noncytokine-injected tumors were also observed as a result of a protective and systemic CD8+ T cell response primed by local therapy. Cytokine collagen-anchoring constitutes a facile, tumor-agnostic strategy to safely potentiate otherwise marginally effective systemic immunotherapies.


Assuntos
Citocinas/administração & dosagem , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Anticorpos Antineoplásicos/imunologia , Linhagem Celular Tumoral , Colágeno , Modelos Animais de Doenças , Interleucina-12/uso terapêutico , Interleucina-2/uso terapêutico , Lumicana/metabolismo , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Terapia Neoadjuvante , PTEN Fosfo-Hidrolase/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Albumina Sérica/metabolismo , Linfócitos T/imunologia , Redução de Peso
20.
J Immunother ; 42(5): 143-161, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31090655

RESUMO

Strategies to stabilize remissions by specific elimination of residual acute myeloid leukemia (AML) blasts are needed. Leukemia-derived dendritic cell (DCleu/DC) generated from myeloid blasts improve antileukemic T-cell reactivity and install T-cell memory. Interferon (IFN)α-DC methods produce DCleu from chronic myeloid leukemia-patients (pts') blood. Various INFα-containing versus other DC methods were studied to produce DCleu (evaluated by flowcytometry) from AML-pts' blast-containing mononuclear (MNC) or whole blood (WB). After DCleu/DC stimulation in mixed lymphocyte cultures, T cells' potential to gain antileukemic cytotoxicity was studied and correlated with different DC methods and DCleu/DC counts. (1) Generation of DCleu/DC: (a) "IFN-GIT" [containing granulocyte macrophage-colony stimulating factor (GM-CSF)+IFNα+ tumor necrosis factor (TNF)-α] produced DC successfully (≥10% DC, ≥5% DCleu/cells) from AML-MNC (WB) in 54 (56%), "MCM-Mimic" in 76 (75%), "Picibanil" in 83 (64%), and "Calcium-ionophore" in 42 (67%) of cases. Proportions of DC subtypes in MNC (WB) were comparable with all DC methods, (b) IFNα combinations containing only GM-CSF+IFNα or only IFNα showed low efficiency to produce DCleu/DC from MNC (WB) compared with "IFN-GIT." (2) Antileukemic functionality: DCleu/DC-stimulated T cells showed improved leukemia cytotoxicity compared with blast cells or unstimulated T cells. The highest blast proliferation (=insufficient T cells) was seen with "IFN-GIT" DC-stimulated T cells. Probability to respond to immunotherapy or to obtain blast lysis of DC-stimulated T cells correlated with high proportions of DCleu/DC after DC culture, independent of DC-generating methods. (3) Cytokine release profiles: levels of interleukin-6, IFN-γ, and interleukin-2 were significantly lower in DC culture supernatants (from MNC/WB) with "IFN-GIT" compared with "MCM," "Pici," and "Ca" DC supernatants. Our data show that (1) WB culture simulates AML-pts' in vivo situation, (2) DC generation is possible from AML-MNC (WB) with IFNα-containing and other DC methods, (3) successful IFNα-DC generation needs GM-CSF+IFNα+TNF-α (IFN-GIT); however, "IFN-GIT" produces less DCleu/DC compared with other (non-IFNα) DC methods, (4) T cells stimulated with "IFN-GIT"-produced DCleu/DC yielded comparable antileukemic cytotoxicity; however, in cases without achieved blast lysis, an increased blast proliferation was observed.


Assuntos
Citocinas/administração & dosagem , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Imunoterapia , Interferon-alfa/administração & dosagem , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Biomarcadores , Células Dendríticas/metabolismo , Feminino , Humanos , Imunofenotipagem , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/metabolismo , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade , Linfócitos T/imunologia , Linfócitos T/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA