Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomed Pharmacother ; 109: 945-956, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30551549

RESUMO

Salvianic acid A (SAA) is an active water-soluble constituent derived from Salvia miltiorrhiza Bge that is used extensively in the treatment of angiocardiopathy in China. However, few reports have investigated the therapeutic effect and the underlying mechanisms of SAA on atherosclerosis (AS). This study examines the protective mechanisms of SAA on AS in vivo and in vitro. SAA treatment (3 and 10 mg/kg/d) prevented the progression of atherosclerotic lesions and decreased 58.2% and 72.8% of the lipid deposition in the aorta of high fat-diet-induced AS rat. Notably, SAA treatment ameliorated serum lipid abnormalities by decreasing 20.4% and 33.8% of triglyceride, 26.1% and 32.7% of total cholesterol, 36.0% and 57.3% of low-density lipoprotein-cholesterol levels and increasing 183.4% and 337.5% of high-density lipoprotein-cholesterol level in the serum of AS rat (all P < 0.05). SAA treatment lowered pro-inflammatory mediators including interleukin-1ß, interleukin-6, tumor necrosis factor-α, intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) (all P < 0.05) by inhibiting the toll-like receptor 4/nuclear factor kappa B pathway. In addition, SAA treatment significantly decreased oxidative stress by increasing antioxidant enzymes activity, upregulating nuclear factor erythroid 2-related factor 2/heme oxygenase-1 pathway and downregulated expression of p47phox and p22phox (all P < 0.05) in vivo. Furthermore, SAA (10-5 and 3 × 10-5 M) suppressed oxidized low-density lipoprotein-induced expression of lectin-like oxidized low-density lipoprotein receptor-1, the phosphorylation of nuclear factor kappa B (p65), ICAM-1 and VCAM-1 (all P < 0.05) and inhibited NADPH oxidase subunit 4-mediated reactive oxygen species generation in human umbilical vein endothelial cells. The experimental data verify the protective role of SAA in AS and the underlying mechanisms are strongly associated with the inhibition of oxidative stress, inflammation, and amelioration of endothelial dysfunction.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Medicamentos de Ervas Chinesas/uso terapêutico , Mediadores da Inflamação/metabolismo , Lactatos/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Animais , Aterosclerose/patologia , Medicamentos de Ervas Chinesas/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Lactatos/farmacologia , Masculino , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo
2.
PLoS One ; 13(10): e0203602, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30273351

RESUMO

In order to determine the role of the adrenergic system in bupivacaine-induced cardiotoxicity, a series of experiments were performed. In an animal experiment, male Sprague-Dawley (SD) rats under chloral hydrate anesthesia received intravenous bupivacaine, followed by an intravenous injection of adrenalin or isoprenalin, and the electrocardiogram (ECG), left ventricular systolic pressure (LVSP), left ventricular end-diastolic pressure (LVEDP), the maximum rate of rise of left ventricular pressure (+dP/dtmax) and the maximum rate of pressure decrease (-dP/dtmax) were continually monitored. In a cellular experiment, freshly isolated adult SD rat ventricular myocytes were perfused with bupivacaine at different concentrations in the presence or absence of isoprenalin, with or without esmolol. The percentage of the sarcomere shortening (bl% peak h), departure velocity (dep v) of sarcomere shortening and time to 50% of the peak speed of myocyte contraction (Tp50) was assessed by a video-based edge-detection system. In an additional experiment, Swiss mice pretreated with saline, isoprenalin, esmolol or dexmedetomidine received bupivacaine to determine the 50% lethal dose (LD50) of bupivacaine. Electron microscopy of myocardial mitochondria was performed to assess damage of these structures. To test mitochondrial reactive oxygen species (ROS) production, freshly isolated SD rat ventricular myocytes were incubated with bupivacaine in the presence of isoprenalin, with or without esmolol. First, our results showed that bupivacaine significantly reduced the LVSP and +dP/dtmax, as well as enhanced the LVEDP and -dP/dtmax (P < 0.05, vs. control, and vs. baseline). Adrenalin and isoprenalin induced a further reduction of LVSP and +dP/dtmax (P < 0.05, vs. before adrenalin or isoprenalin delivery, and vs. control). Second, bupivacaine induced a dose-dependent cardiomyocyte contractile depression. While 5.9 µmol/L or 8.9 µmol/L of bupivacaine resulted in no change, 30.0 µmol/L of bupivacaine prolonged the Tp50 and reduced the bl% peak h and dep v (P < 0.05, vs. control and vs. baseline). Isoprenalin aggravated the bupivacaine-induced cardiomyocyte contractile depression, significantly prolonging the Tp50 (P < 0.05, vs. bupivacaine alone) and reducing the dep v (P < 0.05, vs. bupivacaine alone). Third, esmolol and dexmedetomidine significantly enhanced, while isoprenalin significantly reduced, the LD50 of bupivacaine in mice. Fourth, bupivacaine led to significant mitochondrial swelling, and the extent of myocardial mitochondrial swelling in isoprenalin-pretreated mice was significantly higher than that compared with mice pretreated with saline, as reflected by the higher mitochondrial damage score (P < 0.01). Meanwhile, esmolol pretreatment significantly reduced the mitochondrial damage score (P < 0.01). Fifth, bupivacaine significantly increased the ROS in freshly isolated cardiomyocytes, and added isoprenalin induced a further enhancement of ROS production (P < 0.05, vs. bupivacaine alone). Added esmolol significantly decreased ROS production (P < 0.05, vs. bupivacaine + isoprenalin). Our results suggest that bupivacaine depressed cardiac automaticity, conductivity and contractility, but the predominant effect was contractile dysfunction which resulted from the disruption of mitochondrial energy metabolism. ß-adrenergic activation aggravated the cellular metabolism disorder and therefore contractile dysfunction.


Assuntos
Cardiotoxicidade/fisiopatologia , Epinefrina/administração & dosagem , Isoproterenol/administração & dosagem , Contração Miocárdica/efeitos dos fármacos , Anestesia/efeitos adversos , Animais , Pressão Sanguínea/efeitos dos fármacos , Bupivacaína/administração & dosagem , Bupivacaína/efeitos adversos , Cardiotoxicidade/diagnóstico por imagem , Cardiotoxicidade/etiologia , Hidrato de Cloral/administração & dosagem , Modelos Animais de Doenças , Eletrocardiografia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Humanos , Camundongos , Contração Miocárdica/fisiologia , Propanolaminas/administração & dosagem , Ratos , Espécies Reativas de Oxigênio/metabolismo
3.
J Perianesth Nurs ; 31(2): 154-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27037169

RESUMO

PURPOSE: The purpose of this study was to explore the effect of NaHCO3-buffered lidocaine gel as a topical anesthetic agent for pain relief for rigid cystoscopy. DESIGN: Prospective randomized controlled trial. METHODS: ASA I-II male patients undergoing rigid cystoscopy randomly received 10 mL 2% Carbocaine lidocaine gel with 1 mL 0.9% saline (group 1) or 1 mL 5% NaHCO3 solution (group 2). After 3 minutes exposure, the cystoscope was inserted into the urethra. On receiving the gel, cystoscope insertion, and intravesical observation, pain score was recorded using the visual analog scale. FINDINGS: The gel pH with or without NaHCO3 was 7.20 and 6.41, respectively. The concentration of soluble lidocaine in the gel was stable for 24 hours or more. The visual analog scale score in group 2 was significantly lower (1.3 ± 0.9) than in group 1 (5.28 ± 1.99). No adverse effects were recorded. CONCLUSION: Alkalized lidocaine gel resulted in successful analgesia for rigid cystoscopy in men without adverse effects.


Assuntos
Cistoscopia/métodos , Lidocaína/administração & dosagem , Bicarbonato de Sódio/química , Soluções Tampão , Cistoscopia/efeitos adversos , Humanos , Concentração de Íons de Hidrogênio , Lidocaína/química , Masculino , Estudos Prospectivos
4.
J Anesth ; 30(1): 109-15, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26302692

RESUMO

PURPOSE: KCNQ2/3 channels play an important role in controlling neuronal excitability. Agents that decrease KCNQ2/3 current amplitudes are proconvulsant, whereas KCNQ2/3 current enhancers are anticonvulsant. Levobupivacaine is able to block the KCNQ2/3 channels and enhance neuronal excitation, whereas retigabine is able to reopen the channels and thus reduce overexcitation of neurons. In this study, we aimed to determine if retigabine is able to abolish local-anesthetic-induced seizures. METHODS: Twenty New Zealand rabbits were randomly divided into two groups of ten. Levobupivacaine (0.5 %) was infused into conscious rabbits via the marginal ear vein at 8 ml/kg/h until the rabbits seized, and 5 mg/kg of retigabine were injected intravenously to terminate the seizure. The corresponding volume of saline was used as a control. The behavior of and the electroencephalogram (EEG) for each rabbit were continually monitored. Before levobupivacaine infusion, the rabbits were placed in a prostrate position calmly on the experimental platform, and the EEG pattern exhibited ß waves. Intravenous levobupivacaine induced a typical EEG seizure characterized by multiple spike and slow wave complexes. The EEG changes were accompanied by behavioral convulsions which were characterized by clonic activity and opisthotonus. RESULTS: Retigabine effectively terminated the electrographic and behavioral seizures. After receiving 5 mg/kg of retigabine, the animals became drowsy, and the EEG changed to δ waves. CONCLUSIONS: We propose that KCNQ2/3 channels play an important role in levobupivacaine-induced central nervous system toxicity, and a KCNQ2/3 channel activator may be used to treat levobupivacaine-induced convulsions.


Assuntos
Anticonvulsivantes/farmacologia , Bupivacaína/análogos & derivados , Carbamatos/farmacologia , Fenilenodiaminas/farmacologia , Convulsões/prevenção & controle , Animais , Bupivacaína/toxicidade , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/patologia , Canal de Potássio KCNQ2/antagonistas & inibidores , Levobupivacaína , Neurônios/efeitos dos fármacos , Estudos Prospectivos , Coelhos , Distribuição Aleatória
5.
Pharmacology ; 96(3-4): 124-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26202459

RESUMO

BACKGROUND: Chloroprocaine is a local ester anesthetic, producing excellent sensory block in clinical use. The Kv7/M potassium channel plays an important role in the control of neuronal excitability. In this study, we investigated the effects of the local anesthetic chloroprocaine on Kv7/M channels as well as the effect of retigabine on chloroprocaine-induced seizures. METHODS: A perforated whole-cell patch technique was used to record Kv7 currents from HEK293 cells and M-type currents from rat dorsal root ganglion (DRG) neurons. RESULTS: Chloroprocaine produced a number of effects on Kv7.2/Kv7.3 currents, including a lowering of current amplitudes, a rightward shift in the voltage-dependent activation curves, and a slowing of channel activation. Chloroprocaine had a more selective inhibitory effect on the homomeric Kv7.3 and heteromeric Kv7.2/Kv7.3 channels than on the homomeric Kv7.2 channel. Chloroprocaine also inhibited native M channel currents and induced a depolarization of the DRG neuron membrane potential. CONCLUSION: Taken together, the findings indicate that chloroprocaine concentration dependently inhibited Kv7/M channel currents.


Assuntos
Anestésicos Locais/farmacologia , Canal de Potássio KCNQ2/efeitos dos fármacos , Canal de Potássio KCNQ3/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Procaína/análogos & derivados , Animais , Carbamatos/farmacologia , Relação Dose-Resposta a Droga , Gânglios Espinais/efeitos dos fármacos , Células HEK293 , Humanos , Potenciais da Membrana/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Fenilenodiaminas/farmacologia , Procaína/farmacologia , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente
7.
PLoS One ; 6(1): e16315, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21283706

RESUMO

BACKGROUND: Previously, we identified a major myeloid-derived proinflammatory subpopulation of human blood dendritic cells which we termed slanDCs (e.g. Schäkel et al. (2006) Immunity 24, 767-777). The slan epitope is an O-linked sugar modification (6-sulfo LacNAc, slan) of P-selectin glycoprotein ligand-1 (PSGL-1). As slanDCs can induce neoantigen-specific CD4+ T cells and tumor-reactive CD8+ cytotoxic T cells, they appear as promising targets for an in vivo delivery of antigens for vaccination. However, tools for delivery of antigens to slanDCs were not available until now. Moreover, it is unknown whether or not antigens delivered via the slan epitope can be taken up, properly processed and presented by slanDCs to T cells. METHODOLOGY/PRINCIPAL FINDINGS: Single chain fragment variables were prepared from presently available decavalent monoclonal anti-slan IgM antibodies but failed to bind to slanDCs. Therefore, a novel multivalent anti-slanDC scaffold was developed which consists of two components: (i) a single chain bispecific recombinant diabody (scBsDb) that is directed on the one hand to the slan epitope and on the other hand to a novel peptide epitope tag, and (ii) modular (antigen-containing) linker peptides that are flanked at both their termini with at least one peptide epitope tag. Delivery of a Tetanus Toxin-derived antigen to slanDCs via such a scBsDb/antigen scaffold allowed us to recall autologous Tetanus-specific memory T cells. CONCLUSIONS/SIGNIFICANCE: In summary our data show that (i) the slan epitope can be used for delivery of antigens to this class of human-specific DCs, and (ii) antigens bound to the slan epitope can be taken up by slanDCs, processed and presented to T cells. Consequently, our novel modular scaffold system may be useful for the development of human vaccines.


Assuntos
Amino Açúcares/imunologia , Antígenos/administração & dosagem , Células Dendríticas/imunologia , Sistemas de Liberação de Medicamentos/métodos , Imunoterapia/métodos , Apresentação de Antígeno , Humanos , Glicoproteínas de Membrana , Linfócitos T/imunologia , Vacinas/imunologia
8.
Stem Cells Dev ; 19(5): 607-14, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19824807

RESUMO

Bone marrow-derived mesenchymal stromal cells (MSCs) represent a population of nonhematopoietic cells, which play a crucial role in supporting hematopoiesis and can differentiate into various cell types such as osteocytes, chondrocytes, adipocytes, and myocytes. Due to their differentiation capability, MSCs emerge as promising candidates for therapeutic applications in tissue engineering. In addition, they display immunomodulatory properties that have prompted consideration of their potential use for treatment modalities aimed at the inhibition of immune responses. In this context, MSCs efficiently inhibit maturation, cytokine production, and T-cell stimulatory capacity of dendritic cells (DCs). They also markedly impair proliferation, cytokine secretion, and cytotoxic potential of natural killer cells and T lymphocytes. Furthermore, MSCs are able to inhibit the proliferation of B cells and their capacity to produce antibodies. Various animal models confirm the immunomodulatory properties of MSCs. Thus, administered MSCs prolong the survival of skin and cardiac allografts and ameliorate acute graft-versus-host disease (GVHD) as well as experimental autoimmune encephalomyelitis. Clinical studies enrolling patients with severe acute GVHD reveal that the administration of MSCs results in significant clinical responses. Due to their immunomodulatory capability and their low immunogenicity, MSCs represent promising candidates for the prevention and treatment of immune-mediated diseases.


Assuntos
Doenças do Sistema Imunitário/terapia , Células-Tronco Mesenquimais/imunologia , Humanos , Fatores Imunológicos , Transplante de Células-Tronco Mesenquimais , Células Estromais
9.
Haematologica ; 94(8): 1151-6, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19546436

RESUMO

Mesenchymal stem cells emerged as a promising treatment modality for steroid-refractory graft-versus-host disease, which represents a major complication of allogeneic hematopoietic stem cell transplantation. Dendritic cells (DCs) display an extraordinary capacity to induce T-cell responses and play a crucial role in the pathogenesis of graft-versus-host disease. Here, we investigated the impact of mesenchymal stem cells on the proinflammatory capacity of 6-sulfo LacNAc (slan) dendritic cells, representing a major subpopulation of human blood dendritic cells. Mesenchymal stem cells markedly impair maturation of slanDCs and their ability to secrete proinflammatory cytokines, which was dependent on prostaglandin E(2). In contrast, the release of anti-inflammatory IL-10 was improved by mesenchymal stem cells. Furthermore, mesenchymal stem cells efficiently inhibit slanDC-induced proliferation of CD4(+) and CD8(+) T cells and polarization of naïve CD4(+) T lymphocytes into Th1 cells. These results indicate that mesenchymal stem cells significantly impair the high proinflammatory capacity of slanDCs and further substantiate their potential for the treatment of graft-versus-host disease.


Assuntos
Citocinas/metabolismo , Células Dendríticas/metabolismo , Células-Tronco Mesenquimais/imunologia , Amino Açúcares/imunologia , Antígenos CD/imunologia , Antígeno B7-2/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/imunologia , Citometria de Fluxo , Antígenos HLA-DR/imunologia , Humanos , Imunoglobulinas/imunologia , Interferon gama/metabolismo , Interleucina-4/metabolismo , Glicoproteínas de Membrana/imunologia , Células-Tronco Mesenquimais/citologia , Antígeno CD83
10.
Transplantation ; 87(8): 1134-9, 2009 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-19384158

RESUMO

BACKGROUND: Extracorporeal photopheresis (ECP) emerged as a promising treatment modality for steroid-refractory graft-versus-host disease (GVHD), which represents a major complication of allogeneic hematopoietic stem-cell transplantation. Dendritic cells (DCs) display an extraordinary capacity to induce T-cell responses and play a crucial role in the initiation and maintainance of GVHD. This study evaluated the direct impact of ECP on the proinflammatory capacity of 6-sulfo LacNAc (slan) DCs, representing a major subpopulation of human blood DCs. METHODS: SlanDCs were isolated from ECP-treated or untreated blood of healthy donors or GVHD patients by immunomagnetic isolation. The maturation of slanDC was determined by flow cytometry. Cytokine production of slanDCs was measured by enzyme-linked immunosorbent assay. SlanDC-mediated T-cell proliferation was evaluated by H-thymidine incorporation. SlanDC-mediated T-cell programming was determined by flow cytometry. RESULTS: ECP efficiently impairs the spontaneous maturation and secretion of proinflammatory tumor necrosis factor-alpha, interleukin-1beta, and interleukin-12 by slanDCs. Furthermore, ECP markedly inhibits slanDC-induced proliferation of CD4 and CD8 T cells and polarization of naïve CD4 T lymphocytes into Th1 cells. CONCLUSIONS: These novel findings indicate that ECP efficiently impairs the proinflammatory capacity of slanDCs, which may represent an important mechanism for the therapeutic efficiency of ECP in GVHD.


Assuntos
Sistema ABO de Grupos Sanguíneos/análise , Amino Açúcares/análise , Células Dendríticas/fisiologia , Inflamação/fisiopatologia , Fotoferese , Sistema ABO de Grupos Sanguíneos/imunologia , Proteínas de Transporte/análise , Proteínas de Transporte/imunologia , Células Dendríticas/efeitos da radiação , Humanos , Immunoblotting , Inflamação/prevenção & controle , Proteínas de Membrana/análise , Proteínas de Membrana/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Proteoma , Fator de von Willebrand/análise , Fator de von Willebrand/imunologia
11.
J Immunol ; 174(7): 4127-34, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15778372

RESUMO

Dendritic cells (DCs) are characterized by their unique capacity for primary T cell activation, providing the opportunity for DC-based cancer vaccination protocols. Novel findings reveal that besides their role as potent inducers of tumor-specific T cells, human DCs display additional antitumor effects. Most of these data were obtained with monocyte-derived DCs, whereas studies investigating native blood DCs are limited. In the present study, we analyze the tumoricidal capacity of M-DC8(+) DCs, which represent a major subpopulation of human blood DCs. We demonstrate that IFN-gamma-stimulated M-DC8(+) DCs lyse different tumor cell lines but not normal cells. In addition, we show that tumor cells markedly enhance the production of TNF-alpha by M-DC8(+) DCs via cell-to-cell contact and that this molecule essentially contributes to the killing activity of M-DC8(+) DCs. Furthermore, we illustrate the ability of M-DC8(+) DCs to promote proliferation, IFN-gamma production, and tumor-directed cytotoxicity of NK cells. The M-DC8(+) DC-mediated enhancement of the tumoricidal potential of NK cells is mainly dependent on cell-to-cell contact. These results reveal that, in addition to their crucial role in activating tumor-specific T cells, blood DCs exhibit direct tumor cell killing and enhance the tumoricidal activity of NK cells. These findings point to the pivotal role of DCs in triggering innate and adaptive immune responses against tumors.


Assuntos
Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Neoplasias/patologia , Células Sanguíneas , Antígenos CD8 , Comunicação Celular , Linhagem Celular Tumoral , Células Dendríticas/citologia , Humanos , Imunidade , Ativação Linfocitária/imunologia , Neoplasias/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/fisiologia
12.
Blood ; 100(4): 1502-4, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12149240

RESUMO

Functional studies on native human dendritic cells (DCs) are hampered by technical difficulties in preparing fresh DCs. Recently, with the help of the monoclonal antibody M-DC8, we succeeded in isolating a major subpopulation of human blood DCs by a one-step immunomagnetic separation procedure. These cells strongly express Fc gamma RIII (CD16) and Fc gamma RII (CD32) and are quite efficient in the antigen-specific activation of naive T cells. Because some Fc gamma receptor-bearing cell types are known as effector cells in antibody-dependent cellular cytotoxicity (ADCC), we investigated whether M-DC8(+) DCs are capable of effectuating ADCC. In this report we show that freshly prepared M-DC8(+) DCs efficiently mediate tumor-directed ADCC and that both types of Fc gamma receptors as well as tumor necrosis factor alpha essentially contribute to the cytotoxic activity. The results provide evidence that, in addition to their pivotal role in primary T-cell activation, a subset of blood DCs displays efficient cytotoxicity in ADCC.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Células Dendríticas/imunologia , Adenocarcinoma , Animais , Anticorpos Monoclonais , Neoplasias da Mama , Separação Celular , Neoplasias Colorretais , Humanos , Separação Imunomagnética , Camundongos , Receptores de IgG/análise , Receptores de IgG/imunologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...