Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Reprod Immunol ; 86(5): e13483, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34233077

RESUMO

PROBLEM: Cell-free fetal DNA (cffDNA) shed from the placenta can be detected in maternal blood and increases incrementally during gestation. Concentrations are further elevated with pregnancy complications. Specific activators of cffDNA release in such complications have not been identified. Here, we use trophoblast cells from early and term placenta to examine cffDNA release following apoptosis, infection, and sterile inflammatory stress. METHOD OF STUDY: HTR8/SVneo cells were used to model first-trimester trophoblasts, and term cytotrophoblasts (CTBs) were isolated from placentae collected after uncomplicated deliveries. Trophoblasts were treated with varying concentrations of doxorubicin (DOX), lipopolysaccharide (LPS), or high-mobility group box protein 1 (HMGB1) for 18 h. Cells or supernatants were quantified for caspase-3/7 cleavage, pro-inflammatory cytokine secretion, and cffDNA release. RESULTS: Both HTR8/SVneo and CTBs underwent caspase-3/7 cleavage following DOX treatment, with HTR8/SVneo cells more sensitive to apoptosis than term CTBs. Apoptotic cells released more cffDNA in a dose-dependent manner. Treatment with LPS resulted in an increase in pro-inflammatory IL-6 release, particularly in term CTBs compared to early trophoblasts; however, LPS did not affect cffDNA release. Lastly, while neither cell released more TNF-α following stimulation with HMGB1, both HTR8/SVneo and CTBs released significantly more cffDNA in the presence of HMGB1. CONCLUSIONS: These data show that apoptosis and sterile inflammation induced by DOX and HMGB1, respectively, cause an increase in cffDNA concentrations in both first-trimester and term trophoblasts. Understanding physiologic release of cffDNA during healthy and complicated pregnancy can identify new targets for the diagnosis and treatment of gestational complications.


Assuntos
Apoptose , Ácidos Nucleicos Livres/metabolismo , DNA/metabolismo , Inflamação/metabolismo , Trofoblastos/metabolismo , Adulto , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Ácidos Nucleicos Livres/genética , DNA/genética , Doxorrubicina/toxicidade , Feminino , Proteína HMGB1/toxicidade , Humanos , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Trofoblastos/efeitos dos fármacos , Trofoblastos/patologia , Fator de Necrose Tumoral alfa/metabolismo
2.
Biochem Pharmacol ; 186: 114496, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33667472

RESUMO

High mobility box 1 (HMGB1), a damage-associated molecular pattern, has crucial roles in induction of neuropathic pain. Upregulation of HMGB1 around the injured sciatic nerve contributes to mechanical hypersensitivity following partial sciatic nerve ligation (PSNL) of mice. However, central mechanisms mediating perineural HMGB1-induced nociceptive hypersensitivity, especially within the spinal dorsal horn, have not been determined. The current study shows that perineural treatment of naïve mice with recombinant HMGB1, which mimics increased HMGB1 around the injured sciatic nerve of PSNL mice, significantly induced activation of microglia, but not astrocytes, in the spinal dorsal horn. Intraperitoneal injection of minocycline, a microglial inhibitor, ameliorated perineural rHMGB1-induced mechanical hypersensitivity. In addition, blockade of spinal N-methyl-D-aspartate (NMDA) receptors significantly prevented perineural rHMGB1-induced mechanical hypersensitivity and microglial activation. In contrast, non-NMDA receptors, neurokinin 1 receptor, colony-stimulating factor 1 receptor and P2Y12 receptor were not involved in perineural rHMGB1-induced mechanical hypersensitivity. Furthermore, repeated perineural treatment with an anti-HMGB1 antibody blocked activation of spinal microglia in PSNL mice. Collectively, the current findings demonstrate that increased HMGB1 around injured sciatic nerve might induce nociceptive hypersensitivity through activation of spinal microglia. Thus, HMGB1-dependent mechanisms between the injured sciatic nerve and spinal dorsal horn could be crucial in induction of neuropathic pain.


Assuntos
Glutamatos/metabolismo , Proteína HMGB1/metabolismo , Hiperalgesia/metabolismo , Microglia/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Animais , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Proteína HMGB1/toxicidade , Humanos , Hiperalgesia/induzido quimicamente , Injeções Espinhais , Masculino , Camundongos , Microglia/efeitos dos fármacos , N-Metilaspartato/administração & dosagem , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Corno Dorsal da Medula Espinal/efeitos dos fármacos
3.
Am J Physiol Cell Physiol ; 318(3): C570-C580, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31913693

RESUMO

Cystic fibrosis (CF) lung disease persists and remains life-limiting for many patients. Elevated high-mobility group box-1 protein (HMGB-1) levels and epithelial sodium channel hyperactivity (ENaC) are hallmark features of the CF lung. The objective of this study was to better understand the pathogenic role of HMGB-1 signaling and ENaC in CF airway cells. We hypothesize that HMGB-1 links airway inflammation [via signaling to the receptor for advanced glycation end products (RAGE)] and airway surface liquid dehydration (via upregulation of ENaC) in the CF lung. We calculated equivalent short-current (Isc) and single-channel ENaC open probability (Po) in normal and CF human small airway epithelial cells (SAEC) in the presence and absence of human HMGB-1 peptide (0.5 µg/mL). In normal SAECs, HMGB-1 increased amiloride-sensitive Isc and elevated ENaC Po from 0.15 ± 0.03 to 0.28 ± 0.04 (P < 0.01). In CF SAECs, ENaC Po increased from 0.45 ± 0.06 to 0.73 ± 0.04 (P < 0.01). Pretreatment with 1 µM FPS-ZM1 (a RAGE inhibitor) attenuated all HMGB-1 effects on ENaC current in normal and CF SAECs. Confocal analysis of SAECs indicates that nuclear size and HMBG-1 localization can be impacted by ENaC dysfunction. Masson's trichrome labeling of mouse lung showed that intraperitoneally injected HMGB-1 significantly increased pulmonary fibrosis. Bronchoalveolar lavage fluid from HMGB-1-treated mice showed significant increases in IL-1ß, IL-10, IL-6, IL-27, IL-17A, IFN-ß, and granulocyte-macrophage colony-stimulating factor compared with vehicle-injected mice (P < 0.05). These studies put forth a new model in which HMGB-1 signaling to RAGE plays an important role in perpetuating ENaC dysfunction and inflammation in the CF lung.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Proteína HMGB1/toxicidade , Mediadores da Inflamação/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Mucosa Respiratória/metabolismo , Animais , Células Cultivadas , Feminino , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mucosa Respiratória/citologia , Mucosa Respiratória/efeitos dos fármacos
4.
Arch Pharm Res ; 39(12): 1726-1738, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27778275

RESUMO

A certain nucleosomal protein-high mobility group box-1 (HMGB1)-has recently been established as a late mediator of sepsis, with a relatively wide therapeutic window for pharmacological intervention. Pelargonidin (PEL) is a well-known red pigment found in plants; it has important biological activities that are potentially beneficial for human health. In the present study, we investigated whether PEL can modulate HMGB1-mediated inflammatory responses in human umbilical vein endothelial cells (HUVECs) and in mice. The anti-inflammatory activities of PEL were determined by measuring permeability, leukocyte adhesion and migration, and activation of pro-inflammatory proteins in HMGB1-activated HUVECs and mice, as well as the beneficial effects of PEL on survival rate in the mouse sepsis model. The data showed that PEL had effectively inhibited lipopolysaccharide (LPS)-induced release of HMGB1 and suppressed HMGB1-mediated septic responses, such as hyperpermeability, adhesion and migration of leukocytes, and expression of cell adhesion molecules. Furthermore, PEL inhibited the HMGB1-mediated production of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), as well as the activation of nuclear factor-κB (NF-κB) and extracellular signal-regulated kinases 1 and 2 (ERK1/2). Collectively, these results indicate that PEL could be used to treat various severe vascular inflammatory diseases via the inhibition of the HMGB1 signaling pathway.


Assuntos
Antocianinas/uso terapêutico , Anti-Infecciosos Locais/uso terapêutico , Proteína HMGB1/toxicidade , Pigmentos Biológicos/uso terapêutico , Sepse/tratamento farmacológico , Animais , Antocianinas/química , Antocianinas/farmacologia , Anti-Infecciosos Locais/química , Anti-Infecciosos Locais/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pigmentos Biológicos/química , Pigmentos Biológicos/farmacologia , Sepse/metabolismo , Resultado do Tratamento
5.
J Clin Invest ; 125(10): 3965-80, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26389678

RESUMO

Erythropoiesis is an important response to certain types of stress, including hypoxia, hemorrhage, bone marrow suppression, and anemia, that result in inadequate tissue oxygenation. This stress-induced erythropoiesis is distinct from basal red blood cell generation; however, neither the cellular nor the molecular factors that regulate this process are fully understood. Here, we report that type 1 conventional dendritic cells (cDC1s), which are defined by expression of CD8α in the mouse and XCR1 and CLEC9 in humans, are critical for induction of erythropoiesis in response to stress. Specifically, using murine models, we determined that engagement of a stress sensor, CD24, on cDC1s upregulates expression of the Kit ligand stem cell factor on these cells. The increased expression of stem cell factor resulted in Kit-mediated proliferative expansion of early erythroid progenitors and, ultimately, transient reticulocytosis in the circulation. Moreover, this stress response was triggered in part by alarmin recognition and was blunted in CD24 sensor- and CD8α+ DC-deficient animals. The contribution of the cDC1 subset to the initiation of stress erythropoiesis was distinct from the well-recognized role of macrophages in supporting late erythroid maturation. Together, these findings offer insight into the mechanism of stress erythropoiesis and into disorders of erythrocyte generation associated with stress.


Assuntos
Células Dendríticas/fisiologia , Eritropoese/fisiologia , Estresse Fisiológico/fisiologia , Alarminas/fisiologia , Animais , Antígeno CD24/fisiologia , Antígenos CD8/análise , Cisplatino/toxicidade , Ensaio de Unidades Formadoras de Colônias , Células Dendríticas/classificação , Células Precursoras Eritroides/fisiologia , Feminino , Perfilação da Expressão Gênica , Proteína HMGB1/toxicidade , Transplante de Células-Tronco Hematopoéticas , Xenoenxertos , Humanos , Hipóxia/fisiopatologia , Mesilato de Imatinib/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Flebotomia/efeitos adversos , Quimera por Radiação , Proteínas Recombinantes/toxicidade , Esplenectomia/efeitos adversos , Fator de Células-Tronco/biossíntese , Fator de Células-Tronco/genética
6.
Thromb Haemost ; 114(2): 350-63, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25947626

RESUMO

Sepsis is a life-threatening condition that arises when the body's response to infection causes systemic inflammation. High-mobility group box 1 (HMGB1), as a late mediator of sepsis, enhances hyperpermeability, and it is therefore a therapeutic target. Despite extensive research into the underlying mechanisms of sepsis, the target molecules controlling vascular leakage remain largely unknown. Moesin is a cytoskeletal protein involved in cytoskeletal changes and paracellular gap formation. The objectives of this study were to determine the roles of moesin in HMGB1-mediated vascular hyperpermeability and inflammatory responses and to investigate the mechanisms of action underlying these responses. Using siRNA knockdown of moesin expression in primary human umbilical vein endothelial cells (HUVECs), moesin was found to be required in HMGB1-induced F-actin rearrangement, hyperpermeability, and inflammatory responses. The mechanisms involved in moesin phosphorylation were analysed by blocking the binding of the HMGB1 receptor (RAGE) and inhibiting the Rho and MAPK pathways. HMGB1-treated HUVECs exhibited an increase in Thr558 phosphorylation of moesin. Circulating levels of moesin were measured in patients admitted to the intensive care unit with sepsis, severe sepsis, and septic shock; these patients showed significantly higher levels of moesin than healthy controls, which was strongly correlated with disease severity. High blood moesin levels were also observed in cecal ligation and puncture (CLP)-induced sepsis in mice. Administration of blocking moesin antibodies attenuated CLP-induced septic death. Collectively, our findings demonstrate that the HMGB1-RAGE-moesin axis can elicit severe inflammatory responses, suggesting it to be a potential target for the development of diagnostics and therapeutics for sepsis.


Assuntos
Permeabilidade Capilar/fisiologia , Proteína HMGB1/toxicidade , Sepse/sangue , Actinas/análise , Animais , Ceco/lesões , Adesão Celular , Moléculas de Adesão Celular/biossíntese , Movimento Celular , Citoesqueleto/ultraestrutura , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana , Humanos , Perfuração Intestinal/sangue , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos , Neutrófilos/citologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Índice de Gravidade de Doença , Choque Séptico/sangue , Transdução de Sinais
9.
Anesthesiology ; 120(5): 1160-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24162463

RESUMO

BACKGROUND: Aseptic trauma engages the innate immune response to trigger a neuroinflammatory reaction that results in postoperative cognitive decline. The authors sought to determine whether high-mobility group box 1 protein (HMGB1), an ubiquitous nucleosomal protein, initiates this process through activation and trafficking of circulating bone marrow-derived macrophages to the brain. METHODS: The effects of HMGB1 on memory (using trace fear conditioning) were tested in adult C57BL/6J male mice; separate cohorts were tested after bone marrow-derived macrophages were depleted by clodrolip. The effect of anti-HMGB1 neutralizing antibody on the inflammatory and behavioral responses to tibial surgery were investigated. RESULTS: A single injection of HMGB1 caused memory decline, as evidenced by a decrease in freezing time (52 ± 11% vs. 39 ± 5%; n = 16-17); memory decline was prevented when bone marrow-derived macrophages were depleted (39 ± 5% vs. 50 ± 9%; n = 17). Disabling HMGB1 with a blocking monoclonal antibody, before surgery, reduced postoperative memory decline (52 ± 11% vs. 29 ± 5%; n = 15-16); also, hippocampal expression of monocyte chemotactic protein-1 was prevented by the neutralizing antibody (n = 6). Neither the systemic nor the hippocampal inflammatory responses to surgery occurred in mice pretreated with anti-HMGB1 neutralizing antibody (n = 6). CONCLUSION: Postoperative neuroinflammation and cognitive decline can be prevented by abrogating the effects of HMGB1. Following the earlier characterization of the resolution of surgery-induced memory decline, the mechanisms of its initiation are now described. Together, these data may be used to preoperatively test the risk to surgical patients for the development of exaggerated and prolonged postoperative memory decline that is reflected in delirium and postoperative cognitive dysfunction, respectively.


Assuntos
Células da Medula Óssea/metabolismo , Transtornos Cognitivos/metabolismo , Proteína HMGB1/toxicidade , Macrófagos/metabolismo , Complicações Pós-Operatórias/metabolismo , Animais , Anticorpos Bloqueadores/administração & dosagem , Anticorpos Bloqueadores/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Transtornos Cognitivos/imunologia , Transtornos Cognitivos/prevenção & controle , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/imunologia , Imunidade Inata , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complicações Pós-Operatórias/imunologia , Complicações Pós-Operatórias/prevenção & controle , Distribuição Aleatória
10.
PLoS One ; 8(5): e64375, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691208

RESUMO

BACKGROUND: Acute lung injury (ALI) is considered to be the major cause of respiratory failure in critically ill patients. Clinical studies have found that in patients with sepsis and after hemorrhage, the elevated level of high mobility group box-1(HMGB-1) in their circulation is highly associated with ALI, but the underlying mechanism remains unclear. Extracellular HMGB-1 has cytokine-like properties and can bind to Toll-like Receptor-4 (TLR4), which was reported to play an important role in the pathogenesis of ALI. The aim of this study was to determine whether HMGB-1 directly contributes to ALI and whether TLR4 signaling pathway is involved in this process. METHODS: Recombinant human HMGB-1 (rhHMGB-1) was used to induce ALI in male Sprague-Dawley rats. Lung specimens were collected 2 h after HMGB-1 treatment. The levels of TNF-α, IL-1ß, TLR4 protein, and TLR4 mRNA in lungs as well as pathological changes of lung tissue were assessed. In cell studies, the alveolar macrophage cell line, NR8383, was collected 24 h after rhHMGB-1 treatment and the levels of TNF-α and IL-1ß in cultured medium as well as TLR4 protein and mRNA levels in the cell were examined. TLR4-shRNA-lentivirus was used to inhibit TLR4 expression, and a neutralizing anti-HMGB1 antibody was used to neutralize rhHMGB-1 both in vitro and in vivo. RESULTS: Features of lung injury and significant elevation of IL-1ß and TNF-α levels were found in lungs of rhHMGB-1-treated animals. Cultured NR8383 cells were activated by rhHMGB-1 treatment and resulted in the release of IL-1ß and TNF-α. TLR4 expression was greatly up-regulated by rhHMGB-1. Inhibition of TLR4 or neutralization of HMGB1 with a specific antibody also attenuated the inflammatory response induced by HMGB-1 both in vivo and in vitro. CONCLUSION: HMGB-1 can activate alveolar macrophages to produce proinflammatory cytokines and induce ALI through a mechanism that relies on TLR-4.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Proteína HMGB1/toxicidade , Proteínas Recombinantes/toxicidade , Receptor 4 Toll-Like/metabolismo , Análise de Variância , Animais , Anticorpos Neutralizantes/metabolismo , Western Blotting , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Proteína HMGB1/genética , Proteína HMGB1/imunologia , Técnicas Histológicas , Humanos , Interleucina-1beta/metabolismo , Macrófagos , Masculino , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/genética , Fator de Necrose Tumoral alfa/metabolismo
11.
Shock ; 31(3): 280-4, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19218854

RESUMO

High-mobility group box 1 (HMGB-1) has been reported as a "late" proinflammatory mediator in sepsis. In vitro data have shown that HMGB-1 can induce activation of intracellular signaling pathways via interaction with at least three pattern recognition receptors: Toll-like receptor (TLR) 2, TLR-4, and the receptor for advanced glycation end products (RAGE). The objective of this study was to investigate the role of these receptors in the in vivo response to HMGB-1. Therefore, we first performed a time-series experiment with wild-type (Wt) mice. High-mobility group box 1 induced time-dependent elevations of TNF-alpha, IL-6, monocyte chemoattractant protein 1, and thrombin-antithrombin complex levels in peritoneal lavage fluid and plasma. This inflammatory reaction was accompanied by a prominent and sustained rise in neutrophil counts in the peritoneal cavity. We next administered HMGB-1 to Wt, TLR-2, TLR-4, and RAGE mice. All genotypes showed similar plasma levels of TNF-alpha, IL-6, IL-10, and thrombin-antithrombin complex at 2 h after intraperitoneal injection of HMGB-1. Compared with Wt mice, both TLR-4 and RAGE mice displayed lower TNF-alpha and IL-6 concentrations and lower neutrophil numbers in their peritoneal lavage fluid. In contrast, TLR-2 mice showed increased levels of TNF-alpha and IL-6 in their peritoneal cavity relative to Wt mice. These data indicate that HMGB-1 induces release of cytokines, activation of coagulation, and neutrophil recruitment in vivo via a mechanism that at least in part depends on TLR-4 and RAGE.


Assuntos
Proteína HMGB1/toxicidade , Mediadores da Inflamação/toxicidade , Receptores Imunológicos/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Antitrombina III/genética , Antitrombina III/metabolismo , Citocinas/genética , Citocinas/metabolismo , Feminino , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Inflamação/mortalidade , Masculino , Camundongos , Camundongos Knockout , Neutrófilos/metabolismo , Neutrófilos/patologia , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Fatores de Tempo , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética
12.
Curr Opin Clin Nutr Metab Care ; 6(3): 283-7, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12690260

RESUMO

PURPOSE OF REVIEW: Products of infection, ischemia, and injury stimulate the innate immune system to release proinflammatory cytokines, which act locally to activate specific cellular immune responses and initiate recovery. In pathological cases, however, cytokines are released systemically, resulting in progressive tissue injury, hypotension, organ dysfunction, or death. Observations that animals frequently succumb to systemic inflammation long after the peak activity of tumor necrosis factor and interleukin-1beta suggest that later-acting, downstream inflammatory factors can mediate the pathological sequelae of lethal systemic inflammation. Here, the authors review evidence that the chromosomal protein high mobility group box 1 is a late-acting, downstream mediator of pathological inflammation. RECENT FINDINGS: High mobility group box 1 recently has been identified as a proinflammatory cytokine with significantly delayed release kinetics, as compared with tumor necrosis factor and interleukin-1beta, in animal models of lethal systemic inflammation induced by endotoxin or peritonitis. Administration of exogenous high mobility group box 1 induces acute lung injury, intestinal barrier dysfunction, and lethal systemic inflammatory responses. Its functional cytokine domain has been mapped to the DNA-binding B box, providing structural information that may be useful in the rational design of new therapeutics that target the protein's activity. SUMMARY: Several high mobility group box 1 antagonists have recently been identified. These inhibitors may prove effective in a significantly wider therapeutic window than has been available for previous anti-cytokine strategies, because high mobility group box 1 appears in serum with a significantly delayed kinetics as compared with other cytokines.


Assuntos
Proteína HMGB1/fisiologia , Animais , Citocinas/fisiologia , Proteínas de Ligação a DNA , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/sangue , Proteína HMGB1/toxicidade , Humanos , Inflamação/etiologia , Neoplasias/patologia , Proteínas Recombinantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA