Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
1.
Toxicol Appl Pharmacol ; 484: 116872, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38428465

RÉSUMÉ

Previous studies have demonstrated that tetramethylpyrazine (TMP) can enhance the recovery of motor function in spinal cord injury (SCI) rats. However, the underlying mechanism involved in this therapeutic effect remains to be elucidated. We conducted RNA sequencing with a network pharmacology strategy to predict the targets and mechanism of TMP for SCI. The modified Allen's weight-drop method was used to construct an SCI rat model. The results indicated that the nuclear transfer factor-κB (NF-κB) pathway was identified through the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and an inflammatory response was identified through the Gene Ontology (GO) enrichment analysis. Tumor necrosis factor (TNF) was identified as a crucial target. Western blotting revealed that TMP decreased the protein expression of TNF superfamily receptor 1 (TNFR1), inhibitor κB-α (IκB-α), and NF-κB p65 in spinal cord tissues. Enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) demonstrated that TMP inhibited TNF-α, interleukin-1ß (IL-1ß), reactive oxygen species (ROS), and malondialdehyde (MDA) expression and enhanced superoxide dismutase (SOD) expression. Histopathological observation and behavior assessments showed that TMP improved morphology and motor function. In conclusion, TMP inhibits inflammatory response and oxidative stress, thereby exerting a neuroprotective effect that may be related to the regulation of the TNFR1/IκB-α/NF-κB p65 signaling pathway.


Sujet(s)
Facteur de transcription NF-kappa B , Pyrazines , Traumatismes de la moelle épinière , Animaux , Rats , Facteur de transcription NF-kappa B/métabolisme , Inhibiteur alpha de NF-KappaB , Pyrazines/pharmacologie , Rat Sprague-Dawley , Récepteur au facteur de nécrose tumorale de type I/génétique , Récepteur au facteur de nécrose tumorale de type I/pharmacologie , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Moelle spinale , Traumatismes de la moelle épinière/traitement médicamenteux , Traumatismes de la moelle épinière/anatomopathologie , Facteur de nécrose tumorale alpha/métabolisme
2.
Yonsei Med J ; 65(4): 241-245, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38515362

RÉSUMÉ

Tumor necrosis factor receptor-associated periodic syndrome (TRAPS, OMIM: #142680) is a rare autoinflammatory disease (AID) with recurrent febrile episodes. To our knowledge, we report herein the first case of a patient with TRAPS in South Korea whose symptoms included fever, arthralgia, abdominal pain, rash, myalgia, cough, and lymphadenopathy. A pathogenic de novo mutation, c.175T>C (p.Cys59Arg), in the tumor necrosis factor receptor superfamily member 1A (TNFRSF1A) gene, was confirmed by gene sequencing. The patient has been with tocilizumab (an interleukin-6 inhibitor); tocilizumab administration every other week has completely alleviated the patient's symptoms. Our report further expands the clinical spectrum of patients with TRAPS and reaffirms the use of tocilizumab as a viable alternative treatment option for those patients who are unsatisfactorily responsive to other commonly used biologics, such as canakinumab, anakinra, infliximab, and etanercept. Furthermore, our report may aid in increasing awareness about the existence of mutation-confirmed TRAPS in South Korea in addition to emphasizing the importance of actively pursuing genetic testing to correctly diagnose rare AID.


Sujet(s)
Fièvre , Maladies auto-inflammatoires héréditaires , Humains , Fièvre/complications , Maladies auto-inflammatoires héréditaires/diagnostic , Maladies auto-inflammatoires héréditaires/traitement médicamenteux , Maladies auto-inflammatoires héréditaires/génétique , Récepteur au facteur de nécrose tumorale de type I/génétique , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Mutation , Étanercept/usage thérapeutique
3.
ACS Chem Neurosci ; 15(7): 1596-1608, 2024 04 03.
Article de Anglais | MEDLINE | ID: mdl-38526238

RÉSUMÉ

Multiple sclerosis (MS) is a debilitating autoimmune disease that impacts millions of patients worldwide, disproportionately impacting women (4:1), and often presenting at highly productive stages of life. This disease affects the spinal cord and brain and is characterized by severe neuroinflammation, demyelination, and subsequent neuronal damage, resulting in symptoms like loss of mobility. While untargeted and pan-immunosuppressive therapies have proven to be disease-modifying and manage (or prolong the time between) symptoms in many patients, a significant fraction are unable to achieve remission. Recent work has suggested that targeted neuroinflammation mitigation through selective inflammasome inhibition can offer relief to patients while preserving key components of immune function. Here, we show a screening of potential therapeutic targets using inflammasome-inhibiting Nanoligomers (NF-κB1, TNFR1, TNF-α, IL-6) that meet or far-exceed commercially available small-molecule counterparts like ruxolitinib, MCC950, and deucravacitinib. Using the human brain organoid model, top Nanoligomer combinations (NF-κB1 + TNFR1: NI111, and NF-κB1 + NLRP3: NI112) were shown to significantly reduce neuroinflammation without any observable negative impact on organoid function. Further testing of these top Nanoligomer combinations in an aggressive experimental autoimmune encephalomyelitis (EAE) mouse model for MS using intraperitoneal (IP) injections showed that NF-κB1 and NLRP3 targeting Nanoligomer combination NI112 rescues mice without observable loss of mobility or disability, minimal inflammation in brain and spinal cord histology, and minimal to no immune cell infiltration of the spinal cord and no demyelination, similar to or at par with mice that received no EAE injections (negative control). Mice receiving NI111 (NF-κB1 + TNFR1) also showed reduced neuroinflammation compared to saline (sham)-treated EAE mice and at par/similar to other inflammasome-inhibiting small molecule treatments, although it was significantly higher than NI112 leading to subsequent worsening clinical outcomes. Furthermore, treatment with an oral formulation of NI112 at lower doses showed a significant reduction in EAE severity, albeit with higher variance owing to administration and formulation/fill-and-finish variability. Overall, these results point to the potential of further development and testing of these inflammasome-targeting Nanoliogmers as an effective neuroinflammation treatment for multiple neurodegenerative diseases and potentially benefit several patients suffering from such debilitating autoimmune diseases like MS.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Sclérose en plaques , Humains , Femelle , Souris , Animaux , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Maladies neuro-inflammatoires , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Sclérose en plaques/traitement médicamenteux , Souris de lignée C57BL
4.
Int Immunopharmacol ; 120: 110297, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-37207443

RÉSUMÉ

Overexpression of Staphylococcus aureus mediated CXCL8/CXCR1 axis is a major cause of sepsis and severe inflammatory diseases. This chemokine acts conjointly with various pro-inflammatory and anti-inflammatory cytokines that govern the severity of inflammation. The effects of different combinations of exogenous cytokines on CXCR1 expression in macrophages remain undetermined. Exogenous cytokine and anti-inflammatory cytokine therapy had been used to modulate CXCL8 and CXCR1 expression in peritoneal macrophages. Male Swiss albino mice were inoculated with live S. aureus (106 cells/ mouse) for the development of infection. Exogenous cytokines (TNF-α, IL-12, IFN-γ and IL-10) were administered intraperitoneally (single or combination) 24 h post S. aureus infection. The mice were sacrificed and peritoneal macrophages were isolated three days post infection. CXCL8, IL-12, IL-10 secretion, ROS generation and the bacterial phagocytic process had been evaluated. Western blot was used to study the expressions of TNFR1, IL-1R, CXCR1 and NF-κB. TNF-α, IL-12 and IFN-γ treatments aggravated CXCL8 and CXCR1 expression in the macrophages of infected mice. TNF-α + IFN-γ treatment was a major inducer of nitric oxide release and mediated maximum bacterial killing. IL-12 + TNF-α treatment was most potent in increasing ROS, CXCL8/CXCR1 expression through increased levels of TNFR1, IL-1R and NF-κB activation. IL-10 reversed the effects of exogenous cytokines but also impaired the bacterial clearance phenomenon in peritoneal lavage. Treatment with IL-12 + TNF-α + IL-10 was most effective in ameliorating oxidative stress, reduced CXCL8 release and expression levels of TNFR1, IL-1R, and NF-κB. Concludingly, IL-12 + TNF-α + IL-10 treatment mitigated CXCL8/CXCR1 expression and inflammatory signalling via downregulation of TNFR1-IL-1R-NF-κB pathway in peritoneal macrophages and inflammatory sequelae during S. aureus infection.


Sujet(s)
Récepteur au facteur de nécrose tumorale de type I , Infections à staphylocoques , Animaux , Mâle , Souris , Cytokines/métabolisme , Interleukine-10 , Interleukine-12/usage thérapeutique , Macrophages péritonéaux/métabolisme , Facteur de transcription NF-kappa B , Espèces réactives de l'oxygène , Récepteurs à l'interleukine-8A/métabolisme , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Récepteur au facteur de nécrose tumorale de type I/métabolisme , Infections à staphylocoques/métabolisme , Staphylococcus aureus/métabolisme , Facteur de nécrose tumorale alpha
5.
ACS Biomater Sci Eng ; 9(10): 5843-5854, 2023 Oct 09.
Article de Anglais | MEDLINE | ID: mdl-37043416

RÉSUMÉ

Excessive inflammatory response after severe scalding is an important cause of delayed wound healing and is even life-threatening. Tumor necrosis factor α (TNF-α) is a key pro-inflammatory factor of skin trauma. Interacting with tumor necrosis factor receptor 1 (TNF-R1), TNF-α causes excessive inflammation and poor prognosis by activating NF-κB pathway. Antagonizing high levels of TNF-α is one of the therapeutic approaches for diseases associated with the overactivation of inflammatory responses. However, the available monoclonal antibodies are limited in their application due to their complex preparation process, high price, and the lack of cell targeting ability leading to systemic toxicity and side effects. In this study, by using a genetic bioengineering technique, we modified TNF-R1 on the cell membrane surface-derived nanovesicles (NVs). We confirmed that TNF-R1 NVs stably expressed TNF-R1 on the membrane surface and interacted with its ligand TNF-α. Furthermore, TNF-R1 NVs competitively antagonized the effect of TNF-α in the wound healing assay in vitro. In the scalded mouse model, TNF-R1 NVs were released continuously from the thermosensitive hydrogel Pluronic F-127, resulting in less inflammation and better wound healing. Our results revealed TNF-R1 NVs as promising cell-free therapeutic agents in alleviating TNF-α-mediated pro-inflammatory signaling and promoting wound repair.


Sujet(s)
Récepteur au facteur de nécrose tumorale de type I , Facteur de nécrose tumorale alpha , Animaux , Souris , Hydrogels/composition chimique , Hydrogels/pharmacologie , Inflammation/traitement médicamenteux , Récepteur au facteur de nécrose tumorale de type I/pharmacologie , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Transduction du signal , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme , Nanoparticules/usage thérapeutique , Brûlures/traitement médicamenteux
6.
Drug Discov Today ; 28(6): 103575, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37003513

RÉSUMÉ

Tumor necrosis factor (TNF) is a pleiotropic cytokine with a major role in immune system homeostasis and is involved in many inflammatory and autoimmune diseases, such as rheumatoid arthritis (RA), psoriasis, Alzheimer's disease (AD), and multiple sclerosis (MS). Thus, TNF and its receptors, TNFR1 and TNFR2, are relevant pharmacological targets. Biologics have been developed to block TNF-dependent signaling cascades, but they display serious side effects, and their pharmacological effectiveness decreases over time because of their immunogenicity. In this review, we present recent discoveries in small molecules targeting TNF and its receptors and discuss alternative strategies for modulating TNF signaling.


Sujet(s)
Polyarthrite rhumatoïde , Maladies auto-immunes , Sclérose en plaques , Humains , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Cytokines , Maladies auto-immunes/traitement médicamenteux , Facteur de nécrose tumorale alpha
7.
Cancer Immunol Immunother ; 72(6): 1567-1580, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-36534148

RÉSUMÉ

Obinutuzumab is a therapeutic antibody for B cell non-Hodgkin's Lymphoma (BNHL), which is a glyco-engineered anti-CD20 antibody with enhanced antibody-dependent cellular cytotoxicity (ADCC) and causes binding-induced direct cell death (DCD) through lysosome membrane permeabilization (LMP). Tumour necrosis factor receptor 1 (TNFR1), a pro-inflammatory death receptor, also evokes cell death, partly through lysosomal rupture. As both obinutuzumab- and TNFR1-induced cell deaths are mediated by LMP and combining TNFR1 and obinutuzumab can amplify LMP-mediated cell death, we made dual-targeting antibody for CD20 and TNFR1 to enhance DCD of obinutuzumab.Obinutuzumab treatment-induced CD20 and TNFR1 colocalisation, and TNFR1-overexpressing cells showed increased obinutuzumab-induced DCD. Two targeting modes, anti-CD20/TNFR1 bispecific antibodies (bsAbs), and obinutuzumab-TNFα fusion proteins (OBI-TNFαWT and OBI-TNFαMUT), were designed to cluster CD20 and TNFR1 on the plasma membrane. OBI-TNFαWT and OBI-TNFαMUT showed significantly enhanced LMP, DCD, and ADCC compared with that induced by obinutuzumab. TNFR1 expression is upregulated in many BNHL subtypes compared to that in normal B cells; OBI-TNFαMUT specifically increased DCD and ADCC in a B cell lymphoma cell line overexpressing TNFR1. Further, OBI-TNFαMUT blocked NF-κB activation in the presence of TNF-α, implying that it can antagonise the proliferative role of TNF-α in cancers.Our study suggests that dual targeting of CD20 and TNFR1 can be a new therapeutic strategy for improving BNHL treatment. The OBI-TNFαMUT fusion protein enhances DCD and ADCC and prevents the proliferating effect of TNFα signalling; therefore, it may provide precision treatment for patients with BNHL, especially those with upregulated TNFR1 expression.


Sujet(s)
Lymphome B , Facteur de nécrose tumorale alpha , Humains , Antigènes CD20 , Mort cellulaire , Lymphome B/traitement médicamenteux , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Anticorps bispécifiques/pharmacologie
8.
J Acquir Immune Defic Syndr ; 88(3): 322-327, 2021 11 01.
Article de Anglais | MEDLINE | ID: mdl-34267056

RÉSUMÉ

BACKGROUND: Antiretroviral therapy (ART) is an important hallmark of HIV-1 treatment, enabling viral load suppression to undetectable levels and CD4+ T-cell recovery. However, some individuals do not recover the CD4+ T-cell count to normal levels, despite viral suppression. We hypothesize that variation in genes involved in extrinsic apoptosis pathways may influence interindividual immune recovery during ART. METHODS: We assessed clinical-epidemiological variables and the allelic/genotypic distribution of functional single nucleotide polymorphisms in genes involved in extrinsic apoptosis pathways (TNFRSF1A: rs1800692 and rs767455; TNFAIP3: rs2270926; NFKBIA: rs8904; and TNF-α: rs1800629) and their relationship with immune recovery in ART-treated (1 year) HIV-1-infected individuals. We enrolled 155 HIV-1-infected individuals, with 102 individuals showing immunological success and 53 with immunological failure. RESULTS: Through univariate analysis, we observed that the male sex (60.4%, P = 0.002) showed a higher median of age at treatment onset (34.8 years, P = 0.034) and higher time until virological suppression (6 months, P = 0.035), both risk factors for immune failure. Survival analysis revealed that individuals who started ART treatment with CD4+ T-cell count <200 cells/mm3 took a longer time to immunological recovery (median time = 27 months, P = 0.029). ART containing zidovudine also was associated with immune recovery in univariate e multivariate analysis. Variants in TNFRSF1A (rs767455: T and TT; rs1800692-rs767455: T-T combination) and NFKBIA (rs8904: A) genes were associated with immune failure, whereas NFKBIA (rs8904: GA) and TNF-α (rs1800629: GA) were with CD4+ T-cell recovery. CONCLUSIONS: Clinical-epidemiological variants in genes involved in extrinsic apoptosis pathways might influence the CD4+ T-cell immune recovery.


Sujet(s)
Thérapie antirétrovirale hautement active/effets indésirables , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Infections à VIH/traitement médicamenteux , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Facteur de nécrose tumorale alpha/génétique , Adolescent , Adulte , Numération des lymphocytes CD4 , Lymphocytes T CD4+/métabolisme , Infections à VIH/génétique , Infections à VIH/immunologie , Infections à VIH/virologie , Séropositivité VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/isolement et purification , Humains , Mâle , Adulte d'âge moyen , Récepteur au facteur de nécrose tumorale de type I/génétique , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Charge virale , Jeune adulte
9.
Intensive Care Med ; 46(12): 2265-2283, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-32654006

RÉSUMÉ

ARDS, first described in 1967, is the commonest form of acute severe hypoxemic respiratory failure. Despite considerable advances in our knowledge regarding the pathophysiology of ARDS, insights into the biologic mechanisms of lung injury and repair, and advances in supportive care, particularly ventilatory management, there remains no effective pharmacological therapy for this syndrome. Hospital mortality at 40% remains unacceptably high underlining the need to continue to develop and test therapies for this devastating clinical condition. The purpose of the review is to critically appraise the current status of promising emerging pharmacological therapies for patients with ARDS and potential impact of these and other emerging therapies for COVID-19-induced ARDS. We focus on drugs that: (1) modulate the immune response, both via pleiotropic mechanisms and via specific pathway blockade effects, (2) modify epithelial and channel function, (3) target endothelial and vascular dysfunction, (4) have anticoagulant effects, and (5) enhance ARDS resolution. We also critically assess drugs that demonstrate potential in emerging reports from clinical studies in patients with COVID-19-induced ARDS. Several therapies show promise in earlier and later phase clinical testing, while a growing pipeline of therapies is in preclinical testing. The history of unsuccessful clinical trials of promising therapies underlines the challenges to successful translation. Given this, attention has been focused on the potential to identify biologically homogenous subtypes within ARDS, to enable us to target more specific therapies 'precision medicines.' It is hoped that the substantial number of studies globally investigating potential therapies for COVID-19 will lead to the rapid identification of effective therapies to reduce the mortality and morbidity of this devastating form of ARDS.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Traitement médicamenteux/tendances , /traitement médicamenteux , Antioxydants/usage thérapeutique , Acide ascorbique/usage thérapeutique , Citrulline/usage thérapeutique , Glycoprotéines/usage thérapeutique , Humains , Cellules souches mésenchymateuses , Pandémies , Peptides cycliques/usage thérapeutique , Pyridones/usage thérapeutique , Pyrimidines/usage thérapeutique , Récepteur au facteur de nécrose tumorale de type I/antagonistes et inhibiteurs , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Stéroïdes/usage thérapeutique , Inhibiteurs trypsiques/usage thérapeutique
10.
Brain Behav Immun ; 41: 144-51, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-24880032

RÉSUMÉ

The mechanisms of diabetic painful neuropathy are complicated and comprise of peripheral and central pathophysiological phenomena. A number of proinflammatory cytokines are involved in this process. Tumor necrosis factor α (TNF-α) is considered to be one of the major contributors of neuropathic pain. In order to explore the potential role of inflammation in the peripheral nervous system of Type 1 diabetic animals with painful neuropathy, we investigated whether TNF-α is a key inflammatory mediator to the diabetic neuropathic pain and whether continuous delivery of TNFα soluble receptor from damaged axons achieved by HSV vector mediated transduction of DRG would block or alter the pain perception in animals with diabetic neuropathy. Diabetic animals exhibited changes in threshold of mechanical and thermal pain perception compared to control rats and also demonstrated increases in TNFα in the DRG, spinal cord dorsal horn, sciatic nerve and in the foot skin, 6 weeks after the onset of diabetes. Therapeutic approaches by HSV mediated expression of p55 TNF soluble receptor significantly attenuated the diabetes-induced hyperalgesia and decreased the expression of TNFα with reduction in the phosphorylation of p38MAPK in the spinal cord dorsal horn and DRG. The overall outcome of this study suggests that neuroinflammatory activation in the peripheral nervous system may be involved in the pathogenesis of painful neuropathy in Type 1 diabetes which can be alleviated by local expression of HSV vector expressing p55 TNF soluble receptor.


Sujet(s)
Diabète expérimental/complications , Neuropathies diabétiques/thérapie , Thérapie génétique , Vecteurs génétiques/usage thérapeutique , Hyperalgésie/thérapie , Névralgie/thérapie , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Récepteurs leurres aux facteurs de nécrose tumorale/usage thérapeutique , Animaux , Cellules cultivées , Neuropathies diabétiques/immunologie , Neuropathies diabétiques/physiopathologie , Pied , Ganglions sensitifs des nerfs spinaux/métabolisme , Vecteurs génétiques/génétique , Température élevée/effets indésirables , Hyperalgésie/étiologie , Hyperalgésie/immunologie , Hyperalgésie/physiopathologie , Inflammation , Mâle , Névralgie/étiologie , Névralgie/immunologie , Névralgie/physiopathologie , Seuil nociceptif , Cellules de la corne dorsale/métabolisme , Pression/effets indésirables , Maturation post-traductionnelle des protéines , Rats , Rat Sprague-Dawley , Récepteur au facteur de nécrose tumorale de type I/génétique , Nerf ischiatique/métabolisme , Simplexvirus/génétique , Méthode en simple aveugle , Peau/métabolisme , Transduction génétique , Récepteurs leurres aux facteurs de nécrose tumorale/génétique , p38 Mitogen-Activated Protein Kinases/métabolisme
11.
J Neuroinflammation ; 10: 69, 2013 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-23735240

RÉSUMÉ

BACKGROUND: Tumor necrosis factor-α (TNF-α) is an important inflammatory factor produced by activated macrophages and monocytes and plays an important role in the pathogenesis of diabetic peripheral neuropathy (DPN). To evaluate the effect of TNF-α signaling suppression and the potential of TNF-α in the treatment of DPN, a recombinant human TNF-α receptor-antibody fusion protein (rhTNFR:Fc) was used. We focused on the pathophysiology of the sciatic nerve and examined the expression of myelin basic protein (MBP) under DPN status with or without TNF-α inhibition. METHODS: The DPN rat model was generated by intraperitoneal injection of streptozotocin and by feeding with a high-fat, high-sugar diet. The nerve conduction velocity (NCV) in sciatic nerve of rat was monitored over a period of four weeks. The histopathological changes in nerve tissue were examined through traditional tissue histology and ultrastructure transmission electron microscopy (TEM). The expression of MBP was examined through western blot analysis. RESULTS: The DPN induced rats showed significant signs of nerve damage including lower NCV, demyelination of nerve fibers, disorganization of lamellar and axonal structures, and decreased expression of MBP in the nerve tissue. The inhibition of TNF-α in the DPN rats resulted in a significant recovery from those symptoms compared to the DPN rats. CONCLUSIONS: Our study demonstrates that TNF-α plays a key role in the pathogenesis of DPN and its inhibition by rhTNFR:Fc can prove to be a useful therapeutic strategy for the treatment of and/or prevention from DPN symptoms.


Sujet(s)
Neuropathies diabétiques/traitement médicamenteux , Neuropathies périphériques/traitement médicamenteux , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Animaux , Axones/anatomopathologie , Technique de Western , Diabète expérimental/complications , Diabète expérimental/traitement médicamenteux , Diabète expérimental/anatomopathologie , Humains , Immunohistochimie , Mâle , Microscopie électronique à transmission , Motoneurones/effets des médicaments et des substances chimiques , Protéine basique de la myéline/biosynthèse , Neurofibres/anatomopathologie , Conduction nerveuse/physiologie , Rats , Rat Wistar , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Protéines de fusion recombinantes/usage thérapeutique , Nerf ischiatique/anatomopathologie , Nerf ischiatique/ultrastructure , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques
12.
Diabetes ; 61(8): 2134-45, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22688336

RÉSUMÉ

Diabetic bladder dysfunction (DBD) is common and affects 80% of diabetic patients. However, the molecular mechanisms underlying DBD remain elusive because of a lack of appropriate animal models. We demonstrate DBD in a mouse model that harbors hepatic-specific insulin receptor substrate 1 and 2 deletions (double knockout [DKO]), which develops type 2 diabetes. Bladders of DKO animals exhibited detrusor overactivity at an early stage: increased frequency of nonvoiding contractions during bladder filling, decreased voided volume, and dispersed urine spot patterns. In contrast, older animals with diabetes exhibited detrusor hypoactivity, findings consistent with clinical features of diabetes in humans. The tumor necrosis factor (TNF) superfamily genes were upregulated in DKO bladders. In particular, TNF-α was upregulated in serum and in bladder smooth muscle tissue. TNF-α augmented the contraction of primary cultured bladder smooth muscle cells through upregulating Rho kinase activity and phosphorylating myosin light chain. Systemic treatment of DKO animals with soluble TNF receptor 1 (TNFRI) prevented upregulation of Rho A signaling and reversed the bladder dysfunction, without affecting hyperglycemia. TNFRI combined with the antidiabetic agent, metformin, improved DBD beyond that achieved with metformin alone, suggesting that therapies targeting TNF-α may have utility in reversing the secondary urologic complications of type 2 diabetes.


Sujet(s)
Diabète de type 2/physiopathologie , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Maladies de la vessie/traitement médicamenteux , Vessie hyperactive/traitement médicamenteux , Animaux , Modèles animaux de maladie humaine , Substrats du récepteur à l'insuline/génétique , Metformine/administration et posologie , Souris , Souris knockout , Contraction musculaire/effets des médicaments et des substances chimiques , Contraction musculaire/physiologie , Chaînes légères de myosine/métabolisme , Facteur de nécrose tumorale alpha/sang , Facteur de nécrose tumorale alpha/pharmacologie , Régulation positive/effets des médicaments et des substances chimiques , Vessie urinaire/physiopathologie , rho-Associated Kinases/biosynthèse
13.
Arthroscopy ; 27(10): 1351-7, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21705172

RÉSUMÉ

PURPOSE: The purpose was to determine whether systemic tumor necrosis factor α (TNF-α) blockade can improve rotator cuff healing in a rat model. METHODS: One hundred twenty Lewis rats underwent unilateral detachment and repair of the supraspinatus. Rats were randomized into 2 groups. The experimental group received injections of pegylated soluble tumor necrosis factor receptor type I (3.0 mg/kg every other day for 3 doses). The control group received saline solution on the same dosing schedule. At 2, 4, and 8 weeks, 20 animals in each group were killed (4 for histologic assessment and 16 for biomechanical testing). Outcomes included qualitative histologic assessment to determine new fibrocartilage formation and collagen fiber organization. Immunohistochemical staining was performed to localize TNF-α, ED1 and ED2 macrophages, and tartrate-resistant acidic phosphatase. Biomechanical testing was performed to determine the ultimate load to failure, stiffness, cross-sectional area, and ultimate stress to failure. RESULTS: Qualitative assessments of histology showed that the experimental group had more cartilage formation at 4 weeks but not at 2 or 8 weeks. There was less TNF-α staining in the experimental group at 4 and 8 weeks, and there were fewer ED1 macrophages at 4 weeks compared with controls. The ultimate load to failure was greater in the experimental group compared with controls at 2 weeks (13.3 ± 2.6 N v 11.2 ± 2.7 N, P = .05) and at 4 weeks (21.7 ± 4.6 N v 18.5 ± 2.1 N, P = .04). The experimental group also had a higher stiffness at 2 weeks (7.2 ± 2.3 N/mm v 5.8 ± 1.4 N/mm, P = .04) and at 4 weeks (10.5 ± 2.7 N/mm v 8.4 ± 1.7 N/mm, P = .01). There were no differences in any biomechanical variable at 8 weeks. CONCLUSIONS: TNF-α blockade can improve the biomechanical strength of tendon-bone healing in a rat rotator cuff model at early time points, which corresponded with modest qualitative improvements in histology. However, these differences were not maintained at 8 weeks. CLINICAL RELEVANCE: TNF-α blockade may influence rotator cuff tendon healing.


Sujet(s)
Polyéthylène glycols/usage thérapeutique , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Coiffe des rotateurs/chirurgie , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Animaux , Phénomènes biomécaniques , Cicatrice/prévention et contrôle , Évaluation préclinique de médicament , Ectodysplasines/analyse , Fibrocartilage/physiologie , Humérus/anatomopathologie , Macrophages/composition chimique , Macrophages/anatomopathologie , Polyéthylène glycols/administration et posologie , Répartition aléatoire , Rats , Rats de lignée LEW , Récepteur au facteur de nécrose tumorale de type I/administration et posologie , Régénération/effets des médicaments et des substances chimiques , Coiffe des rotateurs/anatomopathologie , Méthode en simple aveugle , Techniques de suture , Tendons/anatomopathologie , Facteur de nécrose tumorale alpha/analyse
14.
Curr Med Res Opin ; 26(10): 2287-300, 2010 Oct.
Article de Anglais | MEDLINE | ID: mdl-20718590

RÉSUMÉ

BACKGROUND: Tumor necrosis factor (TNF)-α plays a critical role in psoriasis pathogenesis, and several anti-TNF agents have been developed as therapeutic drugs in this indication. SCOPE: To present the preclinical rationale and clinical data for onercept, a novel anti-TNF agent developed for the treatment of moderate-to-severe psoriasis, and to critically evaluate the onercept clinical development program. FINDINGS: Onercept was shown in preclinical studies to inhibit TNF-α and suppress clinical signs in several inflammatory conditions. In phase II studies onercept demonstrated a therapeutic benefit in psoriasis and psoriatic arthritis and no safety issues were identified. Based on these results, a phase III program comprising three multicenter, randomized, double-blind, placebo-controlled studies examining onercept in moderate-to-severe plaque psoriasis was initiated. Following the occurrence of two cases of systemic inflammatory response syndrome (SIRS) and lower than expected efficacy results, an independent Data Safety Monitoring Board (DSMB) determined that the risk-benefit ratio was not sufficiently favorable to justify continued development, and all clinical studies were promptly terminated. Although not initially diagnosed as such by the investigators, two further SIRS events were reported, one after study discontinuation. Although an increased incidence of infection and sepsis-like events has been associated with other anti-TNF therapies, an increased risk of infection was not observed with onercept treatment. Moreover, no infectious etiology was determined in the SIRS cases. The data suggest that the SIRS reactions were due to a systemic inflammatory response. CONCLUSIONS: Despite promising early clinical results, onercept showed many of the expected risks associated with other anti-TNF agents and proved not to have an exceptional efficacy and safety profile. The clinical development of onercept highlights the critical importance of DSMBs and closely monitoring patient safety and evaluating risk-benefit profiles in large clinical programs.


Sujet(s)
Psoriasis/traitement médicamenteux , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Récepteurs leurres aux facteurs de nécrose tumorale/usage thérapeutique , Animaux , Essais cliniques de phase I comme sujet , Essais cliniques de phase II comme sujet , Découverte de médicament , Antihormones/effets indésirables , Antihormones/synthèse chimique , Antihormones/pharmacologie , Antihormones/usage thérapeutique , Humains , Liaison aux protéines , Essais contrôlés randomisés comme sujet , Récepteur au facteur de nécrose tumorale de type I/effets indésirables , Récepteur au facteur de nécrose tumorale de type I/synthèse chimique , Récepteur au facteur de nécrose tumorale de type I/pharmacologie , Syndrome de réponse inflammatoire généralisée/induit chimiquement , Syndrome de réponse inflammatoire généralisée/épidémiologie , Résultat thérapeutique , Récepteurs leurres aux facteurs de nécrose tumorale/effets indésirables , Récepteurs leurres aux facteurs de nécrose tumorale/synthèse chimique , Récepteurs leurres aux facteurs de nécrose tumorale/pharmacologie , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Facteur de nécrose tumorale alpha/métabolisme
15.
Zhonghua Yi Xue Za Zhi ; 90(11): 768-71, 2010 Mar 23.
Article de Chinois | MEDLINE | ID: mdl-20627024

RÉSUMÉ

OBJECTIVE: To construct a latent human soluble tumor necrosis factor receptor I (hsTNFRI) using the latency associated protein (LAP) of transforming growth factor-beta1 (TGF-beta1) fused via a matrix metalloproteinase (MMP) cleavage site to hsTNFRI so as to detect the latent biological activity of LAP-MMP-hsTNFRI fusion protein. METHODS: A double-stranded deoxyoligonucleotide coding for MMP cleavage site was cloned into plasmid pcDNA3.1. LAP and hsTNFRI cDNA were then inserted into both two sides of MMP cleavage site. After being transferred by LAP-MMP-hsTNFRI fusion gene with liposome, the expression of fusion protein in COS-7 cells was detected by RT-PCR and Western blot. The inhibitory effect of fusion protein upon cytotoxicity of TNF-alpha was detected by methyl thiazolyl tetrazolium (MTT) assay before and after the fusion protein incubated in MMP or peritoneal fluid from endometriosis patients. RESULTS: The recombinant plasmid LAP-MMP-hsTNFRI-pcDNA3.1 was constructed successfully and was expressed effectively in COS-7 cells. The MTT assay showed that there was no difference in the mortality rate of L929 cells between LAP-MMP-hsTNFRI-pcDNA3.1 and empty vector transfection groups (P > 0.05). The mortality rates of L929 cells with 800 ng/L TNF-alpha in LAP-MMP-hsTNFRI-pcDNA3.1 transfection group after incubation with MMP or peritoneal fluid from endometriosis patients were (44.5 +/- 2.4)% and (33.8 +/- 1.9)% respectively. And it was lower than the pre-incubation period (58.1 +/- 2.4)% (P < 0.05). CONCLUSION: The biological activity of LAP-MMP-hsTNFRI fusion protein can be made latent by LAP and activated by peritoneal fluid from endometriosis. Thus a new method has been provided for a targeted therapy of endometriosis.


Sujet(s)
Endométriose/thérapie , Récepteur au facteur de nécrose tumorale de type I/génétique , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Liquide d'ascite/cytologie , Lignée cellulaire , Femelle , Vecteurs génétiques , Humains , Plasmides , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/usage thérapeutique , Transfection
16.
Mol Cancer Ther ; 9(6): 1719-29, 2010 Jun.
Article de Anglais | MEDLINE | ID: mdl-20515949

RÉSUMÉ

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered an attractive anticancer agent due to its tumor cell-specific cytotoxicity. However, its low stability, solubility, unexpected side effects, and weak pharmacokinetic profiles restrict its successful clinical application. To develop efficient TRAIL-based anticancer biotherapeutics, a new version of trimeric TRAIL was constructed by incorporating trimer-forming zipper sequences (HZ-TRAIL), and then NH(2)-terminal-specific PEGylation was done to produce PEGylated TRAIL (PEG-HZ-TRAIL). The biological, physicochemical, and pharmaceutical characteristics of PEG-HZ-TRAIL were then investigated using various in vitro and in vivo experiments, including a cell-based cytotoxicity test, a solubility test, pharmacokinetic analysis, and antitumor efficacy evaluations. Although slight activity loss occurred after PEGylation, PEG-HZ-TRAIL showed excellent tumor cell-specific cytotoxic effects via apoptotic pathways with negligible normal cell toxicity. The stability and pharmacokinetic problems of HZ-TRAIL were successfully overcome by PEGylation. Furthermore, in vivo antitumor tests revealed that PEG-HZ-TRAIL treatment enhanced therapeutic potentials compared with HZ-TRAIL in tumor xenograft animal models, and these enhancements were attributed to its better pharmacokinetic properties and tumor-targeting performance. These findings show that PEG-HZ-TRAIL administration provides an effective antitumor treatment, which exhibits superior tumor targeting and better inhibits tumor growth, and suggest that PEG-HZ-TRAIL should be considered a potential candidate for antitumor biotherapy.


Sujet(s)
Tumeurs/traitement médicamenteux , Polyéthylène glycols/usage thérapeutique , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Ligand TRAIL/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Lignée cellulaire tumorale , Humains , Mâle , Souris , Imagerie moléculaire , Tumeurs/anatomopathologie , Polyéthylène glycols/composition chimique , Rats , Rat Sprague-Dawley , Récepteur au facteur de nécrose tumorale de type I/composition chimique , Ligand TRAIL/composition chimique , Ligand TRAIL/pharmacocinétique , Ligand TRAIL/pharmacologie , Distribution tissulaire/effets des médicaments et des substances chimiques
17.
Pharmacotherapy ; 30(4): 339-53, 2010 Apr.
Article de Anglais | MEDLINE | ID: mdl-20334454

RÉSUMÉ

STUDY OBJECTIVE: To evaluate the efficacy and safety of using the anti-tumor necrosis factor-alpha (anti-TNF-alpha) drugs adalimumab, etanercept, and infliximab for the treatment of rheumatoid arthritis. DESIGN: Systematic review and meta-analysis of 21 randomized, placebo-controlled trials (eight adalimumab, seven infliximab, six etanercept). PATIENTS: Adults with rheumatoid arthritis who received adalimumab (1524 patients), infliximab (1116 patients), etanercept (1029 patients), or placebo (2834 patients) with or without concomitant methotrexate in all groups. MEASUREMENTS AND MAIN RESULTS: A literature search of several databases from January 1995-December 2008 was performed. There were no restrictions based on language or date of publication, and low-quality studies (based on Jadad score) were excluded. American College of Rheumatology (ACR) 20% improvement criteria (ACR20), 50% improvement criteria (ACR50), and 70% improvement criteria (ACR70) were used to compare treatment efficacy. Safety was compared based on frequency of serious adverse events, serious infections, malignancy, and death. Withdrawals due to adverse events and lack of efficacy were also evaluated. With short-term treatment (12-30 wks), etanercept demonstrated the highest risk ratios (RRs) for reaching ACR20 and ACR50: 2.94 (95% confidence interval [CI] 2.27-3.81) and 5.28 (95% CI 3.12-8.92), respectively. Adalimumab demonstrated the highest RR for achieving ACR70 (5.36, 95% CI 3.76- 7.64). Over a long-term treatment course (1-3 yrs), adalimumab demonstrated the highest RRs (95% CIs) for these parameters: 1.85 (1.07-3.19), 2.80 (1.16-6.77), and 3.23 (1.37-7.61) for ACR20, ACR50, and ACR70, respectively. No statistically significant differences were noted in the safety of any of the three drugs compared with placebo. Infliximab had the highest RRs for withdrawing from the study due to lack of efficacy (2.05, 95% CI 1.33-3.16) and adverse events (0.41, 95% CI 0.18-0.95). CONCLUSION: With short-term treatment, etanercept and adalimumab had higher efficacy results; with long-term treatment, adalimumab appeared to be the most effective. Clinicians should be aware that each of the three drugs has different rates of efficacy and different safety considerations that must be taken into account when selecting the best treatment for an individual with rheumatoid arthritis.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Polyarthrite rhumatoïde/traitement médicamenteux , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Sécurité , Adalimumab , Adulte , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux humanisés , Étanercept , Humains , Immunoglobuline G/administration et posologie , Immunoglobuline G/usage thérapeutique , Infliximab , Essais contrôlés randomisés comme sujet , Récepteurs aux facteurs de nécrose tumorale/administration et posologie , Récepteurs aux facteurs de nécrose tumorale/usage thérapeutique , Résultat thérapeutique
18.
Minerva Ginecol ; 62(1): 17-31, 2010 Feb.
Article de Anglais | MEDLINE | ID: mdl-20186112

RÉSUMÉ

Endometriosis is an enigmatic, debilitating disease which affects as many as 15% of all women of reproductive age and is characterized by pelvic pain and infertility. Current treatment regimes used to manage the disease do so by inducing a hypoestrogenic state. While the absence of circulating estrogen levels lead to a regression of the disease, this hypoestrogenism also induces many unpleasant and unwanted side-effects. As such, these and other shortcomings of current drug therapies emphasize their limitations and the necessity for the development of novel endometriosis treatments. In this review, current therapies for medical management of endometriosis are discussed as are their shortcomings. Potential target areas which may be attractive alternatives to current therapies are also reviewed and include aromatase inhibitors, angiogenesis disruptors and anti-tumor necrosis factor-alpha inhibitors.


Sujet(s)
Endométriose/traitement médicamenteux , Adalimumab , Anastrozole , Inhibiteurs de l'angiogenèse/usage thérapeutique , Anti-inflammatoires non stéroïdiens/usage thérapeutique , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux humanisés , Inhibiteurs de l'aromatase/usage thérapeutique , Endométriose/chirurgie , Modulateurs des récepteurs des oestrogènes/effets indésirables , Modulateurs des récepteurs des oestrogènes/usage thérapeutique , Étanercept , Femelle , Hormone de libération des gonadotrophines/antagonistes et inhibiteurs , Antihormones/usage thérapeutique , Humains , Immunoglobuline G/usage thérapeutique , Infliximab , Isoxazoles/usage thérapeutique , Léflunomide , Létrozole , Nitriles/usage thérapeutique , Pentoxifylline/usage thérapeutique , Progestérone/antagonistes et inhibiteurs , Récepteurs aux facteurs de nécrose tumorale/usage thérapeutique , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Triazoles/usage thérapeutique , Récepteurs leurres aux facteurs de nécrose tumorale/usage thérapeutique , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs
19.
Ann Rheum Dis ; 69(4): 706-14, 2010 Apr.
Article de Anglais | MEDLINE | ID: mdl-19460761

RÉSUMÉ

OBJECTIVES: To evaluate synovial tissue and serum biomarkers of disease activity, therapeutic response and radiographic progression during biological therapy for rheumatoid arthritis (RA). METHODS: Patients with active RA entered a randomised study of anakinra 100 mg/day, administered as monotherapy or in combination with pegsunercept 800 microg/kg twice a week. Arthroscopic synovial tissue biopsies were obtained at baseline and two further time points. Following immunohistochemical staining, selected mediators of RA pathophysiology were quantified using digital image analysis. Selected mediators were also measured in the serum. RESULTS: Twenty-two patients were randomly assigned: 11 received monotherapy and 11 combination therapy. American College of Rheumatology 20, 50 and 70 response rates were 64%, 64% and 46% with combination therapy and 36%, 9% and 0% with monotherapy, respectively. In synovial tissue, T-cell infiltration, vascularity and transforming growth factor beta (TGFbeta) expression demonstrated significant utility as biomarkers of disease activity and therapeutic response. In serum, interleukin 6 (IL-6), matrix metalloproteinase (MMP) 1, MMP-3 and tissue inhibitor of metalloproteinase 1 (TIMP-1) were most useful in this regard. An early decrease in serum levels of TIMP-1 was predictive of the later therapeutic outcome. Pretreatment tissue levels of T-cell infiltration and the growth factors vascular endothelial growth factor/TGFbeta, and serum levels of IL-6, IL-8, MMP-1, TIMP-1, soluble tumour necrosis factor receptor types I and II and IL-18 correlated with radiographic progression. CONCLUSIONS: Synovial tissue analysis identified biomarkers of disease activity, therapeutic response and radiographic progression. Biomarker expression in tissue was independent of the levels measured in the serum.


Sujet(s)
Antirhumatismaux/usage thérapeutique , Polyarthrite rhumatoïde/traitement médicamenteux , Membrane synoviale/métabolisme , Antirhumatismaux/effets indésirables , Polyarthrite rhumatoïde/imagerie diagnostique , Polyarthrite rhumatoïde/immunologie , Arthroscopie , Marqueurs biologiques/sang , Marqueurs biologiques/métabolisme , Évolution de la maladie , Association de médicaments , Femelle , Humains , Antagoniste du récepteur à l'interleukine-1/effets indésirables , Antagoniste du récepteur à l'interleukine-1/usage thérapeutique , Mâle , Adulte d'âge moyen , Polyéthylène glycols/effets indésirables , Polyéthylène glycols/usage thérapeutique , Radiographie , Récepteur au facteur de nécrose tumorale de type I/effets indésirables , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Résultat thérapeutique , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs
20.
Ann Rheum Dis ; 69(9): 1687-90, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-19640853

RÉSUMÉ

BACKGROUND: Adiponectin is an anti-inflammatory and potentially antiatherogenic molecule. Some recent reports suggest that tumour necrosis factor alpha (TNFalpha) blockade therapy increases circulating adiponectin levels, but data are sparse and inconsistent. METHODS: Data from a double-blind placebo controlled study of onercept in 126 patients with psoriatic arthritis (PsA) and from pre- and post-adalimumab treatment in 171 patients with rheumatoid arthritis (RA) were used to examine the effect of TNFalpha blockade therapy on adiponectin. RESULTS: Despite expected associations of adiponectin with gender and baseline high-density lipoprotein cholesterol and triglyceride, adiponectin levels did not change over time with TNFalpha blockade therapy in either group. The mean+/-SD absolute change in adiponectin levels was -0.23+/-4.6 microg/ml in patients with PsA treated with combined onercept 50 mg and onercept 100 mg (vs placebo, p=0.60) and 0.28+/-3.23 microg/ml in patients with RA treated with adalimumab (vs baseline, p=0.66). CONCLUSION: These results do not support a significant effect of TNFalpha blockade therapy on circulating adiponectin levels in patients with autoimmune disease.


Sujet(s)
Adiponectine/sang , Antirhumatismaux/pharmacologie , Arthrite psoriasique/sang , Maladies auto-immunes/sang , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Adalimumab , Adulte , Sujet âgé , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux humanisés , Antirhumatismaux/usage thérapeutique , Arthrite psoriasique/complications , Arthrite psoriasique/traitement médicamenteux , Polyarthrite rhumatoïde/sang , Polyarthrite rhumatoïde/complications , Polyarthrite rhumatoïde/traitement médicamenteux , Maladies auto-immunes/complications , Maladies auto-immunes/traitement médicamenteux , Maladies cardiovasculaires/étiologie , Méthodes épidémiologiques , Femelle , Humains , Lipides/sang , Mâle , Adulte d'âge moyen , Récepteur au facteur de nécrose tumorale de type I/pharmacologie , Récepteur au facteur de nécrose tumorale de type I/usage thérapeutique , Récepteurs leurres aux facteurs de nécrose tumorale/pharmacologie , Récepteurs leurres aux facteurs de nécrose tumorale/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...