Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell ; 161(7): 1633-43, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26091040

RESUMEN

Lipid biology continues to emerge as an area of significant therapeutic interest, particularly as the result of an enhanced understanding of the wealth of signaling molecules with diverse physiological properties. This growth in knowledge is epitomized by lysophosphatidic acid (LPA), which functions through interactions with at least six cognate G protein-coupled receptors. Herein, we present three crystal structures of LPA1 in complex with antagonist tool compounds selected and designed through structural and stability analyses. Structural analysis combined with molecular dynamics identified a basis for ligand access to the LPA1 binding pocket from the extracellular space contrasting with the proposed access for the sphingosine 1-phosphate receptor. Characteristics of the LPA1 binding pocket raise the possibility of promiscuous ligand recognition of phosphorylated endocannabinoids. Cell-based assays confirmed this hypothesis, linking the distinct receptor systems through metabolically related ligands with potential functional and therapeutic implications for treatment of disease.


Asunto(s)
Cristalografía por Rayos X , Sitios de Unión , Cromatografía en Gel , Humanos , Ligandos , Modelos Moleculares , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/química , Bibliotecas de Moléculas Pequeñas
2.
Adv Exp Med Biol ; 1274: 223-258, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32894513

RESUMEN

G protein-coupled receptors (GPCRs) have seven transmembrane spanning domains and comprise the largest superfamily with ~800 receptors in humans. GPCRs are attractive targets for drug discovery because they transduce intracellular signaling in response to endogenous ligands via heterotrimeric G proteins or arrestins, resulting in a wide variety of physiological and pathophysiological responses. The endogenous ligands for GPCRs are highly chemically diverse and include ions, biogenic amines, nucleotides, peptides, and lipids. In this review, we follow the KonMari method to better understand druggable lipid GPCRs. First, we have a comprehensive tidying up of lipid GPCRs including receptors for prostanoids, leukotrienes, specialized pro-resolving mediators (SPMs), lysophospholipids, sphingosine 1-phosphate (S1P), cannabinoids, platelet-activating factor (PAF), free fatty acids (FFAs), and sterols. This tidying up consolidates 46 lipid GPCRs and declutters several perplexing lipid GPCRs. Then, we further tidy up the lipid GPCR-directed drugs from the literature and databases, which identified 24 clinical drugs targeting 16 unique lipid GPCRs available in the market and 44 drugs under evaluation in more than 100 clinical trials as of 2019. Finally, we introduce drug designs for GPCRs that spark joy, such as positive or negative allosteric modulators (PAM or NAM), biased agonism, functional antagonism like fingolimod, and monoclonal antibodies (MAbs). These strategic drug designs may increase the efficacy and specificity of drugs and reduce side effects. Technological advances will help to discover more endogenous lipid ligands from the vast number of remaining orphan GPCRs and will also lead to the development novel lipid GPCR drugs to treat various diseases.


Asunto(s)
Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Arrestinas/metabolismo , Enfermedad , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Ligandos , Lípidos/farmacología , Lípidos/uso terapéutico , Receptores Acoplados a Proteínas G/metabolismo
3.
J Lipid Res ; 60(1): 212-217, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30463988

RESUMEN

Lysophosphatidic acid (LPA) activates cognate G protein-coupled receptors (GPCRs) to initiate biological signaling cascades. Lysophospholipid (LP) receptor binding properties remain incompletely assessed because of difficulties with ligand lipophilicity and lipid "stickiness." These inherent attributes produce high levels of nonspecific binding within cell-membrane preparations used to assess GPCRs, as has been shown in classical binding assays using radiolabeled ligands, making accurate measurements of lipid binding kinetics difficult to achieve. Backscattering interferometry (BSI) is an optical technology that measures molecular binding interactions by reporting changes in the refractive index of a solution after binding events. Here, we report the use of BSI to assess LPA1 for its ability to bind to naturally occurring lipids and a synthetic LPA1 antagonist (ONO-9780307), under both primary- and competition-binding conditions. Assessment of 12 different lipids demonstrated that the known LP ligand, 1-oleoyl-LPA, as well as an endocannabinoid metabolite, anandamide phosphate, are specific ligands for LPA1, whereas other LPs tested were not. Newly determined dissociation constants (Kd values) for orthosteric lipid ligands approximated 10-9 M, substantially lower (i.e., with higher affinity) than measured Kd values in classical binding or cell-based assays. These results demonstrate that BSI may have particular utility in assessing binding interactions between lipid receptors and their lipid ligands and could provide new screening approaches for lipid receptor identification and drug discovery.


Asunto(s)
Interferometría/métodos , Luz , Lisofosfolípidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Unión Competitiva , Línea Celular , Ligandos , Unión Proteica , Dispersión de Radiación , Especificidad por Sustrato
4.
Exp Cell Res ; 333(2): 171-177, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25499971

RESUMEN

Lysophospholipids (LPs), including lysophosphatidic acid (LPA), sphingosine 1-phospate (S1P), lysophosphatidylinositol (LPI), and lysophosphatidylserine (LysoPS), are bioactive lipids that transduce signals through their specific cell-surface G protein-coupled receptors, LPA1-6, S1P1-5, LPI1, and LysoPS1-3, respectively. These LPs and their receptors have been implicated in both physiological and pathophysiological processes such as autoimmune diseases, neurodegenerative diseases, fibrosis, pain, cancer, inflammation, metabolic syndrome, bone formation, fertility, organismal development, and other effects on most organ systems. Advances in the LP receptor field have enabled the development of novel small molecules targeting LP receptors for several diseases. Most notably, fingolimod (FTY720, Gilenya, Novartis), an S1P receptor modulator, became the first FDA-approved medicine as an orally bioavailable drug for treating relapsing forms of multiple sclerosis. This success is currently being followed by multiple, mechanistically related compounds targeting S1P receptor subtypes, which are in various stages of clinical development. In addition, an LPA1 antagonist, BMS-986020 (Bristol-Myers Squibb), is in Phase 2 clinical development for treating idiopathic pulmonary fibrosis, as a distinct compound, SAR100842 (Sanofi) for the treatment of systemic sclerosis and related fibrotic diseases. This review summarizes the current state of drug discovery in the LP receptor field.


Asunto(s)
Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Animales , Descubrimiento de Drogas , Humanos , Lisofosfolípidos/fisiología , Terapia Molecular Dirigida , Esclerosis Múltiple/tratamiento farmacológico , Receptores del Ácido Lisofosfatídico/fisiología , Receptores de Lisoesfingolípidos/fisiología , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/fisiología
5.
Pharmacology ; 92(1-2): 99-107, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23949095

RESUMEN

Ten mammalian diacylglycerol kinase (DGK) isozymes (α-κ) have been identified. Recent studies have revealed that DGK isozymes play pivotal roles in a wide variety of pathophysiological functions. Thus, it is important to be able to easily check DGK activity in each pathophysiological event. Moreover, the conventional DGK assay is quite laborious because it requires the use of a radioisotope and thin-layer chromatography including multiple extraction steps. In order to minimize the laborious procedures, we established a non-radioactive, single well, two-step DGK assay system. We demonstrated that, compared to the conventional method, the new assay system has comparable sensitivity and much higher efficiency, and is effective in detecting potential agents with high reliability (Z'-factor = 0.69 ± 0.12; n = 3). Using the newly developed assay, we comprehensively evaluated the DGK isozyme selectivities of commercially available DGK inhibitors, R59022 and R59949, in vitro. We found that among 10 isozymes, R59022 strongly inhibited type I DGKα and moderately attenuated type III DGKε and type V DGKθ, and that R59949 strongly inhibited type I DGK α and γ, and moderately attenuated type II DGK δ and κ.


Asunto(s)
Diacilglicerol Quinasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Piperidinas/farmacología , Pirimidinonas/farmacología , Quinazolinonas/farmacología , Tiazoles/farmacología , Animales , Células COS , Chlorocebus aethiops , Isoenzimas/antagonistas & inhibidores
6.
Bioorg Med Chem Lett ; 22(1): 144-8, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22153936

RESUMEN

Structure-activity relationship (SAR) of sphingosine-1-phosphate receptor agonists with a dihydronaphthalene scaffold was investigated. Compound 1 was modified to improve S1P(1) agonistic activity and in vivo peripheral lymphocyte lowering (PLL) activity without impairing selectivity over S1P(3) agonistic activity. A detailed SAR study of the terminal lipophilic part revealed that the introduction of substituents on the propylene linker and the terminal benzene ring influences in vitro and PLL activities. Compound 6n bearing a (S)-methyl group at the 2-position on the propylene linker and chlorine at the para-position on the terminal benzene ring showed potent hS1P(1) agonistic activity with excellent selectivity over hS1P(3) and in vivo PLL activity in mice.


Asunto(s)
Química Farmacéutica/métodos , Lisofosfolípidos/antagonistas & inhibidores , Naftalenos/química , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Esfingosina/análogos & derivados , Administración Oral , Animales , Benceno/química , Cloro/química , Diseño de Fármacos , Humanos , Ligandos , Ratones , Modelos Químicos , Ratas , Esfingosina/antagonistas & inhibidores , Relación Estructura-Actividad
7.
Bioorg Med Chem Lett ; 21(5): 1390-3, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21295477

RESUMEN

Structure-activity relationship of sphingosine-1-phosphate receptor agonist was examined. In terms of reducing the flexibility of molecule, hit compound 1 was modified to improve S1P(1) agonistic activity as well as selectivity over S1P(3) agonistic activity. Novel S1P agonists with cinnamyl scaffold or 1,2,5,6-tetrahydropyridine scaffold were identified.


Asunto(s)
Cinamatos/síntesis química , Receptores de Lisoesfingolípidos/agonistas , beta-Alanina/síntesis química , Animales , Cinamatos/química , Clorhidrato de Fingolimod , Glicoles de Propileno/química , Glicoles de Propileno/farmacología , Ratas , Esfingosina/análogos & derivados , Esfingosina/química , Esfingosina/farmacología , Relación Estructura-Actividad , beta-Alanina/química
8.
Bioorg Med Chem Lett ; 21(13): 3885-9, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21641216

RESUMEN

Structure-activity relationship of sphingosine-1-phosphate receptor agonists was examined. Cinnamyl derivative 1 was modified to improve S1P(1) agonistic activity as well as selectivity over S1P(3) agonistic activity. Dihydronaphthalene derivative 10d was identified as a potent S1P(1) receptor agonist with high selectivity against S1P(3) and enhanced efficacy in lowering peripheral lymphocyte counts in mice.


Asunto(s)
Naftalenos/síntesis química , Propanoles/química , Receptores de Lisoesfingolípidos/agonistas , Administración Oral , Animales , Células CHO , Cricetinae , Cricetulus , Clorhidrato de Fingolimod , Humanos , Linfocitos/efectos de los fármacos , Ratones , Estructura Molecular , Naftalenos/administración & dosificación , Naftalenos/farmacología , Propanoles/administración & dosificación , Propanoles/farmacología , Glicoles de Propileno , Esfingosina/análogos & derivados , Relación Estructura-Actividad
9.
EBioMedicine ; 65: 103255, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33676899

RESUMEN

BACKGROUND: Antivirals are needed to combat the COVID-19 pandemic, which is caused by SARS-CoV-2. The clinically-proven protease inhibitor Camostat mesylate inhibits SARS-CoV-2 infection by blocking the virus-activating host cell protease TMPRSS2. However, antiviral activity of Camostat mesylate metabolites and potential viral resistance have not been analyzed. Moreover, antiviral activity of Camostat mesylate in human lung tissue remains to be demonstrated. METHODS: We used recombinant TMPRSS2, reporter particles bearing the spike protein of SARS-CoV-2 or authentic SARS-CoV-2 to assess inhibition of TMPRSS2 and viral entry, respectively, by Camostat mesylate and its metabolite GBPA. FINDINGS: We show that several TMPRSS2-related proteases activate SARS-CoV-2 and that two, TMPRSS11D and TMPRSS13, are robustly expressed in the upper respiratory tract. However, entry mediated by these proteases was blocked by Camostat mesylate. The Camostat metabolite GBPA inhibited recombinant TMPRSS2 with reduced efficiency as compared to Camostat mesylate. In contrast, both inhibitors exhibited similar antiviral activity and this correlated with the rapid conversion of Camostat mesylate into GBPA in the presence of serum. Finally, Camostat mesylate and GBPA blocked SARS-CoV-2 spread in human lung tissue ex vivo and the related protease inhibitor Nafamostat mesylate exerted augmented antiviral activity. INTERPRETATION: Our results suggest that SARS-CoV-2 can use TMPRSS2 and closely related proteases for spread in the upper respiratory tract and that spread in the human lung can be blocked by Camostat mesylate and its metabolite GBPA. FUNDING: NIH, Damon Runyon Foundation, ACS, NYCT, DFG, EU, Berlin Mathematics center MATH+, BMBF, Lower Saxony, Lundbeck Foundation, Novo Nordisk Foundation.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Ésteres/farmacología , Guanidinas/farmacología , Inhibidores de Proteasas/farmacología , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Células HEK293 , Humanos , Pulmón/patología , Pulmón/virología , Proteínas de la Membrana/biosíntesis , Simulación de Dinámica Molecular , Serina Endopeptidasas/biosíntesis , Serina Proteasas/biosíntesis , Células Vero , Activación Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
10.
bioRxiv ; 2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32793911

RESUMEN

Antiviral therapy is urgently needed to combat the coronavirus disease 2019 (COVID-19) pandemic, which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The protease inhibitor camostat mesylate inhibits SARS-CoV-2 infection of lung cells by blocking the virus-activating host cell protease TMPRSS2. Camostat mesylate has been approved for treatment of pancreatitis in Japan and is currently being repurposed for COVID-19 treatment. However, potential mechanisms of viral resistance as well as camostat mesylate metabolization and antiviral activity of metabolites are unclear. Here, we show that SARS-CoV-2 can employ TMPRSS2-related host cell proteases for activation and that several of them are expressed in viral target cells. However, entry mediated by these proteases was blocked by camostat mesylate. The camostat metabolite GBPA inhibited the activity of recombinant TMPRSS2 with reduced efficiency as compared to camostat mesylate and was rapidly generated in the presence of serum. Importantly, the infection experiments in which camostat mesylate was identified as a SARS-CoV-2 inhibitor involved preincubation of target cells with camostat mesylate in the presence of serum for 2 h and thus allowed conversion of camostat mesylate into GBPA. Indeed, when the antiviral activities of GBPA and camostat mesylate were compared in this setting, no major differences were identified. Our results indicate that use of TMPRSS2-related proteases for entry into target cells will not render SARS-CoV-2 camostat mesylate resistant. Moreover, the present and previous findings suggest that the peak concentrations of GBPA established after the clinically approved camostat mesylate dose (600 mg/day) will result in antiviral activity.

11.
J Med Chem ; 60(23): 9508-9530, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29120624

RESUMEN

The discovery of 1-({6-[(2-methoxy-4-propylbenzyl)oxy]-1-methyl-3,4-dihydronaphthalen-2-yl}methyl)azetidine-3-carboxylic acid 13n (ceralifimod, ONO-4641), a sphingosine-1-phosphate (S1P) receptor agonist selective for S1P1 and S1P5, is described. While it has been revealed that the modulation of the S1P1 receptor is an effective way to treat autoimmune diseases such as relapsing-remitting multiple sclerosis (RRMS), it was also reported that activation of the S1P3 receptor is implicated in some undesirable effects. We carried out a structure-activity relationship (SAR) study of hit compound 6 with an amino acid moiety in the hydrophilic head region. Following identification of a lead compound with a dihydronaphthalene central core by inducing conformational constraint, optimization of the lipophilic tail region led to the discovery of 13n as a clinical candidate that exhibited >30 000-fold selectivity for S1P1 over S1P3 and was potent in a peripheral lymphocyte lowering (PLL) test in mice (ED50 = 0.029 mg/kg, 24 h after oral dosing).


Asunto(s)
Azetidinas/farmacología , Linfocitos/efectos de los fármacos , Naftalenos/farmacología , Receptores de Lisoesfingolípidos/agonistas , Administración Oral , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Azetidinas/administración & dosificación , Azetidinas/química , Azetidinas/farmacocinética , Células CHO , Cricetulus , Femenino , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos BALB C , Naftalenos/administración & dosificación , Naftalenos/química , Naftalenos/farmacocinética , Ratas Endogámicas Lew , Ratas Sprague-Dawley
12.
PLoS One ; 9(4): e93230, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24747415

RESUMEN

Autotaxin, also known as ectonucleotide pyrophosphatase/phosphodiesterase 2 (ENPP2), is a secreted enzyme that has lysophospholipase D activity, which converts lysophosphatidylcholine to bioactive lysophosphatidic acid. Lysophosphatidic acid activates at least six G-protein coupled recpetors, which promote cell proliferation, survival, migration and muscle contraction. These physiological effects become dysfunctional in the pathology of cancer, fibrosis, and pain. To date, several autotaxin/ENPP2 inhibitors have been reported; however, none were able to completely and continuously inhibit autotaxin/ENPP2 in vivo. In this study, we report the discovery of a highly potent autotaxin/ENPP2 inhibitor, ONO-8430506, which decreased plasma lysophosphatidic acid formation. The IC50 values of ONO-8540506 for lysophospholipase D activity were 6.4-19 nM for recombinant autotaxin/ENPP2 proteins and 4.7-11.6 nM for plasma from various animal species. Plasma lysophosphatidic acid formation during 1-h incubation was almost completely inhibited by the addition of >300 nM of the compound to human plasma. In addition, when administered orally to rats at a dose of 30 mg/kg, the compound demonstrated good pharmacokinetics in rats and persistently inhibited plasma lysophosphatidic acid formation even at 24 h after administration. Smooth muscle contraction is a known to be promoted by lysophosphatidic acid. In this study, we showed that dosing rats with ONO-8430506 decreased intraurethral pressure accompanied by urethral relaxation. These findings demonstrate the potential of this autotaxin/ENPP2 inhibitor for the treatment of various diseases caused by lysophosphatidic acid, including urethral obstructive disease such as benign prostatic hyperplasia.


Asunto(s)
Carbolinas/farmacología , Inhibidores Enzimáticos/farmacología , Lisofosfolípidos/biosíntesis , Lisofosfolípidos/sangre , Hidrolasas Diéster Fosfóricas/metabolismo , Uretra/efectos de los fármacos , Uretra/fisiología , Animales , Carbolinas/farmacocinética , Inhibidores Enzimáticos/farmacocinética , Femenino , Humanos , Concentración 50 Inhibidora , Masculino , Ratones , Relajación Muscular/efectos de los fármacos , Presión , Ratas , Factores de Tiempo
13.
Transpl Immunol ; 26(2-3): 163-70, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22210496

RESUMEN

BACKGROUND: In small bowel transplantation (SBTx), inhibition of both graft-versus-host disease (GVHD) and allograft rejection is necessary. METHODS: We investigated the potency of a new sphingosine-1-phosphate receptor agonist, W-061, for these two immune responses in SBTx. W-061 has a completely different molecular structure from FTY720. Heterotopic SBTx was performed from Wistar-Furth (WF) into (WF×ACI) F1 rats as a GVHD model or F1 to WF rats as a rejection model. Recipients were orally given 3 mg/kg/day W-061 for 14 days after SBTx. Recipient survival, body weight, histopathology, lymphocyte subpopulations, and the cytokine profile were evaluated. RESULTS: W-061 treatment significantly prolonged graft survival over 100 days in four out of six recipients in the GVHD group and over 60 days in three out of six recipients in the rejection group. W-061 strongly inhibited GVHD and rejection as seen histopathologically in comparison with untreated control rats. W-061 caused a significant reduction in donor-derived T cells in target organs and infiltrating T cells in allografts by promoting these cells to home into the secondary lymphoid tissues and sequestrating those cells there. W-061 significantly decreased production of interferon-γ in target organs and allografts. CONCLUSION: Therefore, these data suggest that W-061 has considerable potential as a new therapeutic immunosuppressant in patients with SBTx.


Asunto(s)
Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/farmacología , Intestino Delgado/trasplante , Receptores de Lisoesfingolípidos/agonistas , Animales , Células CHO , Cricetinae , Cricetulus , Citocinas , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Humanos , Intestino Delgado/inmunología , Glicoles de Propileno/farmacología , Ratas , Ratas Wistar , Esfingosina/análogos & derivados , Esfingosina/farmacología , Linfocitos T , Factores de Tiempo , Trasplante Homólogo
14.
PLoS One ; 6(9): e23933, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931623

RESUMEN

Although IL-17 is a pro-inflammatory cytokine reportedly involved in various autoimmune inflammatory disorders, its role remains unclear in murine models of colitis. Acute colitis was induced by 2.5% dextran sodium sulfate (DSS) treatment for 5 days. A novel sphingosine-1-phosphate receptor agonist W-061, a prototype of ONO-4641, was orally administered daily, and histopathological analysis was performed on the colon. The number of lymphocytes and their cytokine production were also evaluated in spleen, mesenteric lymph node, Peyer's patch and lamina propria of the colon. Daily administration of W-061 resulted in improvement of DSS-induced colitis, and significantly reduced the number of CD4+ T cells in the colonic lamina propria. Numbers of both Th17 and Th1 cells were reduced by W-061 treatment. W-061, however, had no influence on the number of Treg cells in lamina propria. Thus, Th17 and Th1 cells in lamina propria were thought to be the key subsets in the pathogenesis of DSS-induced colitis. In conclusion, W-061 may be a novel therapeutic strategy to ameliorate acute aggravation of inflammatory bowel diseases.


Asunto(s)
Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Sulfato de Dextran/efectos adversos , Receptores de Lisoesfingolípidos/agonistas , Animales , Peso Corporal/efectos de los fármacos , Células CHO , Movimiento Celular/efectos de los fármacos , Colitis/inmunología , Colitis/patología , Colon/efectos de los fármacos , Colon/patología , Cricetinae , Cricetulus , Esquema de Medicación , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Membrana Mucosa/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Especificidad por Sustrato , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/citología , Células Th17/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA