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










Base de datos
Intervalo de año de publicación
1.
Vaccines (Basel) ; 9(8)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34451996

RESUMEN

Chlamydia trachomatis causes most bacterial sexually transmitted diseases worldwide. Different major outer membrane proteins (MOMPs) define various serovars of this intracellular pathogen: In women, D to L3 can cause urethritis, cervicitis, salpingitis, and oophoritis, and, thus, infertility. Protective immunity might be serovar-specific since chlamydial infection does not appear to induce an effective acquired immunity and reinfections occur. A better understanding of induced cross-serovar protection is essential for the selection of suitable antigens in vaccine development. In our mouse lung infection screening model, we evaluated the urogenital serovars D, E, and L2 in this regard. Seven weeks after primary infection or mock-infection, respectively, mice were infected a second time with the identical or one of the other serovars. Body weight and clinical score were monitored for 7 days. Near the peak of the second lung infection, bacterial load, myeloperoxidase, IFN-γ, and TNF-α in lung homogenate, as well as chlamydia-specific IgG levels in blood were determined. Surprisingly, compared with mice that were infected then for the first time, almost independent of the serovar combination used, all acquired parameters of disease were similarly diminished. Our reinfection study suggests that efficient cross-serovar protection could be achieved by a vaccine combining chlamydial antigens that do not include nonconserved MOMP regions.

2.
Vaccines (Basel) ; 9(6)2021 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-34204170

RESUMEN

Chlamydia trachomatis is the most frequent sexually-transmitted disease-causing bacterium. Urogenital serovars of this intracellular pathogen lead to urethritis and cervicitis. Ascending infections result in pelvic inflammatory disease, salpingitis, and oophoritis. One of 200 urogenital infections leads to tubal infertility. Serovars A-C cause trachoma with visual impairment. There is an urgent need for a vaccine. We characterized a new five-component subunit vaccine in a mouse vaccination-lung challenge infection model. Four recombinant Pmp family-members and Ctad1 from C. trachomatis serovar E, all of which participate in adhesion and binding of chlamydial elementary bodies to host cells, were combined with the mucosal adjuvant cyclic-di-adenosine monophosphate. Intranasal application led to a high degree of cross-serovar protection against urogenital and ocular strains of C. trachomatis, which lasted at least five months. Critical evaluated parameters were body weight, clinical score, chlamydial load, a granulocyte marker and the cytokines IFN-γ/TNF-α in lung homogenate. Vaccine antigen-specific antibodies and a mixed Th1/Th2/Th17 T cell response with multi-functional CD4+ and CD8+ T cells correlate with protection. However, serum-transfer did not protect the recipients suggesting that circulating antibodies play only a minor role. In the long run, our new vaccine might help to prevent the feared consequences of human C. trachomatis infections.

3.
Front Immunol ; 12: 580594, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33767691

RESUMEN

The zoonotic intracellular bacterium Chlamydia psittaci causes life-threatening pneumonia in humans. During mouse lung infection, complement factor C3 and the anaphylatoxin C3a augment protection against C. psittaci by a so far unknown mechanism. To clarify how complement contributes to the early, innate and the late, specific immune response and resulting protection, this study addresses the amount of C3, the timing when its presence is required as well as the anaphylatoxin receptor(s) mediating its effects and the complement-dependent migration of dendritic cells. Challenge experiments with C. psittaci on various complement KO mice were combined with transient decomplementation by pharmacological treatment, as well as the analysis of in vivo dendritic cells migration. Our findings reveal that a plasma concentration of C3 close to wildtype levels was required to achieve full protection. The diminished levels of C3 of heterozygote C3+/- mice permitted already relative effective protection and improved survival as compared to C3-/- mice, but overall recovery of these animals was delayed. Complement was in particular required during the first days of infection. However, additionally, it seems to support protection at later stages. Migration of CD103+ dendritic cells from the infected lung to the draining lymph node-as prerequisite of antigen presentation-depended on C3 and C3aR and/or C5aR. Our results provide unique mechanistic insight in various aspects of complement-dependent immune responses under almost identical, rather physiological experimental conditions. Our study contributes to an improved understanding of the role of complement, and C3a in particular, in infections by intracellular bacteria.


Asunto(s)
Movimiento Celular/inmunología , Infecciones por Chlamydiaceae/inmunología , Chlamydophila psittaci/inmunología , Complemento C3a/inmunología , Células Dendríticas/inmunología , Pulmón/inmunología , Anafilatoxinas/inmunología , Anafilatoxinas/metabolismo , Animales , Línea Celular , Infecciones por Chlamydiaceae/metabolismo , Infecciones por Chlamydiaceae/microbiología , Chlamydophila psittaci/fisiología , Activación de Complemento/inmunología , Complemento C3a/genética , Complemento C3a/metabolismo , Células Dendríticas/citología , Células Dendríticas/microbiología , Pulmón/metabolismo , Pulmón/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Transducción de Señal/inmunología , Análisis de Supervivencia
4.
Front Immunol ; 12: 626627, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746963

RESUMEN

Recent advances in complement research have revolutionized our understanding of its role in immune responses. The immunomodulatory features of complement in infections by intracellular pathogens, e.g., viruses, are attracting increasing attention. Thereby, local production and activation of complement by myeloid-derived cells seem to be crucial. We could recently show that C3, a key player of the complement cascade, is required for effective defense against the intracellular bacterium Chlamydia psittaci. Avian zoonotic strains of this pathogen cause life-threatening pneumonia with systemic spread in humans; closely related non-avian strains are responsible for less severe diseases of domestic animals with economic loss. To clarify how far myeloid- and non-myeloid cell-derived complement contributes to immune response and resulting protection against C. psittaci, adoptive bone marrow transfer experiments focusing on C3 were combined with challenge experiments using a non-avian (BSL 2) strain of this intracellular bacterium. Surprisingly, our data prove that for C. psittaci-induced pneumonia in mice, non-myeloid-derived, circulating/systemic C3 has a leading role in protection, in particular on the development of pathogen-specific T- and B- cell responses. In contrast, myeloid-derived and most likely locally produced C3 plays only a minor, mainly fine-tuning role. The work we present here describes authentic, although less pronounced, antigen directed immune responses.


Asunto(s)
Inmunidad Adaptativa , Infecciones por Chlamydia/microbiología , Chlamydophila psittaci/patogenicidad , Complemento C3/metabolismo , Pulmón/microbiología , Neumonía Bacteriana/microbiología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/microbiología , Trasplante de Médula Ósea , Células Cultivadas , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/metabolismo , Chlamydophila psittaci/inmunología , Complemento C3/genética , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/microbiología , Quimera por Trasplante
5.
Science ; 352(6292): aad1210, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27313051

RESUMEN

The NLRP3 inflammasome controls interleukin-1ß maturation in antigen-presenting cells, but a direct role for NLRP3 in human adaptive immune cells has not been described. We found that the NLRP3 inflammasome assembles in human CD4(+) T cells and initiates caspase-1-dependent interleukin-1ß secretion, thereby promoting interferon-γ production and T helper 1 (T(H)1) differentiation in an autocrine fashion. NLRP3 assembly requires intracellular C5 activation and stimulation of C5a receptor 1 (C5aR1), which is negatively regulated by surface-expressed C5aR2. Aberrant NLRP3 activity in T cells affects inflammatory responses in human autoinflammatory disease and in mouse models of inflammation and infection. Our results demonstrate that NLRP3 inflammasome activity is not confined to "innate immune cells" but is an integral component of normal adaptive T(H)1 responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proteínas Portadoras/metabolismo , Complemento C5a/inmunología , Inflamasomas/inmunología , Interferón gamma/biosíntesis , Células TH1/inmunología , Inmunidad Adaptativa , Animales , Comunicación Autocrina , Proteínas Portadoras/genética , Activación de Complemento , Síndromes Periódicos Asociados a Criopirina/inmunología , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inmunidad Innata , Inflamación/inmunología , Proteína Cofactora de Membrana/inmunología , Ratones , Ratones Mutantes , Proteína con Dominio Pirina 3 de la Familia NLR , Especies Reactivas de Oxígeno/metabolismo , Receptor de Anafilatoxina C5a/agonistas , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Antígenos de Linfocitos T/agonistas , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Quimiocina/agonistas , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/metabolismo
6.
Pathog Dis ; 74(2)2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26676260

RESUMEN

Chlamydia trachomatis causes sexually transmitted diseases with infertility, pelvic inflammatory disease and neonatal pneumonia as complications. The duration of urogenital mouse models with the strict mouse pathogen C. muridarum addressing vaginal shedding, pathological changes of the upper genital tract or infertility is rather long. Moreover, vaginal C. trachomatis application usually does not lead to the complications feared in women. A fast-to-perform mouse model is urgently needed to analyze new antibiotics, vaccine candidates, immune responses (in gene knockout animals) or mutants of C. trachomatis. To complement the valuable urogenital model with a much faster and quantifiable screening method, we established an optimized lung infection model for the human intracellular bacterium C. trachomatis serovar D (and L2) in immunocompetent C57BL/6J mice. We demonstrated its usefulness by sensitive determination of antibiotic effects characterizing advantages and limitations achievable by early or delayed short tetracycline treatment and single-dose azithromycin application. Moreover, we achieved partial acquired protection in reinfection with serovar D indicating usability for vaccine studies, and showed a different course of disease in absence of complement factor C3. Sensitive monitoring parameters were survival rate, body weight, clinical score, bacterial load, histological score, the granulocyte marker myeloperoxidase, IFN-γ, TNF-α, MCP-1 and IL-6.


Asunto(s)
Antibacterianos/uso terapéutico , Vacunas Bacterianas/inmunología , Chlamydia trachomatis/efectos de los fármacos , Chlamydia trachomatis/fisiología , Neumonía por Clamidia/tratamiento farmacológico , Neumonía por Clamidia/prevención & control , Interacciones Huésped-Patógeno , Animales , Antibacterianos/farmacología , Carga Bacteriana , Biopsia , Línea Celular , Neumonía por Clamidia/microbiología , Neumonía por Clamidia/mortalidad , Complemento C3/genética , Complemento C3/inmunología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunoglobulina G/inmunología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Noqueados , Peroxidasa/metabolismo
7.
J Infect Dis ; 209(8): 1269-78, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24273177

RESUMEN

BACKGROUND: The complement system protects against extracellular pathogens and links innate and adaptive immunity. In this study, we investigated the anaphylatoxin C3a receptor (C3aR) in Chlamydia psittaci lung infection and elucidated C3a-dependent adaptive immune mechanisms. METHODS: Survival, body weight, and clinical score were monitored in primary mouse infection and after serum transfer. Bacterial load, histology, cellular distribution, cytokines, antibodies, and lymphocytes were analyzed. RESULTS: C3aR(-/-) mice showed prolonged pneumonia with decreased survival, lower weight, and higher clinical score. Compared to wild-type mice bacterial clearance was impaired, and inflammatory parameters were increased. In lung-draining lymph nodes of C3aR(-/-) mice the total number of B cells, CD4(+) T cells, and Chlamydia-specific IFN-γ(+) (CD4(+) or CD8(+)) cells was reduced upon infection, and the mice were incapable of Chlamydia-specific immunoglobulin M or immunoglobulin G production. Performed before infection, transfer of hyperimmune serum prolonged survival of C3aR(-/-) mice. CONCLUSIONS: C3a and its receptor are critical for defense against C. psittaci in mouse lung infection. In this model, C3a acts via its receptor as immune modulator. Enhancement of specific B and T cell responses upon infection with an intracellular bacterium were identified as hitherto unknown features of C3a/C3aR. These new functions might be of general immunological importance.


Asunto(s)
Inmunidad Adaptativa/inmunología , Infecciones por Chlamydophila/prevención & control , Chlamydophila psittaci/patogenicidad , Pulmón/microbiología , Neumonía Bacteriana/prevención & control , Receptores de Complemento/fisiología , Linfocitos T/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Infecciones por Chlamydophila/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Masculino , Ratones , Neumonía Bacteriana/inmunología
8.
Inflamm Bowel Dis ; 15(12): 1812-23, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19714742

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) is a critical public health issue; more and more people are affected, but treatment options remain limited. Complement activation and the anaphylatoxin C5a have been shown to play a role in IBD. In this study, mouse models of acute and chronic dextran sulfate-induced colitis were used to further elucidate the impact of C5a and its receptor (C5aR) on disease development. METHODS: In C57BL/6J wildtype and C5aR(-/-) mice the extent of complement activation, changes in weight, and water/food consumption were determined. Disease severity was evaluated via a clinical score, histology, cytokine- and myeloperoxidase-determination as well as real-time reverse-transcriptase polymerase chain reaction (RT-PCR) for expression of anaphylatoxin receptors and inflammatory mediators. RESULTS: C5aR(-/-) mice showed milder disease symptoms, less histological damage, and a lower expression of inflammatory mediators in acute colitis, a setting where complement was activated. In chronic colitis the knockout mice exhibited aggravated weight loss, a higher degree of histological damage and granulocyte infiltration. Intriguingly, increases in C3a-receptor and C5L2 mRNA were dependent on C5aR. Compared to wildtype mice, C5aR(-/-) animals displayed smaller lymph nodes in acute colitis, but extensive swelling and diminished IL-4 and IFN-γ responses in the chronic disease, demonstrating that C5aR modifies T-helper cell polarization. CONCLUSIONS: C5aR exerts detrimental functions in acute colitis, strongly supporting the idea that a C5aR-antagonist might be useful for IBD treatment. However, since the absence of C5aR was no longer protective and in some regards disadvantageous in chronic IBD, future studies should address the efficacy and the possible side effects of a sustained antagonist treatment.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Receptor de Anafilatoxina C5a/inmunología , Enfermedad Aguda , Animales , Polaridad Celular/inmunología , Activación de Complemento/inmunología , Sulfato de Dextran/inmunología , Sulfato de Dextran/toxicidad , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/patología , Interferón gamma/inmunología , Interleucina-4/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a/genética , Índice de Severidad de la Enfermedad , Linfocitos T Colaboradores-Inductores/inmunología
9.
J Biol Chem ; 281(51): 39088-95, 2006 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-17068344

RESUMEN

During complement activation the pro-inflammatory anaphylatoxins C3a and C5a are generated, which interact with the C3a receptor and C5a receptor (CD88), respectively. C5a and its degradation product C5a-des-Arg(74) also bind to the C5a receptor-like 2 (C5L2). C3a and C3a-des-Arg(77), also called acylation-stimulating protein, augment triglyceride synthesis and glucose uptake in adipocytes and skin fibroblasts. Based on data obtained using transfected HEK293 and RBL cells, C5L2 is additionally proposed as a functional receptor for C3a and C3a-des-Arg(77). Here we use (125)I-ligand binding assays and flow cytometry with fluorescently labeled ligands to demonstrate that neither C3a nor C3a-des-Arg(77) binds to C5L2. C5L2 expression and its regulation are investigated on various cell lines by a novel C5L2-restricted binding assay and quantitative real time PCR. Dibutyryl cAMP and interferon-gamma induce up-regulation of this receptor on myeloblastic cell lines (U937 and HL-60), whereas tumor necrosis factor-alpha (TNF-alpha) has no effect. In contrast, epithelial HeLa cells are found to constitutively express C5L2 but not the C5a receptor. In HeLa cells, interferon-gamma and TNF-alpha drastically reduce C5L2 expression. No C5a-dependent Ca(2+) signaling is observed even in these cells endogenously expressing C5L2. Taken together, C5L2 is not a receptor for C3a or C3a-des-Arg(77). Thus, this receptor is unlikely to be directly involved in lipid metabolism. Instead, the identification of stimuli modifying C5L2 expression indicates that C5L2 is a highly regulated scavenger receptor for C5a and C5a-des-Arg(74).


Asunto(s)
Células Epiteliales/citología , Células Mieloides/metabolismo , Receptores de Quimiocina/fisiología , Anafilatoxinas/química , Animales , Complemento C3a/química , Complemento C5a/química , Células HL-60 , Células HeLa , Humanos , Ligandos , Lípidos/química , Ratas , Receptor de Anafilatoxina C5a , Receptores de Quimiocina/química , Transfección , Células U937
10.
J Biol Chem ; 280(51): 42113-23, 2005 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-16253992

RESUMEN

The anaphylatoxin C3a is a proinflammatory mediator generated during complement activation. The tight control of C3a receptor (C3aR) expression is crucial for the regulation of anaphylatoxin-mediated effects. Key factors regulating constitutive expression of the C3aR in the mast cell line HMC-1 and receptor induction by dibutyryl-cAMP in monomyeloblastic U937 cells were determined by functional characterization of the C3aR promoter. Nucleotides -18 to -285 upstream of the translational start site proved to be critical for promoter activity in HMC-1 cells. Binding sites for the transcription factors AP-1 and Ets could be located. Overexpressed c-Jun/c-Fos (AP-1) and Ets-1 led synergistically to increased promoter activity that was substantially reduced by site-directed mutagenesis of the corresponding elements within the C3aR promoter. In HMC-1 cells, Ets interacted directly with the predicted binding motif of the C3aR promoter as determined by electromobility shift assays. AP-1 binding to the C3aR promoter was augmented during C3aR induction in U937 cells. A retroviral gene transfer system was used to express a dominant negative mutant of Ets-1 in these cells. The resulting cells failed to up-regulate the C3aR after stimulation with dibutyryl-cAMP and showed decreased AP-1 binding, suggesting that Ets acts here indirectly. Thus, it was established that Ets and the AP-1 element mediates dibutyryl-cAMP induction of C3aR promoter activity, hence providing a mechanistic explanation of dibutyryl-cAMP-dependent up-regulation of C3aR expression. In conclusion, this study demonstrates an important role of AP-1 and a member of the Ets family in the transcriptional regulation of C3aR expression, a prerequisite for the ability of C3a to participate in immunomodulation and inflammation.


Asunto(s)
Complemento C3a/metabolismo , Proteína Proto-Oncogénica c-ets-1/fisiología , Receptores de Complemento/metabolismo , Factor de Transcripción AP-1/fisiología , Regiones no Traducidas 5' , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Regiones Promotoras Genéticas , Receptores de Complemento/genética , Secuencias Reguladoras de Ácidos Nucleicos
11.
Cell Microbiol ; 5(11): 785-95, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14531894

RESUMEN

Chlamydophila pneumoniae and Chlamydia trachomatis cause infections of the respiratory or urogenital tract. In addition, both species have been associated with atherosclerosis or reactive arthritis respectively. For these intracellular pathogens the interaction with their host-cells is of particular importance. To get insight into this relationship, we conducted a comparative analysis of the host-cell gene regulation of human epithelial cells during infection with Chlamydia. In a screening of HeLa cells by Affymetrix-microchips, numerous regulated host-genes were identified. A detailed expression profile was obtained for 14 genes by real-time RT-PCR - comparing C. pneumoniae, C. trachomatis and intracellular S. typhimurium. The transcriptional responses induced by C. pneumoniae were similar (but usually smaller) compared to C. trachomatis, some were absent. UV-inactivated bacteria induced no differential gene expression suggesting that pathomechanisms other than those associated with innate immunity play here an important role. The expression pattern induced by Salmonella differed substantially. These genus- or group-specific transcriptional response patterns elicited by viable intracellular pathogens may considerably contribute to the different pathologies encountered in the clinic.


Asunto(s)
Chlamydia trachomatis/metabolismo , Chlamydophila pneumoniae/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inmunidad Innata/fisiología , Células Cultivadas , Infecciones por Chlamydia , Chlamydia trachomatis/genética , Infecciones por Chlamydophila , Chlamydophila pneumoniae/genética , Células HeLa , Humanos , Monocitos/citología , Monocitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos
12.
Eur J Immunol ; 33(4): 920-7, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12672058

RESUMEN

The anaphylatoxic peptide C3a is a pro-inflammatory mediator generated during complement activation, whose specific G protein coupled receptor is expressed on granulocytes, monocytes, mast cells, activated lymphocytes, and in the nervous tissue. We have generated RBL-2H3 cell clones stably expressing mutants of the human C3a-receptor (C3aR) with combined alanine (Ala) substitutions of ten C-terminal serine (Ser) or threonine (Thr) residues, which may represent putative phosphorylation sites to characterize their role in ligand-induced C3aR internalization and signaling. Ser475/479 and Thr480/481 as well as Ser449 seemed not to be involved in ligand-induced receptor internalization. Either directly or by a conformational change they even "inhibit" C3aR internalization. In contrast, mutants with Ala substitutions at Ser465/470 and Thr463/466 were poorly internalized, and Thr463 seemed to be the most important C-terminal Thr or Ser residue directly effecting receptor internalization. However, it is likely that other C3aR regions additionally participate in this negative feed-back mechanism since even mutants with multiple Ala substitutions still internalized to a limited degree. Interestingly, in a mutant with a single exchange of Ser449 to Ala, the signal transduction assessed by a Ca(2+) assay and [(35)S]GTP gamma S-binding on HEK cells transiently co-transfected with G-alpha 16 or G-alpha O, respectively, was severely impaired, indicating that this residue of C3aR is involved in G protein coupling.


Asunto(s)
Proteínas de la Membrana , Receptores de Complemento/química , Receptores de Complemento/fisiología , Serina/fisiología , Transducción de Señal , Treonina/fisiología , Animales , Calcio/metabolismo , Línea Celular , Endocitosis , Subunidades alfa de la Proteína de Unión al GTP , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Cinética , Mutación , Ratas , Receptores de Complemento/genética , Serina/genética , Relación Estructura-Actividad , Treonina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...