Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(1): e2213715120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36577072

RESUMEN

The nuclear long non-coding RNA LUCAT1 has previously been identified as a negative feedback regulator of type I interferon and inflammatory cytokine expression in human myeloid cells. Here, we define the mechanistic basis for the suppression of inflammatory gene expression by LUCAT1. Using comprehensive identification of RNA-binding proteins by mass spectrometry as well as RNA immunoprecipitation, we identified proteins important in processing and alternative splicing of mRNAs as LUCAT1-binding proteins. These included heterogeneous nuclear ribonucleoprotein C, M, and A2B1. Consistent with this finding, cells lacking LUCAT1 have altered splicing of selected immune genes. In particular, upon lipopolysaccharide stimulation, the splicing of the nuclear receptor 4A2 (NR4A2) gene was particularly affected. As a consequence, expression of NR4A2 was reduced and delayed in cells lacking LUCAT1. NR4A2-deficient cells had elevated expression of immune genes. These observations suggest that LUCAT1 is induced to control the splicing and stability of NR4A2, which is in part responsible for the anti-inflammatory effect of LUCAT1. Furthermore, we analyzed a large cohort of patients with inflammatory bowel disease as well as asthma and chronic obstructive pulmonary disease. In these patients, LUCAT1 levels were elevated and in both diseases, positively correlated with disease severity. Collectively, these studies define a key molecular mechanism of LUCAT1-dependent immune regulation through post-transcriptional regulation of mRNAs highlighting its role in the regulation of inflammatory disease.


Asunto(s)
Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , ARN Largo no Codificante , Humanos , Movimiento Celular , Proliferación Celular , Inflamación/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Receptores Citoplasmáticos y Nucleares , ARN Largo no Codificante/metabolismo , Empalme del ARN , Estabilidad del ARN
2.
Semin Cell Dev Biol ; 111: 126-134, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32580911

RESUMEN

Tight regulation of the immune response is fundamental for efficient pathogen clearance and to prevent excessive inflammation. Long non-coding RNAs (lncRNAs) have emerged as potent regulators of the innate and adaptive immune responses to viral pathogens. Host-derived lncRNAs control the differentiation and polarization of immune cell populations and the production of cytokines, interferons and antiviral factors. This review provides an updated overview of lncRNAs that modulate viral replication or pathogenesis. Beyond that, viruses have developed lncRNA-based strategies to mask themselves from immune detection and evade antiviral immunity. A deeper understanding of lncRNA biology in the context of host-pathogen interactions may unveil new treatment strategies in the near future.


Asunto(s)
Inmunidad Innata/genética , ARN Largo no Codificante/genética , ARN Viral/genética , Proteínas de Unión al ARN/genética , Virosis/genética , Virus/genética , Regulación de la Expresión Génica , Humanos , Evasión Inmune/genética , Interferones/genética , Interferones/inmunología , Interleucinas/genética , Interleucinas/inmunología , MicroARNs/genética , MicroARNs/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , ARN Largo no Codificante/inmunología , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Viral/inmunología , Proteínas de Unión al ARN/inmunología , Transducción de Señal , Virosis/inmunología , Virosis/patología , Virosis/virología , Replicación Viral , Virus/crecimiento & desarrollo , Virus/patogenicidad
3.
RNA ; 25(7): 869-880, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31019095

RESUMEN

Bacterial RNA has emerged as an important activator of innate immune responses by stimulating Toll-like receptors TLR7 and TLR8 in humans. Guanosine 2'-O-methylation at position 18 (Gm18) in bacterial tRNA was shown to antagonize tRNA-induced TLR7/8 activation, suggesting a potential role of Gm18 as an immune escape mechanism. This modification also occurs in eukaryotic tRNA, yet a physiological immune function remained to be tested. We therefore set out to investigate the immune modulatory role of Gm18 in both prokaryotic and eukaryotic microorganisms, Escherichia coli and Saccharomyces cerevisiae, and in human cells. Using RiboMethSeq analysis we show that mutation of trmH in E. coli, trm3 in S. cereviase, and CRISPR/Cas9-induced knockout of TARBP1 in H. sapiens results in loss of Gm18 within tRNA. Lack of Gm18 across the kingdoms resulted in increased immunostimulation of peripheral blood mononuclear cells when activated by tRNA preparations. In E. coli, lack of 2'-O-methyltransferase trmH also enhanced immune stimulatory properties by whole cellular RNA. In contrast, lack of Gm18 in yeasts and human cells did not affect immunostimulation by whole RNA preparations. When using live E. coli bacteria, lack of trmH did not affect overall immune stimulation although we detected a defined TLR8/RNA-dependent gene expression signature upon E. coli infection. Together, these results demonstrate that Gm18 is a global immune inhibitory tRNA modification across the kingdoms and contributes to tRNA recognition by innate immune cells, but as an individual modification has insufficient potency to modulate recognition of the investigated microorganisms.


Asunto(s)
Endosomas/metabolismo , Células Eucariotas/inmunología , Guanosina/química , Inmunidad Innata/inmunología , Células Procariotas/inmunología , ARN de Transferencia/metabolismo , Receptores Toll-Like/metabolismo , Células Eucariotas/metabolismo , Humanos , Metilación , Células Procariotas/metabolismo , ARN de Transferencia/genética , Receptores Toll-Like/genética , ARNt Metiltransferasas/genética , ARNt Metiltransferasas/metabolismo
4.
Allergy ; 74(2): 223-235, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30475385

RESUMEN

RNA-sensing Toll-like receptors (TLRs) are often described as antiviral receptors of the innate immune system. However, the past decade has shown that the function and relevance of these receptors are far more complex. They were found to be essential for the detection of various bacterial, archaeal, and eukaryotic microorganisms and facilitate the discrimination between dead and living microbes. The cytokine and interferon response profile that is triggered has the potential to improve the efficacy of next-generation vaccines and may prevent the development of asthma and allergy. Nevertheless, the ability to recognize foreign RNA comes with a cost as also damaged host cells can release nucleic acids that might induce an inappropriate immune response. Thus, it is not surprising that RNA-sensing TLRs play a key role in various autoimmune diseases. However, promising new inhibitors and antagonists are on the horizon to improve their treatment.


Asunto(s)
ARN/genética , Receptores Toll-Like/metabolismo , Animales , Asma/etiología , Asma/metabolismo , Autoinmunidad , Biomarcadores , Susceptibilidad a Enfermedades , Endosomas/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Hipersensibilidad/etiología , Hipersensibilidad/metabolismo , Inmunidad Innata , Ligandos , ARN/química , ARN/metabolismo , Relación Estructura-Actividad , Receptores Toll-Like/química
5.
JCI Insight ; 9(3)2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38329124

RESUMEN

The role of long noncoding RNAs (lncRNAs) in disease is incompletely understood, but their regulation of inflammation is increasingly appreciated. We addressed the extent of lncRNA involvement in inflammatory bowel disease (IBD) using biopsy-derived RNA-sequencing data from a large cohort of deeply phenotyped patients with IBD. Weighted gene correlation network analysis revealed gene modules of lncRNAs coexpressed with protein-coding genes enriched for biological pathways, correlated with epithelial and immune cell signatures, or correlated with distal colon expression. Correlation of modules with clinical features uncovered a module correlated with disease severity, with an enriched interferon response signature containing the hub lncRNA IRF1-AS1. Connecting genes to IBD-associated single nucleotide polymorphisms (SNPs) revealed an enrichment of SNP-adjacent lncRNAs in biologically relevant modules. Ulcerative colitis-specific SNPs were enriched in distal colon-related modules, suggesting that disease-specific mechanisms may result from altered lncRNA expression. The function of the IBD-associated SNP-adjacent lncRNA IRF1-AS1 was explored in human myeloid cells, and our results suggested IRF1-AS1 promoted optimal production of TNF-α, IL-6, and IL-23. A CRISPR/Cas9-mediated activation screen in THP-1 cells revealed several lncRNAs that modulated LPS-induced TNF-α responses. Overall, this study uncovered the expression patterns of lncRNAs in IBD that identify functional, disease-relevant lncRNAs.


Asunto(s)
Colitis Ulcerosa , ARN Largo no Codificante , Humanos , Redes Reguladoras de Genes , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Necrosis Tumoral alfa/genética , Colitis Ulcerosa/genética , Inflamación
6.
Cell Metab ; 35(8): 1441-1456.e9, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37494932

RESUMEN

This study reveals a previously uncharacterized mechanism to restrict intestinal inflammation via a regulatory RNA transcribed from a noncoding genomic locus. We identified a novel transcript of the lncRNA HOXA11os specifically expressed in the distal colon that is reduced to undetectable levels in colitis. HOXA11os is localized to mitochondria under basal conditions and interacts with a core subunit of complex 1 of the electron transport chain (ETC) to maintain its activity. Deficiency of HOXA11os in colonic myeloid cells results in complex I deficiency, dysfunctional oxidative phosphorylation (OXPHOS), and the production of mitochondrial reactive oxygen species (mtROS). As a result, HOXA11os-deficient mice develop spontaneous intestinal inflammation and are hypersusceptible to colitis. Collectively, these studies identify a new regulatory axis whereby a lncRNA maintains intestinal homeostasis and restricts inflammation in the colon through the regulation of complex I activity.


Asunto(s)
Colitis , ARN Largo no Codificante , Animales , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Colitis/genética , Colitis/metabolismo , Inflamación/metabolismo , Mitocondrias/genética , Homeostasis , Mucosa Intestinal/metabolismo
7.
J Exp Med ; 219(8)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35792863

RESUMEN

Hepatocyte nuclear factor 4 α (HNF4A) is a highly conserved nuclear receptor that has been associated with ulcerative colitis. In mice, HNF4A is indispensable for the maintenance of intestinal homeostasis, yet the underlying mechanisms are poorly characterized. Here, we demonstrate that the expression of HNF4A in intestinal epithelial cells (IECs) is required for the proper development and composition of the intraepithelial lymphocyte (IEL) compartment. HNF4A directly regulates expression of immune signaling molecules including butyrophilin-like (Btnl) 1, Btnl6, H2-T3, and Clec2e that control IEC-IEL crosstalk. HNF4A selectively enhances the expansion of natural IELs that are TCRγδ+ or TCRαß+CD8αα+ to shape the composition of IEL compartment. In the small intestine, HNF4A cooperates with its paralog HNF4G, to drive expression of immune signaling molecules. Moreover, the HNF4A-BTNL regulatory axis is conserved in human IECs. Collectively, these findings underscore the importance of HNF4A as a conserved transcription factor controlling IEC-IEL crosstalk and suggest that HNF4A maintains intestinal homeostasis through regulation of the IEL compartment.


Asunto(s)
Linfocitos Intraepiteliales , Animales , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Mucosa Intestinal , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
8.
Cell Rep ; 39(2): 110691, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417716

RESUMEN

The innate immune recognition of the malaria-causing pathogen Plasmodium falciparum (P. falciparum) is not fully explored. Here, we identify the nucleoside 5'-methylthioinosine (MTI), a Plasmodium-specific intermediate of the purine salvage pathway, as a pathogen-derived Toll-like receptor 8 (TLR8) agonist. Co-incubation of MTI with the TLR8 enhancer poly(dT) as well as synthetic or P. falciparum-derived RNA strongly increase its stimulatory activity. Of note, MTI generated from methylthioadenosine (MTA) by P. falciparum lysates activates TLR8 when MTI metabolism is inhibited by immucillin targeting the purine nucleoside phosphorylase (PfPNP). Importantly, P. falciparum-infected red blood cells incubated with MTI or cultivated with MTA and immucillin lead to TLR8-dependent interleukin-6 (IL-6) production in human monocytes. Our data demonstrate that the nucleoside MTI is a natural human TLR8 ligand with possible in vivo relevance for innate sensing of P. falciparum.


Asunto(s)
Malaria Falciparum , Metiltioinosina , Receptor Toll-Like 8 , Humanos , Metiltioinosina/análogos & derivados , Nucleósidos , Plasmodium falciparum/metabolismo , Purina-Nucleósido Fosforilasa/metabolismo , Purinas , Receptor Toll-Like 8/metabolismo
9.
J Clin Invest ; 131(16)2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34255743

RESUMEN

In view of emerging drug-resistant tuberculosis (TB), host-directed adjunct therapies are urgently needed to improve treatment outcomes with currently available anti-TB therapies. One approach is to interfere with the formation of lipid-laden "foamy" macrophages in the host, as they provide a nutrient-rich host cell environment for Mycobacterium tuberculosis (Mtb). Here, we provide evidence that Wnt family member 6 (WNT6), a ligand of the evolutionarily conserved Wingless/Integrase 1 (WNT) signaling pathway, promotes foam cell formation by regulating key lipid metabolic genes including acetyl-CoA carboxylase 2 (ACC2) during pulmonary TB. Using genetic and pharmacological approaches, we demonstrated that lack of functional WNT6 or ACC2 significantly reduced intracellular triacylglycerol (TAG) levels and Mtb survival in macrophages. Moreover, treatment of Mtb-infected mice with a combination of a pharmacological ACC2 inhibitor and the anti-TB drug isoniazid (INH) reduced lung TAG and cytokine levels, as well as lung weights, compared with treatment with INH alone. This combination also reduced Mtb bacterial numbers and the size of mononuclear cell infiltrates in livers of infected mice. In summary, our findings demonstrate that Mtb exploits WNT6/ACC2-induced storage of TAGs in macrophages to facilitate its intracellular survival, a finding that opens new perspectives for host-directed adjunctive treatment of pulmonary TB.


Asunto(s)
Acetil-CoA Carboxilasa/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Proteínas Proto-Oncogénicas/metabolismo , Triglicéridos/metabolismo , Proteínas Wnt/metabolismo , Acetil-CoA Carboxilasa/antagonistas & inhibidores , Animales , Antituberculosos/administración & dosificación , Inhibidores Enzimáticos/administración & dosificación , Células Espumosas/metabolismo , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/fisiología , Humanos , Isoniazida/administración & dosificación , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/efectos de los fármacos , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/efectos de los fármacos , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/metabolismo , Tuberculosis Pulmonar/microbiología , Proteínas Wnt/deficiencia , Proteínas Wnt/genética
10.
Nat Commun ; 11(1): 6348, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33311506

RESUMEN

Long non-coding RNAs are important regulators of biological processes including immune responses. The immunoregulatory functions of lncRNAs have been revealed primarily in murine models with limited understanding of lncRNAs in human immune responses. Here, we identify lncRNA LUCAT1 which is upregulated in human myeloid cells stimulated with lipopolysaccharide and other innate immune stimuli. Targeted deletion of LUCAT1 in myeloid cells increases expression of type I interferon stimulated genes in response to LPS. By contrast, increased LUCAT1 expression results in a reduction of the inducible ISG response. In activated cells, LUCAT1 is enriched in the nucleus where it associates with chromatin. Further, LUCAT1 limits transcription of interferon stimulated genes by interacting with STAT1 in the nucleus. Together, our study highlights the role of the lncRNA LUCAT1 as a post-induction feedback regulator which functions to restrain the immune response in human cells.


Asunto(s)
Regulación de la Expresión Génica , Interferones/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Animales , Fenómenos Biológicos , Cromatina/metabolismo , Citocinas/metabolismo , Retroalimentación , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata/efectos de los fármacos , Lipopolisacáridos/efectos adversos , Ratones , Células Mieloides/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Proteínas , Factor de Transcripción STAT1/metabolismo , Células THP-1
11.
Nat Commun ; 11(1): 105, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31913271

RESUMEN

Psoriasis is an inflammatory skin disease with strong neutrophil (PMN) infiltration and high levels of the antimicrobial peptide, LL37. LL37 in complex with DNA and RNA is thought to initiate disease exacerbation via plasmacytoid dendritic cells. However, the source of nucleic acids supposed to start this initial inflammatory event remains unknown. We show here that primary murine and human PMNs mount a fulminant and self-propagating neutrophil extracellular trap (NET) and cytokine response, but independently of the canonical NET component, DNA. Unexpectedly, RNA, which is abundant in NETs and psoriatic but not healthy skin, in complex with LL37 triggered TLR8/TLR13-mediated cytokine and NET release by PMNs in vitro and in vivo. Transfer of NETs to naive human PMNs prompts additional NET release, promoting further inflammation. Our study thus uncovers a self-propagating vicious cycle contributing to chronic inflammation in psoriasis, and NET-associated RNA (naRNA) as a physiologically relevant NET component.


Asunto(s)
Trampas Extracelulares/inmunología , Neutrófilos/inmunología , Psoriasis/inmunología , Adulto , Animales , Péptidos Catiónicos Antimicrobianos , Citocinas/genética , Citocinas/inmunología , Trampas Extracelulares/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Infiltración Neutrófila , Psoriasis/genética , ARN/genética , ARN/inmunología , Receptor Toll-Like 8/genética , Receptor Toll-Like 8/inmunología , Adulto Joven , Catelicidinas
12.
PLoS One ; 12(10): e0185919, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28982164

RESUMEN

The methanogenic archaeon Methanomassiliicoccus luminyensis strain B10T was isolated from human feces just a few years ago. Due to its remarkable metabolic properties, particularly the degradation of trimethylamines, this strain was supposed to be used as "Archaebiotic" during metabolic disorders of the human intestine. However, there is still no data published regarding adaptations to the natural habitat of M. luminyensis as it has been shown for the other two reported mucosa-associated methanoarchaea. This study aimed at unraveling susceptibility of M. luminyensis to antimicrobial peptides as well as its immunogenicity. By using the established microtiter plate assay adapted to the anaerobic growth requirements of methanogenic archaea, we demonstrated that M. luminyensis is highly sensitive against LL32, a derivative of human cathelicidin (MIC = 2 µM). However, the strain was highly resistant against the porcine lysin NK-2 (MIC = 10 µM) and the synthetic antilipopolysaccharide peptide (Lpep) (MIC>10 µM) and overall differed from the two other methanoarchaea, Methanobrevibacter smithii and Methanosphaera stadtmanae in respect to AMP sensitivity. Moreover, only weak immunogenic potential of M. luminyensis was demonstrated using peripheral blood mononuclear cells (PBMCs) and monocyte-derived dendritic cells (moDCs) by determining release of pro-inflammatory cytokines. Overall, our findings clearly demonstrate that the archaeal gut inhabitant M. luminyensis is susceptible to the release of human-derived antimicrobial peptides and exhibits low immunogenicity towards human immune cells in vitro-revealing characteristics of a typical commensal gut microbe.


Asunto(s)
Antiinfecciosos/farmacología , Intestinos/microbiología , Methanomicrobiaceae/inmunología , Péptidos/farmacología , Humanos , Methanomicrobiaceae/efectos de los fármacos , Methanomicrobiaceae/crecimiento & desarrollo
13.
Front Immunol ; 8: 1535, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29181003

RESUMEN

The archaeon Methanosphaera stadtmanae is a member of the gut microbiota; yet, the molecular cross-talk between archaea and the human immune system and its potential contribution to inflammatory diseases has not been evaluated. Although archaea are as bacteria prokaryotes, they form a distinct domain having unique features such as different cell wall structures and membrane lipids. So far, no microbe-associated molecular patterns of archaea which activate innate immune receptors have been identified. By stimulating human myeloid cells with M. stadtmanae and purified archaeal nucleic acids, we identified both the microorganism and its RNA as potent stimuli for the innate immune system. To dissect the recognition and activation pathways induced by M. stadtmanae, human monocytic BLaER1 knockout cells were generated using the CRISPR/Cas9 system targeting components of TLR and inflammasome signaling. While the recognition of M. stadtmanae is mediated by TLR7 and TLR8, activation of the NLRP3 inflammasome depends solely on TLR8 engagement. Notably, this process resembles hallmarks of both the canonical and the recently described alternative inflammasome activation. Thus, we have demonstrated for the first time the specific recognition of and response to an archaeon by human cells at the molecular level.

14.
J Immunother ; 38(3): 85-95, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25710248

RESUMEN

Human cytolytic fusion proteins (hCFPs) are therapeutically efficacious recombinant polypeptides comprising a target cell-specific binding component and a human effector domain that induces apoptosis. Compared with former generations of immunotoxins, which contain immunogenic cytotoxic domains derived from bacteria or plants, hCFPs contain solely human proteins that do not induce an immune response, thus avoiding the development of neutralizing antibodies. Here, we investigated the suitability of human angiogenin (Ang) mutants as effector domains. We engineered 3 different Ang variants that outperformed the wild-type enzyme by replacing amino acid residues with key roles in the protein's catalytic activity and its interaction with the ribonuclease inhibitor RNH1. The cytotoxic potential of these mutants was compared with wild-type Ang by fusing each to the CD64-specific single-chain variable fragment H22. All hCFPs were successfully expressed in HEK293T cells and purified from the cell culture supernatant by immobilized metal ion affinity chromatography. The Ang mutant-based hCFPs showed normal binding activity towards human interferon-γ-stimulated CD64 HL-60 cells and activated human macrophages isolated from peripheral blood mononuclear cells, but increased cytotoxicity based on reduced affinity towards RNH1 and higher ribonucleolytic activity.


Asunto(s)
Proteínas Mutantes , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/toxicidad , Ribonucleasa Pancreática/genética , Apoptosis/efectos de los fármacos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Expresión Génica , Variación Genética , Células HL-60 , Humanos , Inmunotoxinas/farmacología , Cinética , Activación de Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Unión Proteica , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleasa Pancreática/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA