Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Hepatol ; 76(5): 1127-1137, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35074474

RESUMEN

BACKGROUND & AIMS: Myeloid cells are key regulators of cirrhosis, a major cause of mortality worldwide. Because stromal cells can modulate the functionality of myeloid cells in vitro, targeting stromal-myeloid interactions has become an attractive potential therapeutic strategy. We aimed to investigate how human liver stromal cells impact myeloid cell properties and to understand the utility of a stromal-myeloid coculture system to study these interactions in the context of cirrhosis. METHODS: Single-cell RNA-sequencing analyses of non-cirrhotic (n = 7) and cirrhotic (n = 5) human liver tissue were correlated to the bulk RNA-sequencing results of in vitro cocultured human CD14+ and primary liver stromal cells. Complimentary mechanistic experiments and flow cytometric analysis were performed on human liver stromal-myeloid coculture systems. RESULTS: We found that stromal-myeloid coculture reduces the frequency CD14+ cell subsets transcriptionally similar to liver macrophages, showing that stromal cells inhibit the maturation of monocytes into macrophages. Stromal cells also influenced in vitro macrophage differentiation by skewing away from cirrhosis-linked CD9+ scar-associated macrophage-like cells and towards CD163+ Kupffer cell-like macrophages. We identify IL-6 production as a mechanism by which stromal cells limit CD9+ macrophage differentiation and find that local IL-6 levels are decreased in early-stage human liver disease compared to healthy liver tissue, suggesting a protective role for local IL-6 in the healthy liver. CONCLUSIONS: Our work reveals an unanticipated role for liver stromal cells in impeding the maturation and altering the differentiation of macrophages and should prompt investigations into the role of local IL-6 production in the pathogenesis of liver disease. These studies provide a framework for investigating macrophage-stromal interactions during cirrhosis. LAY SUMMARY: The impact of human liver stromal cells on myeloid cell maturation and differentiation in liver disease is incompletely understood. In this study, we present a mechanistic analysis using a primary in vitro human liver stromal-myeloid coculture system that is translated to liver disease using single-cell RNA sequencing analysis of cirrhotic and non-cirrhotic human liver tissue. Our work supports a role for stromal cell contact in restricting macrophage maturation and for stromal-derived IL-6 in limiting the differentiation of a cirrhotic macrophage subset.


Asunto(s)
Interleucina-6 , Hepatopatías , Diferenciación Celular , Humanos , Cirrosis Hepática/etiología , Hepatopatías/patología , Macrófagos/patología , Monocitos/patología , ARN , Células del Estroma/patología
2.
Nat Commun ; 10(1): 2712, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221971

RESUMEN

Clostridium difficile (C. difficile) incidence has tripled over the past 15 years and is attributed to the emergence of hypervirulent strains. While it is clear that C. difficile toxins cause damaging colonic inflammation, the immune mechanisms protecting from tissue damage require further investigation. Through a transcriptome analysis, we identify IL-33 as an immune target upregulated in response to hypervirulent C. difficile. We demonstrate that IL-33 prevents C. difficile-associated mortality and epithelial disruption independently of bacterial burden or toxin expression. IL-33 drives colonic group 2 innate lymphoid cell (ILC2) activation during infection and IL-33 activated ILC2s are sufficient to prevent disease. Furthermore, intestinal IL-33 expression is regulated by the microbiota as fecal microbiota transplantation (FMT) rescues antibiotic-associated depletion of IL-33. Lastly, dysregulated IL-33 signaling via the decoy receptor, sST2, predicts C. difficile-associated mortality in human patients. Thus, IL-33 signaling to ILC2s is an important mechanism of defense from C. difficile colitis.


Asunto(s)
Clostridioides difficile/inmunología , Enterocolitis Seudomembranosa/inmunología , Inmunidad Innata , Interleucina-33/metabolismo , Linfocitos/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antibacterianos/efectos adversos , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Clostridioides difficile/patogenicidad , Colon/citología , Colon/inmunología , Colon/microbiología , Colon/patología , Modelos Animales de Enfermedad , Enterocolitis Seudomembranosa/microbiología , Enterocolitis Seudomembranosa/mortalidad , Enterocolitis Seudomembranosa/terapia , Trasplante de Microbiota Fecal , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Perfilación de la Expresión Génica , Humanos , Interleucina-33/inmunología , Linfocitos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Virulencia/inmunología , Adulto Joven
3.
Cell Host Microbe ; 25(5): 756-765.e5, 2019 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-31003940

RESUMEN

Clostridium difficile infection (CDI) is the number one hospital-acquired infection in the United States. CDI is more common and severe in inflammatory bowel disease patients. Here, we studied the mechanism by which prior colitis exacerbates CDI. Mice were given dextran sulfate sodium (DSS) colitis, recovered for 2 weeks, and then were infected with C. difficile. Mortality and CDI severity were increased in DSS-treated mice compared to controls. Severe CDI is dependent on CD4+ T cells, which persist after colitis-associated inflammation subsides. Adoptive transfer of Th17 cells to naive mice is sufficient to increase CDI-associated mortality through elevated IL-17 production. Finally, in humans, the Th17 cytokines IL-6 and IL-23 associate with severe CDI, and patients with high serum IL-6 are 7.6 times more likely to die post infection. These findings establish a central role for Th17 cells in CDI pathogenesis following colitis and identify them as a potential target for preventing severe disease.


Asunto(s)
Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/inmunología , Colitis/complicaciones , Colitis/patología , Susceptibilidad a Enfermedades , Células Th17/inmunología , Adolescente , Traslado Adoptivo , Adulto , Anciano , Animales , Niño , Infecciones por Clostridium/mortalidad , Infecciones por Clostridium/patología , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/patología , Subunidad p19 de la Interleucina-23/sangre , Interleucina-6/sangre , Masculino , Ratones , Persona de Mediana Edad , Medición de Riesgo , Análisis de Supervivencia , Adulto Joven
5.
JAMA Surg ; 153(12): 1127-1133, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30208386

RESUMEN

Importance: Recent evidence from an animal model suggests that peripheral loss of eosinophils in Clostridium difficile infection (CDI) is associated with severe disease. The ability to identify high-risk patients with CDI as early as the time of admission could improve outcomes by guiding management decisions. Objective: To construct a model using clinical indices readily available at the time of hospital admission, including peripheral eosinophil counts, to predict inpatient mortality in patients with CDI. Design, Setting, and Participants: In a cohort study, a total of 2065 patients admitted for CDI through the emergency department of 2 tertiary referral centers from January 1, 2005, to December 31, 2015, formed a training and a validation cohort. The sample was stratified by admission eosinophil count (0.0 cells/µL or >0.0 cells/µL), and multivariable logistic regression was used to construct a predictive model for inpatient mortality as well as other disease-related outcomes. Main Outcomes and Measures: Inpatient mortality was the primary outcome. Secondary outcomes included the need for a monitored care setting, need for vasopressors, and rates of inpatient colectomy. Results: Of the 2065 patients in the study, 1092 (52.9%) were women and patients had a mean (SD) age of 63.4 (18.4) years. Those with an undetectable eosinophil count at admission had increased in-hospital mortality in both the training (odds ratio [OR], 2.01; 95% CI, 1.08-3.73; P = .03) and validation (OR, 2.26; 95% CI, 1.33-3.83; P = .002) cohorts in both univariable and multivariable analysis. Undetectable eosinophil counts were also associated with indicators of severe sepsis, such as admission to monitored care settings (OR, 1.40; 95% CI, 1.06-1.86), the need for vasopressors (OR, 2.08; 95% CI, 1.32-3.28), and emergency total colectomy (OR, 2.56; 95% CI, 1.12-5.87). Other significant predictors of mortality at admission included increasing comorbidity burden (for each 1-unit increase: OR, 1.13; 95% CI, 1.05-1.22) and lower systolic blood pressures (for each 1-mm Hg increase: OR, 0.99; 95% CI, 0.98-1.00). In a subgroup analysis of patients presenting without initial tachycardia or hypotension, only patients with undetectable admission eosinophil counts, but not those with an elevated white blood cell count, had significantly increased odds of inpatient mortality (OR, 5.76; 95% CI, 1.99-16.64). Conclusions and Relevance: This study describes a simple, widely available, inexpensive model predicting CDI severity and mortality to identify at-risk patients at the time of admission.


Asunto(s)
Infecciones por Clostridium/patología , Eosinófilos/patología , Adulto , Anciano , Infecciones por Clostridium/mortalidad , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Oportunidad Relativa , Valor Predictivo de las Pruebas , Factores de Riesgo , Adulto Joven
6.
mBio ; 8(1)2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28246365

RESUMEN

The parasite Entamoeba histolytica is a cause of diarrhea in infants in low-income countries. Previously, it was shown that tumor necrosis factor alpha (TNF-α) production was associated with increased risk of E. histolytica diarrhea in children. Interleukin-25 (IL-25) is a cytokine that is produced by intestinal epithelial cells that has a role in maintenance of gut barrier function and inhibition of TNF-α production. IL-25 expression was decreased in humans and in the mouse model of amebic colitis. Repletion of IL-25 blocked E. histolytica infection and barrier disruption in mice, increased gut eosinophils, and suppressed colonic TNF-α. Depletion of eosinophils with anti-Siglec-F antibody prevented IL-25-mediated protection. In contrast, depletion of TNF-α resulted in resistance to amebic infection. We concluded that IL-25 provides protection from amebiasis, which is dependent upon intestinal eosinophils and suppression of TNF-α.IMPORTANCE The intestinal epithelial barrier is important for protection from intestinal amebiasis. We discovered that the intestinal epithelial cytokine IL-25 was suppressed during amebic colitis in humans and that protection could be restored in the mouse model by IL-25 administration. IL-25 acted via eosinophils and suppressed TNF-α. This work illustrates a previously unrecognized pathway of innate mucosal immune response.


Asunto(s)
Disentería Amebiana/inmunología , Entamoeba histolytica/inmunología , Eosinófilos/inmunología , Interleucina-17/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Interleucinas/metabolismo , Ratones
7.
Nat Microbiol ; 1(8): 16108, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27573114

RESUMEN

Clostridium difficile is the most common hospital acquired pathogen in the USA, and infection is, in many cases, fatal. Toxins A and B are its major virulence factors, but expression of a third toxin, known as C. difficile transferase (CDT), is increasingly common. An adenosine diphosphate (ADP)-ribosyltransferase that causes actin cytoskeletal disruption, CDT is typically produced by the major, hypervirulent strains and has been associated with more severe disease. Here, we show that CDT enhances the virulence of two PCR-ribotype 027 strains in mice. The toxin induces pathogenic host inflammation via a Toll-like receptor 2 (TLR2)-dependent pathway, resulting in the suppression of a protective host eosinophilic response. Finally, we show that restoration of TLR2-deficient eosinophils is sufficient for protection from a strain producing CDT. These findings offer an explanation for the enhanced virulence of CDT-expressing C. difficile and demonstrate a mechanism by which this binary toxin subverts the host immune response.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Proteínas Bacterianas/metabolismo , Clostridioides difficile/inmunología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/patología , Colon/inmunología , Eosinófilos/inmunología , Factores de Virulencia/metabolismo , Animales , Clostridioides difficile/clasificación , Clostridioides difficile/genética , Infecciones por Clostridium/microbiología , Modelos Animales de Enfermedad , Ratones , Ribotipificación
8.
Cell Rep ; 16(2): 432-443, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27346351

RESUMEN

Clostridium difficile infection (CDI) is the most common cause of hospital-acquired infection in the United States. Host susceptibility and the severity of infection are influenced by disruption of the microbiota and the immune response. However, how the microbiota regulate immune responses to mediate CDI outcome remains unclear. Here, we have investigated the role of the microbiota-linked cytokine IL-25 during infection. Intestinal IL-25 was suppressed during CDI in humans and mice. Restoration of IL-25 reduced CDI-associated mortality and tissue pathology even though equivalent levels of C. difficile bacteria and toxin remained in the gut. IL-25 protection was mediated by gut eosinophils, as demonstrated by an increase in intestinal eosinophils and a loss of IL-25 protection upon eosinophil depletion. These findings support a mechanism whereby the induction of IL-25-mediated eosinophilia can reduce host mortality during active CDI. This work may provide targets for future development of microbial or immune-based therapies.


Asunto(s)
Clostridioides difficile/inmunología , Enterocolitis Seudomembranosa/metabolismo , Eosinófilos/inmunología , Interleucinas/fisiología , Animales , Enterocolitis Seudomembranosa/inmunología , Enterocolitis Seudomembranosa/microbiología , Mucinas Gástricas/biosíntesis , Microbioma Gastrointestinal , Humanos , Interleucina-4/metabolismo , Interleucinas/farmacología , Recuento de Leucocitos , Masculino , Ratones Endogámicos C57BL , Membrana Mucosa/inmunología , Membrana Mucosa/patología , Factores Protectores
9.
Anaerobe ; 41: 79-84, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27212111

RESUMEN

Clostridium difficile is a gram-positive, spore forming anaerobe that infects the gut when the normal microbiota has been disrupted. C. difficile infection (CDI) is the most common cause of hospital acquired infection in the United States, and the leading cause of death due to gastroenteritis. Patients suffering from CDI have varying symptoms which range from mild diarrhea to pseudomembranous colitis and death. The involvement of the immune response to influence disease severity is just beginning to be investigated. There is evidence that the immune response can facilitate either protective or pathogenic phenotypes, suggesting it plays a multifaceted role during CDI. In addition to the immune response, the microbiota is pivotal in dictating the pathogenesis to CDI. A healthy microbiota effectively inhibits infection by restricting the ability of C. difficile to expand in the colon. Thus, understanding which immune mediators and components of the microbiota play beneficial roles during CDI will be important to future therapeutic developments. This review outlines how the microbiota can modulate specific immune mediators, such as IL-23 and others, to influence disease outcome.


Asunto(s)
Clostridioides difficile/inmunología , Enterocolitis Seudomembranosa/microbiología , Microbioma Gastrointestinal , Inmunidad Adaptativa , Animales , Colitis/inmunología , Colitis/microbiología , Colitis/terapia , Susceptibilidad a Enfermedades , Enterocolitis Seudomembranosa/inmunología , Enterocolitis Seudomembranosa/terapia , Trasplante de Microbiota Fecal , Humanos , Inmunidad Innata
10.
Cell Host Microbe ; 18(1): 5-6, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26159713

RESUMEN

The contribution of the innate immune response to the resolution of Clostridium difficile infection (CDI) remains incompletely defined. In this issue of Cell Host & Microbe, Abt et al. demonstrated that innate lymphoid cells and the effector cytokine IFN-γ are important for recovery from the acute phase of CDI.


Asunto(s)
Clostridioides difficile/inmunología , Infecciones por Clostridium/inmunología , Resistencia a la Enfermedad , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Animales
11.
mBio ; 6(1)2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25626905

RESUMEN

UNLABELLED: Clostridium difficile is the most common hospital-acquired pathogen, causing antibiotic-associated diarrhea in over 250,000 patients annually in the United States. Disease is primarily mediated by toxins A and B, which induce potent proinflammatory signaling in host cells and can activate an ASC-containing inflammasome. Recent findings suggest that the intensity of the host response to infection correlates with disease severity. Our lab has identified the proinflammatory cytokine interleukin-23 (IL-23) as a pathogenic mediator during C. difficile infection (CDI). The mechanisms by which C. difficile induces IL-23, however, are not well understood, and the role of toxins A and B in this process is unclear. Here, we show that toxins A and B alone are not sufficient for IL-23 production but synergistically increase the amount of IL-23 produced in response to MyD88-dependent danger signals, including pathogen-associated molecular patterns (PAMPs) and host-derived damage associated molecular patterns (DAMPs). Danger signals also enhanced the secretion of IL-1ß in response to toxins A and B, and subsequent IL-1 receptor signaling accounted for the majority of the increase in IL-23 that occurred in the presence of the toxins. Inhibition of inflammasome activation in the presence of extracellular K(+) likewise decreased IL-23 production. Finally, we found that IL-1ß was increased in the serum of patients with CDI, suggesting that this systemic response could influence downstream production of pathogenic IL-23. Identification of the synergy of danger signals with toxins A and B via inflammasome signaling represents a novel finding in the mechanistic understanding of C. difficile-induced inflammation. IMPORTANCE: Clostridium difficile is among the leading causes of death due to health care-associated infection, and factors determining disease severity are not well understood. C. difficile secretes toxins A and B, which cause inflammation and tissue damage, and recent findings suggest that some of this tissue damage may be due to an inappropriate host immune response. We have found that toxins A and B, in combination with both bacterium- and host-derived danger signals, can induce expression of the proinflammatory cytokines IL-1ß and IL-23. Our results demonstrate that IL-1ß signaling enhances IL-23 production and could lead to increased pathogenic inflammation during CDI.


Asunto(s)
Clostridioides difficile/inmunología , Infecciones por Clostridium/inmunología , Inflamasomas/inmunología , Interleucina-23/inmunología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Clostridioides difficile/genética , Clostridioides difficile/fisiología , Infecciones por Clostridium/genética , Infecciones por Clostridium/microbiología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Enterotoxinas/inmunología , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-23/genética
12.
PLoS One ; 9(2): e87740, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24503860

RESUMEN

Peripheral self-tolerance eliminates lymphocytes specific for tissue-specific antigens not encountered in the thymus. Recently, we demonstrated that lymphatic endothelial cells in mice directly express peripheral tissue antigens, including tyrosinase, and induce deletion of specific CD8 T cells via Programmed Death Ligand-1 (PD-L1). Here, we demonstrate that high-level expression of peripheral tissue antigens and PD-L1 is confined to lymphatic endothelial cells in lymph nodes, as opposed to tissue (diaphragm and colon) lymphatics. Lymphatic endothelial cells in the lymph node medullary sinus express the highest levels of peripheral tissue antigens and PD-L1, and are the only subpopulation that expresses tyrosinase epitope. The representation of lymphatic endothelial cells in the medullary sinus expressing high-level PD-L1, which is necessary for normal CD8 T cell deletion kinetics, is controlled by lymphotoxin-ß receptor signaling and B cells. Lymphatic endothelial cells from neonatal mice do not express high-level PD-L1 or present tyrosinase epitope. This work uncovers a critical role for the lymph node microenvironment in endowing lymphatic endothelial cells with potent tolerogenic properties.


Asunto(s)
Microambiente Celular , Células Endoteliales/metabolismo , Endotelio Linfático/metabolismo , Tolerancia Inmunológica , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Animales , Animales Recién Nacidos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Microambiente Celular/inmunología , Receptor beta de Linfotoxina/metabolismo , Ratones , Ratones Noqueados , Monofenol Monooxigenasa/metabolismo , Mucoproteínas , Fenotipo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
13.
Infect Immun ; 82(1): 341-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24166957

RESUMEN

The role of leptin in the mucosal immune response to Clostridium difficile colitis, a leading cause of nosocomial infection, was studied in humans and in a murine model. Previously, a mutation in the receptor for leptin (LEPR) was shown to be associated with susceptibility to infectious colitis and liver abscess due to Entamoeba histolytica as well as to bacterial peritonitis. Here we discovered that European Americans homozygous for the same LEPR Q223R mutation (rs1137101), known to result in decreased STAT3 signaling, were at increased risk of C. difficile infection (odds ratio, 3.03; P = 0.015). The mechanism of increased susceptibility was studied in a murine model. Mice lacking a functional leptin receptor (db/db) had decreased clearance of C. difficile from the gut lumen and diminished inflammation. Mutation of tyrosine 1138 in the intracellular domain of LepRb that mediates signaling through the STAT3/SOCS3 pathway also resulted in decreased mucosal chemokine and cell recruitment. Collectively, these data support a protective mucosal immune function for leptin in C. difficile colitis partially mediated by a leptin-STAT3 inflammatory pathway that is defective in the LEPR Q223R mutation. Identification of the role of leptin in protection from C. difficile offers the potential for host-directed therapy and demonstrates a connection between metabolism and immunity.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Colitis/microbiología , Leptina/fisiología , Receptores de Leptina/genética , Animales , Quimiocinas/metabolismo , Clostridioides difficile/genética , Clostridioides difficile/inmunología , Infecciones por Clostridium/genética , Infecciones por Clostridium/inmunología , Colitis/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Predisposición Genética a la Enfermedad , Mucosa Intestinal/inmunología , Intestinos/microbiología , Leptina/inmunología , Modelos Logísticos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oportunidad Relativa , Polimorfismo Genético , Receptores de Leptina/deficiencia , Factor de Transcripción STAT3 , Transducción de Señal/fisiología , Tirosina/genética
14.
J Infect Dis ; 208(6): 917-20, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23776194

RESUMEN

Clostridium difficile is currently the leading cause of hospital-acquired infections in the United States. Here, we observed increased interleukin 23 (IL-23) protein levels in human colon biopsy specimens positive for C. difficile toxins, compared with levels in negative controls (P = .008) We also investigated the role of IL-23 during C. difficile infection, using 2 distinct murine models. Mice lacking IL-23 signaling had a significant increase in survival (100% [12 mice]), compared with control mice (16.7%-50% [12 mice]). These data suggest a new potential drug target for human C. difficile treatment and indicate the first link between IL-23 and disease severity during murine infection.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium/inmunología , Colitis/inmunología , Interleucina-23/inmunología , Transducción de Señal , Animales , Biopsia , Colitis/microbiología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Manejo de Especímenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...