Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Prog Retin Eye Res ; 102: 101286, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38969166

RESUMEN

Single-cell RNA sequencing (scRNA-seq) has enabled the identification of novel gene signatures and cell heterogeneity in numerous tissues and diseases. Here we review the use of this technology for Fuchs' Endothelial Corneal Dystrophy (FECD). FECD is the most common indication for corneal endothelial transplantation worldwide. FECD is challenging to manage because it is genetically heterogenous, can be autosomal dominant or sporadic, and progress at different rates. Single-cell RNA sequencing has enabled the discovery of several FECD subtypes, each with associated gene signatures, and cell heterogeneity. Current FECD treatments are mainly surgical, with various Rho kinase (ROCK) inhibitors used to promote endothelial cell metabolism and proliferation following surgery. A range of emerging therapies for FECD including cell therapies, gene therapies, tissue engineered scaffolds, and pharmaceuticals are in preclinical and clinical trials. Unlike conventional disease management methods based on clinical presentations and family history, targeting FECD using scRNA-seq based precision-medicine has the potential to pinpoint the disease subtypes, mechanisms, stages, severities, and help clinicians in making the best decision for surgeries and the applications of therapeutics. In this review, we first discuss the feasibility and potential of using scRNA-seq in clinical diagnostics for FECD, highlight advances from the latest clinical treatments and emerging therapies for FECD, integrate scRNA-seq results and clinical notes from our FECD patients and discuss the potential of applying alternative therapies to manage these cases clinically.

2.
Clin Exp Ophthalmol ; 51(7): 663-672, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37608637

RESUMEN

BACKGROUND: To compare graft survival of endothelial keratoplasty (EK) versus penetrating keratoplasty (PK) in patients with iridocorneal endothelial (ICE) syndrome and identify ocular features associated with graft survival. METHODS: Observational, prospective, cohort study. A total of 30 806 first grafts performed between 1985 and 2020 were identified through the Australian Corneal Graft Registry and included in this observational, prospective cohort study. A total of 196 eyes underwent a primary corneal graft for ICE syndrome. Kaplan-Meier graft survival plots and Chi-squared tests were performed to identify graft survival rates for EK and PK. A history of raised intraocular pressure (IOP) was also recorded and analysed. Graft survival of eyes with ICE syndrome were compared to that of other indications. RESULTS: Grafts performed for ICE syndrome increased to 0.8% of all cases during the 2005 to 2020 period compared with 0.5% between 1985 to 2004 (χ2 =9.35, p = 0.002). From 2010, EK surpassed PK as the preferred graft type. Survival of primary grafts in eyes with ICE syndrome was lower than for other indications (log-rank = 56.62, p < 0.001). Graft survival was higher following PK than Descemet stripping (automated) endothelial keratoplasty (DS(A)EK) (log-rank = 10.56, p = 0.001). Graft survival was higher in eyes without a history of raised IOP compared to those with a reported history of raised IOP (log-rank = 13.06, p < 0.001). CONCLUSIONS: ICE syndrome carries a poor prognosis for graft survival. DS(A)EK had a poorer prognosis than PK. A history of raised IOP is associated with higher risk of graft failure.


Asunto(s)
Enfermedades de la Córnea , Queratoplastia Endotelial de la Lámina Limitante Posterior , Distrofia Endotelial de Fuchs , Glaucoma , Síndrome Endotelial Iridocorneal , Humanos , Síndrome Endotelial Iridocorneal/diagnóstico , Síndrome Endotelial Iridocorneal/cirugía , Síndrome Endotelial Iridocorneal/complicaciones , Estudios Prospectivos , Distrofia Endotelial de Fuchs/cirugía , Estudios de Cohortes , Agudeza Visual , Australia , Endotelio Corneal/cirugía , Queratoplastia Penetrante , Glaucoma/cirugía , Sistema de Registros , Supervivencia de Injerto , Estudios Retrospectivos , Enfermedades de la Córnea/cirugía , Enfermedades de la Córnea/complicaciones
3.
Pharmacol Ther ; 242: 108349, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36682466

RESUMEN

Corneal diseases are one of the leading causes of moderate-to-severe visual impairment and blindness worldwide, after glaucoma, cataract, and retinal disease in overall importance. Given its tendency to affect people at a younger age than other blinding conditions such as cataract and glaucoma, corneal scarring poses a huge burden both on the individuals and society. Furthermore, corneal scarring and fibrosis disproportionately affects people in poorer and remote areas, making it a significant ophthalmic public health problem. Traditional medical strategies, such as topical corticosteroids, are not effective in preventing fibrosis or scars. Corneal transplantation, the only effective sight-restoring treatment for corneal scars, is curbed by challenges including a severe shortage of tissue, graft rejection, secondary conditions, cultural barriers, the lack of well-trained surgeons, operating rooms, and well-equipped infrastructures. Thanks to tremendous research efforts, emerging therapeutic options including gene therapy, protein therapy, cell therapy and novel molecules are in development to prevent the progression of corneal scarring and compliment the surgical options currently available for treating established corneal scars in clinics. In this article, we summarise the most relevant preclinical and clinical studies on emerging therapies for corneal scarring in recent years, showing how these approaches may prevent scarring in its early development.


Asunto(s)
Catarata , Enfermedades de la Córnea , Lesiones de la Cornea , Glaucoma , Humanos , Cicatriz/terapia , Cicatriz/complicaciones , Lesiones de la Cornea/terapia , Lesiones de la Cornea/complicaciones , Enfermedades de la Córnea/tratamiento farmacológico , Enfermedades de la Córnea/complicaciones , Glaucoma/complicaciones , Catarata/complicaciones
4.
Int J Mol Sci ; 23(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35806191

RESUMEN

Strategies that alter the pH of wounds to improve healing outcomes are an emerging area of interest. Currently, there is limited understanding of the effect of hydrogen (H+) on the functionality of skin cells during proliferation and migration, highlighting the need for research to determine the effect of pH during wound healing. This study aimed to determine the effect of acidification on the metabolic activity and migration of human immortalized keratinocytes (HaCaT) and human foreskin fibroblasts (HFF). In vitro models were used with phosphoric and citric acid buffers at a pH range between 3 and 7. Our results showed that cells were more viable in buffers with low rather than high ionic strength. A time-dependent effect of the acidification treatment was also observed with cell metabolic activity varying with treatment duration and frequency. Our results showed that a 24 h treatment and subsequent resting phase significantly improved cell proliferation and migration. This in vitro study is the first to establish a correlation between the role of acidic pH, molarity and treatment regimen in cellular activity. Our data demonstrated a positive effect of acidic pH on cell metabolic activity and migration rate, suggesting a clinical potential in indications such as wound healing.


Asunto(s)
Piel , Cicatrización de Heridas , Movimiento Celular , Proliferación Celular , Fibroblastos , Humanos , Queratinocitos/metabolismo , Piel/lesiones
5.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34948000

RESUMEN

Cutaneous squamous cell carcinoma (cSCC) accounts for 25% of cutaneous malignancies diagnosed in Caucasian populations. Surgical removal in combination with radiation and chemotherapy are effective treatments for cSCC. Nevertheless, the aggressive metastatic forms of cSCC still have a relatively poor patient outcome. Studies have linked actin cytoskeletal dynamics and the Wnt/ß-catenin signaling pathway as important modulators of cSCC pathogenesis. Previous studies have also shown that the actin-remodeling protein Flightless (Flii) is a negative regulator of cSCC. The aim of this study was to investigate if the functional effects of Flii on cSCC involve the Wnt/ß-catenin signaling pathway. Flii knockdown was performed using siRNA in a human late stage aggressive metastatic cSCC cell line (MET-1) alongside analysis of Flii genetic murine models of 3-methylcholanthrene induced cSCC. Flii was increased in a MET-1 cSCC cell line and reducing Flii expression led to fewer PCNA positive cells and a concomitant reduction in cellular proliferation and symmetrical division. Knockdown of Flii led to decreased ß-catenin and a decrease in the expression of the downstream effector of ß-catenin signaling protein SOX9. 3-Methylcholanthrene (MCA)-induced cSCC in Flii overexpressing mice showed increased markers of cancer metastasis including talin and keratin-14 and a significant increase in SOX9 alongside a reduction in Flii associated protein (Flap-1). Taken together, this study demonstrates a role for Flii in regulating proteins involved in cSCC proliferation and tumor progression and suggests a potential role for Flii in aggressive metastatic cSCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Proteínas de Microfilamentos/genética , Neoplasias Cutáneas/genética , Transactivadores/genética , Regulación hacia Arriba , Vía de Señalización Wnt , Animales , Carcinoma de Células Escamosas/inducido químicamente , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Metilcolantreno/efectos adversos , Ratones , Neoplasias Cutáneas/inducido químicamente
6.
Int J Mol Sci ; 22(15)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34361001

RESUMEN

Epidermal progenitor cells divide symmetrically and asymmetrically to form stratified epidermis and hair follicles during late embryonic development. Flightless I (Flii), an actin remodelling protein, is implicated in Wnt/ß-cat and integrin signalling pathways that govern cell division. This study investigated the effect of altering Flii on the divisional orientation of epidermal progenitor cells (EpSCs) in the basal layer during late murine embryonic development and early adolescence. The effect of altering Flii expression on asymmetric vs. symmetric division was assessed in vitro in adult human primary keratinocytes and in vivo at late embryonic development stages (E16, E17 and E19) as well as adolescence (P21 day-old) in mice with altered Flii expression (Flii knockdown: Flii+/-, wild type: WT, transgenic Flii overexpressing: FliiTg/Tg) using Western blot and immunohistochemistry. Flii+/- embryonic skin showed increased asymmetrical cell division of EpSCs with an increase in epidermal stratification and elevated talin, activated-Itgb1 and Par3 expression. FliiTg/Tg led to increased symmetrical cell division of EpSCs with increased cell proliferation rate, an elevated epidermal SOX9, Flap1 and ß-cat expression, a thinner epidermis, but increased hair follicle number and depth. Flii promotes symmetric division of epidermal progenitor cells during murine embryonic development.


Asunto(s)
División Celular , Proteínas de Microfilamentos/genética , Células Madre Embrionarias de Ratones/metabolismo , Piel/metabolismo , Transactivadores/genética , Animales , Células Cultivadas , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/metabolismo , Células Madre Embrionarias de Ratones/citología , Factor de Transcripción SOX9/metabolismo , Piel/embriología , Transactivadores/metabolismo , beta Catenina/metabolismo
7.
Adv Wound Care (New Rochelle) ; 9(4): 161-173, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32117580

RESUMEN

Objective: Activation of epidermal stem cells (EpSCs) from their quiescent niche is an integral component of wound reepithelialization and involves Wnt/ß-catenin (ß-Cat) signaling and remodeling of the actin cytoskeleton. The aim of this study was to investigate the effect of Flightless I (Flii), a cytoskeletal protein and inhibitor of wound healing, on EpSC activation during wound repair. Approach: Genetically modified Flii mice (Flii knockdown: Flii+/- , wild type: WT, Flii overexpressing: FliiTg/Tg ) received two incisional wounds along the lateral axis of the dorsal skin. Indicators of EpSC activation (epidermal growth factor receptor 1 [EGFR1], leucine-rich repeats and immunoglobulin-like domains-1 [Lrig1], K14), Wnt/ß-Cat signaling (Lgr6, Flap2, ß-Cat, and axis inhibition protein 2 [Axin2]), and cell proliferation (proliferating cell nuclear antigen [PCNA]) were assessed using immunohistochemistry. ß-Cat stabilization was examined using western blotting with cell cycling and differentiation of isolated CD34+ITGA6high EpSCs examined using real time-quantitative polymerase chain reaction after treatment with wound-conditioned media. Results:Flii+/- led to increased numbers of activated EpSCs expressing PCNA, elevated EGFR1, and decreased Lrig1. EpSCs in Flii+/- hair follicle niches adjacent to the wounds also showed expression of Wnt-activation markers including increased ß-Cat and Lgr6, and decreased Axin2. EpSCs (CD34+ITGA6high) isolated from Flii+/- unwounded skin showed elevated expression of cell-cycling genes including ΔNp63, filaggrin (Fila), involucrin (Invo), cyclin D1 (Ccnd1), and cell-division cycle protein-20 (Cdc20); and elevated ΔNp63 and Invo after treatment with wound-conditioned media compared with WT and FliiTg/Tg counterparts. Innovation: Flii was identified as an inhibitor of EpSC activation that may explain its negative effects on wound reepithelialization. Conclusion: Flii may inhibit EpSC activation by interrupting Wnt/ß-Cat signaling. Strategies that reduce Flii may increase activation of EpSCs and promote reepithelialization of wounds.


Asunto(s)
Células Epidérmicas/metabolismo , Proteínas de Microfilamentos/metabolismo , Nicho de Células Madre/genética , Células Madre/metabolismo , Transactivadores/metabolismo , Cicatrización de Heridas/genética , Animales , Proliferación Celular/genética , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Piel/lesiones , Piel/metabolismo , Transactivadores/genética , Vía de Señalización Wnt/genética , beta Catenina/metabolismo
9.
Front Immunol ; 9: 1833, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30147695

RESUMEN

Atopic dermatitis (AD) is a chronic pruritic inflammatory skin disease characterized by excessive inflammation and disrupted skin barrier function. Although the etiology of AD is not completely understood, clinical and basic studies suggest increasing involvement of autoantibodies against intracellular proteins. An actin remodeling protein, Flightless I (Flii), has been shown to promote development of inflammatory mediated skin conditions and impairment of skin barrier development and function. Here, we sought to determine the effect of altering Flii expression on the development of AD and its contribution to autoimmune aspects of inflammatory skin conditions. Ovalbumin (OVA)-induced AD skin-like disease was induced in Flii heterozygous (Flii+/- ), wild-type (Flii+/+ ), and Flii transgenic (FliiTg/Tg ) mice by epicutaneous exposure to OVA for 3 weeks; each week was separated by 2-week resting period. Reduced Flii expression resulted in decreased disease severity and tissue inflammation as determined by histology, lymphocytic, and mast cell infiltrate and increased anti-inflammatory IL-10 cytokine levels and a marked IFN-γ Th1 response. In contrast, Flii over-expression lead to a Th2 skewed response characterized by increased pro-inflammatory TNF-α cytokine production, Th2 chemokine levels, and Th2 cell numbers. Sera from OVA-induced AD skin-like disease Flii+/- mice showed a decreased level of autoreactivity while sera from FliiTg/Tg mice counterparts showed an altered autoantibody profile with strong nuclear localization favoring development of a more severe disease. These findings demonstrate autoimmune responses in this model of OVA-induced AD-like skin disease and suggest that Flii is a novel target, whose manipulation could be a potential approach for the treatment of patients with AD.


Asunto(s)
Dermatitis Atópica/inmunología , Inflamación/inmunología , Proteínas de Microfilamentos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Piel/inmunología , Células TH1/inmunología , Células Th2/inmunología , Alérgenos/inmunología , Animales , Autoanticuerpos/sangre , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas de Microfilamentos/genética , Ovalbúmina/inmunología , Receptores Citoplasmáticos y Nucleares/genética , Transactivadores
10.
Int J Mol Sci ; 19(7)2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29996558

RESUMEN

Wound healing is an increasing clinical problem involving substantial morbidity, mortality, and rising health care costs. Leucine-rich repeat flightless-interacting protein-1 (LRRFIP-1) regulates toll-like receptor (TLR)-mediated inflammation, suggesting a potential role in the healing of wounds. We sought to determine the role of LRRFIP-1 in wound repair and whether the exogenous addition of recombinant LRRFIP-1 (rLRRFIP-1) affected healing responses. Using a model of full-thickness incisional acute wounds in BALB/c mice, we investigated the effect of wounding on LRRFIP-1 expression. The effect of rLRRFIP-1 on cellular proliferation, inflammation, and collagen deposition was also investigated. LRRFIP-1 was upregulated in response to wounding, was found to directly associate with flightless I (Flii), and significantly increased cellular proliferation both in vitro and in vivo. rLRRFIP-1 reduced Flii expression in wounds in vivo and resulted in significantly improved healing with a concurrent dampening of TLR4-mediated inflammation and improved collagen deposition. Additionally, decreased levels of TGF-ß1 and increased levels of TGF-ß3 were observed in rLRRFIP-1-treated wounds suggesting a possible antiscarring effect of rLRRFIP-1. Further studies are required to elucidate if the mechanisms behind LRRFIP-1 action in wound repair are independent of Flii. However, these results identify rLRRFIP-1 as a possible treatment modality for improved healing of acute wounds.


Asunto(s)
Inflamación/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Piel/lesiones , Cicatrización de Heridas , Animales , Línea Celular , Proliferación Celular , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/metabolismo , Receptor Toll-Like 4/metabolismo , Receptores Toll-Like/metabolismo , Transactivadores , Regulación hacia Arriba
11.
Stem Cells Dev ; 25(10): 749-59, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27021878

RESUMEN

Cutaneous stem cells (CSCs) orchestrate the homeostasis and regeneration of mammalian skin. Epithelial CSCs have been isolated and characterized from the skin and hold great potential for tissue engineering and clinical applications. The actin cytoskeleton is known to regulate cell adhesion and motility through its intricate participation in signal transduction and structural modifications. The dynamics of actin cytoskeleton can directly influence CSCs behaviors including tissue morphogenesis, homeostasis, niche maintenance, activation, and wound repair. Various regulators of the actin cytoskeleton including kinases, actin-remodeling proteins, paracrine signals, and micro-RNAs collaborate and contribute to epithelial CSC proliferation, adhesion, and differentiation. This review brings together the latest mechanistic insights into how the actin cytoskeleton participates in the regulation of epithelial CSCs during development, homeostasis, and wound repair.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Células Epiteliales/citología , Piel/citología , Células Madre/citología , Animales , Humanos , Trasplante de Células Madre
12.
Oncotarget ; 6(34): 36426-40, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26497552

RESUMEN

Flightless I (Flii) is an actin remodeling protein that affects cellular processes including adhesion, proliferation and migration. In order to determine the role of Flii during carcinogenesis, squamous cell carcinomas (SCCs) were induced in Flii heterozygous (Flii+/-), wild-type and Flii overexpressing (FliiTg/Tg) mice by intradermal injection of 3-methylcholanthrene (MCA). Flii levels were further assessed in biopsies from human SCCs and the human SCC cell line (MET-1) was used to determine the effect of Flii on cellular invasion. Flii was highly expressed in human SCC biopsies particularly by the invading cells at the tumor edge. FliiTg/Tg mice developed large, aggressive SCCs in response to MCA. In contrast Flii+/- mice had significantly smaller tumors that were less invasive. Intradermal injection of Flii neutralizing antibodies during SCC initiation and progression significantly reduced the size of the tumors and, in vitro, decreased cellular sphere formation and invasion. Analysis of the tumors from the Flii overexpressing mice showed reduced caspase I and annexin V expression suggesting Flii may negatively regulate apoptosis within these tumors. These studies therefore suggest that Flii enhances SCC tumor progression by decreasing apoptosis and enhancing tumor cell invasion. Targeting Flii may be a potential strategy for reducing the severity of SCCs.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Microfilamentos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Animales , Apoptosis/fisiología , Carcinoma de Células Escamosas/genética , Proteínas Portadoras , Proteínas del Citoesqueleto/genética , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/genética , Receptores Citoplasmáticos y Nucleares/genética , Neoplasias Cutáneas/genética , Transactivadores
13.
J Pathol ; 232(5): 541-52, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24375017

RESUMEN

Development of an intact epidermis is critical for maintaining the integrity of the skin. Patients with epidermolysis bullosa (EB) experience multiple erosions, which breach the epidermal barrier and lead to increased microbial colocalization of wounds, infections and sepsis. The cytoskeletal protein Flightless I (Flii) is a known regulator of both development and wound healing. Using Flii(+/-), WT and Flii(Tg/Tg) mice, we investigated the effect of altering Flii levels in embryos and adult mice on the development of the epidermal barrier and, consequently, how this affects the integrity of the skin in EB. Flii over-expression resulted in delayed formation of the epidermal barrier in embryos and decreased expression of tight junction (TJ) proteins Claudin-1 and ZO-2. Increased intercellular space and transepidermal water loss was observed in Flii(Tg)(/Tg) adult mouse skin, while Flii(Tg/Tg) keratinocytes showed altered TJ protein localization and reduced transepithelial resistance. Flii is increased in the blistered skin of patients with EB, and over-expression of Flii in experimental EBA showed impaired Claudin-1 and -4 TJ protein expression and delayed recovery of functional barrier post-blistering. Immunoprecipitation confirmed Flii associated with TJ proteins and in vivo actin assays showed that the effect of Flii on actin polymerization underpinned the impaired barrier function observed in Flii(Tg/Tg) mice. These results therefore demonstrate an important role for Flii in the development and regulation of the epidermal barrier, which may contribute to the impaired healing and skin fragility of EB patients.


Asunto(s)
Vesícula/metabolismo , Epidermis/metabolismo , Epidermólisis Ampollosa/metabolismo , Proteína Proto-Oncogénica c-fli-1/deficiencia , Proteína Proto-Oncogénica c-fli-1/metabolismo , Cicatrización de Heridas , Actinas/metabolismo , Animales , Vesícula/genética , Vesícula/patología , Células Cultivadas , Modelos Animales de Enfermedad , Impedancia Eléctrica , Epidermis/patología , Epidermólisis Ampollosa/genética , Epidermólisis Ampollosa/patología , Genotipo , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Permeabilidad , Fenotipo , Polimerizacion , Proteína Proto-Oncogénica c-fli-1/genética , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Factores de Tiempo , Regulación hacia Arriba , Pérdida Insensible de Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...