Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Reprod Med Biol ; 22(1): e12544, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37810897

RESUMEN

The contribution of Prominin-1 (aka CD133) to male fertility has recently been (re)investigated, with contradictory results. Early findings, essential for deciphering its role, have unfortunately been neglected. Here, the authors present what is currently known about its expression in the male reproductive system of rodents and men so that its involvement in male fertility can be re-examined and discussed in the light of these elements.

2.
Ther Apher Dial ; 26 Suppl 1: 29-34, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36468302

RESUMEN

Low-density lipoprotein (LDL) apheresis is effective and safe for patients with diabetes, proteinuria, and dyslipidemia. Diabetes mellitus is accompanied by ocular microvascular complications like retinal neovascularization or diabetic macular edema. These are leading causes of blindness and can be mediated by abnormal vessel growth and increased vascular permeability due to elevated levels of vascular endothelial growth factor (VEGF) in diabetic patients. In this study, we established methods to study the expression of different VEGF isoforms in human retinal and endothelial cells. The VEGF-A165a isoform is much higher expressed in retinal cells, compared to endothelial cells. Stimulation with glyoxal as a model of oxidative stress under diabetic conditions lead to a pronounced induction of VEGF-A165a in human retinal and endothelial cells. These data suggest that diabetes and oxidative stress induce VEGF-A isoforms which could be relevant in regulating the ingrowths of novel blood vessels into the retina in diabetic patients.


Asunto(s)
Retinopatía Diabética , Edema Macular , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Retinopatía Diabética/terapia , Retinopatía Diabética/etiología , Retinopatía Diabética/metabolismo , Células Endoteliales/metabolismo , Glioxal/farmacología , Glioxal/metabolismo , Retina/metabolismo , Isoformas de Proteínas/metabolismo
3.
Biomedicines ; 10(2)2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-35203414

RESUMEN

Anti-Vascular Endothelial Growth Factor (VEGF) agents are the first-line treatment for retinal neovascular diseases, which represent the most prevalent causes of acquired vision loss world-wide. VEGF-Trap (Aflibercept, AFL), a recombinant decoy receptor recognizing ligands of both VEGFR-1 and -2, was recently reported to be highly efficient in improving visual acuity and preserving retinal anatomy in individuals affected by diabetic macular edema. However, the precise molecular and cell biological mechanisms underlying the beneficial effects of this novel tool have yet to be elucidated. Using the mouse oxygen-induced retinopathy (OIR) model as a surrogate of retinopathies with sterile post-ischemic inflammation, such as late proliferative diabetic retinopathy (PDR), retinopathy of prematurity (ROP), and diabetic macular edema (DME), we provide evidence that AFL modulates inflammation in response to hypoxia by regulating the morphology of microglial cells, a parameter commonly used as a proxy for changes in their activation state. We show that AFL administration during the hypoxic period of OIR leads to an increased number of ramified Iba1+ microglial cells/macrophages while subsequently limiting the accumulation of these cells in particular retinal layers. Our results suggest that, beyond its well-documented beneficial effects on microvascular regeneration, AFL might exert important modulatory effects on post-ischemic retinal inflammation.

4.
Sci Rep ; 11(1): 15313, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321516

RESUMEN

Ischemic retinal dystrophies are leading causes of acquired vision loss. Although the dysregulated expression of the hypoxia-responsive VEGF-A is a major driver of ischemic retinopathies, implication of additional VEGF-family members in their pathogenesis has led to the development of multivalent anti-angiogenic tools. Designed as a decoy receptor for all ligands of VEGFR1 and VEGFR2, Aflibercept is a potent anti-angiogenic agent. Notwithstanding, the molecular mechanisms mediating Aflibercept's efficacy remain only partially understood. Here, we used the oxygen-induced retinopathy (OIR) mouse as a model system of pathological retinal vascularization to investigate the transcriptional response of the murine retina to hypoxia and of the OIR retina to Aflibercept. While OIR severely impaired transcriptional changes normally ensuing during retinal development, analysis of gene expression patterns hinted at alterations in leukocyte recruitment during the recovery phase of the OIR protocol. Moreover, the levels of Angiopoietin-2, a major player in the progression of diabetic retinopathy, were elevated in OIR tissues and consistently downregulated by Aflibercept. Notably, GO term, KEGG pathway enrichment, and expression dynamics analyses revealed that, beyond regulating angiogenic processes, Aflibercept also modulated inflammation and supported synaptic transmission. Altogether, our findings delineate novel mechanisms potentially underlying Aflibercept's efficacy against ischemic retinopathies.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Proteínas del Ojo/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Isquemia/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Retina/efectos de los fármacos , Vasos Retinianos , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Quimiotaxis de Leucocito/genética , Retinopatía Diabética , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Proteínas del Ojo/genética , Ontología de Genes , Redes Reguladoras de Genes , Isquemia/genética , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/genética , Oxígeno/metabolismo , Oxígeno/toxicidad , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Retina/metabolismo , Retinopatía de la Prematuridad , Transcripción Genética/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/fisiología
5.
J Biol Chem ; 295(18): 6007-6022, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32201384

RESUMEN

Prominins (proms) are transmembrane glycoproteins conserved throughout the animal kingdom. They are associated with plasma membrane protrusions, such as primary cilia, as well as extracellular vesicles derived thereof. Primary cilia host numerous signaling pathways affected in diseases known as ciliopathies. Human PROM1 (CD133) is detected in both somatic and cancer stem cells and is also expressed in terminally differentiated epithelial and photoreceptor cells. Genetic mutations in the PROM1 gene result in retinal degeneration by impairing the proper formation of the outer segment of photoreceptors, a modified cilium. Here, we investigated the impact of proms on two distinct examples of ciliogenesis. First, we demonstrate that the overexpression of a dominant-negative mutant variant of human PROM1 (i.e. mutation Y819F/Y828F) significantly decreases ciliary length in Madin-Darby canine kidney cells. These results contrast strongly to the previously observed enhancing effect of WT PROM1 on ciliary length. Mechanistically, the mutation impeded the interaction of PROM1 with ADP-ribosylation factor-like protein 13B, a key regulator of ciliary length. Second, we observed that in vivo knockdown of prom3 in zebrafish alters the number and length of monocilia in the Kupffer's vesicle, resulting in molecular and anatomical defects in the left-right asymmetry. These distinct loss-of-function approaches in two biological systems reveal that prom proteins are critical for the integrity and function of cilia. Our data provide new insights into ciliogenesis and might be of particular interest for investigations of the etiologies of ciliopathies.


Asunto(s)
Antígeno AC133/metabolismo , Cilios/metabolismo , Pez Cebra , Antígeno AC133/química , Antígeno AC133/genética , Animales , Perros , Regulación hacia Abajo , Regulación del Desarrollo de la Expresión Génica , Espacio Intracelular/metabolismo , Macrófagos del Hígado/citología , Células de Riñón Canino Madin Darby , Mutación , Transporte de Proteínas , Tirosina
6.
J Neurochem ; 153(3): 390-412, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31550048

RESUMEN

Retinal hypoxia triggers abnormal vessel growth and microvascular hyper-permeability in ischemic retinopathies. Whereas vascular endothelial growth factor A (VEGF-A) inhibitors significantly hinder disease progression, their benefits to retinal neurons remain poorly understood. Similar to humans, oxygen-induced retinopathy (OIR) mice exhibit severe retinal microvascular malformations and profound neuronal dysfunction. OIR mice are thus a phenocopy of human retinopathy of prematurity, and a proxy for investigating advanced stages of proliferative diabetic retinopathy. Hence, the OIR model offers an excellent platform for assessing morpho-functional responses of the ischemic retina to anti-angiogenic therapies. Using this model, we investigated the retinal responses to VEGF-Trap (Aflibercept), an anti-angiogenic agent recognizing ligands of VEGF receptors 1 and 2 that possesses regulatory approval for the treatment of neovascular age-related macular degeneration, macular edema secondary to retinal vein occlusion and diabetic macular edema. Our results indicate that Aflibercept not only reduces the severity of retinal microvascular aberrations but also significantly improves neuroretinal function. Aflibercept administration significantly enhanced light-responsiveness, as revealed by electroretinographic examinations, and led to increased numbers of dopaminergic amacrine cells. Additionally, retinal transcriptional profiling revealed the concerted regulation of both angiogenic and neuronal targets, including transcripts encoding subunits of transmitter receptors relevant to amacrine cell function. Thus, Aflibercept represents a promising therapeutic alternative for the treatment of further progressive ischemic retinal neurovasculopathies beyond the set of disease conditions for which it has regulatory approval. Cover Image for this issue: doi: 10.1111/jnc.14743.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Microvasos/efectos de los fármacos , Red Nerviosa/efectos de los fármacos , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Degeneración Retiniana/tratamiento farmacológico , Vasos Retinianos/efectos de los fármacos , Animales , Animales Recién Nacidos , Neuronas Dopaminérgicas/patología , Femenino , Isquemia/tratamiento farmacológico , Isquemia/patología , Masculino , Ratones , Microvasos/patología , Red Nerviosa/patología , Proteínas Recombinantes de Fusión/farmacología , Degeneración Retiniana/patología , Vasos Retinianos/patología , Sistema Vasomotor/efectos de los fármacos , Sistema Vasomotor/patología
8.
Cells ; 8(9)2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31540455

RESUMEN

Excessive abnormal angiogenesis plays a pivotal role in tumor progression and is a hallmark of solid tumors. This process is driven by an imbalance between pro- and anti-angiogenic factors dominated by the tissue hypoxia-triggered overproduction of vascular endothelial growth factor (VEGF). VEGF-mediated signaling has quickly become one of the most promising anti-angiogenic therapeutic targets in oncology. Nevertheless, the clinical efficacy of this approach is severely limited in certain tumor types or shows only transient efficacy in patients. Acquired or intrinsic therapy resistance associated with anti-VEGF monotherapeutic approaches indicates the necessity of a paradigm change when targeting neoangiogenesis in solid tumors. In this context, the elaboration of the conceptual framework of "vessel normalization" might be a promising approach to increase the efficacy of anti-angiogenic therapies and the survival rates of patients. Indeed, the promotion of vessel maturation instead of regressing tumors by vaso-obliteration could result in reduced tumor hypoxia and improved drug delivery. The implementation of such anti-angiogenic strategies, however, faces several pitfalls due to the potential involvement of multiple pro-angiogenic factors and modulatory effects of the innate and adaptive immune system. Thus, effective treatments bypassing relapses associated with anti-VEGF monotherapies or breaking the intrinsic therapy resistance of solid tumors might use combination therapies or agents with a multimodal mode of action. This review enumerates some of the current approaches and possible future directions of treating solid tumors by targeting neovascularization.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Factores de Crecimiento Endotelial Vascular/farmacología , Animales , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología
10.
Int J Radiat Biol ; 90(8): 700-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24512568

RESUMEN

PURPOSE: In this work we examined the presence of the neural stem cell biomarker Hairy and Enhancer of Split 3 (Hes3) in the anterior eye segment and in the aberrant growth condition of the conjunctiva pterygium. Further, we studied the response of Hes3 to irradiation. MATERIALS AND METHODS: Adult mouse and human corneoscleral junction and conjunctiva, as well as human pterygium were prepared for immunohistochemical detection of Hes3 and other markers. Total body irradiation was used to study the changes in the pattern of Hes3 expression. RESULTS: The adult rodent and human eye as well as pterygium, contain a population of cells expressing Hes3. In the human eye, Hes3-expressing (Hes3+) cells are found predominantly in the subconjunctival space spanning over the limbus where they physically associate with blood vessels. The cytoarchitecture of Hes3 + cells is similar to those previously observed in the adult central nervous system. Furthermore, irradiation reduces the number of Hes3 + cells in the subconjunctival space. In contrast, irradiation strongly promotes the nuclear localization of Hes3 in the ciliary body epithelium. CONCLUSIONS: Our results suggest that a recently identified signal transduction pathway that regulates neural stem cells and glioblastoma cancer stem cells also operates in the ocular surface, ciliary body, and in pterygium.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al ADN/metabolismo , Ojo/metabolismo , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Pterigion/metabolismo , Factores de Transcripción/metabolismo , Animales , Conjuntiva/efectos de los fármacos , Conjuntiva/metabolismo , Conjuntiva/efectos de la radiación , Ojo/irrigación sanguínea , Ojo/efectos de los fármacos , Ojo/efectos de la radiación , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Ratones , Terapia Molecular Dirigida , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/efectos de la radiación , Pterigion/tratamiento farmacológico , Pterigion/fisiopatología , Proteínas Represoras
11.
PLoS One ; 8(5): e63457, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23723983

RESUMEN

BACKGROUND: In mammals, embryonic neural progenitors as well as adult neural stem cells can be prospectively isolated based on the cell surface expression of prominin-1 (CD133), a plasma membrane glycoprotein. In contrast, characterization of neural progenitors in non-mammalian vertebrates endowed with significant constitutive neurogenesis and inherent self-repair ability is hampered by the lack of suitable cell surface markers. Here, we have investigated whether prominin-1-orthologues of the major non-mammalian vertebrate model organisms show any degree of conservation as for their association with neurogenic geminative zones within the central nervous system (CNS) as they do in mammals or associated with activated neural progenitors during provoked neurogenesis in the regenerating CNS. METHODS: We have recently identified prominin-1 orthologues from zebrafish, axolotl and chicken. The spatial distribution of prominin-1-positive cells--in comparison to those of mice--was mapped in the intact brain in these organisms by non-radioactive in situ hybridization combined with detection of proliferating neural progenitors, marked either by proliferating cell nuclear antigen or 5-bromo-deoxyuridine. Furthermore, distribution of prominin-1 transcripts was investigated in the regenerating spinal cord of injured axolotl. RESULTS: Remarkably, a conserved association of prominin-1 with germinative zones of the CNS was uncovered as manifested in a significant co-localization with cell proliferation markers during normal constitutive neurogenesis in all species investigated. Moreover, an enhanced expression of prominin-1 became evident associated with provoked, compensatory neurogenesis during the epimorphic regeneration of the axolotl spinal cord. Interestingly, significant prominin-1-expressing cell populations were also detected at distinct extraventricular (parenchymal) locations in the CNS of all vertebrate species being suggestive of further, non-neurogenic neural function(s). CONCLUSION/INTERPRETATION: Collectively, our work provides the first data set describing a comparative analysis of prominin-1-positive progenitor cells across species establishing a framework for further functional characterization in the context of regeneration.


Asunto(s)
Antígenos CD/metabolismo , Encéfalo/citología , Glicoproteínas/metabolismo , Células-Madre Neurales/metabolismo , Péptidos/metabolismo , Antígeno AC133 , Ambystoma mexicanum , Animales , Antígenos CD/genética , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Embrión de Pollo , Femenino , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/genética , Ratones , Placa Neural/citología , Tubo Neural/citología , Especificidad de Órganos , Péptidos/genética , Regeneración , Médula Espinal/fisiología , Regulación hacia Arriba , Pez Cebra
12.
Cells Tissues Organs ; 197(5): 344-59, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23571553

RESUMEN

Perivascular cells of microvascular niches are the prime candidates for being a reservoire of mesenchymal stem cell (MSC)-like cells in many tissues and organs that could serve as a potential source of cells and a target of novel cell-based therapeutic approaches. In the present study, by utilising typical markers of pericytes (neuronal-glial antigen 2, NG2, a chondroitin sulphate proteoglycan) and those of MSCs (CD146 and CD105) and primitive pluripotent cells (sex-determining region Y-box 2, Sox2), the phenotypic traits and the distribution of murine and rat retinal perivascular cells were investigated in situ. Our findings indicate that retinal microvessels of juvenile rodents are highly covered by NG2-positive branching processes of pericytic (perivascular) cells that are less prominent in mature capillary networks of the adult retina. In the adult rodent retinal vascular bed, NG2 labeling is mainly confined to membranes of the cell body resulting in a pearl-chain-like distribution along the vessels. Retinal pericytes, which were identified by their morphology and NG2 expression, simultaneously express CD146. Furthermore, CD146-positive cells located at small arteriole-to-capillary branching points appear more intensely stained than elsewhere. Evidence for a differential expression of the two markers around capillaries that would hint at a clonal heterogeneity among pericytic cells, however, is lacking. In contrast, the expression of CD105 is exclusively restricted to vascular endothelial cells and Sox2 is detected neither in perivascular nor in endothelial cells. In dissociated retinal cultures, however, simultaneous expression of NG2 and CD105 was observed. Collectively, our data indicate that vascular wall resident retinal pericytes share some phenotypic features (i.e. CD146 expression) with archetypal MSCs, which is even more striking in dissociated retinal cultures (i.e. CD105 expression). These findings might have implications for the treatment of retinal pathologies.


Asunto(s)
Capilares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Vasos Retinianos/citología , Envejecimiento/metabolismo , Animales , Antígenos/metabolismo , Biomarcadores/metabolismo , Antígeno CD146/metabolismo , Capilares/citología , Adhesión Celular , Linaje de la Célula , Crioultramicrotomía , Endotelio/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Pericitos/citología , Pericitos/metabolismo , Proteoglicanos/metabolismo , Ratas , Ratas Wistar , Factores de Transcripción SOXB1/metabolismo
13.
Adv Exp Med Biol ; 777: 3-24, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23161072

RESUMEN

Our knowledge of the first member of the prominin family is growing rapidly as the clinical value of prominin-1 (CD133) increases with its ever-wider use as a stem cell marker in normal and cancer tissues. Although the physiological function of this evolutionally conserved pentaspan membrane glycoprotein remains elusive, several studies have revealed new biological features regarding stem cells, cancer stem cells, and photoreceptors. The wide expression of CD133 in terminally differentiated epithelial cells, long overlooked by many authors, has attracted significant interest through the extensive investigation of human PROMININ-1 as a potential target for cancer therapies in various organs. Biochemically, this cholesterol-binding protein is selectively concentrated in plasma membrane protrusions, where it is associated with cholesterol-driven membrane microdomains. Clinically, mutations in the PROM1 gene are associated with various forms of retinal degeneration, which are mimicked in genetically modified mice carrying either a null allele or mutated form of PROMININ-1. In this introductory chapter, we attempted to review 15 years of prominin-1 study, focusing on its unique protein characteristics across species and the recent developments regarding its cell biology that may shed new light on its intriguing involvement in defining cancer-initiating cells.


Asunto(s)
Células Epiteliales , Glicoproteínas de Membrana , Animales , Diferenciación Celular , Células Epiteliales/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Microdominios de Membrana/metabolismo , Células Madre Neoplásicas/metabolismo
14.
World J Gastroenterol ; 17(36): 4149-52, 2011 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22039332

RESUMEN

Understanding all facets of membrane microdomains in normal and cancerous cells within the digestive tract is highly important, not only from a clinical point of view, but also in terms of our basic knowledge of cellular transformation. By studying the normal and cancer stem cell-associated molecule CD133 (prominin-1), novel aspects of the organization and dynamics of polarized epithelial cells have been revealed during the last decade. Its association with particular membrane microdomains is highly relevant in these contexts and might also offer new avenues in diagnosis and/or targeting of cancer stem cells.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Microdominios de Membrana/metabolismo , Humanos
15.
PLoS One ; 6(3): e17590, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21407811

RESUMEN

Besides being a marker of various somatic stem cells in mammals, prominin-1 (CD133) plays a role in maintaining the photoreceptor integrity since mutations in the PROM1 gene are linked with retinal degeneration. In spite of that, little information is available regarding its distribution in eyes of non-mammalian vertebrates endowed with high regenerative abilities. To address this subject, prominin-1 cognates were isolated from axolotl, zebrafish and chicken, and their retinal compartmentalization was investigated and compared to that of their mammalian orthologue. Interestingly, prominin-1 transcripts--except for the axolotl--were not strictly restricted to the outer nuclear layer (i.e., photoreceptor cells), but they also marked distinct subdivisions of the inner nuclear layer (INL). In zebrafish, where the prominin-1 gene is duplicated (i.e., prominin-1a and prominin-1b), a differential expression was noted for both paralogues within the INL being localized either to its vitreal or scleral subdivision, respectively. Interestingly, expression of prominin-1a within the former domain coincided with Pax-6-positive cells that are known to act as progenitors upon injury-induced retino-neurogenesis. A similar, but minute population of prominin-1-positive cells located at the vitreal side of the INL was also detected in developing and adult mice. In chicken, however, prominin-1-positive cells appeared to be aligned along the scleral side of the INL reminiscent of zebrafish prominin-1b. Taken together our data indicate that in addition to conserved expression of prominin-1 in photoreceptors, significant prominin-1-expressing non-photoreceptor retinal cell populations are present in the vertebrate eye that might represent potential sources of stem/progenitor cells for regenerative therapies.


Asunto(s)
Antígenos CD/metabolismo , Secuencia Conservada , Glicoproteínas/metabolismo , Péptidos/metabolismo , Retina/citología , Retina/metabolismo , Antígeno AC133 , Ambystoma mexicanum/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos CD/química , Embrión de Pollo , Pollos/metabolismo , Clonación Molecular , Glicoproteínas/química , Ratones , Datos de Secuencia Molecular , Péptidos/química , Estructura Terciaria de Proteína , Transporte de Proteínas , Especificidad de la Especie , Pez Cebra/metabolismo
16.
Histochem Cell Biol ; 133(5): 527-39, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20333396

RESUMEN

Prominin-1 (CD133) and its paralogue, prominin-2, are pentaspan membrane glycoproteins that are strongly expressed in the kidney where they have been originally cloned from. Previously, we have described the localization of prominin-1 in proximal tubules of the nephron. The spatial distribution of prominin-2, however, has not yet been documented in the kidney. We therefore examined the expression of this molecule along distinct tubular segments of the human and murine nephron using in situ hybridization and immunohistochemistry. Our findings indicated that human prominin-2 transcripts and protein were confined to distal tubules of the nephron including the thick ascending limb of Henle's loop and the distal convoluted tubule, the connecting duct and to the collecting duct system. Therein, this glycoprotein was enriched at the basolateral plasma membrane of the tubular epithelial cells with exception of the thick ascending limb where it was also found in the apical domain. This is in contrast with the exclusive apical localization of prominin-1 in epithelial cells of proximal nephron tubules. The distribution of murine prominin-2 transcripts was reminiscent of its human orthologue. In addition, a marked enrichment in the epithelium covering the papilla and in the urothelium of the renal pelvis was noted in mice. Finally, our biochemical analysis revealed that prominin-2 was released into the clinically healthy human urine as a constituent of small membrane vesicles. Collectively our data show the distribution and subcellular localization of prominin-2 within the kidney in situ and its release into the urine. Urinary detection of this protein might offer novel diagnostic approaches for studying renal diseases affecting distal segments of the nephron.


Asunto(s)
Túbulos Renales Colectores/metabolismo , Túbulos Renales Distales/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Antígeno AC133 , Animales , Antígenos CD/orina , Acuaporina 2/metabolismo , Calbindinas , Células Epiteliales/metabolismo , Expresión Génica/genética , Glicoproteínas/orina , Humanos , Corteza Renal/crecimiento & desarrollo , Corteza Renal/metabolismo , Médula Renal/crecimiento & desarrollo , Médula Renal/metabolismo , Pelvis Renal/crecimiento & desarrollo , Pelvis Renal/metabolismo , Glicoproteínas de Membrana/orina , Ratones , Ratones Endogámicos , Mucoproteínas/metabolismo , Nefronas/metabolismo , Péptidos/orina , Receptores de Droga/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12 , Miembro 3 de la Familia de Transportadores de Soluto 12 , Simportadores/metabolismo , Uromodulina , Urotelio/crecimiento & desarrollo , Urotelio/metabolismo
17.
J Neurosci ; 29(7): 2297-308, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19228982

RESUMEN

Prominin-1/CD133 (Prom-1) is a commonly used marker of neuronal, vascular, hematopoietic and other stem cells, yet little is known about its biological role and importance in vivo. Here, we show that loss of Prom-1 results in progressive degeneration of mature photoreceptors with complete loss of vision. Despite the expression of Prom-1 on endothelial progenitors, photoreceptor degeneration was not attributable to retinal vessel defects, but caused by intrinsic photoreceptor defects in disk formation, outer segment morphogenesis, and associated with visual pigment sorting and phototransduction abnormalities. These findings shed novel insight on how Prom-1 regulates neural retinal development and phototransduction in vertebrates.


Asunto(s)
Antígenos CD/genética , Glicoproteínas/genética , Péptidos/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/anomalías , Retina/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Antígeno AC133 , Animales , Colesterol/metabolismo , Regulación hacia Abajo/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Predisposición Genética a la Enfermedad/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Células Fotorreceptoras de Vertebrados/patología , Retina/fisiopatología , Arteria Retiniana/anomalías , Arteria Retiniana/fisiopatología , Degeneración Retiniana/fisiopatología , Pigmentos Retinianos/metabolismo , Células Madre/citología , Células Madre/metabolismo , Visión Ocular/genética
18.
Glia ; 57(8): 860-74, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19053060

RESUMEN

Prominin-1 (CD133) is a cholesterol-interacting pentaspan membrane glycoprotein specifically associated with plasma membrane protrusions. Prominin-1 is expressed by various stem and progenitor cells, notably neuroepithelial progenitors found in the developing embryonic brain. Here, we further investigated its expression in the murine brain. Biochemical analyses of brain membranes at early stages of development revealed the expression of two distinct splice variants of prominin-1, s1 and s3, which have different cytoplasmic C-terminal domains. The relative abundance of the s3 variant increased toward adulthood, whereas the opposite was observed for the s1 variant. Our combined in situ hybridization and immunohistochemistry revealed the expression of prominin-1 in a subpopulation of Olig-2-positive oligodendroglial cells present within white matter tracts of postnatal and adult brain. Furthermore, immunohistological and biochemical characterization suggested strongly that the s3 variant is a novel component of myelin. Consistent with this, the expression of prominin-1.s3 was significantly reduced in the brain of myelin-deficient mice. Finally, oligodendrocytes expressed selectively the s3 variant whereas GFAP-positive astrocytes expressed the s1 variant in primary glial cell cultures derived from embryonic brains. Collectively, our data demonstrate a complex expression pattern of prominin-1 molecules in developing adult brain. Given that prominin-1 is thought to act as an organizer of plasma membrane protrusions, they further suggest that a specific prominin-1 splice variant might play a role in morphogenesis and/or maintenance of the myelin sheath.


Asunto(s)
Antígenos CD/genética , Antígenos CD/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Neuroglía/metabolismo , Péptidos/genética , Péptidos/metabolismo , Antígeno AC133 , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Chlorocebus aethiops , Regulación del Desarrollo de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Riñón/citología , Ratones , Ratones Jimpy , Ratones Mutantes , Microscopía Inmunoelectrónica/métodos , Proteína Básica de Mielina/deficiencia , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/ultraestructura , Factor de Transcripción 2 de los Oligodendrocitos , Nervio Óptico/metabolismo , Nervio Óptico/ultraestructura , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transfección
19.
J Histochem Cytochem ; 56(11): 977-93, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18645205

RESUMEN

Human prominin-1 (CD133) is expressed by various stem and progenitor cells originating from diverse sources. In addition to stem cells, its mouse ortholog is expressed in a broad range of adult epithelial cells, where it is selectively concentrated in their apical domain. The lack of detection of prominin-1 in adult human epithelia might be explained, at least in part, by the specificity of the widely used AC133 antibody, which recognizes an epitope that seems dependent on glycosylation. Here we decided to re-examine its expression in adult human tissues, particularly in glandular epithelia, using a novel monoclonal antibody (80B258) generated against the human prominin-1 polypeptide. In examined tissues, we observed 80B258 immunoreactivity at the apical or apicolateral membranes of polarized cells. For instance, we found expression in secretory serous and mucous cells as well as intercalated ducts of the large salivary and lacrimal glands. In sweat glands including the gland of Moll, 80B258 immunoreactivity was found in the secretory (eccrine and apocrine glands) and duct (eccrine glands) portion. In the liver, 80B258 immunoreactivity was identified in the canals of Hering, bile ductules, and small interlobular bile ducts. In the uterus, we detected 80B258 immunoreactivity in endometrial and cervical glands. Together these data show that the overall expression of human prominin-1 is beyond the rare primitive cells, and it seems to be a general marker of apical or apicolateral membrane of glandular epithelia. This manuscript contains online supplemental material at http://www.jhc.org. Please visit this article online to view these materials.


Asunto(s)
Antígenos CD/biosíntesis , Glándulas Exocrinas/metabolismo , Glicoproteínas/biosíntesis , Hígado/metabolismo , Páncreas/metabolismo , Útero/metabolismo , Antígeno AC133 , Animales , Anticuerpos Monoclonales , Antígenos CD/inmunología , Células CHO , Células CACO-2 , Cricetinae , Cricetulus , Epitelio/metabolismo , Femenino , Citometría de Flujo , Glicoproteínas/inmunología , Humanos , Inmunohistoquímica , Microscopía Confocal , Especificidad de Órganos , Péptidos/inmunología
20.
Histochem Cell Biol ; 130(4): 749-59, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18536929

RESUMEN

Although the male reproductive system seems to be enriched in transcripts encoding for both Prominin genes, little is known about their spatial distribution in distinct segments of this organ system. This is especially true for the less-characterized second Prominin paralogue, Prominin-2. The present study, therefore, mainly examines the expression of Prominin-2 in male mice and reveals the existence of some crucial differences in the tissue compartmentalization of the two Prominin paralogues in the testis, epididymis, seminal vesicle, prostate and urinary bladder. Our in situ hybridization analysis demonstrates that the major domains of overlapping expression between the two Prominin genes are those compartments that are derived ontogenetically from the epigonadal mesonephric tubules, i.e. ductuli efferentes, or from the Wolffian-tube/ductus mesonephricus, for instance the corpus epididymidis and vesicula seminalis. In contrast, the sinus urogenitalis derivative urinary bladder epithelium expresses exclusively Prominin-2, but not Prominin-1 (CD133). The testis expresses only Prominin-1, not Prominin-2. In human prostate, we finally demonstrate that the expression of Prominin-2 (transcript and protein) is highly enriched in cells located in the basal compartment of the glandular epithelium where only a minute population was recently reported to be Prominin-1 positive. Taken together our data indicate that, except for the gonad, Prominin-2 is widely and abundantly expressed along the epithelia of various segments of the adult male genitourinary tract.


Asunto(s)
Genitales Masculinos/metabolismo , Glicoproteínas de Membrana/biosíntesis , Vejiga Urinaria/metabolismo , Antígeno AC133 , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Genitales Masculinos/citología , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Humanos , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Péptidos/genética , Vejiga Urinaria/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA