Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Reprod Med Biol ; 22(1): e12544, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37810897

RESUMO

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.
J Biol Chem ; 295(18): 6007-6022, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32201384

RESUMO

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.


Assuntos
Antígeno AC133/metabolismo , Cílios/metabolismo , Peixe-Zebra , Antígeno AC133/química , Antígeno AC133/genética , Animais , Cães , Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Espaço Intracelular/metabolismo , Células de Kupffer/citologia , Células Madin Darby de Rim Canino , Mutação , Transporte Proteico , Tirosina
3.
J Neurochem ; 153(3): 390-412, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31550048

RESUMO

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.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Degeneração Retiniana/tratamento farmacológico , Vasos Retinianos/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Neurônios Dopaminérgicos/patologia , Feminino , Isquemia/tratamento farmacológico , Isquemia/patologia , Masculino , Camundongos , Microvasos/patologia , Rede Nervosa/patologia , Proteínas Recombinantes de Fusão/farmacologia , Degeneração Retiniana/patologia , Vasos Retinianos/patologia , Sistema Vasomotor/efeitos dos fármacos , Sistema Vasomotor/patologia
5.
Cells Tissues Organs ; 197(5): 344-59, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23571553

RESUMO

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.


Assuntos
Capilares/metabolismo , Células-Tronco Mesenquimais/metabolismo , Vasos Retinianos/citologia , Envelhecimento/metabolismo , Animais , Antígenos/metabolismo , Biomarcadores/metabolismo , Antígeno CD146/metabolismo , Capilares/citologia , Adesão Celular , Linhagem da Célula , Crioultramicrotomia , Endotélio/metabolismo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Pericitos/citologia , Pericitos/metabolismo , Proteoglicanas/metabolismo , Ratos , Ratos Wistar , Fatores de Transcrição SOXB1/metabolismo
6.
Adv Exp Med Biol ; 777: 3-24, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23161072

RESUMO

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.


Assuntos
Células Epiteliais , Glicoproteínas de Membrana , Animais , Diferenciação Celular , Células Epiteliais/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/metabolismo , Células-Tronco Neoplásicas/metabolismo
8.
Biomedicines ; 10(2)2022 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-35203414

RESUMO

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.

9.
Ther Apher Dial ; 26 Suppl 1: 29-34, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36468302

RESUMO

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.


Assuntos
Retinopatia Diabética , Edema Macular , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Retinopatia Diabética/terapia , Retinopatia Diabética/etiologia , Retinopatia Diabética/metabolismo , Células Endoteliais/metabolismo , Glioxal/farmacologia , Glioxal/metabolismo , Retina/metabolismo , Isoformas de Proteínas/metabolismo
10.
Sci Rep ; 11(1): 15313, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321516

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Proteínas do Olho/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Isquemia/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Retina/efeitos dos fármacos , Vasos Retinianos , Inibidores da Angiogênese/uso terapêutico , Animais , Quimiotaxia de Leucócito/genética , Retinopatia Diabética , Modelos Animais de Doenças , Metabolismo Energético/genética , Proteínas do Olho/genética , Ontologia Genética , Redes Reguladoras de Genes , Isquemia/genética , Isquemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/genética , Oxigênio/metabolismo , Oxigênio/toxicidade , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Retina/metabolismo , Retinopatia da Prematuridade , Transcrição Gênica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/fisiologia
11.
J Neurosci ; 29(7): 2297-308, 2009 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-19228982

RESUMO

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.


Assuntos
Antígenos CD/genética , Glicoproteínas/genética , Peptídeos/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/anormalidades , Retina/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Antígeno AC133 , Animais , Colesterol/metabolismo , Regulação para Baixo/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Predisposição Genética para Doença/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Células Fotorreceptoras de Vertebrados/patologia , Retina/fisiopatologia , Artéria Retiniana/anormalidades , Artéria Retiniana/fisiopatologia , Degeneração Retiniana/fisiopatologia , Pigmentos da Retina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Visão Ocular/genética
12.
Histochem Cell Biol ; 133(5): 527-39, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20333396

RESUMO

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.


Assuntos
Túbulos Renais Coletores/metabolismo , Túbulos Renais Distais/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Antígeno AC133 , Animais , Antígenos CD/urina , Aquaporina 2/metabolismo , Calbindinas , Células Epiteliais/metabolismo , Expressão Gênica/genética , Glicoproteínas/urina , Humanos , Córtex Renal/crescimento & desenvolvimento , Córtex Renal/metabolismo , Medula Renal/crescimento & desenvolvimento , Medula Renal/metabolismo , Pelve Renal/crescimento & desenvolvimento , Pelve Renal/metabolismo , Glicoproteínas de Membrana/urina , Camundongos , Camundongos Endogâmicos , Mucoproteínas/metabolismo , Néfrons/metabolismo , Peptídeos/urina , Receptores de Droga/metabolismo , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto , Membro 3 da Família 12 de Carreador de Soluto , Simportadores/metabolismo , Uromodulina , Urotélio/crescimento & desenvolvimento , Urotélio/metabolismo
13.
Glia ; 57(8): 860-74, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19053060

RESUMO

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.


Assuntos
Antígenos CD/genética , Antígenos CD/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/genética , Glicoproteínas/metabolismo , Neuroglia/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Antígeno AC133 , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Chlorocebus aethiops , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/metabolismo , Rim/citologia , Camundongos , Camundongos Jimpy , Camundongos Mutantes , Microscopia Imunoeletrônica/métodos , Proteína Básica da Mielina/deficiência , Proteína Básica da Mielina/metabolismo , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/ultraestrutura , Fator de Transcrição 2 de Oligodendrócitos , Nervo Óptico/metabolismo , Nervo Óptico/ultraestrutura , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transfecção
14.
Stem Cells ; 26(3): 698-705, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18096722

RESUMO

Cerebrospinal fluid (CSF) is routinely used for diagnosing and monitoring neurological diseases. The CSF proteins used so far for diagnostic purposes (except for those associated with whole cells) are soluble. Here, we show that human CSF contains specific membrane particles that carry prominin-1/CD133, a neural stem cell marker implicated in brain tumors, notably glioblastoma. Differential and equilibrium centrifugation and detergent solubility analyses showed that these membrane particles were similar in physical properties and microdomain organization to small membrane vesicles previously shown to be released from neural stem cells in the mouse embryo. The levels of membrane particle-associated prominin-1/CD133 declined during childhood and remained constant thereafter, with a remarkably narrow range in healthy adults. Glioblastoma patients showed elevated levels of membrane particle-associated prominin-1/CD133, which decreased dramatically in the final stage of the disease. Hence, analysis of CSF for membrane particles carrying the somatic stem cell marker prominin-1/CD133 offers a novel approach for studying human central nervous system disease.


Assuntos
Antígenos CD/líquido cefalorraquidiano , Doenças do Sistema Nervoso Central/líquido cefalorraquidiano , Glicoproteínas/líquido cefalorraquidiano , Peptídeos/líquido cefalorraquidiano , Células-Tronco/metabolismo , Antígeno AC133 , Biomarcadores/metabolismo , Células CACO-2 , Glioblastoma/líquido cefalorraquidiano , Humanos , Padrões de Referência , Fatores de Tempo
15.
Cells ; 8(9)2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31540455

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Fatores de Crescimento do Endotélio Vascular/farmacologia , Animais , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
16.
J Histochem Cytochem ; 56(11): 977-93, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18645205

RESUMO

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.


Assuntos
Antígenos CD/biossíntese , Glândulas Exócrinas/metabolismo , Glicoproteínas/biossíntese , Fígado/metabolismo , Pâncreas/metabolismo , Útero/metabolismo , Antígeno AC133 , Animais , Anticorpos Monoclonais , Antígenos CD/imunologia , Células CHO , Células CACO-2 , Cricetinae , Cricetulus , Epitélio/metabolismo , Feminino , Citometria de Fluxo , Glicoproteínas/imunologia , Humanos , Imuno-Histoquímica , Microscopia Confocal , Especificidade de Órgãos , Peptídeos/imunologia
17.
Cells Tissues Organs ; 188(1-2): 127-38, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18160824

RESUMO

Prominin-1 (alias CD133) has received considerable interest because of its expression by several stem and progenitor cells originating from various sources, including the neural and hematopoietic systems. As a cell surface marker, prominin-1 is now used for somatic stem cell isolation. Its expression in cancer stem cells has broadened its clinical value, as it might be useful to outline new prospects for more effective cancer therapies by targeting tumor-initiating cells. Cell biological studies of this molecule have demonstrated that it is specifically concentrated in various membrane structures that protrude from the planar areas of the plasmalemma. Prominin-1 binds to the plasma membrane cholesterol and is associated with a particular membrane microdomain in a cholesterol-dependent manner. Although its physiological function is not yet determined, it is becoming clear that this cell surface protein, as a unique marker of both plasma membrane protrusions and membrane microdomains, might reveal new aspects of the cell biology of rare stem and cancer stem cells. The aim of this review is to outline the recent discoveries regarding the dynamic reorganization of the plasma membrane of rare CD133+ hematopoietic progenitor cells during cell migration and division.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Células-Tronco Hematopoéticas/citologia , Peptídeos/metabolismo , Antígeno AC133 , Humanos
18.
Mech Dev ; 113(1): 73-7, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11900977

RESUMO

In Drosophila melanogaster, the seven-pass transmembrane protein Rhomboid (Rho) is a crucial positive modulator of EGF signaling playing a substantial role in patterning of the ventral neuroectoderm and fate specification of neuroblasts. Here, we describe the cloning and expression pattern of Ventrhoid (Vrho), the novel evolutionarily conserved vertebrate cDNA related to fruit fly rho. Most importantly, like rho in Drosophila, Vrho is also expressed in a spatially restricted manner. Vrho expression is most prominent along the developing ventral neural tube, and is also detectable in the ventral forebrain, prospective diencephalon, otic vesicles, mandibular arches, cranial sensory placodes, last formed pair of somites and hindgut in midgestational mouse embryos.


Assuntos
Proteínas de Drosophila , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Sequência de Aminoácidos , Animais , Sistema Nervoso Central/embriologia , Clonagem Molecular , DNA/biossíntese , DNA Complementar/metabolismo , Drosophila , Éxons , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Íntrons , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Neurônios/fisiologia , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Distribuição Tecidual , Regulação para Cima
19.
Int J Radiat Biol ; 90(8): 700-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24512568

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Olho/metabolismo , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/metabolismo , Pterígio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Túnica Conjuntiva/efeitos dos fármacos , Túnica Conjuntiva/metabolismo , Túnica Conjuntiva/efeitos da radiação , Olho/irrigação sanguínea , Olho/efeitos dos fármacos , Olho/efeitos da radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Camundongos , Terapia de Alvo Molecular , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/efeitos da radiação , Pterígio/tratamento farmacológico , Pterígio/fisiopatologia , Proteínas Repressoras
20.
PLoS One ; 8(5): e63457, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23723983

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Encéfalo/citologia , Glicoproteínas/metabolismo , Células-Tronco Neurais/metabolismo , Peptídeos/metabolismo , Antígeno AC133 , Ambystoma mexicanum , Animais , Antígenos CD/genética , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Embrião de Galinha , Feminino , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/genética , Camundongos , Placa Neural/citologia , Tubo Neural/citologia , Especificidade de Órgãos , Peptídeos/genética , Regeneração , Medula Espinal/fisiologia , Regulação para Cima , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA