Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
PLoS Genet ; 18(2): e1009994, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35143487

RESUMEN

Alzheimer's Disease (AD) is a neuroinflammatory disease characterized partly by the inability to clear, and subsequent build-up, of amyloid-beta (Aß). AD has a bi-directional relationship with circadian disruption (CD) with sleep disturbances starting years before disease onset. However, the molecular mechanism underlying the relationship of CD and AD has not been elucidated. Myeloid-based phagocytosis, a key component in the metabolism of Aß, is circadianly-regulated, presenting a potential link between CD and AD. In this work, we revealed that the phagocytosis of Aß42 undergoes a daily circadian oscillation. We found the circadian timing of global heparan sulfate proteoglycan (HSPG) biosynthesis was the molecular timer for the clock-controlled phagocytosis of Aß and that both HSPG binding and aggregation may play a role in this oscillation. These data highlight that circadian regulation in immune cells may play a role in the intricate relationship between the circadian clock and AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Ritmo Circadiano/fisiología , Proteoglicanos de Heparán Sulfato/metabolismo , Fagocitosis/fisiología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Relojes Circadianos , Modelos Animales de Enfermedad , Proteoglicanos de Heparán Sulfato/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Agregación Patológica de Proteínas/metabolismo
2.
FEBS J ; 289(1): 279-293, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34324261

RESUMEN

Kashin-Beck disease (KBD) is an endemic osteochondropathy. Due to a lack of suitable animal or cellular disease models, the research progress on KBD has been limited. Our goal was to establish the first disease-specific human induced pluripotent stem cell (hiPSC) cellular disease model of KBD, and to explore its etiology and pathogenesis exploiting transcriptome sequencing. HiPSCs were reprogrammed from dermal fibroblasts of two KBD and one healthy control donor via integration-free vectors. Subsequently, hiPSCs were differentiated into chondrocytes through three-week culture. Gene expression profiles in KBD, normal primary chondrocytes, and hiPSC-derived chondrocytes were defined by RNA sequencing. A Venn diagram was constructed to show the number of shared differentially expressed genes (DEGs) between KBD and normal. Gene oncology and Kyoto Encyclopedia of Genes and Genomes annotations were performed, and six DEGs were further validated in other individuals by RT-qPCR. KBD cellular disease models were successfully established by generation of hiPSC lines. Seventeen consistent and significant DEGs present in all compared groups (KBD and normal) were identified. RT-qPCR validation gave consistent results with the sequencing data. Glycosaminoglycan biosynthesis-heparan sulfate/heparin; PPAR signaling pathway; and cell adhesion molecules (CAMs) were identified to be significantly altered in KBD. Differentiated chondrocytes derived from KBD-origin hiPSCs provide the first cellular disease model for etiological studies of KBD. This study also provides new sights into the pathogenesis and etiology of KBD and is likely to inform the development of targeted therapeutics for its treatment.


Asunto(s)
Proteoglicanos de Heparán Sulfato/genética , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Kashin-Beck/genética , Transcriptoma/genética , Condrocitos/citología , Condrocitos/metabolismo , Regulación de la Expresión Génica/genética , Proteoglicanos de Heparán Sulfato/biosíntesis , Humanos , Células Madre Pluripotentes Inducidas/citología , Enfermedad de Kashin-Beck/metabolismo , Enfermedad de Kashin-Beck/patología , Receptores Activados del Proliferador del Peroxisoma/genética , Cultivo Primario de Células , Biosíntesis de Proteínas/genética , Transducción de Señal/genética
3.
Cell Death Dis ; 13(1): 11, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930890

RESUMEN

TMEM165 deficiency leads to skeletal disorder characterized by major skeletal dysplasia and pronounced dwarfism. However, the molecular mechanisms involved have not been fully understood. Here, we uncover that TMEM165 deficiency impairs the synthesis of proteoglycans by producing a blockage in the elongation of chondroitin-and heparan-sulfate glycosaminoglycan chains leading to the synthesis of proteoglycans with shorter glycosaminoglycan chains. We demonstrated that the blockage in elongation of glycosaminoglycan chains is not due to defect in the Golgi elongating enzymes but rather to availability of the co-factor Mn2+. Supplementation of cell with Mn2+ rescue the elongation process, confirming a role of TMEM165 in Mn2+ Golgi homeostasis. Additionally, we showed that TMEM165 deficiency functionally impairs TGFß and BMP signaling pathways in chondrocytes and in fibroblast cells of TMEM165 deficient patients. Finally, we found that loss of TMEM165 impairs chondrogenic differentiation by accelerating the timing of Ihh expression and promoting early chondrocyte maturation and hypertrophy. Collectively, our results indicate that TMEM165 plays an important role in proteoglycan synthesis and underline the critical role of glycosaminoglycan chains structure in the regulation of chondrogenesis. Our data also suggest that Mn2+ supplementation may be a promising therapeutic strategy in the treatment of TMEM165 deficient patients.


Asunto(s)
Antiportadores/deficiencia , Antiportadores/metabolismo , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/metabolismo , Diferenciación Celular/genética , Condrocitos/metabolismo , Condrocitos/patología , Sulfatos de Condroitina/biosíntesis , Enanismo/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Transducción de Señal/genética , Animales , Antiportadores/genética , Estudios de Casos y Controles , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Condrogénesis/genética , Enanismo/patología , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes/métodos , Glicosilación , Células HEK293 , Humanos , Hipertrofia/metabolismo , Ratones , Transfección
4.
Transl Stroke Res ; 12(1): 72-86, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32253702

RESUMEN

The extracellular matrix fragment perlecan domain V is neuroprotective and functionally restorative following experimental stroke. As neurogenesis is an important component of chronic post-stroke repair, and previous studies have implicated perlecan in developmental neurogenesis, we hypothesized that domain V could have a broad therapeutic window by enhancing neurogenesis after stroke. We demonstrated that domain V is chronically increased in the brains of human stroke patients, suggesting that it is present during post-stroke neurogenic periods. Furthermore, perlecan deficient mice had significantly less neuroblast precursor cells after experimental stroke. Seven-day delayed domain V administration enhanced neurogenesis and restored peri-infarct excitatory synaptic drive to neocortical layer 2/3 pyramidal neurons after experimental stroke. Domain V's effects were inhibited by blockade of α2ß1 integrin, suggesting the importance of α2ß1 integrin to neurogenesis and domain V neurogenic effects. Our results demonstrate that perlecan plays a previously unrecognized role in post-stroke neurogenesis and that delayed DV administration after experimental stroke enhances neurogenesis and improves recovery in an α2ß1 integrin-mediated fashion. We conclude that domain V is a clinically relevant neuroprotective and neuroreparative novel stroke therapy with a broad therapeutic window.


Asunto(s)
Encéfalo/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Neurogénesis/fisiología , Neuroprotección/fisiología , Accidente Cerebrovascular/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Células Cultivadas , Proteoglicanos de Heparán Sulfato/administración & dosificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Técnicas de Cultivo de Órganos , Dominios Proteicos , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/prevención & control
5.
Molecules ; 25(18)2020 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-32937952

RESUMEN

Glycosylation is a common and widespread post-translational modification that affects a large majority of proteins. Of these, a small minority, about 20, are specifically modified by the addition of heparan sulfate, a linear polysaccharide from the glycosaminoglycan family. The resulting molecules, heparan sulfate proteoglycans, nevertheless play a fundamental role in most biological functions by interacting with a myriad of proteins. This large functional repertoire stems from the ubiquitous presence of these molecules within the tissue and a tremendous structural variety of the heparan sulfate chains, generated through both biosynthesis and post synthesis mechanisms. The present review focusses on how proteoglycans are "gagosylated" and acquire structural complexity through the concerted action of Golgi-localized biosynthesis enzymes and extracellular modifying enzymes. It examines, in particular, the possibility that these enzymes form complexes of different modes of organization, leading to the synthesis of various oligosaccharide sequences.


Asunto(s)
Aparato de Golgi/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Heparitina Sulfato/metabolismo , Animales , Membrana Celular/metabolismo , Matriz Extracelular/metabolismo , Glicosaminoglicanos/metabolismo , Glicosilación , Proteoglicanos de Heparán Sulfato/química , Humanos , Mutación , Oligosacáridos/química , Biosíntesis de Proteínas , Dominios Proteicos , Procesamiento Proteico-Postraduccional
6.
J Toxicol Sci ; 45(2): 109-115, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32062617

RESUMEN

Proteoglycans that are synthesized by vascular endothelial cells contribute to the proliferation, migration, and blood coagulation-fibrinolytic system in vascular endothelial cells. Clarification of the molecular mechanisms for proteoglycan synthesis allows understanding of the regulation of endothelial functions. The research strategy of bioorganometallics analyzes biological systems using organic-inorganic hybrid molecules as tools. The present study found dichloro(2,9-dimethyl-1,10-phenanthroline)zinc(II) and its ligand-modulated perlecan expression in vascular endothelial cells, which depends on the cell density.


Asunto(s)
Recuento de Células , Células Endoteliales/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Animales , Bovinos , Células Cultivadas
7.
Biotechnol Bioeng ; 116(10): 2730-2741, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31282995

RESUMEN

It is widely believed that the differentiation of embryonic stem cells (ESCs) into viable endothelial cells (ECs) for use in vascular tissue engineering can be enhanced by mechanical forces. In our previous work, we reported that shear stress enhanced important EC functional genes on a CD31+ /CD45- cell population derived from mouse ESC committed to the EC lineage. In the present study, in contrast to the effects of shear stress on this cell population, we observed that cyclic strain significantly reduced the expression of EC-specific marker genes (vWF, VE-cadherin, and PECAM-1), tight junction protein genes (ZO-1, OCLD, and CLD5), and vasoactive genes (eNOS and ET1), while it did not alter the expression of COX2. Taken together, these studies indicate that only shear stress, not cyclic strain, is a useful mechanical stimulus for enhancing the properties of CD31+ /CD45- cells for use as EC in vascular tissue engineering. To begin examining the mechanisms controlling cyclic strain-induced suppression of gene expression in CD31+ /CD45- cells, we depleted the heparan sulfate (HS) component of the glycocalyx, blocked integrins, and silenced the HS proteoglycan syndecan-4 in separate experiments. All of these treatments resulted in the reversal of cyclic strain-induced gene suppression. The current study and our previous work provide a deeper understanding of the mechanisms that balance the influence of cyclic strain and shear stress in endothelial cells.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Proteoglicanos de Heparán Sulfato/biosíntesis , Integrinas/biosíntesis , Mecanotransducción Celular , Células Madre Embrionarias de Ratones/metabolismo , Sindecano-4/biosíntesis , Animales , Células Endoteliales/citología , Glicocálix/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Ingeniería de Tejidos
8.
Biochem Soc Trans ; 46(4): 789-796, 2018 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-29934302

RESUMEN

The leishmaniases are a group of neglected tropical diseases caused by parasites from the Leishmania genus. More than 20 Leishmania species are responsible for human disease, causing a broad spectrum of symptoms ranging from cutaneous lesions to a fatal visceral infection. There is no single safe and effective approach to treat these diseases and resistance to current anti-leishmanial drugs is emerging. New drug targets need to be identified and validated to generate novel treatments. Host heparan sulfates (HSs) are abundant, heterogeneous polysaccharides displayed on proteoglycans that bind various ligands, including cell surface proteins expressed on Leishmania promastigote and amastigote parasites. The fine chemical structure of HS is formed by a plethora of specific enzymes during biosynthesis, with various positions (N-, 2-O-, 6-O- and 3-O-) on the carbon sugar backbone modified with sulfate groups. Post-biosynthesis mechanisms can further modify the sulfation pattern or size of the polysaccharide, altering ligand affinity to moderate biological functions. Chemically modified heparins used to mimic the heterogeneous nature of HS influence the affinity of different Leishmania species, demonstrating the importance of specific HS chemical sequences in parasite interaction. However, the endogenous structures of host HSs that might interact with Leishmania parasites during host invasion have not been elucidated, nor has the role of HSs in host-parasite biology. Decoding the structure of HSs on target host cells will increase understanding of HS/parasite interactions in leishmaniasis, potentiating identification of new opportunities for the development of novel treatments.


Asunto(s)
Heparitina Sulfato/fisiología , Leishmania/metabolismo , Leishmania/patogenicidad , Macrófagos/parasitología , Animales , Antiprotozoarios/uso terapéutico , Moléculas de Adhesión Celular/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/metabolismo , Interacciones Huésped-Parásitos , Humanos , Leishmaniasis/tratamiento farmacológico , Unión Proteica , Proteínas Protozoarias/metabolismo
9.
J Biol Regul Homeost Agents ; 32(3): 479-487, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29921372

RESUMEN

Alterations in cellular and extracellular matrix components play an important role during tumorigenesis; proteoglycans are included among these components. Ameloblastomas are odontogenic tumors distinguished as invasive and infiltrative neoplasms and are divided into different histological types, the most common of which are the unicystic ameloblastoma and the conventional ameloblastoma. The aim of this study was to identify the presence of two proteoglycans, perlecan and biglycan, in different types of ameloblastoma. Using immunohistochemistry, we determined the presence of both proteins in 28 unicystic ameloblastomas and 23 conventional ameloblastomas. We identified the cytoplasmic and nuclear presence of perlecan and the cytoplasmic presence of biglycan in both types of ameloblastoma. The mean values of immunoexpression were higher in the conventional type compared to the unicystic type. Neither the presence of biglycan in ameloblastomas nor the nuclear presence of perlecan in any odontogenic tumor has previously been reported. The differential immunoexpression of perlecan and biglycan in these types of ameloblastomas suggests their participation in the developmental process of these tumors.


Asunto(s)
Ameloblastoma , Biglicano/biosíntesis , Regulación Neoplásica de la Expresión Génica , Proteoglicanos de Heparán Sulfato/biosíntesis , Neoplasias Maxilomandibulares , Proteínas de Neoplasias/biosíntesis , Adulto , Ameloblastoma/clasificación , Ameloblastoma/metabolismo , Ameloblastoma/patología , Femenino , Humanos , Inmunohistoquímica , Neoplasias Maxilomandibulares/clasificación , Neoplasias Maxilomandibulares/metabolismo , Neoplasias Maxilomandibulares/patología , Masculino
10.
Respirology ; 23(1): 68-75, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28834088

RESUMEN

BACKGROUND AND OBJECTIVE: Involvement of pulmonary vascular remodelling is a characteristic sign in COPD. Vascular mediators such as vascular endothelial growth factor (VEGF) and prostacyclin may regulate fibroblast activity. The objective was to study the synthesis of VEGF and interactions with prostacyclin and transforming growth factor (TGF)-ß1 in lung fibroblasts from patients with COPD and healthy control subjects. To further explore the autocrine role of synthesized VEGF on fibroblast activity, studies were performed in human lung fibroblasts (HFL-1). METHODS: Primary distal lung fibroblast cultures were established from healthy individuals and from COPD patients (GOLD stage IV). Lung fibroblasts were stimulated with the prostacyclin analogue iloprost and the profibrotic stimuli TGF-ß1 . VEGF synthesis was measured in the cell culture medium. Changes in proliferation rate, migration and synthesis of the extracellular matrix (ECM) proteins proteoglycans were analysed after stimulations with VEGF-A isoform 165 (VEGF165 ; 1-10 000 pg/mL) in HFL-1. RESULTS: Iloprost and TGF-ß1 significantly increased VEGF synthesis in both fibroblasts from COPD patients and control subjects. TGF-ß1 -induced VEGF synthesis was significantly reduced by the cyclooxygenase inhibitor indomethacin in fibroblasts from COPD patients. VEGF significantly increased proliferation rate and migration capacity in HFL-1. VEGF also significantly increased synthesis of the ECM proteins biglycan and perlecan. The VEGF receptors (VEGFR), VEGFR1, VEGFR2 and VEGFR3, were all expressed in primary lung fibroblasts and HFL-1. CONCLUSION: VEGF is synthesized in high amounts by distal lung fibroblasts and may have a crucial role in ongoing vascular remodelling processes in the distal lung compartments.


Asunto(s)
Fibroblastos/efectos de los fármacos , Iloprost/farmacología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Remodelación Vascular , Anciano , Biglicano/biosíntesis , Movimiento Celular , Proliferación Celular , Células Cultivadas , Inhibidores de la Ciclooxigenasa/farmacología , Fibroblastos/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Humanos , Indometacina/farmacología , Pulmón/citología , Pulmón/metabolismo , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Autophagy ; 13(8): 1262-1279, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28402693

RESUMEN

Heparan sulfate-modified proteoglycans (HSPGs) are important regulators of signaling and molecular recognition at the cell surface and in the extracellular space. Disruption of HSPG core proteins, HS-synthesis, or HS-degradation can have profound effects on growth, patterning, and cell survival. The Drosophila neuromuscular junction provides a tractable model for understanding the activities of HSPGs at a synapse that displays developmental and activity-dependent plasticity. Muscle cell-specific knockdown of HS biosynthesis disrupted the organization of a specialized postsynaptic membrane, the subsynaptic reticulum (SSR), and affected the number and morphology of mitochondria. We provide evidence that these changes result from a dysregulation of macroautophagy (hereafter referred to as autophagy). Cellular and molecular markers of autophagy are all consistent with an increase in the levels of autophagy in the absence of normal HS-chain biosynthesis and modification. HS production is also required for normal levels of autophagy in the fat body, the central energy storage and nutritional sensing organ in Drosophila. Genetic mosaic analysis indicates that HS-dependent regulation of autophagy occurs non-cell autonomously, consistent with HSPGs influencing this cellular process via signaling in the extracellular space. These findings demonstrate that HS biosynthesis has important regulatory effects on autophagy and that autophagy is critical for normal assembly of postsynaptic membrane specializations.


Asunto(s)
Autofagia , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Animales , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Regulación hacia Abajo , Drosophila/genética , Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/ultraestructura , Cuerpo Adiposo/metabolismo , Cuerpo Adiposo/ultraestructura , Proteoglicanos de Heparán Sulfato/biosíntesis , Homocigoto , Larva/metabolismo , Larva/ultraestructura , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Músculos/metabolismo , Músculos/ultraestructura , Mutación/genética , Unión Neuromuscular/metabolismo , Fenotipo , Interferencia de ARN , Sinapsis/metabolismo , Sinapsis/ultraestructura
12.
Int Rev Cell Mol Biol ; 325: 275-93, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27241223

RESUMEN

Heparan sulfate proteoglycans (HSPGs) are a class of carbohydrate-modified proteins involved in key biological processes, including growth factor signaling, cell adhesion, and enzymatic catalysis. HSPGs serve as coreceptors for a number of ligand molecules to regulate their signaling and distribution. These HS-dependent factors include fibroblast growth factors, bone morphogenetic proteins, Wnt-related factors, hedgehog, and cytokines. Several classes of HSPGs are evolutionarily conserved from humans to the genetically tractable model organism Drosophila. Sophisticated molecular genetic tools available in Drosophila provide for a powerful system to address unanswered questions regarding in vivo functions of HSPGs. These studies have highlighted the functions of HSPGs in the regulation of significant developmental events, such as morphogen gradient formation, nervous system formation, and the stem cell niche. Drosophila genetics has also established HSPGs as key factors in feedback controls that ensure robustness in developmental systems.


Asunto(s)
Drosophila/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Animales , Glipicanos/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Modelos Animales , Sindecanos/metabolismo
13.
BMC Cancer ; 15: 742, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26482785

RESUMEN

BACKGROUND: Heparan sulfate proteoglycans (HSPGs) are complex molecules involved in the growth, invasion and metastatic properties of cancerous cells. This study analyses the alterations in the expression patterns of these molecules in right sided colorectal cancer (CRC), both metastatic and non-metastatic. METHODS: Twenty right sided CRCs were studied. A transcriptomic approach was used, employing qPCR to analyze both the expression of the enzymes involved in heparan sulfate (HS) chains biosynthesis, as well as the proteoglycan core proteins. Since some of these proteoglycans can also carry chondroitin sulfate (CS) chains, we include the study of the genes involved in the biosynthesis of these glycosaminoglycans. Immunohistochemical techniques were also used to analyze tissue expression of particular genes showing significant expression differences, of potential interest. RESULTS: Changes in proteoglycan core proteins differ depending on their location; those located intracellularly or in the extracellular matrix show very similar alteration patterns, while those located on the cell surface vary greatly depending on the nature of the tumor: glypicans 1, 3, 6 and betaglycan are affected in the non-metastatic tumors, whereas in the metastatic, only glypican-1 and syndecan-1 are modified, the latter showing opposing alterations in levels of RNA and of protein, suggesting post-transcriptional regulation in these tumors. Furthermore, in non-metastatic tumors, polymerization of glycosaminoglycan chains is modified, particularly affecting the synthesis of the tetrasaccharide linker and the initiation and elongation of CS chains, HS chains being less affected. Regarding the enzymes responsible for the modificaton of the HS chains, alterations were only found in non-metastatic tumors, affecting N-sulfation and the isoforms HS6ST1, HS3ST3B and HS3ST5. In contrast, synthesis of the CS chains suggests changes in epimerization and sulfation of the C4 and C2 in both types of tumor. CONCLUSIONS: Right sided CRCs show alterations in the expression of HSPGs, including the expression of the cell surface core proteins, many glycosiltransferases and some enzymes that modify the HS chains depending on the metastatic nature of the tumor, resulting more affected in non-metastatic ones. However, matrix proteoglycans and enzymes involved in CS fine structure synthesis are extensively modified independetly of the presence of lymph node metastasis.


Asunto(s)
Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Proteoglicanos de Heparán Sulfato/genética , ARN Neoplásico/genética , Anciano , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/secundario , Femenino , Proteoglicanos de Heparán Sulfato/biosíntesis , Humanos , Inmunohistoquímica , Masculino , Metástasis de la Neoplasia , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Tumorales Cultivadas
14.
Metab Brain Dis ; 30(1): 1-5, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24964971

RESUMEN

Vascular dementia (VaD) is the second most common cause of dementia and leads to a decline in cognitive thinking via conditions that lead to blockage or reduced blood flow to the brain. It is a poorly understood disease, and the changes that occur are often linked to other types of dementia such as Alzheimer's disease. To date, there are no approved therapies or drugs to treat the symptoms of VaD, even though there is some evidence of drugs approved for Alzheimer's that might have some benefit in patients diagnosed with VaD. The altered blood flow that precedes VaD may result in compensatory mechanisms, such as angiogenesis, to increase blood flow in the brain. Angiogenesis, the process of new blood vessel formations from pre-existing ones, involves several pro-angiogenic factors such as vascular endothelial growth factor (VEGF) and is regulated by a variety of growth factors from neurons, astrocytes, and pericytes in the brain as well the extracellular matrix (ECM). The ECM highly regulates angiogenesis and other processes in the brain. One such ECM component is the heparan sulfate proteoglycan perlecan and its bioactive region, Domain V (DV). Here we discuss the potential role of DV as a novel therapy to treat VaD.


Asunto(s)
Inductores de la Angiogénesis/uso terapéutico , Demencia Vascular/tratamiento farmacológico , Proteoglicanos de Heparán Sulfato/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Estenosis Carotídea/complicaciones , Angiopatía Amiloide Cerebral/tratamiento farmacológico , Angiopatía Amiloide Cerebral/fisiopatología , Demencia Vascular/etiología , Angiopatías Diabéticas/tratamiento farmacológico , Angiopatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Proteoglicanos de Heparán Sulfato/biosíntesis , Proteoglicanos de Heparán Sulfato/química , Humanos , Ratones Transgénicos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Estructura Terciaria de Proteína , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/fisiopatología
15.
Philos Trans R Soc Lond B Biol Sci ; 369(1657)2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25349453

RESUMEN

During mouse embryogenesis, diffusible growth factors, i.e. fibroblast growth factors, Wnt, bone morphogenetic protein and Hedgehog family members, emanating from localized areas can travel through the extracellular space and reach their target cells to specify the cell fate and form tissue architectures in coordination. However, the mechanisms by which these growth factors travel great distances to their target cells and control the signalling activity as morphogens remain an enigma. Recent studies in mice and other model animals have revealed that heparan sulfate proteoglycans (HSPGs) located on the cell surface (e.g. syndecans and glypicans) and in the extracellular matrix (ECM; e.g. perlecan and agrin) play crucial roles in the extracellular distribution of growth factors. Principally, the function of HSPGs depends primarily on the fine features and localization of their heparan sulfate glycosaminoglycan chains. Cell-surface-tethered HSPGs retain growth factors as co-receptors and/or endocytosis mediators, and enzymatic release of HSPGs from the cell membrane allows HSPGs to transport or move multiple growth factors. By contrast, ECM-associated HSPGs function as a reservoir or barrier in a context-dependent manner. This review is focused on our current understanding of the extracellular distribution of multiple growth factors controlled by HSPGs in mammalian development.


Asunto(s)
Desarrollo Embrionario/fisiología , Espacio Extracelular/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Transducción de Señal/fisiología , Animales , Transporte Biológico/fisiología , Proteoglicanos de Heparán Sulfato/biosíntesis , Proteoglicanos de Heparán Sulfato/química , Ratones , Modelos Biológicos
16.
Yakugaku Zasshi ; 134(7): 789-91, 2014.
Artículo en Japonés | MEDLINE | ID: mdl-24989466

RESUMEN

  Organometallic compounds and complexes possibly show novel bioactivities unprecedented with organic molecules that consist of C, H, O, and N atoms. We have already reported that an organoantimony compound selectively induces the expression of perlecan, a large heparin sulfate proteoglycan, in cultured vascular endothelial cells. Perlecan consists of a core protein with Mr of approximately 400 kDa and three anticoagulant heparan sulfate glycosaminoglycan chains. However, little is known about the molecular mechanisms of perlecan expression in endothelial cells. Since an organoantimony compound induces the expression of perlecan, we prepared a variety of its derivatives and tried to identify the binding proteins for the organoantimony compound by the drug affinity responsive target stability method, a negative selection method to identify the target proteins. As the results, we found glucocoriticoid receptor as binding protein of the organoantimony compound, which mediates the expression of perlecan in endothelial cells.


Asunto(s)
Antimonio/farmacología , Células Endoteliales/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Animales , Antimonio/química , Células Endoteliales/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Humanos , Fenómenos Químicos Orgánicos , Relación Estructura-Actividad
17.
J Biol Chem ; 289(22): 15231-43, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24753252

RESUMEN

Degradation of heparan sulfate (HS) in the extracellular matrix by heparanase is linked to the processes of tumor invasion and metastasis. Thus, a heparanase inhibitor can be a potential anticancer drug. Because HS with unsubstituted glucosamine residues accumulates in heparanase-expressing breast cancer cells, we assumed that these HS structures are resistant to heparanase and can therefore be utilized as a heparanase inhibitor. As expected, chemically synthetic HS-tetrasaccharides containing unsubstituted glucosamine residues, GlcAß1-4GlcNH3 (+)(6-O-sulfate)α1-4GlcAß1-4GlcNH3 (+)(6-O-sulfate), inhibited heparanase activity and suppressed invasion of breast cancer cells in vitro. Bifunctional NDST-1 (N-deacetylase/N-sulfotransferase-1) catalyzes the modification of N-acetylglucosamine residues within HS chains, and the balance of N-deacetylase and N-sulfotransferase activities of NDST-1 is thought to be a determinant of the generation of unsubstituted glucosamine. We also report here that EXTL3 (exostosin-like 3) controls N-sulfotransferase activity of NDST-1 by forming a complex with NDST-1 and contributes to generation of unsubstituted glucosamine residues.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glucosamina/metabolismo , Glucuronidasa/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Sulfotransferasas/metabolismo , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/secundario , Femenino , Fibroblastos/citología , Glucuronidasa/antagonistas & inhibidores , Glicosaminoglicanos/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , Humanos , Células MCF-7 , Ratones , N-Acetilglucosaminiltransferasas/metabolismo , Invasividad Neoplásica
18.
J Cell Biochem ; 115(7): 1322-33, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24700612

RESUMEN

Perlecan/HSPG2, a heparan sulfate proteoglycan typically found at tissue borders including those separating epithelia and connective tissue, increases near sites of invasion of primary prostatic tumors as previously shown for other proteins involved in desmoplastic tissue reaction. Studies of prostate cancer cells and stromal cells from both prostate and bone, the major site for prostate cancer metastasis, showed that cancer cells and a subset of stromal cells increased production of perlecan in response to cytokines present in the tumor microenvironment. In silico analysis of the HSPG2 promoter revealed two conserved NFκB binding sites, in addition to the previously reported SMAD3 binding sites. By systematically transfecting cells with a variety of reporter constructs including sequences up to 2.6 kb from the start site of transcription, we identified an active cis element in the distal region of the HSPG2 promoter, and showed that it functions in regulating transcription of HSPG2. Treatment with TNF-α and/or TGFß1 identified TNF-α as a major cytokine regulator of perlecan production. TNF-α treatment also triggered p65 nuclear translocation and binding to the HSPG2 regulatory region in stromal cells and cancer cells. In addition to stromal induction of perlecan production in the prostate, we identified a matrix-secreting bone marrow stromal cell type that may represent the source for increases in perlecan in the metastatic bone marrow environment. These studies implicate perlecan in cytokine-mediated, innate tissue responses to cancer cell invasion, a process we suggest reflects a modified wound healing tissue response co-opted by prostate cancer cells.


Asunto(s)
Proteoglicanos de Heparán Sulfato/biosíntesis , Neoplasias de la Próstata/genética , Células del Estroma/citología , Factor de Transcripción ReIA/metabolismo , Activación Transcripcional , Transporte Activo de Núcleo Celular , Sitios de Unión , Línea Celular Tumoral , Proteínas de Unión al ADN , Proteoglicanos de Heparán Sulfato/genética , Humanos , Masculino , Regiones Promotoras Genéticas , Próstata/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
19.
J Cell Biochem ; 115(5): 967-76, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24357546

RESUMEN

Heparan sulfate proteoglycans (HSPGs) are key components of the extracellular matrix that mediate cell proliferation, invasion, and cellular signaling. The biological functions of HSPGs are linked to their co-stimulatory effects on extracellular ligands (e.g., WNTs) and the resulting activation of transcription factors that control mammalian development but also associated with tumorigenesis. We examined the expression profile of HSPG core protein syndecans (SDC1-4) and glypicans (GPC1-6) along with the enzymes that initiate or modify their glycosaminoglycan chains in human breast cancer (HBC) epithelial cells. Gene expression in relation to cell proliferation was examined in the HBC cell lines MCF-7 and MDA-MB-231 following treatment with the HS agonist heparin. Heparin increased gene expression of chain initiation and modification enzymes including EXT1 and NDST1, as well as core proteins SDC2 and GPC6. With HS/Wnt interactions established, we next investigated WNT pathway components and observed that increased proliferation of the more invasive MDA-MB-231 cells is associated with activation of the Wnt signaling pathway. Specifically, there was substantial upregulation (>5-fold) of AXIN1, WNT4A, and MYC in MDA-MB-231 but not in MCF-7 cells. The changes in gene expression observed for HSPG core proteins and related enzymes along with the associated Wnt signaling components suggest coordinated interactions. The influence of HSPGs on cellular proliferation and invasive potential of breast cancer epithelial cells are cell and niche specific. Further studies on the interactions between HSPGs and WNT ligands may yield clinically relevant molecular targets, as well as new biomarkers for characterization of breast cancer progression.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/efectos de los fármacos , Proteoglicanos de Heparán Sulfato/biosíntesis , Vía de Señalización Wnt/genética , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteoglicanos de Heparán Sulfato/genética , Heparina/administración & dosificación , Humanos , Células MCF-7 , Vía de Señalización Wnt/efectos de los fármacos
20.
Science ; 341(6152): 1404-8, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-24052309

RESUMEN

An appropriate balance in glycosylation of proteoglycans is crucial for their ability to regulate animal development. Here, we report that the Caenorhabditis elegans microRNA mir-79, an ortholog of mammalian miR-9, controls sugar-chain homeostasis by targeting two proteins in the proteoglycan biosynthetic pathway: a chondroitin synthase (SQV-5; squashed vulva-5) and a uridine 5'-diphosphate-sugar transporter (SQV-7). Loss of mir-79 causes neurodevelopmental defects through SQV-5 and SQV-7 dysregulation in the epidermis. This results in a partial shutdown of heparan sulfate biosynthesis that impinges on a LON-2/glypican pathway and disrupts neuronal migration. Our results identify a regulatory axis controlled by a conserved microRNA that maintains proteoglycan homeostasis in cells.


Asunto(s)
Caenorhabditis elegans/fisiología , Movimiento Celular , Epidermis/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , MicroARNs/fisiología , Neuronas/fisiología , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/biosíntesis , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Glicosilación , Glicosiltransferasas/metabolismo , Glipicanos/biosíntesis , Glipicanos/genética , Proteoglicanos de Heparán Sulfato/genética , MicroARNs/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Portadoras de Nucleobases, Nucleósidos, Nucleótidos y Ácidos Nucleicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA