Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Bioorg Med Chem ; 90: 117335, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37257254

RESUMEN

Heparanase-1 (HPSE) is a promising yet challenging therapeutic target. It is the only known enzyme that is responsible for cleavage of heparan sulfate (HS) side chains from heparan sulfate proteoglycans (HSPGs), and is the key enzyme involved in the remodeling and degradation of the extracellular matrix (ECM). Overexpression of HPSE is found in various types of diseases, including cancers, inflammations, diabetes, and viral infections. Inhibiting HPSE can restore ECM functions and integrity, making the development of HPSE inhibitors a highly sought-after topic. So far, all HPSE inhibitors that have entered clinical trials belong to the category of HS mimetics, and no small-molecule or drug-like HPSE inhibitors have made similar progress. None of the HS mimetics have been approved as drugs, with some clinical trials discontinued due to poor bioavailability, side effects, and unfavorable pharmacokinetics characteristics. Small-molecule HPSE inhibitors are, therefore, particularly appealing due to their drug-like characteristics. Advances in the chemical spaces and drug design technologies, including the increasing use of in vitro and in silico screening methods, have provided new opportunities in drug discovery. This article aims to review the discovery and development of small-molecule HPSE inhibitors via screening strategies to shed light on the future endeavors in the development of novel HPSE inhibitors.


Asunto(s)
Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Proteoglicanos de Heparán Sulfato/metabolismo , Proteoglicanos de Heparán Sulfato/uso terapéutico , Heparitina Sulfato/metabolismo , Heparitina Sulfato/uso terapéutico , Glucuronidasa/metabolismo , Glucuronidasa/uso terapéutico
2.
Adv Cancer Res ; 157: 251-291, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36725112

RESUMEN

The heparan sulfate proteoglycans (HSPGs) are glycoproteins that consist of a proteoglycan "core" protein and covalently attached heparan sulfate (HS) chain. HSPGs are ubiquitously expressed in mammalian cells on the cell surface and in the extracellular matrix (ECM) and secretory vesicles. Within HSPGs, the protein cores determine when and where HSPG expression takes place, and the HS chains mediate most of HSPG's biological roles through binding various protein ligands, including cytokines, chemokines, growth factors and receptors, morphogens, proteases, protease inhibitors, and ECM proteins. Through these interactions, HSPGs modulate cell proliferation, adhesion, migration, invasion, and angiogenesis to display essential functions in physiology and pathology. Under physiological conditions, the expression and localization of HSPGs are finely regulated to orchestrate their physiological functions, and this is disrupted in cancer. The HSPG dysregulation elicits multiple oncogenic signaling, including growth factor signaling, ECM and Integrin signaling, chemokine and immune signaling, cancer stem cell, cell differentiation, apoptosis, and senescence, to prompt cell transformation, proliferation, tumor invasion and metastasis, tumor angiogenesis and inflammation, and immunotolerance. These oncogenic roles make HSPGs an attractive pharmacological target for anti-cancer therapy. Several therapeutic strategies have been under development, including anti-HSPG antibodies, peptides and HS mimetics, synthetic xylosides, and heparinase inhibitors, and shown promising anti-cancer efficacy. Therefore, much progress has been made in this line of study. However, it needs to bear in mind that the roles of HSPGs in cancer can be either oncogenic or tumor-suppressive, depending on the HSPG and the cancer cell type with the underlying mechanisms that remain obscure. Further studies need to address these to fill the knowledge gap and rationalize more efficient therapeutic targeting.


Asunto(s)
Proteoglicanos de Heparán Sulfato , Neoplasias , Animales , Humanos , Proteoglicanos de Heparán Sulfato/metabolismo , Proteoglicanos de Heparán Sulfato/uso terapéutico , Neoplasias/patología , Heparitina Sulfato/metabolismo , Heparitina Sulfato/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Diferenciación Celular , Mamíferos/metabolismo
3.
Biomolecules ; 11(1)2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33450893

RESUMEN

Articular cartilage is considered to have limited regenerative capacity, which has led to the search for therapies to limit or halt the progression of its destruction. Perlecan, a multifunctional heparan sulphate (HS) proteoglycan, promotes embryonic cartilage development and stabilises the mature tissue. We investigated the immunolocalisation of perlecan and collagen between donor-matched biopsies of human articular cartilage defects (n = 10 × 2) that were repaired either naturally or using autologous cell therapy, and with age-matched normal cartilage. We explored how the removal of HS from perlecan affects human chondrocytes in vitro. Immunohistochemistry showed both a pericellular and diffuse matrix staining pattern for perlecan in both natural and cell therapy repaired cartilage, which related to whether the morphology of the newly formed tissue was hyaline cartilage or fibrocartilage. Immunostaining for perlecan was significantly greater in both these repair tissues compared to normal age-matched controls. The immunolocalisation of collagens type III and VI was also dependent on tissue morphology. Heparanase treatment of chondrocytes in vitro resulted in significantly increased proliferation, while the expression of key chondrogenic surface and genetic markers was unaffected. Perlecan was more prominent in chondrocyte clusters than in individual cells after heparanase treatment. Heparanase treatment could be a means of increasing chondrocyte responsiveness to cartilage injury and perhaps to improve repair of defects.


Asunto(s)
Cartílago Articular/patología , Tratamiento Basado en Trasplante de Células y Tejidos , Proteoglicanos de Heparán Sulfato/uso terapéutico , Regeneración , Adulto , Biomarcadores/metabolismo , Biopsia , Agregación Celular , Proliferación Celular , Condrocitos/patología , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/metabolismo , Femenino , Regulación de la Expresión Génica , Glucuronidasa/metabolismo , Humanos , Cinética , Masculino , Persona de Mediana Edad , Células Madre/metabolismo , Donantes de Tejidos
4.
Cancer Biol Ther ; 21(12): 1087-1094, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33180600

RESUMEN

Heparan sulfate proteoglycans (HSPGs) play important roles in cancer initiation and progression, by interacting with the signaling pathways that affect proliferation, adhesion, invasion and angiogenesis. These roles suggest the possibility of various strategies of regulation of these molecules. In this review, we demonstrated that the anticancer drugs can regulate the heparan sulfate proteoglycans activity in different ways: some act directly in core protein, and can bind to a specific type of HSPG. Others drugs interact with glycosaminoglycans chains, and others can act directly in enzymes that regulate HSPGs levels. We also demonstrated that the HSPGs drug targets can be divided into four groups: monoclonal antibodies, antitumor antibiotic, natural products, and mimetics peptide. Interestingly, many drugs demonstrated in this review are approved by FDA and is used in cancer therapy (Food and Drug Administration) like trastuzumab, panitumumab, bleomycin and bisphosphonate zoledronic acid (ASCO) or are in clinical trials like codrituzumab and genistein. This review should help researchers to understand the mechanism of action of anticancer drugs existing and also may inspire the discovery of new drugs that regulate the heparan sulfate proteoglycans activity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Proteoglicanos de Heparán Sulfato/uso terapéutico , Neoplasias/terapia , Anticuerpos Monoclonales/farmacología , Proteoglicanos de Heparán Sulfato/farmacología , Humanos
5.
Cells ; 9(11)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33238404

RESUMEN

Perlecan is an extracellular matrix molecule anchored to the sarcolemma by a dystrophin-glycoprotein complex. Perlecan-deficient mice are tolerant to muscle atrophy, suggesting that perlecan negatively regulates mechanical stress-dependent skeletal muscle mass. Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma to the cytosol triggers protein degradation, thereby initiating skeletal muscle atrophy. We hypothesized that perlecan regulates nNOS delocalization and activates protein degradation during this process. To determine the role of perlecan in nNOS-mediated mechanotransduction, we used sciatic nerve transection as a denervation model of gastrocnemius muscles. Gastrocnemius muscle atrophy was significantly lower in perinatal lethality-rescued perlecan-knockout (Hspg2-/--Tg) mice than controls (WT-Tg) on days 4 and 14 following surgery. Immunofluorescence microscopy showed that cell membrane nNOS expression was reduced by denervation in WT-Tg mice, with marginal effects in Hspg2-/--Tg mice. Moreover, levels of atrophy-related proteins-i.e., FoxO1a, FoxO3a, atrogin-1, and Lys48-polyubiquitinated proteins-increased in the denervated muscles of WT-Tg mice but not in Hspg2-/--Tg mice. These findings suggest that during denervation, perlecan promotes nNOS delocalization from the membrane and stimulates protein degradation and muscle atrophy by activating FoxO signaling and the ubiquitin-proteasome system.


Asunto(s)
Proteoglicanos de Heparán Sulfato/uso terapéutico , Atrofia Muscular/inducido químicamente , Óxido Nítrico Sintasa de Tipo I/efectos de los fármacos , Animales , Proteoglicanos de Heparán Sulfato/farmacología , Humanos , Ratones , Ratones Noqueados
6.
Molecules ; 20(4): 6342-88, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25867824

RESUMEN

Angiogenesis, the process of formation of new blood vessel from pre-existing ones, is involved in various intertwined pathological processes including virus infection, inflammation and oncogenesis, making it a promising target for the development of novel strategies for various interventions. To induce angiogenesis, angiogenic growth factors (AGFs) must interact with pro-angiogenic receptors to induce proliferation, protease production and migration of endothelial cells (ECs). The action of AGFs is counteracted by antiangiogenic modulators whose main mechanism of action is to bind (thus sequestering or masking) AGFs or their receptors. Many sugars, either free or associated to proteins, are involved in these interactions, thus exerting a tight regulation of the neovascularization process. Heparin and heparan sulfate proteoglycans undoubtedly play a pivotal role in this context since they bind to almost all the known AGFs, to several pro-angiogenic receptors and even to angiogenic inhibitors, originating an intricate network of interaction, the so called "angiogenesis glycomic interactome". The decoding of the angiogenesis glycomic interactome, achievable by a systematic study of the interactions occurring among angiogenic modulators and sugars, may help to design novel antiangiogenic therapies with implications in the cure of angiogenesis-dependent diseases.


Asunto(s)
Glicómica , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/metabolismo , Neovascularización Fisiológica , Moduladores de la Angiogénesis/metabolismo , Moduladores de la Angiogénesis/farmacología , Moduladores de la Angiogénesis/uso terapéutico , Animales , Proteoglicanos de Heparán Sulfato/uso terapéutico , Heparina/uso terapéutico , Humanos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Unión Proteica
7.
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
8.
Transl Stroke Res ; 4(5): 515-23, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24323378

RESUMEN

With the failure of so many pre-clinical stroke studies to translate into the clinic, there is a need to find new therapeutics to minimize the extent of cellular damage and aid in functional recovery. Domain V (DV), the c-terminal protein fragment of the vascular basement membrane component, perlecan, was recently shown to afford significant protection in multiple transient middle cerebral artery occlusion stroke models. We sought here to determine whether DV might have similar therapeutic properties in a focal photothrombosis stroke model in both young and aged mice. Young (3-month old) and aged (24-month old) mice underwent photothrombotic stroke to the motor cortex and were then treated with DV or phosphate buffered saline vehicle at different initial time points up to 7 days. Stroke volume was analyzed histologically using cresyl violet and functional recovery assessed behaviorally on both the grid-walking and cylinder tasks. In young mice, DV administration resulted in a significant decrease in infarct volume when treatment started 3 or 6 h post-stroke. In aged mice, DV administration was only protective when started 3 h post-stroke. In addition to a decrease in the area of infarction, DV treatment was effective in significantly decreasing the number of foot-faults on the grid-walking task and improving use of the stroke-affected limb in the cylinder task in both young and aged. Previously, we have shown that DV can alter the expression profile of various astroglial markers. Consistent with our previous finding, treatment groups that showed therapeutic potential in both young and aged mice also showed an elevation in glial fibrillary acidic protein (GFAP) expression in peri-infarct regions. We conclude that DV is neuroprotective and affords significant improvements in functional recovery in both young and aged mice after focal ischemia. These data also highlight a therapeutic time-window shift that is narrower in aged compared with young mice and is associated with an elevation in GFAP expression and heightened astrogliosis.


Asunto(s)
Proteoglicanos de Heparán Sulfato/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Factores de Edad , Animales , Benzofuranos , Modelos Animales de Enfermedad , Ratones , Quinolinas , Resultado del Tratamiento
9.
ACS Chem Neurosci ; 4(3): 370-4, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23509972

RESUMEN

The sad reality is that in the year 2012, people are still dying or suffering from the extreme morbidity of ischemic stroke. This tragedy is only compounded by the graveyard full of once promising new therapies. While it is indeed true that the overall mortality from stroke has declined in the United States, perhaps due to increased awareness of stroke symptoms by both the lay public and physicians, it is clear that better therapies are needed. In this regard, progress has been tremendously slowed by the simple fact that experimental models of stroke and the animals that they typically employ, rats and mice, do not adequately represent human stroke. Furthermore, the neuroprotective therapeutic approach, in which potential treatments are administered with the hope of preventing the spread of dying neurons that accompanies a stroke, typically fail for a number of reasons such as there is simply more brain matter to protect in a human than there is in a rodent! For this reason, there has been somewhat of a shift in stroke research away from neuroprotection and toward a neurorepair approach. This too may be problematic in that agents that might foster brain repair could be acutely deleterious or neurotoxic and vice versa, making the timing of treatment administration after stroke critical. Therefore, in our efforts to discover a new stroke therapy, we decided to focus on identifying brain repair elements that were (1) endogenously and actively generated in response to stroke in both human and experimental animal brains, (2) present acutely and chronically after ischemic stroke, suggesting that they could have a role in acute neuroprotection and chronic neurorepair, and (3) able to be administered peripherally and reach the site of stroke brain injury. In this review, I will discuss the evidence that suggests that perlecan domain V may be just that substance, a potential beacon of hope for stroke patients.


Asunto(s)
Proteoglicanos de Heparán Sulfato/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología , Enfermedad Aguda , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiología , Modelos Animales de Enfermedad , Proteoglicanos de Heparán Sulfato/metabolismo , Proteoglicanos de Heparán Sulfato/fisiología , Humanos , Ratones , Fármacos Neuroprotectores/metabolismo , Ratas , Accidente Cerebrovascular/metabolismo
10.
Crit Rev Eukaryot Gene Expr ; 21(1): 1-12, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21967329

RESUMEN

This review summarizes the emerging strategies that exploit the glycosaminoglycan sugar, heparan sulfate (HS), either as a substitute for, or as a supplement to growth factor (GF) therapy for regenerative medicine. Excluding autograft, the administration of GFs is currently the most effective treatment for critical bone repair and restoration. However, major hurdles in the clinical development of GF therapies include the high cost, the unwanted side effects, and the toxicity associated with the physiological overdosing required to achieve a successful outcome. These drawbacks may be overcome with the application of particular HS fractions that have been optimized to bind, recruit and enhance the biological activity of endogenous GF at the site of injury. Three HS-based treatments are discussed here: first, the single, localized, and sustained delivery of HS as a stand-alone therapeutic agent; then, the inclusion of an HS component within a delivery device so as to stabilize and potentiate the bioactivity of the incorporated GF; and finally, the growing use of HS mimetics, particularly for bone repair.


Asunto(s)
Heparitina Sulfato/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Medicina Regenerativa , Proteína Morfogenética Ósea 2/uso terapéutico , Regeneración Ósea/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Sustitución de Medicamentos , Proteoglicanos de Heparán Sulfato/uso terapéutico , Heparina/farmacología , Heparitina Sulfato/administración & dosificación , Heparitina Sulfato/análogos & derivados , Humanos , Péptidos y Proteínas de Señalización Intercelular/efectos adversos , Cicatrización de Heridas
11.
Plast Reconstr Surg ; 127(2): 541-550, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21285759

RESUMEN

BACKGROUND: ReGeneraTing Agents (RGTAs) are biodegradable polymers engineered to mimic heparan-sulfate in the extracellular matrix of damaged tissue. RGTAs improve tissue healing in several animal models by stabilizing and protecting heparin-binding growth factors and matrix proteins. RGTA restores the normal matrix architecture and supports tissue regeneration. In this study, the authors evaluated the effects of RGTA on epidermal repair and dermal remodeling in a rat burn model. METHODS: Deep second-degree burns were induced in 156 hairless rats, of which half (n = 78) received topical and intramuscular RGTA immediately after the burn followed by intramuscular RGTA weekly for 1 month. The controls (n = 78) received saline according to the same protocol. Rats were killed starting on each day of the first week and on days 14, 28, 60, 120, 240, and 365. The burns were evaluated by photography, histology, and immunohistochemistry. RESULTS: Coagulation necrosis involved the entire epidermis and superficial adnexa. Compared with the controls, speed of epidermal repair, as assessed between days 3 and 7 based on cell-layer number and anticytokeratin-14 staining, was faster in the RGTA group; and the zone of stasis, as assessed based on secondary vascular lesions in the dermis, was smaller. On day 7, reepithelialization was complete in both groups. On days 14 and 28, the remodeled dermal zone was smaller in the RGTA group. CONCLUSION: RGTA accelerated epidermal repair and protected the dermis from secondary effects of heat as quantified by zone-of-stasis size and extent of dermal remodeling.


Asunto(s)
Quemaduras/tratamiento farmacológico , Glicosaminoglicanos/uso terapéutico , Proteoglicanos de Heparán Sulfato/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos , Animales , Materiales Biomiméticos/uso terapéutico , Quemaduras/patología , Quemaduras/fisiopatología , Modelos Animales de Enfermedad , Epidermis/patología , Epidermis/fisiología , Matriz Extracelular/efectos de los fármacos , Femenino , Glicosaminoglicanos/farmacología , Proteoglicanos de Heparán Sulfato/farmacología , Queratinocitos , Necrosis , Ratas , Ratas Endogámicas , Regeneración/efectos de los fármacos
12.
J Biol Chem ; 285(10): 7312-23, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20053999

RESUMEN

Heparin-like glycans with diverse disaccharide composition and high anticoagulant activity have been described in several families of marine mollusks. The present work focused on the structural characterization of a new heparan sulfate (HS)-like polymer isolated from the mollusk Nodipecten nodosus (Linnaeus, 1758) and on its anticoagulant and antithrombotic properties. Total glycans were extracted from the mollusk and fractionated by ethanol precipitation. The main component (>90%) was identified as HS-like glycosaminoglycan, representing approximately 4.6 mg g(-1) of dry tissue. The mollusk HS resists degradation with heparinase I but is cleaved by nitrous acid. Analysis of the mollusk glycan by one-dimensional (1)H, two-dimensional correlated spectroscopy, and heteronuclear single quantum coherence nuclear magnetic resonance revealed characteristic signals of glucuronic acid and glucosamine residues. Signals corresponding to anomeric protons of nonsulfated, 3- or 2-sulfated glucuronic acid as well as N-sulfated and/or 6-sulfated glucosamine were also observed. The mollusk HS has an anticoagulant activity of 36 IU mg(-1), 5-fold lower than porcine heparin (180 IU mg(-1)), as measured by the activated partial thromboplastin time assay. It also inhibits factor Xa (IC(50) = 0.835 microg ml(-1)) and thrombin (IC(50) = 9.3 microg ml(-1)) in the presence of antithrombin. In vivo assays demonstrated that at the dose of 1 mg kg(-1), the mollusk HS inhibited thrombus growth in photochemically injured arteries. No bleeding effect, factor XIIa-mediated kallikrein activity, or toxic effect on fibroblast cells was induced by the invertebrate HS at the antithrombotic dose.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/metabolismo , Arterias , Trombosis de las Arterias Carótidas/prevención & control , Endotelio Vascular , Matriz Extracelular/química , Proteoglicanos de Heparán Sulfato/metabolismo , Proteoglicanos de Heparán Sulfato/uso terapéutico , Animales , Anticoagulantes/aislamiento & purificación , Anticoagulantes/uso terapéutico , Antitrombinas/metabolismo , Arterias/efectos de los fármacos , Arterias/patología , Arterias/efectos de la radiación , Bivalvos/metabolismo , Conformación de Carbohidratos , Línea Celular , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/efectos de la radiación , Matriz Extracelular/metabolismo , Factor Xa/metabolismo , Inhibidores del Factor Xa , Femenino , Proteoglicanos de Heparán Sulfato/química , Proteoglicanos de Heparán Sulfato/aislamiento & purificación , Heparina/metabolismo , Heparina/uso terapéutico , Cofactor II de Heparina/metabolismo , Liasa de Heparina/metabolismo , Humanos , Masculino , Ácido Nitroso/metabolismo , Ratas , Análisis Espectral/métodos , Porcinos , Trombina/antagonistas & inhibidores , Trombina/metabolismo
13.
Neurodegener Dis ; 5(3-4): 206-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18322391

RESUMEN

The beta-secretase (BACE1) is initially synthesized as a partially active zymogen containing a prodomain which can be further activated through proteolytic cleavage of the prodomain by a furin-like protease. The active site of BACE1 is large and although a number of high-affinity active-site inhibitors of BACE1 have been described, most of these compounds are large, polar and do not cross the blood-brain barrier. However, it may be possible to target other regions of the protein which regulate BACE1 allosterically. We have found that proBACE1 can be stimulated by relatively low concentrations (e.g. 1 microg/ml) of heparin. Heparin initially increases proBACE1 activity, probably by binding to the prodomain, which decreases steric inhibition at the active site. However, the heparin-activated zymogen also undergoes autocatalysis, which ultimately leads to a loss of enzyme activity. We speculate that proBACE1 can be regulated by endogenous heparan sulfate proteoglycans and that drugs which target this interaction may have value in the treatment of Alzheimer's disease.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Precursores Enzimáticos/antagonistas & inhibidores , Precursores Enzimáticos/metabolismo , Proteoglicanos de Heparán Sulfato/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Animales , Proteoglicanos de Heparán Sulfato/uso terapéutico , Humanos
14.
Curr Opin Mol Ther ; 9(4): 364-77, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17694449

RESUMEN

Heparan sulfate (HS) is a member of the family of glycosaminoglycans (GAGs) that is generally bound to a core protein to form a proteoglycan (PG). HSPGs may be cell-membrane associated (glypicans and syndecans) or located within the extracellular matrix (agrin, perlecan and type XVIII collagen). The sulfate and carboxylic groups in HS are responsible for the negative charge of the sugar chain. HS is abundantly present in the filter unit of the kidney, especially in the glomerular basement membrane (GBM), and is assumed to repel negatively charged proteins, including albumin, thereby preventing their filtration. Alterations in HS expression in the GBM have been reported in a number of renal pathologies, including diabetic nephropathy, minimal change nephropathy and membranous glomerulopathy.A decreased HS expression in the GBM generally correlates with an increase in the level of proteinuria. Progressive proteinuria may result in end-stage renal failure when untreated. Based on these findings, GAG-based drugs have been used to treat proteinuria and some, notably sulodexide, have shown beneficial effects. The biosynthesis of HS and its possible role in renal filtration are discussed, an overview of GAG-based drugs and their effect on proteinuria is provided, and possible mechanisms by which GAG-based drugs ameliorate proteinuria are discussed.


Asunto(s)
Proteoglicanos de Heparán Sulfato/uso terapéutico , Riñón , Proteinuria , Animales , Conformación de Carbohidratos , Secuencia de Carbohidratos , Citocinas/metabolismo , Fibrinolíticos/uso terapéutico , Glucuronidasa/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Proteoglicanos de Heparán Sulfato/química , Heparina/química , Heparina/uso terapéutico , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/anatomía & histología , Riñón/metabolismo , Riñón/fisiología , Glomérulos Renales/metabolismo , Glomérulos Renales/ultraestructura , Datos de Secuencia Molecular , Proteinuria/tratamiento farmacológico , Proteinuria/fisiopatología
15.
J Natl Cancer Inst ; 98(22): 1634-46, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17105986

RESUMEN

BACKGROUND: The antiangiogenic approach to controlling cancer requires a better understanding of angiogenesis and the discovery of new compounds that modulate this key biological process. Here we investigated the role of endorepellin, an angiostatic protein fragment that is derived from the C-terminus of perlecan, a heparan sulfate proteoglycan, in controlling tumor angiogenesis in vivo. METHODS: We administered human recombinant endorepellin systemically to mice bearing orthotopic squamous carcinoma xenografts or syngeneic Lewis lung carcinoma tumors. We monitored tumor growth, angiogenesis, metabolism, hypoxia, and mitotic index by using quantitative immunohistochemistry and positron emission tomography scan imaging. In addition, we determined the localization of injected endorepellin using near-infrared labeling and immunohistochemistry of frozen tumor sections. Finally, we isolated tumor-derived endothelial cells and tested whether endorepellin could interact with these cells and disrupt in vitro capillary morphogenesis. All statistical tests were two-sided. RESULTS: Endorepellin specifically targeted the tumor vasculature as determined by immunohistochemical analysis and accumulated in the tumor perivascular zones where it persisted for several days as discrete deposits. This led to inhibition of tumor angiogenesis (as measured by decreased CD31-positive cells, mean control = 1902 CD31-positive pixels, mean endorepellin treated = 343.9, difference between means = 1558, 95% confidence interval [CI] = 1296 to 1820, P<.001), enhanced tumor hypoxia, and a statistically significant decrease in tumor metabolism and mitotic index (as measured by decreased Ki67-positive cells, mean control Ki67 pixels = 5970, mean endorepellin-treated Ki67 pixels = 3644, difference between means = 2326, 95% CI = 1904 to 2749, P<.001) compared to untreated controls. Endorepellin was actively internalized by tumor-derived endothelial cells causing a redistribution of alpha2beta1 integrin such that both proteins colocalized to punctate deposits in the perivascular region. Endorepellin treatment inhibited in vitro capillary morphogenesis of both normal and tumor-derived endothelia. CONCLUSIONS: Our results provide support for the hypothesis that endorepellin is an effective antitumor vasculature agent that could be used as a therapeutic modality to combat cancer.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Células Endoteliales/efectos de los fármacos , Proteoglicanos de Heparán Sulfato/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Células Endoteliales/metabolismo , Femenino , Proteoglicanos de Heparán Sulfato/administración & dosificación , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Inmunohistoquímica , Infusiones Parenterales , Integrina alfa2beta1/metabolismo , Masculino , Ratones , Ratones Desnudos , Índice Mitótico , Neoplasias/irrigación sanguínea , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/metabolismo , Tomografía de Emisión de Positrones , Distribución Aleatoria , Proteínas Recombinantes/uso terapéutico , Tomografía Computarizada por Rayos X , Trasplante Heterólogo
16.
Trends Cardiovasc Med ; 12(2): 71-7, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11852254

RESUMEN

Heparan sulfate glycosaminoglycans (HSGAGs) are an important subset of complex polysaccharides that represent the third major class of biopolymer, along with polynucleic acids and polypeptides. However, the importance of HSGAGs in biological processes is underappreciated because of a lack of effective molecular tools to correlate specific structures with functions. Only recently have significant strides been made in understanding the steps of HSGAG biosynthesis that lead to the formation of unique structures of functional importance. Such advances now create possibilities for intervening in numerous clinical situations, creating much-needed novel therapies for a variety of pathophysiological conditions including atherosclerosis, thromboembolic disorders, and unstable angina.


Asunto(s)
Glicosaminoglicanos/biosíntesis , Proteoglicanos de Heparán Sulfato/biosíntesis , Angina Inestable/tratamiento farmacológico , Angina Inestable/metabolismo , Arteriosclerosis/tratamiento farmacológico , Arteriosclerosis/metabolismo , Membrana Celular/metabolismo , Matriz Extracelular/metabolismo , Predicción , Glicosaminoglicanos/uso terapéutico , Hemostasis , Proteoglicanos de Heparán Sulfato/uso terapéutico , Humanos , Neovascularización Fisiológica , Polisacáridos/metabolismo , Relación Estructura-Actividad
17.
Minerva Cardioangiol ; 45(6): 279-84, 1997 Jun.
Artículo en Italiano | MEDLINE | ID: mdl-9432569

RESUMEN

BACKGROUND: Antithrombotics and profibrinolytics are indicated in several clinical thrombophilic conditions such as postphlebitic syndrome. Heparansulphate, a glycosaminoglycan, shows an antithrombotic activity with low anticoagulant effect. The aim of the study was to evaluate the efficacy and the safety of heparansulphate (100 mg b.i.d.) versus mesoglycan (50 mg b.i.d.), both administered for three months in patients with postphlebitic syndrome. METHODS: The trial was performed in an open-label, controlled, with parallel and randomized groups, design. Thirty patients, with chronic venous insufficiency and a history of venous thrombosis were enrolled. Coagulative and fibrinolytic parameters (PT, aPTT, euglobulin lysis time, fibrinogen, D-dimer, t-PA, PAI-1) and signs and symptoms (cramps, paresthesia, itch, edema, local pain, skin trophism) were assessed at enrollment, 15 days later after the pharmacological washout period and after 1, 2, 3 months of treatment. Safety was evaluated by monitoring any adverse event during the study and performing clinical laboratory tests at the beginning and at the end of treatment. RESULTS: The two drugs showed a superimposable efficacy and very good tolerability. Coagulative and fibrinolytic parameters were positively affected by both treatments and the clinical benefit was particularly evident in the heparansulphate group with a significant decrease "between times" of local pain, edema, paresthesia and itching. CONCLUSIONS: These data support the use of heparansulphate, and in general of glycosaminoglycans, in the postflebitic syndrome.


Asunto(s)
Fibrinolíticos/uso terapéutico , Glicosaminoglicanos/uso terapéutico , Proteoglicanos de Heparán Sulfato/uso terapéutico , Síndrome Posflebítico/tratamiento farmacológico , Tromboembolia/prevención & control , Evaluación de Medicamentos , Tolerancia a Medicamentos , Femenino , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...