Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 216
Filtrar
1.
Exp Mol Pathol ; 124: 104740, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34998864

RESUMEN

Aloin, an anthraquinone glycoside, is one of other C-glycosides found in the leaf exudate of Aloe plant. Aloin possesses several biologic activities, including antitumor activity in vitro and in vivo. However, aloin treatment has shown iron deficiency anemia and erythropoiesis in vivo. The present study was undertaken to verify if iron supplementation could alleviate these perturbations, compared to doxorubicin, an anthracycline analog. Oral iron supplementation (20.56 mg elemental Fe/kg bw) to aloin-treated rats normalized red blood corpuscles count, hemoglobin concentration, and serum levels of total iron binding capacity and saturated transferrin, as well as hepatic iron content, hepcidin level, and mRNA expression of ferritin heavy chain (Ferr-H) and transferrin receptor-1 (TfR-1) genes. Although, serum hyperferremia, and leukocytosis were maintained, yet the spleen iron overload was substantially modulated. However, combined aloin and iron treatment increased iron storage levels in the heart and bone marrow, compared to aloin treatment per se. On other hand, oral iron supplementation to rats treated with doxorubicin (15 mg/kg bw) lessened the increase in the spleen iron content concomitantly with hepatic hepcidin level, rebound hepatic iron content to normal level, and by contrast augmented serum levels of iron and transferrin saturation. Also, activated Ferr-H mRNA expression and repressed TfR-1 mRNA expression were recorded, compared to doxorubicin treatment per se. Histopathological examination of the major body iron stores in rats supplemented with iron along with aloin or doxorubicin showed an increase in extramedullary hematopoiesis. In conclusion, iron supplementation restores the disturbances in iron homeostasis and erythropoiesis induced by aloin treatment.


Asunto(s)
Anemia Ferropénica , Suplementos Dietéticos , Emodina/análogos & derivados , Hierro , Anemia Ferropénica/tratamiento farmacológico , Anemia Ferropénica/metabolismo , Animales , Emodina/efectos adversos , Emodina/farmacología , Eritropoyesis/efectos de los fármacos , Glicósidos/efectos adversos , Glicósidos/farmacología , Hepcidinas/sangre , Hepcidinas/efectos de los fármacos , Hierro/metabolismo , Hierro/uso terapéutico , Deficiencias de Hierro/tratamiento farmacológico , Deficiencias de Hierro/metabolismo , Hígado/metabolismo , Ratas , Receptores de Transferrina/sangre , Receptores de Transferrina/efectos de los fármacos , Bazo/metabolismo
2.
J Nanobiotechnology ; 19(1): 115, 2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33892746

RESUMEN

BACKGROUND: Many studies have found that ruthenium complexes possess unique biochemical characteristics and inhibit tumor growth or metastasis. RESULTS: Here, we report the novel dual-targeting ruthenium candidate 2b, which has both antitumor and antimetastatic properties and targets tumor sites through the enhanced permeability and retention (EPR) effect and transferrin/transferrin receptor (TF/TFR) interaction. The candidate 2b is composed of ruthenium-complexed carboline acid and four chloride ions. In vitro, 2b triggered DNA cleavage and thus blocked cell cycle progression and induced apoptosis via the PARP/ATM pathway. In vivo, 2b inhibited not only Lewis lung cancer (LLC) tumor growth but also lung metastasis. We detected apoptosis and decreased CD31 expression in tumor tissues, and ruthenium accumulated in the primary tumor tissue of C57BL/6 mice implanted with LLC cells. CONCLUSIONS: Thus, we conclude that 2b targets tumors, inhibits tumor growth and prevents lung metastasis.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Nanomedicina/métodos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Rutenio/química , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/uso terapéutico , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/patología , Ciclo Celular , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Receptores de Transferrina/efectos de los fármacos , Transferrina/farmacología
3.
Drug Deliv ; 27(1): 917-926, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32597244

RESUMEN

Trojan horse technology institutes a potentially promising strategy to bring together a diagnostic or cell-based drug design and a delivery platform. It provides the opportunity to re-engineer a novel multimodal, neurovascular detection probe, or medicine to fuse with blood-brain barrier (BBB) molecular Trojan horse. In Alzheimer's disease (AD) this could allow the targeted delivery of detection or therapeutic probes across the BBB to the sites of plaques and tangles development to image or decrease amyloid load, enhance perivascular Aß clearance, and improve cerebral blood flow, owing principally to the significantly improved cerebral permeation. A Trojan horse can also be equipped with photosensitizers, nanoparticles, quantum dots, or fluorescent molecules to function as multiple targeting theranostic compounds that could be activated following changes in disease-specific processes of the diseased tissue such as pH and protease activity, or exogenous stimuli such as, light. This concept review theorizes the use of receptor-mediated transport-based platforms to transform such novel ideas to engineer systemic and smart Trojan detection or therapeutic probes to advance the neurodegenerative field.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Anticuerpos Monoclonales/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Receptores de Transferrina/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Concentración de Iones de Hidrógeno , Oxígeno Singlete/administración & dosificación , Oxígeno Singlete/farmacología
4.
J Mol Biol ; 432(14): 3989-4009, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32304700

RESUMEN

The impenetrability of the blood-brain barrier (BBB) to most conventional drugs impedes the treatment of central nervous system (CNS) disorders. Interventions for diseases like brain cancer, neurodegeneration, or age-associated inflammatory processes require varied approaches to CNS drug delivery. Cystine-dense peptides (CDPs) have drawn recent interest as drugs or drug-delivery vehicles. Found throughout the phylogenetic tree, often in drug-like roles, their size, stability, and protein interaction capabilities make CDPs an attractive mid-size biologic scaffold to complement conventional antibody-based drugs. Here, we describe the identification, maturation, characterization, and utilization of a CDP that binds to the transferrin receptor (TfR), a native receptor and BBB transporter for the iron chaperone transferrin. We developed variants with varying binding affinities (KD as low as 216 pM), co-crystallized it with the receptor, and confirmed murine cross-reactivity. It accumulates in the mouse CNS at ~25% of blood levels (CNS blood content is only ~1%-6%) and delivers neurotensin, an otherwise non-BBB-penetrant neuropeptide, at levels capable of modulating CREB signaling in the mouse brain. Our work highlights the utility of CDPs as a diverse, easy-to-screen scaffold family worthy of inclusion in modern drug discovery strategies, demonstrated by the discovery of a candidate CNS drug delivery vehicle ready for further optimization and preclinical development.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Péptidos/farmacología , Animales , Antígenos CD/química , Antígenos CD/efectos de los fármacos , Antígenos CD/genética , Antígenos CD/farmacología , Sistema Nervioso Central/efectos de los fármacos , Cistina/química , Cistina/genética , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Ratones , Neuropéptidos/química , Neuropéptidos/farmacología , Neurotensina/química , Neurotensina/farmacología , Péptidos/química , Unión Proteica/efectos de los fármacos , Receptores de Transferrina/química , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/genética
5.
Behav Brain Res ; 367: 194-200, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30953658

RESUMEN

Subarachnoid hemorrhage (SAH) may lead to brain atrophy and cognitive dysfunction. This study aimed to compare the efficacy of nimodipine and deferoxamine on these sequelae of SAH. A rat model of SAH was established by the double-hemorrhage method. These rats were injected with saline (intraperitoneal, IP), nimodipine (IP), or deferoxamine (IP and intranasal) every 12 h for 5 days after SAH. The MRI scanning, including magnetic resonance angiography, diffusion tensor imaging, T2-weighted imaging, was performed to detect the brain structure. The levels of iron metabolism-related proteins were examined by Western blot analysis. The Morris water maze (MWM) test was used to assess the cognitive function. Then, then neurons in the cortex and hippocampus were counted on hematoxylin and eosin-stained brain sections. Significant cerebral vasospasm (CVS) was found in the saline and deferoxamine groups, but not in the nimodipine group. Cerebral peduncle injury was detected in the saline and nimodipine groups, but not significantly in the deferoxamine group. Compared with nimodipine, deferoxamine reduced transferrin (Tf), Tf receptor, and ferritin levels after SAH. The MWM performances were significantly worse in the saline and nimodipine groups than that in the deferoxamine group. Brain atrophy and neuronal losses were more significant in the saline and nimodipine groups than in the deferoxamine group. Nimodipine significantly ameliorated CVS, but it did not improve the late changes in brain structure and cognitive function. Deferoxamine effectively reduced neuronal cell death and ameliorated cognitive function after SAH.


Asunto(s)
Disfunción Cognitiva/prevención & control , Deferoxamina/farmacología , Ferritinas/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Nimodipina/farmacología , Receptores de Transferrina/efectos de los fármacos , Sideróforos/farmacología , Hemorragia Subaracnoidea/tratamiento farmacológico , Transferrina/efectos de los fármacos , Vasodilatadores/farmacología , Vasoespasmo Intracraneal/prevención & control , Animales , Atrofia/prevención & control , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Masculino , Nimodipina/administración & dosificación , Ratas , Ratas Sprague-Dawley , Solución Salina/farmacología , Sideróforos/administración & dosificación , Hemorragia Subaracnoidea/complicaciones , Vasodilatadores/administración & dosificación , Vasoespasmo Intracraneal/etiología
6.
Am J Physiol Endocrinol Metab ; 316(5): E922-E930, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30888858

RESUMEN

Doxorubicin (DOX) is an effective chemotherapeutic treatment with lasting side effects in heart and skeletal muscle. DOX is known to bind with iron, contributing to oxidative damage resulting in cardiac and skeletal muscle toxicity. However, major cellular changes to iron regulation in response to DOX are poorly understood in liver, heart, and skeletal muscle. Additionally, two cotreatments, exercise (EX) and metformin (MET), were studied for their effectiveness in reducing DOX toxicity by ameliorating iron dysregulation and preventing oxidative stress. The purposes of this study were to 1) characterize the DOX-induced changes of the major iron regulation pathway in liver, heart, and skeletal muscle and 2) to determine whether EX and MET exert their benefits by minimizing DOX-induced iron dysregulation. Mice were assigned to receive saline or DOX (15 mg/kg) treatments, paired with either EX (5 days) or MET (500 mg/kg), and were euthanized 3 days after DOX treatment. Results suggest that the cellular response to DOX is protective against oxidative stress by reducing iron availability. DOX increased iron storage capacity through elevated ferritin levels in liver, heart, and skeletal muscle. DOX reduced iron transport capacity through reduced transferrin receptor levels in heart and skeletal muscle. EX and MET cotreatments had protective effects in the liver through reduced transferrin receptor levels. At 3 days after DOX, oxidative stress was mild, as shown by normal glutathione and lipid peroxidation levels. Together these results suggest that the cellular response to reduce iron availability in response to DOX treatment is sufficient to match oxidative stress.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Hipoglucemiantes/farmacología , Hierro/metabolismo , Metformina/farmacología , Condicionamiento Físico Animal , Animales , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Corazón/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/metabolismo
7.
Blood Cells Mol Dis ; 61: 37-45, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27667164

RESUMEN

Hepcidin is the key regulator of systemic iron homeostasis. The iron-sensing mechanisms and the role of intracellular iron in modulating hepatic hepcidin secretion are unclear. Therefore, we created a novel cell line, recombinant-TfR1 HepG2, expressing iron-response-element-independent TFRC mRNA to promote cellular iron-overload and examined the effect of excess holotransferrin (5g/L) on cell-surface TfR1, iron content, hepcidin secretion and mRNA expressions of TFRC, HAMP, SLC40A1, HFE and TFR2. Results showed that the recombinant cells exceeded levels of cell-surface TfR1 in wild-type cells under basal (2.8-fold; p<0.03) and holotransferrin-supplemented conditions for 24h and 48h (4.4- and 7.5-fold, respectively; p<0.01). Also, these cells showed higher intracellular iron content than wild-type cells under basal (3-fold; p<0.03) and holotransferrin-supplemented conditions (6.6-fold at 4h; p<0.01). However, hepcidin secretion was not higher than wild-type cells. Moreover, holotransferrin treatment to recombinant cells did not elevate HAMP responses compared to untreated or wild-type cells. In conclusion, increased intracellular iron content in recombinant cells did not increase hepcidin responses compared to wild-type cells, resembling hemochromatosis. Furthermore, TFR2 expression altered within 4h of treatment, while HFE expression altered later at 24h and 48h, suggesting that TFR2 may function prior to HFE in HAMP regulation.


Asunto(s)
Hepcidinas/sangre , Transferrina/farmacología , Antígenos CD/efectos de los fármacos , Antígenos CD/genética , Proteína de la Hemocromatosis/sangre , Proteína de la Hemocromatosis/efectos de los fármacos , Células Hep G2 , Hepcidinas/efectos de los fármacos , Humanos , Hierro/sangre , Sobrecarga de Hierro , ARN Mensajero/sangre , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/genética , Proteínas Recombinantes , Proteína 2 de Unión a Repeticiones Teloméricas/sangre , Proteína 2 de Unión a Repeticiones Teloméricas/efectos de los fármacos , Factores de Tiempo
8.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(5): 501-507, 2016 05 25.
Artículo en Chino | MEDLINE | ID: mdl-28087910

RESUMEN

Artemisinin is an anti-malarial drug with poor water solubility and oral absorption; so a variety of derivatives based on the parent nucleus have been developed. Compared with artemisinin, dihydroartemisinin (DHA) has a stronger anti-malaria activity, and has the advantages of high metabolic rate and better water solubility. Recent studies have discovered that DHA has a good inhibitory effect on tumor cells, which is closely related to the peroxide bridge in its molecular structure. Since tumor cells need more Fe3+ than normal cells, there are a large number of transferrin receptors on the tumor cell membrane. DHA can break the peroxide bridge in the presence of Fe2+, and the free radicals generated can play its lethal effect on tumor cells. In addition, DHA can promote endocytosis of transferrin receptor, and thus prevent cancer cells from taking Fe3+ from microenvironment. This article reviews the anti-tumor molecular mechanism of DHA, including accelerating oxidative damage, inducing apoptosis, inhibiting the growth, proliferation and invasion of tumor cells, reversing tumor multidrug resistance.


Asunto(s)
Antígenos CD/efectos de los fármacos , Antineoplásicos/farmacología , Artemisininas/farmacología , Artemisininas/farmacocinética , Radicales Libres/síntesis química , Hierro/metabolismo , Neoplasias/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Receptores de Transferrina/efectos de los fármacos , Antígenos CD/metabolismo , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Artemisininas/metabolismo , Endocitosis/efectos de los fármacos , Radicales Libres/farmacología , Humanos , Neoplasias/tratamiento farmacológico , Receptores de Transferrina/metabolismo
9.
J Med Chem ; 59(1): 294-312, 2016 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-26645570

RESUMEN

Selenosemicarbazones show marked antitumor activity. However, their mechanism of action remains unknown. We examined the medicinal chemistry of the selenosemicarbazone, 2-acetylpyridine 4,4-dimethyl-3-selenosemicarbazone (Ap44mSe), and its iron and copper complexes to elucidate its mechanisms of action. Ap44mSe demonstrated a pronounced improvement in selectivity toward neoplastic relative to normal cells compared to its parent thiosemicarbazone. It also effectively depleted cellular Fe, resulting in transferrin receptor-1 up-regulation, ferritin down-regulation, and increased expression of the potent metastasis suppressor, N-myc downstream regulated gene-1. Significantly, Ap44mSe limited deleterious methemoglobin formation, highlighting its usefulness in overcoming toxicities of clinically relevant thiosemicarbazones. Furthermore, Cu-Ap44mSe mediated intracellular reactive oxygen species generation, which was attenuated by the antioxidant, N-acetyl-L-cysteine, or Cu sequestration. Notably, Ap44mSe forms redox active Cu complexes that target the lysosome to induce lysosomal membrane permeabilization. This investigation highlights novel structure-activity relationships for future chemotherapeutic design and underlines the potential of Ap44mSe as a selective anticancer/antimetastatic agent.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Proteínas de Membrana de los Lisosomas/efectos de los fármacos , Lisosomas/efectos de los fármacos , Piridinas/síntesis química , Piridinas/farmacología , Semicarbazonas/síntesis química , Semicarbazonas/farmacología , Antioxidantes/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Ferritinas/efectos de los fármacos , Genes myc/efectos de los fármacos , Humanos , Hierro/metabolismo , Quelantes del Hierro/farmacología , Metahemoglobina/metabolismo , Modelos Moleculares , Conformación Molecular , Permeabilidad , Especies Reactivas de Oxígeno/metabolismo , Receptores de Transferrina/efectos de los fármacos , Relación Estructura-Actividad
10.
J Clin Neurosci ; 22(7): 1071-6, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25891893

RESUMEN

The current standard treatment for glioblastoma multiforme (GBM) is surgery followed by chemotherapy and external radiation. Even with the standard treatment, the 2 year survival rate for GBM is less than 20%, making research for alternative treatments necessary. Transferrin receptor 1 (TfR1) controls the rate of cellular iron uptake by tuning the amount of iron delivered to the cells to meet metabolic needs. Kawabata et al. (J Biol Chem 1999;274:20826-32) cloned a second TfR molecule known as transferrin receptor 2 (TfR2) in 1999. Multiple experimental studies have documented increased expression of TfR1 on both proliferating cells and cells that have undergone malignant transformation. Calzolari et al. concluded that TfR2 is frequently expressed in human cell lines in 2007 (Blood Cells Mol Dis 2007;39:82-91) and in GBM in particular in 2010 (Transl Oncol 2010;3:123-34). In GBM, a highly significant correlation (p<0.0001) was found between the expression level of TfR2 and overall survival, showing that higher levels of TfR2 expression were associated with an overall longer survival. The data on which of the two transferrin receptors is the better target is also unclear and should be studied. The transferrin pathway may be a promising target, but more research should be completed on the antigenicity to discern the viability of it as an immunotherapy target.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Receptores de Transferrina/genética , Transferrina/genética , Humanos , Inmunoterapia , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/metabolismo , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Transferrina/efectos de los fármacos , Transferrina/metabolismo
11.
Angew Chem Int Ed Engl ; 54(13): 3967-72, 2015 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-25650865

RESUMEN

The blood-brain barrier (BBB) is a formidable physical and enzymatic barrier that tightly controls the passage of molecules from the blood to the brain. In fact, less than 2 % of all potential neurotherapeutics are able to cross it. Here, by applying the retro-enantio approach to a peptide that targets the transferrin receptor, a full protease-resistant peptide with the capacity to act as a BBB shuttle was obtained and thus enabled the transport of a variety of cargos into the central nervous system.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Péptidos/síntesis química , Péptidos/farmacocinética , Animales , Transporte Biológico , Bovinos , Fármacos del Sistema Nervioso Central/farmacocinética , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Ratones , Péptido Hidrolasas/química , Permeabilidad , Ratas , Receptores de Transferrina/efectos de los fármacos , Estereoisomerismo
12.
Br J Pharmacol ; 172(9): 2286-99, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25537422

RESUMEN

BACKGROUND AND PURPOSE: Recently, we have described the use of caerulomycin A (CaeA) as a potent novel immunosuppressive agent. Immunosuppressive drugs are crucial for long-term graft survival following organ transplantation and treatment of autoimmune diseases, inflammatory disorders, hypersensitivity to allergens, etc. The objective of this study was to identify cellular targets of CaeA and decipher its mechanism of action. EXPERIMENTAL APPROACH: Jurkat cells were treated with CaeA and cellular iron content, iron uptake/release, DNA content and deoxyribonucleoside triphosphate pool determined. Activation of MAPKs; expression level of transferrin receptor 1, ferritin and cell cycle control molecules; reactive oxygen species (ROS) and cell viability were measured using Western blotting, qRT-PCR or flow cytometry. KEY RESULTS: CaeA caused intracellular iron depletion by reducing its uptake and increasing its release by cells. CaeA caused cell cycle arrest by (i) inhibiting ribonucleotide reductase (RNR) enzyme, which catalyses the rate-limiting step in the synthesis of DNA; (ii) stimulating MAPKs signalling transduction pathways that play an important role in cell growth, proliferation and differentiation; and (iii) by targeting cell cycle control molecules such as cyclin D1, cyclin-dependent kinase 4 and p21(CIP1/WAF1) . The effect of CaeA on cell proliferation was reversible. CONCLUSIONS AND IMPLICATIONS: CaeA exerts its immunosuppressive effect by targeting iron. The effect is reversible, which makes CaeA an attractive candidate for development as a potent immunosuppressive drug, but also indicates that iron chelation can be used as a rationale approach to selectively suppress the immune system, because compared with normal cells, rapidly proliferating cells require a higher utilization of iron.


Asunto(s)
Inmunosupresores/farmacología , Quelantes del Hierro/farmacología , Hierro/metabolismo , Piridinas/farmacología , Linfocitos T/efectos de los fármacos , Antígenos CD/efectos de los fármacos , Antígenos CD/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ferritinas/metabolismo , Humanos , Células Jurkat , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/metabolismo , Ribonucleósido Difosfato Reductasa/antagonistas & inhibidores , Ribonucleósido Difosfato Reductasa/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/patología
13.
Ther Deliv ; 4(3): 369-94, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23442082

RESUMEN

Receptor-based targeting of therapeutics may be a fascinating proposition to improve the therapeutic efficacy of encapsulated drugs. The development of safe and effective nanomedicines is a prerequisite in the current nanotechnological scenario. Currently, the surface engineering of nanocarriers has attracted great attention for targeted therapeutic delivery by selective binding of targeting ligand to the specific receptors present on the surface of cells. In this review, we have discussed the current status of various receptors such as transferrin, lectoferrin, lectin, folate, human EGF receptor, scavenger, nuclear and integrin, which are over-expressed on the surface of cancer cells; along with the relevance of targeted delivery systems such as nanoparticles, polymersomes, dendrimers, liposomes and carbon nanotubes. The review also focuses on the effective utilization of receptor-based targeted delivery systems for the management of cancer in effective ways by minimizing the drug-associated side effects and improving the therapeutic efficacy of developed nano-architectures.


Asunto(s)
Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Receptores de Superficie Celular/efectos de los fármacos , Animales , Receptores ErbB/efectos de los fármacos , Humanos , Receptores de Hialuranos/efectos de los fármacos , Nanopartículas , Receptores de Leptina/efectos de los fármacos , Receptores de Transferrina/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos
14.
Cancer Chemother Pharmacol ; 71(3): 799-807, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23328867

RESUMEN

PURPOSE: Transferrin receptor (TfR) is a cell membrane-associated glycoprotein involved in the cellular uptake of iron and the regulation of cell growth. Recent studies have shown elevated expression levels of TfR on cancer cells compared with normal cells. We previously designed a TfR-lytic hybrid peptide, which combines the TfR-binding peptide and a lytic peptide, and reported that it bound specifically to TfR and selectively killed cancer cells. Furthermore, the intravenous administration of TfR-lytic peptide in an athymic mouse model significantly inhibited tumor progression. To evaluate the immunogenicity of this peptide as a novel and potent anticancer agent, we investigated whether TfR-lytic hybrid peptide elicits cellular and humoral immune responses to produce antibodies. We also examined the toxicity of this peptide in syngeneic mice. METHODS: We performed hematologic and blood chemistry test and histological analysis and assessed hemolytic activity to check toxicity. To evaluate the immunogenicity, measurement of murine interferon-gamma and detection of TfR-lytic-specific antibody by ELISA were demonstrated. RESULTS: No T cell immune response or antibodies were detected in the group treated with TfR-lytic hybrid peptide. No hematologic toxicity, except for a decrease in leukocytes, was observed, and no remarkable influence on metabolic parameters and organs (liver, kidney, and spleen) was noted. CONCLUSIONS: Therefore, TfR-lytic hybrid peptide might provide an alternative therapeutic option for patients with cancer.


Asunto(s)
Antineoplásicos/farmacología , Péptidos/farmacología , Receptores de Transferrina/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Antineoplásicos/inmunología , Línea Celular Tumoral , Eritrocitos/efectos de los fármacos , Femenino , Citometría de Flujo , Hemólisis/efectos de los fármacos , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Interferón gamma/farmacología , Ganglios Linfáticos/citología , Ganglios Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Trasplante de Neoplasias , Péptidos/inmunología , Trasplante Isogénico
15.
Front Biosci (Landmark Ed) ; 16(4): 1388-412, 2011 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-21196238

RESUMEN

Within the broad spectrum of nanoparticulate carriers, polymeric and lipid-core micelles, liposomes, solid nanoparticles and many others have demonstrated great biological properties which make them excellent pharmaceutical delivery systems. In particular, micelles and liposomes have been shown to have good longevity in the blood that allows their accumulation in pathological areas with a compromised vasculature; can possess specific targeting to disease sites when various targeting ligands are attached to the surface of the nanocarriers or to surface-attached cell-penetrating molecules (like TAT peptide) to enhance intracellular penetration; possess stimulus-sensitivity allowing for drug release from the carriers under certain pathological conditions; and show contrast properties with carrier loading of various contrast materials that allow for direct carrier visualization in vivo. The engineering of "multifunctional pharmaceutical nanocarriers" based on the combination of several useful properties in the same system can significantly enhance the efficacy of many therapeutic and diagnostic protocols. This review considers the current status and next future directions in the emerging area of nanomedicine with particular attention to two lipid-based nanoparticulate systems: liposomes and micelles.


Asunto(s)
Portadores de Fármacos/química , Liposomas/administración & dosificación , Nanomedicina/métodos , Anticuerpos Monoclonales/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Transportadores de Ácido Fólico/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Ligandos , Micelas , Polietilenglicoles/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Receptores de Transferrina/efectos de los fármacos
16.
Transpl Int ; 24(2): 167-74, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20840666

RESUMEN

The expression of TfR/CD71 in T-cell surface plays a pivotal role in T-cell activation and proliferation. Anti-human-TfR monoclonal antibody could be used as an immunosuppressant in transplant therapy because of their potential to suppress T-cell responses to alloantigens. We therefore examined the feasibility of an anti-human-TfR chimeric antibody (D2C) in suppression of T-cell activation in vitro and graft-versus-host reaction (GVHR) in animals. D2C is a chimeric antibody produced by introducing the human Fc fragment. This antibody showed low antigenicity but high suppressive effect manifested by high potency to block the activation and proliferation of lymphocytes in response to alloantigens. D2C also showed capability to mediate complement-dependent cytotoxicity, which could be correlated with TfR expression in peripheral blood mononuclear cells (PBMCs). Importantly, administration of D2C significantly prolonged survival time of nude mice transplanted with human PBMCs when compared with that of control IgG-treated animals (61.2 ± 4.46 vs. 22.1 ± 5.5 days), which is associated with inhibited GVHR characterized by decreased interleukin-1 and tumor necrosis factor α production derived from transplanted PBMCs. Human-TfR chimeric antibody such as D2C could be a valuable option for the treatment of acute form of graft-versus-host disease.


Asunto(s)
Antígenos CD/farmacología , Rechazo de Injerto/inmunología , Receptores de Transferrina/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Proliferación Celular/efectos de los fármacos , Rechazo de Injerto/patología , Humanos , Interleucina-1/biosíntesis , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Ratones , Ratones Desnudos , Receptores de Transferrina/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Br J Pharmacol ; 159(7): 1497-510, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20233216

RESUMEN

BACKGROUND AND PURPOSE: Andrographolide is the active component of Andrographis paniculata, a plant used in both Indian and Chinese traditional medicine, and it has been demonstrated to induce apoptosis in different cancer cell lines. However, not much is known about how it may affect the key receptors implicated in cancer. Knowledge of how andrographolide affects receptor trafficking will allow us to better understand new mechanisms by which andrographolide may cause death in cancer cells. EXPERIMENTAL APPROACH: We utilized the well-characterized epidermal growth factor receptor (EGFR) and transferrin receptor (TfR) expressed in epidermoid carcinoma (A-431) cells as a model to study the effect of andrographolide on receptor trafficking. Receptor distribution, the total number of receptors and surface receptors were analysed by immunofluorescence, Western blot as well as flow-cytometry respectively. KEY RESULTS: Andrographolide treatment inhibited cell growth, down-regulated EGFRs on the cell surface and affected the degradation of EGFRs and TfRs. The EGFR was internalized into the cell at an increased rate, and accumulated in a compartment that co-localizes with the lysosomal-associated membrane protein in the late endosomes. CONCLUSION AND IMPLICATIONS: This study sheds light on how andrographolide may affect receptor trafficking by inhibiting receptor movement from the late endosomes to lysosomes. The down-regulation of EGFR from the cell surface also indicates a new mechanism by which andrographolide may induce cancer cell death.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Diterpenos/farmacología , Receptores ErbB/efectos de los fármacos , Receptores de Transferrina/efectos de los fármacos , Western Blotting , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Regulación hacia Abajo , Receptores ErbB/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Receptores de Transferrina/metabolismo
18.
Blood Cells Mol Dis ; 42(1): 5-13, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19019709

RESUMEN

In a recent study we have explored TfR2 expression in a panel of cancer cell lines and we observed that about 40% of these cell lines clearly express TfR2. Taking advantage of this observation and considering the frequent overexpression of c-Myc in cancer cells we have explored the existence of a possible relationship between c-Myc and TfR2 in these cell lines. Our results provided evidence that TfR2(+) cell lines express low c-Myc levels and low TfR1 levels, while TfR2(-) cell lines express high c-Myc and TfR1 levels. Using the erythroleukemic K562 TfR2(+) cells as a model, we observed that agents that enhance c-Myc expression, such as iron, determine a decrease of TfR2 expression, while molecules that induce a decreased c-Myc expression, such as the iron chelator desferoxamine or the kinase inhibitor ST 1571, induce an enhanced TfR2 expression. On the other hand, we have evaluated a possible effect of hypoxia and nitric oxide on TfR2 expression in erythroleukemia K526 and hepatoma HepG2 cells, providing evidence that: (i) agents inducing cellular hypoxia, such as CoCl(2), elicited a marked upmodulation of TfR1, but a downmodulation of TfR2 expression; (ii) NO(+) donors, such as sodium nitroprusside (SNP), induced a moderate decrease of TfR1, associated with a marked decline of TfR2 expression; (iii) NO donors, such as S-Nitroso-N-Acetylpenicillamine (SNAP), induced a clear increase of TfR1, associated with a moderate upmodulation of TfR2 expression. The ensemble of these observations suggests that in cancer cell lines TfR2 expression can be modulated through stimuli similar to those known to act on TfR1 and these findings may have important implications for our understanding of the role of TfR2 in the regulation of iron homeostasis.


Asunto(s)
Antígenos CD/biosíntesis , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Transferrina/biosíntesis , Antígenos CD/efectos de los fármacos , Antimutagênicos/farmacología , Apoferritinas/biosíntesis , Benzamidas , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Cobalto/farmacología , Deferoxamina/farmacología , Humanos , Mesilato de Imatinib , Hierro/farmacología , Proteína 2 Reguladora de Hierro/biosíntesis , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Penicilamina/análogos & derivados , Penicilamina/farmacología , Piperazinas , Proteínas Proto-Oncogénicas c-myc/efectos de los fármacos , Pirimidinas/farmacología , Receptores de Transferrina/efectos de los fármacos , Sideróforos/farmacología
19.
Anticancer Drugs ; 19(3): 247-55, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18510170

RESUMEN

Dihydroartemisinin (DHA), a water-soluble active metabolite of artemisinin derivatives, is the safest and most effective antimalarial analog of artemisinin. In the present investigation, we assessed the apoptotic effect of DHA on leukemia HL60 cells and its regulation of transferrin receptor (TfR). Cell growth inhibition was assessed by Trypan blue exclusive staining; the expression of caspase-3, Bcl-2, and Bax in HL60 cells was evaluated by Western blotting; DHA-induced apoptosis was determined by AO/EB double staining, DNA fragmentation assay, and flow cytometric analysis; the expression of TfR in HL60 cells was examined by real-time PCR assays, Western blotting, and flow cytometric analysis. DHA could specifically reduce the mRNA and protein expression of TfR in HL60 cells, and the flow cytometric analysis presented the unity tendency that the TfR content decreased progressively in a dose-dependent manner. Consequently, DHA exhibited high anticancer activity in HL60 cells; MTT assay and growth inhibition assay showed that DHA could specifically inhibit the growth of HL60 cells in a dose-dependent (0.25-8 micromol/l) and time-dependent (12-72 h) manner. DHA-induced DNA fragmentation also induced the activation of caspase-3 and influenced the expression of Bcl-2 and Bax. Taken together, these data from our study show that DHA can induce HL60 cell apoptosis via the effect of downregulation TfR expression resulting in an induction of apoptosis through the mitochondrial pathway, and it might be a potential antileukemia strategy for leukemia therapy.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Receptores de Transferrina/efectos de los fármacos , Sesquiterpenos/farmacología , Antimaláricos/administración & dosificación , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Artemisininas/administración & dosificación , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Fragmentación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HL-60 , Humanos , Mitocondrias/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Transferrina/genética , Sesquiterpenos/administración & dosificación , Coloración y Etiquetado , Factores de Tiempo , Azul de Tripano , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
20.
Exp Neurol ; 206(2): 209-19, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17580085

RESUMEN

A 25-35% reduction of brain cytochrome oxidase (COx) activity found in Alzheimer's disease (AD) could contribute to neuronal dysfunction and cognitive impairment. The present study replicated the reduction in brain COx activity in rats by administering sodium azide (NaN(3)) for 4 weeks via Alzet minipumps at the rate of 1 mg/kg/h, and determined its effect on hippocampal cholinergic transmission, spatial and episodic memory. NaN(3) caused a selective reduction in choline acetyltransferase (ChAT) immunoreactivity in the diagonal band, a major source of cholinergic input to the hippocampus and cingulate cortex, without altering the number of cholinergic neurons. NaN(3) also induced a significant increase in vesicular acetylcholine transporter (VAChT)-immunoreactive varicosities, GAP-43 in the subgranular layer and of transferrin receptors (TfR) in the hilus of the dentate gyrus. These neurochemical changes were associated with impairment in spatial learning in the Morris water maze and in episodic memory in the object recognition test. Chronic treatment with ladostigil, a novel cholinesterase and monoamine oxidase inhibitor, prevented the decrease in ChAT in the diagonal band, the compensatory increase in synaptic plasticity and TfR and the memory deficits without restoring COx activity. Ladostigil had no significant effect on ChAT activity, synaptic plasticity or TfR in control rats. Ladostigil may have a beneficial effect on cognitive deficits in AD patients that have a reduction in cortical COx activity and cholinergic hypofunction.


Asunto(s)
Acetilcolina/metabolismo , Fibras Colinérgicas/efectos de los fármacos , Complejo IV de Transporte de Electrones/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Indanos/farmacología , Trastornos de la Memoria/enzimología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/fisiopatología , Animales , Fibras Colinérgicas/metabolismo , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Inhibidores Enzimáticos/toxicidad , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Hipocampo/fisiopatología , Indanos/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/metabolismo , Núcleos Septales/efectos de los fármacos , Núcleos Septales/enzimología , Núcleos Septales/fisiopatología , Azida Sódica/toxicidad , Resultado del Tratamiento , Proteínas de Transporte Vesicular de Acetilcolina/efectos de los fármacos , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...