Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Omega ; 9(1): 67-80, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38222554

RESUMEN

Tissue engineering is an emerging technological field that aims to restore and replace human tissues. A significant number of individuals require bone replacement annually as a result of skeletal abnormalities or accidents. In recent decades, notable progress has been made in the field of biomedical research, specifically in the realm of sophisticated and biocompatible materials. The purpose of these biomaterials is to facilitate bone tissue regeneration. Carbon nanomaterial-based scaffolds are particularly notable due to their accessibility, mechanical durability, and biofunctionality. The scaffolds exhibit the capacity to enhance cellular proliferation, mitigate cell damage, induce bone tissue growth, and maintain biological compatibility. Therefore, they play a crucial role in the development of the bone matrix and the necessary cellular interactions required for bone tissue restoration. The attachment, growth, and specialization of osteogenic stem cells on biomaterial scaffolds play critical roles in bone tissue engineering. The optimal biomaterial should facilitate the development of bone tissue in a manner that closely resembles that of human bone. This comprehensive review encompasses the examination of graphene oxide (GO), carbon nanotubes (CNTs), fullerenes, carbon dots (CDs), nanodiamonds, and their respective derivatives. The biomaterial frameworks possess the ability to replicate the intricate characteristics of the bone microenvironment, thereby rendering them suitable for utilization in tissue engineering endeavors.

2.
Cell Tissue Res ; 394(2): 245-255, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37548764

RESUMEN

Recent years have seen the emergence of tissue engineering strategies as a means to overcome some of the limits of conventional medical treatment. A biomaterial with tailored physio-chemical characteristics is used in this sophisticated method to transport stem cells and growth factors/bioactive substances, or to attract local endogenous cells, enabling new tissue formation. Biomaterials might serve as a biomimetic structure inspired by the natural milieu, assisting the cells in establishing their natural relationships. Such a method would benefit from having ready access to an abundant reservoir of stem cells with strong tissue regeneration capacity, in addition to using biological compatible material to promote new tissue creation. Teeth may have a plethora of self-renewing, multipotent mesenchymal stem cell (MSC) populations. Recent advancements and promising directions for cell transplantation and homing techniques using dental MSCs for tissue regeneration are discussed in this review paper. Overall, this research paints a picture of the present landscape of new approaches to using tooth-derived MSCs in conjunction with biomaterials and bioactive substances for tissue regeneration.


Asunto(s)
Células Madre Mesenquimatosas , Diente , Ingeniería de Tejidos/métodos , Materiales Biocompatibles/farmacología , Células Madre , Pulpa Dental
3.
Peptides ; 163: 170974, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36775021

RESUMEN

Bone cells express the glucagon-like peptide 1 receptor (GLP-1R). However, its presence and role in human dental pulp derived stem cells (hDPSCs) remains elusive. Hence, in the current study, we isolated hDPSCs and differentiated them into osteoblasts, where GLP-1R expression was found to be upregulated during osteoblast differentiation. GLP-1 receptor agonist, liraglutide peptide treatment, increased osteoblast differentiation in hDPSCs by increasing calcium deposition, ALP activity, and osteoblast marker genes, Runx2, type 1 col, osteonectin, and osteocalcin. Furthermore, activation of long non-coding RNA (LncRNA) LINC00968 and microRNA-3658 signalling increased Runx2 expression. Specifically, liraglutide increased LncRNA-LINC00968 expression while decreasing miR-3658 expression. LINC00968 targets miR-3658, and miR-3658 targets Runx2. Additionally, in an in-vivo study, zebrafish scale regeneration model, liraglutide promoted calcium deposition, osteoblastic cell count, collagen 1α, osteonectin, osteocalcin, runx2a MASNA isoform expression (transcribed from promoter P1), and Ca/P ratio in scales. Overall, GLP-1R activation promotes osteoblast differentiation via Runx2/LncRNA-LINC00968/miR-3658 signalling in hDPSCs and promotes bone formation in zebrafish scale regeneration.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Animales , Humanos , Osteogénesis/genética , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Osteonectina/metabolismo , Osteonectina/farmacología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/farmacología , Osteocalcina/genética , Liraglutida/farmacología , Calcio/metabolismo , Pulpa Dental/metabolismo , Diferenciación Celular/genética , MicroARNs/genética , MicroARNs/metabolismo , Células Madre , Osteoblastos/metabolismo
4.
Chem Biol Interact ; 349: 109674, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34562440

RESUMEN

We have assessed the molecular role of Rutin and rutin-Zn(II) complex on osteoblast differentiation and mineralization in human dental pulp cells and zebrafish model. The biocompatibility of the rutin-Zn(II) complex was determined using MTT and chick embryotoxicity assays. Alizarin red staining and ALP measurements were performed to study the osteogenic role of Rutin and rutin-Zn(II) complex at the cellular level in hDPSCs. At molecular level, following rutin and rutin-Zn(II) exposure, the mRNA expression profile of osteoblast markers such Runx2, type 1 col, OC, and ON were investigated. In addition to this, the expression of negative regulators of osteoblast development such Smad7, Smurf1, and HDAC7 waere studied by Real time RT-PCR analysis. The osteogenic role of prepared complex under in vivo was studied by an in-house zebrafish scale model followed by osteoblast differentiation markers expression profiling and Ca:P level measurement by ICP-MS. Rutin and the rutin-Zn(II) complex were found to be non-toxic till 10 µM and increased the expression of osteoblast differentiation marker genes. It also enhanced calcium deposition in both in vitro and in vivo models. Osteogenic property of rutin-Zn(II) in hDPSCs was found be mediated by Smad7, Smurf1, and HDAC7 and enhancing Runx2 expression. Our study warrants the possible use of rutin-Zn(II) as naïve agent or in combination with other bone scaffolding systems/materials for bone tissue engineering applications.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Complejos de Coordinación/farmacología , Osteogénesis/efectos de los fármacos , Rutina/química , Zinc/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Pulpa Dental/citología , Humanos , Osteocalcina/genética , Osteocalcina/metabolismo , Proteína smad7/genética , Proteína smad7/metabolismo , Células Madre/citología , Células Madre/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Pez Cebra/metabolismo
5.
Mater Sci Eng C Mater Biol Appl ; 126: 111856, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34082925

RESUMEN

Treatment of bone disease and disorders is often challenging due to its complex structure. Each year millions of people needs bone substitution materials with quick recovery from diseases conditions. Synthetic bone substitutes mimicking structural, chemical and biological properties of bone matrix structure will be very obliging and of copious need. In this work, we reported on the fabrication of bioinspired, biomimetic, multifunctional bone-like three-dimensional (3D) membranes made up of inorganic bioactive glass fibers matrixed organic collagen structure. The 3D structure is arranged as a stacked-layer similar to the order of apatite and neotissue formation. Comparative studies on collagen, collagen with hollow and solid bioactive glass fibers evidenced that, collagen/hollow bioactive glass is mechanically robust, has optimal hydrophilicity, simultaneously promotes bioactivity and in situ forming drug delivery. The 3D membrane displays outstanding mechanical properties apropos to the bioactive glass fibers arrangement, with its Youngs modulus approaching the modulus of cortical bone. The in vitro cell culture studies with fibroblast cells (3T3) on the membranes display enhanced cell adhesion and proliferation with the cell alignment similar to anisotropic cell alignment found in the native bone extracellular matrix. The membranes also support 3D cell culturing and exhibits cell proliferation on the membrane surface, which extends the possibility of its bone tissue engineering application. The alkaline phosphatase assessment and alizarin red staining of osteoblast cells (MG63) depicted an enhanced osteogenic activity of the membranes. Notable Runx2, Col-Type-1 mRNA, osteocalcin, and osteonectin levels were found to be significantly increased in cells grown on the collagen/hollow bioactive glass membrane. This membrane also promotes vascularization in the chick chorioallantoic membrane model. The results altogether evidence this multifunctional 3D membrane could potentially be utilized for treatment of bone defects.


Asunto(s)
Huesos , Vidrio , Osteoblastos , Ingeniería de Tejidos , Línea Celular , Colágeno , Humanos , Osteogénesis , Andamios del Tejido
6.
Lab Invest ; 101(8): 1011-1025, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33846539

RESUMEN

Osteosarcoma (OS) is a type of bone tumor conferred with high metastatic potential. Attainable growth of tumors necessitates functional vasculature mediated by sprouting angiogenesis (SA) and intussusceptive angiogenesis (IA). However, the regulation of IA and SA is still unclear in OS. To understand the mechanisms adopted by OS to induce angiogenesis, initially, we assessed the expression profile of a set of miRNAs' in both OS cells (SaOS2 and MG63) and normal bone cells. Amongst them, miR-432-5p was found to be highly downregulated in OS. The functional role of miR-432-5p in OS was further analyzed using miR-432-5p mimic/inhibitor. Platelet-derived growth factor-B (PDGFB) was found to be a putative target of miR-432-5p and it was further confirmed that the PDGFB 3'UTR is directly targeted by miR-432-5p using the luciferase reporter gene system. PDGFB was found to be secreted by OS to regulate angiogenesis by targeting the cells in its microenvironment. The conditioned medium obtained from miR-432-5p mimic transfected MG63 and SaOS2 cells decreased cell viability, proliferation, migration, and aorta ring formation in endothelial cells. The miRNA mimic/inhibitor transfected MG63 and SaOS2 cells were placed on SA (day 6) and IA (day 9) phase of CAM development to analyze SA and IA mechanisms. It was found that miR-432-5p mimic transfection in OS promotes the transition of SA to IA which was documented by the angiogenic parameters and SA and IA-associated gene expression. Interestingly, this outcome was also supported by the zebrafish tumor xenograft model. Corroborating these results, it is clear that miR-432-5p expression in OS cells regulates SA and IA by targeting PDGFB genes. We conclude that targeting miR-432-5p/PDGFB signaling can be a potential therapeutic strategy to treat OS along with other existing strategies.


Asunto(s)
MicroARNs/metabolismo , Neovascularización Patológica/metabolismo , Osteosarcoma , Proteínas Proto-Oncogénicas c-sis/metabolismo , Microambiente Tumoral/genética , Animales , Línea Celular Tumoral , Humanos , MicroARNs/genética , Neovascularización Patológica/genética , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Proteínas Proto-Oncogénicas c-sis/genética , Pez Cebra
7.
Eur J Pharm Sci ; 160: 105768, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33607242

RESUMEN

Bone tissue regeneration is augmented by biocompatible nanofiber scaffolds, that supports reliable and enhanced bone formation. Zinc is an essential mineral that is vital for routine skeletal growth and it emerges to be able to improve bone regeneration. Phytochemicals, particularly flavonoids have achieved prominent interest for their therapeutic ability, they have demonstrated promising effects on bone by encouraging osteoblastogenesis, which finally leads to bone formation. In this study, we have synthesized bioactive zinc(II) quercetin complex material and used for nanofibers scaffold fabrication to enhance bone tissue regeneration property. Two derivatives of zinc(II) quercetin complexes [(Zn(quercetin) (H2O)2) (Zn+Q), and Zn(quercetin)(phenanthroline) (Zn+Q(PHt)) have been synthesized and characterized using UV-Visible spectrophotometer and Fourier Transform-IR spectroscopy. The UV-Visible absorption and IR spectra prove the B-ring chelation of the flavonoid quercetin to zinc(II) rather C-ring chelation. The potential ability of the above synthesized metal complexes on osteogenesis and angiogenesis have been studied. Besides the bioactivity of the metal complexes, the control quercetin has also been examined. The chick embryo chorioallantoic membrane (CAM) assay demonstrated that the angiogenic parameters were increased by the (Zn+Q(PHt)) complex. Amongst, (Zn+Q(PHt)) complex showed significant activity and thereby this complex has been further examined for the bone tissue activity by incorporating the complex into a nanofiber through electrospinning method. At the molecular level, Runx2, mRNA and protein, ALP and type 1 collagen mRNAs, and osteoblast-specific microRNA, pre-mir-15b were examined using real time RT-PCR and Western blot assay. Histology studies showed that the (PCL/gelatin/Zn+Q(PHt)) was biocompatibility in-ovo. Overall, the present study showed that quercetin-zinc complex (Zn+Q(PHt)) incorporated into PCL/gelatin nanofiber can act as a pharmacological agent for treating bone associated defects and promote bone regeneration.


Asunto(s)
Nanofibras , Animales , Regeneración Ósea , Huesos , Proliferación Celular , Embrión de Pollo , Gelatina , Poliésteres , Ingeniería de Tejidos , Andamios del Tejido , Zinc
8.
Life Sci ; 256: 117993, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32574664

RESUMEN

Flavonoids are known for their wide range of bioactive properties including beneficial effect on bone formation. Their intense metal ion chelating capacity endorsed their nomination as a new biomaterial for biomedical applications. The present study examined the functional role of Kaemferal-Zinc(II) (Kaem-Zn) complex in bone formation, in vitro and in vivo. The cyto-compatibility assay confirmed that upto 25 µM of Kaem and Kaem-Zn complex was non-toxic. In fact, it facilitates ALP activity and accumulation of calcium in osteoblast; it was confirmed by Alizarin red and von Kossa staining. In addition to this, osteoblast markers, Runx2, type 1 col., ALP mRNAs expression, and osteocalcin and osteonectin secretory proteins level were also induced by the Kaem-Zn complex. Furthermore, bone forming ability of Kaem and Kaem-Zn was assessed by zebrafish model. The optimal concentration of Kaem and Kaem-Zn was determined by the viability assay of Zebrafish larvae. Osteoblasts distribution in scale, vertebrae and caudal fin ossification was studied by alizarin red staining accompanied by confocal imaging were carried out in adult zebrafish exposed to Kaem and Kaem-Zn complex. To sum up, our findings showed that Kaem promotes bone growth, and Kaem-Zn complex has further strengthened it. Kaem-Zn complex could be an effectively explored and used use in bone tissue engineering.


Asunto(s)
Quempferoles/administración & dosificación , Quempferoles/síntesis química , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Zinc/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Pez Cebra , Zinc/química
9.
Life Sci ; 256: 118011, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32592723

RESUMEN

Melatonin is recognized as an anti-angiogenic agent, but its function in the tumor microenvironment especially in osteosarcoma remains uncertain. Among the selected miRNAs, miR-205, miR-424, miR-140, miR-106, and miR-519 were upregulated by melatonin in osteosarcoma cells. The functional role of miR-424-5p in osteosarcoma was further analyzed using miR-424-5p mimic/inhibitor. VEGFA mRNA and protein expression were altered by miR-424-5p mimic/inhibitor transfection with and without melatonin treatment and it was further identified that the VEGFA 3'UTR is directly targeted by miR-424-5p using the luciferase reporter gene system. The conditioned medium from SaOS2 and MG63 cells treated with melatonin and/or transfected with miR-424-5p mimic/inhibitor was exposed to endothelial cells, and cell proliferation and migration was analyzed. MG-63 and SaOS2 cells are also transfected with miR-424-5p inhibitors and positioned on CAM vascular bed to study the angiogenic activity at both morphological and molecular level under melatonin treatment. Our observations demonstrate for the first time that, melatonin upregulated the expression of miR-424-5p in osteosarcoma inhibiting VEGFA. Furthermore, it suppresses tumor angiogenesis, modulating surrounding endothelial cell proliferation and migration as well as the morphology of blood vessels, and angiogenic growth factors. These findings suggest that melatonin could play a pivotal role in tumor suppression via miR-424-5p/VEGFA axis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias Óseas/tratamiento farmacológico , Melatonina/farmacología , Osteosarcoma/tratamiento farmacológico , Animales , Neoplasias Óseas/irrigación sanguínea , Línea Celular Tumoral , Pollos , Yema de Huevo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , MicroARNs/genética , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Osteosarcoma/irrigación sanguínea , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Life Sci ; 252: 117670, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32298741

RESUMEN

Deregulation of angiogenesis is a key reason for tumor growth and progression. Several anti-angiogenic drugs in clinical practice attempt to normalize abnormal tumor vasculature. Unfortunately, these drugs are ineffective due to the development of resistance in patients after drug holidays. A sizable literature suggests that resistance to these anti-angiogenic drugs occurs due to various compensatory mechanisms of tumor angiogenesis. Therefore, we describe different compensatory mechanisms of tumor angiogenesis, and explain why intussusceptive angiogenesis (IA), is a crucial mechanism of compensatory angiogenesis in tumors which resist anti-VEGF (vascular endothelial growth factor) therapies. IA is often overlooked due to the scarcity of experimental models. Therefore, we examine data from existing experimental models and our novel ex-ovo model of angiogenesis in chick embryos, and explain the important genes and signaling pathways driving IA. Using bio-informatic analyses of major genes regulating conventional sprouting angiogenesis (SA) and intussusceptive angiogenesis, we provide fresh insights on the 'angiogenic switch' which regulates the transition from SA to IA. Finally, we examine the interplay between molecules regulating SA, IA, and molecules known to promote tumor progression. Based on these analyses, we conclude that intussusceptive angiogenesis (IA) is a promising therapeutic target for developing effective anti-cancer treatment regimes.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Embrión de Pollo , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , Neoplasias/irrigación sanguínea , Neovascularización Patológica/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
11.
Am J Physiol Heart Circ Physiol ; 317(2): H213-H225, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31125258

RESUMEN

Ischemic heart disease is a growing worldwide epidemic. Improvements in medical and surgical therapies have reduced early mortality after acute myocardial infarction and increased the number of patients living with chronic heart failure. The irreversible loss of functional cardiomyocytes puts these patients at significant risk of ongoing morbidity and mortality after their index event. Recent evidence suggests that inflammation is a key mediator of postinfarction adverse remodeling in the heart. In this review, we discuss the cardioprotective and deleterious effects of inflammation and its mediators during acute myocardial infarction. We also explore the role of mesenchymal stem cell therapy to limit secondary injury and promote myocardial healing after myocardial infarction.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Infarto del Miocardio/cirugía , Miocarditis/cirugía , Miocitos Cardíacos/inmunología , Regeneración , Animales , Humanos , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocarditis/inmunología , Miocarditis/metabolismo , Miocarditis/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Recuperación de la Función , Cicatrización de Heridas
12.
Int J Biol Macromol ; 121: 38-54, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30291931

RESUMEN

Tissue engineering (TE) is a promising approach for repairing diseased and damaged bone tissue. Injectable hydrogel based strategies offer a wide range of applications in rapid recovery of bone defects by acting as filler materials and depots for delivering various bioactive molecules and averting the need for surgical intervention. Chitosan (CS), a natural polysaccharide, forms a thermosensitive injectable hydrogel through the addition of beta-glycerophosphate (ß-GP). This hybrid hydrogel possesses numerous advantages namely mimicking native extracellular matrix (ECM) and providing an amenable microenvironment for cell growth. In this review, a brief insight into the gelation mechanism of CS/GP hydrogels, modifications, bioactive additives and their applications in treating bone defects are presented.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Quitosano/química , Glicerofosfatos/química , Hidrogeles/química , Hidrogeles/farmacología , Ingeniería de Tejidos/métodos , Animales , Humanos , Hidrogeles/administración & dosificación , Inyecciones
13.
Int J Biol Macromol ; 123: 1229-1240, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30468812

RESUMEN

Angiogenesis is a distinct process which follows sprouting angiogenesis (SA) and intussusceptive angiogenesis (IA) forming the basis for various physiological and pathological scenarios. Angiogenesis is a double edged sword exerting both desirable and discernible effects owing to the referred microenvironment. Therapeutic interventions to promote angiogenesis in regenerative medicine is essential to achieve functional syncytium of tissue constructs while, angiogenic inhibition is a key therapeutic target to suppress tumor growth. In the recent years, clustered regularly interspaced short palindromic repeats associated 9 (CRISPR-Cas9) based gene editing approaches have been gaining considerable attention in the field of biomedical research owing to its ease in tailoring targeted genome in living organisms. The Zebrafish model, with adequately high-throughput fitness, is a likely option for genome editing and angiogenesis research. In this review, we focus on the implication of Zebrafish as a model to study IA and furthermore enumerate CRISPR/Cas9 based genome editing in Zebrafish as a candidate for modeling different types of angiogenesis and support its candidature as a model organism.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Modelos Biológicos , Neovascularización Fisiológica , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente
14.
Compr Physiol ; 9(1): 75-125, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30549015

RESUMEN

Cardiovascular disease leading to heart failure (HF) remains a leading cause of morbidity and mortality worldwide. Improved pharmacological and interventional coronary procedures have led to improved outcomes following acute myocardial infarction. This success has translated into an unforeseen increased incidence in HF. This review summarizes the signaling pathways implicated in the transition to HF following cardiac injury. In addition, we provide an update on cell death signaling and discuss recent advances in cardiac fibrosis as an independent event leading to HF. Finally, we discuss cell-based therapies and their possible use to avert the deteriorating nature of HF. © 2019 American Physiological Society. Compr Physiol 9:75-125, 2019.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Medicina Regenerativa/métodos , Transducción de Señal , Ingeniería de Tejidos/métodos , Animales , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/terapia , Humanos , Miocardio/metabolismo , Miocardio/patología
15.
Sci Rep ; 8(1): 15069, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305684

RESUMEN

Abnormal conduction and improper electrical impulse propagation are common in heart after myocardial infarction (MI). The scar tissue is non-conductive therefore the electrical communication between adjacent cardiomyocytes is disrupted. In the current study, we synthesized and characterized a conductive biodegradable scaffold by incorporating graphene oxide gold nanosheets (GO-Au) into a clinically approved natural polymer chitosan (CS). Inclusion of GO-Au nanosheets in CS scaffold displayed two fold increase in electrical conductivity. The scaffold exhibited excellent porous architecture with desired swelling and controlled degradation properties. It also supported cell attachment and growth with no signs of discrete cytotoxicity. In a rat model of MI, in vivo as well as in isolated heart, the scaffold after 5 weeks of implantation showed a significant improvement in QRS interval which was associated with enhanced conduction velocity and contractility in the infarct zone by increasing connexin 43 levels. These results corroborate that implantation of novel conductive polymeric scaffold in the infarcted heart improved the cardiac contractility and restored ventricular function. Therefore, our approach may be useful in planning future strategies to construct clinically relevant conductive polymer patches for cardiac patients with conduction defects.


Asunto(s)
Medicamentos Herbarios Chinos/química , Oro/química , Grafito/química , Contracción Miocárdica , Infarto del Miocardio , Nanoestructuras/química , Andamios del Tejido/química , Animales , Masculino , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Ratas , Ratas Wistar
16.
Biomed Pharmacother ; 107: 908-917, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30257403

RESUMEN

Chitosan (CS), glycerophosphate (GP) based injectable hydrogels are explored for its implications in bone defect healing and regeneration. Both acellular and cell laden CS based hydrogels are widely investigated and improved through the inclusion of various nanoparticles, polymers and bioactive molecules. In order to improve its applicability for bone tissue repair, we developed an injectable, thermosensitive CS hydrogel containing graphene oxide (GO) and investigated its properties. The hydrogels were investigated for its porous architecture using scanning electron microscopy (SEM), swelling property, protein adsorption ability, degradation rate and exogenous biomineralization. GO addition improved the physico-chemical properties with notable betterment. The CS/GP/GO hydrogel was biocompatible to mesenchymal stem cells and they were metabolically active upon encapsulation. The hydrogel promoted osteogenic differentiation of mouse mesenchymal stem cells by upregualtion of Runt-related transcription factor 2 (Runx2), Alkaline phosphatase (ALP), Type -1 collagen (COL-1), and osteocalcin (OC) under osteogenic conditions. The hydrogel proves to be an amenable platform for carrying cells and exhibited suitable properties to be a potential candidate for bone tissue regeneration.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Quitosano/química , Grafito/farmacología , Osteogénesis/efectos de los fármacos , Animales , Regeneración Ósea/efectos de los fármacos , Células Cultivadas , Glicerofosfatos/química , Grafito/administración & dosificación , Humanos , Hidrogeles , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Microscopía Electrónica de Rastreo , Porosidad , Ratas
17.
Biomed Pharmacother ; 105: 440-448, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29879628

RESUMEN

During the last few decades, gold nanoparticles (AuNP's) have gained considerable attention in nanomedicine and expanded its application in clinical diagnosis and as therapeutics. Employing plant extract for synthesising gold nanoparticles proves to be an eco-friendly technology for large scale production. It is highly economical and suitable for biological applications by negating the use of chemicals involved in conventional route. In this study, AuNP's was prepared by a simple one step method of employing aqueous Mangifera indica seed extract as a reducing agent. Scanning electron microscopy and transmission electron microscopy revealed spherical shaped nanoparticles and dynamic light scattering analysis indicated the AuNP's to be approximately 46.8 nm in size. AuNP's efficiently inhibited the growth of E. coli and S. aureus by its inherent ability to generate reactive oxygen species (ROS) and exhibited detrimental effects towards the tested bacterial species. Biocompatibility assessment indicated the non-toxic nature of AuNP's towards mesenchymal stem cells at 25 µg/ml and interestingly, suppressed the growth of human gastric cancer cells under in vitro culture conditions. AuNP's significantly exhibited anti-angiogenic property in chick chorioallantoic membrane model (CAM) by downregulating Ang-1/Tie2 pathway. Overall, the synthesized AuNP's exhibited antibacterial and anti-angiogenic properties with high biocompatibility thereby supporting its candidature for various biomedical applications. It can be employed in suppressing tumor growth, combat inflammatory diseases that necessitate the involvement of angiogenesis suppression, and antibacterial activity is suitable for its clinical translation to negate surgery associated infections.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antibacterianos/uso terapéutico , Antineoplásicos/uso terapéutico , Materiales Biocompatibles/química , Oro/química , Mangifera/química , Nanopartículas del Metal/química , Extractos Vegetales/uso terapéutico , Inhibidores de la Angiogénesis/farmacología , Angiopoyetina 1/metabolismo , Animales , Antibacterianos/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Pollos , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Humanos , Nanopartículas del Metal/ultraestructura , Pruebas de Sensibilidad Microbiana , Neovascularización Fisiológica/efectos de los fármacos , Extractos Vegetales/farmacología , Receptor TIE-2/metabolismo , Semillas/química , Espectrofotometría Ultravioleta , Staphylococcus aureus/efectos de los fármacos
18.
J Biol Inorg Chem ; 23(5): 753-762, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29779062

RESUMEN

In the recent decades, flavonoid metal complexes have been widely investigated for their multifaceted role in enabling osteoblast differentiation and bone formation. Silibinin complexed with copper(II) ion has been synthesized along with two mixed ligand complexes, namely copper(II) silibinin-phenanthroline and neocuproine as co-ligands, and their positive role in promoting neovacularization and osteoblast differentiation was investigated. Silibinin mono complex [Cu(sil)(H2O)2] and [Cu(sil)(phen)] showed similar UV-visible absorption in the region of 315 and 222 nm, whereas Cu(silibinin)(neocuproine) [Cu(sil)(neo)] showed a blueshift in the 320 nm transition. The involvement of carbonyl group present in the C-ring in metal ion chelation was identified by FT-IR analysis. Thermal gravimetric analysis (TGA) depicted that [Cu(sil)(neo)] has higher thermal stability when compared with the control silibinin and Cu-silibinin mono, and phen complexes. Cu-silibinin complexes were found to be non-toxic to human MG-63 cells and mouse mesenchymal stem cells (MSCs). Our investigations identified the positive role of these complexes in promoting osteoblast differentiation by enhancing calcium deposition and alkaline phosphatase (ALP) activity at the cellular level and stimulation of osteoblastic marker genes such as Runx2, ALP, type 1 collagen, and OCN mRNAs expression at the molecular level. These complexes also supported angiogenesis by upregulation of VEGF and Ang 1 expression in mouse MSCs. Hence, our results suggest that the potential of these metal complexes along with mixed ligand complexes promoted osteoblast differentiation, thus warranting its candidature for bone tissue regeneration application.


Asunto(s)
Regeneración Ósea , Complejos de Coordinación/química , Compuestos Organometálicos/química , Fenantrolinas/química , Silibina/química , Animales , Diferenciación Celular , Línea Celular , Humanos , Técnicas In Vitro , Células Madre Mesenquimatosas/citología , Ratones , Osteoblastos/citología , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier , Termogravimetría
19.
Colloids Surf B Biointerfaces ; 167: 134-143, 2018 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-29635136

RESUMEN

Zinc silibinin complex [Zn(sil)(H2O)2] and mixed ligand zinc complexes such as Zn(silibinin)(phenanthroline) [Zn(sil)(phen)], and Zn(silibinin)(neocuproine) [Zn(sil)(neo)] have been synthesized and characterized. The UV-vis spectra of the Zn(II) complexes showed a considerable shift in the intra-ligand transition. From the IR spectra, it is clear that carbonyl group in the C-ring is involved in the metal chelation besides A/C-ring hydroxyl group. Thermal gravimetric analysis showed that [Zn(sil)(neo)] has higher thermal stability compared to the other two Zn(II) complexes. The potential biological activities of the synthesized complexes were studied systematically. In osteoblast differentiation, silibinin and Zn-silibinin complexes enhanced osteoblast differentiation at the cellular level by increasing calcium deposition and ALP activity, and at molecular level increased osteoblast markers include Runx2, type 1 col, ALP and OC mRNAs expression. Additionally, Zn-silibinin complexes showed promising effects on osteoblast differentiation by regulating miR-590/Smad7 signaling pathway. Among the complexes, Zn(sil)(phen) showed more stimulatory effect on osteoblastic differentiation. These complexes also exhibited angiogenic property by increasing VEGF and Ang 1 expression in mouse MSCs and antibacterial activity against E. coli (Gram-negative) and S. aureus (Gram-positive) strains. Thus, the present study demonstrated that the Zn-silibinin complexes exhibit great potential as a pharmacological agent for bone tissue engineering.


Asunto(s)
Inductores de la Angiogénesis/química , Antibacterianos/química , Huesos/metabolismo , Silimarina/química , Ingeniería de Tejidos/métodos , Zinc/química , Inductores de la Angiogénesis/síntesis química , Inductores de la Angiogénesis/farmacología , Antibacterianos/síntesis química , Antibacterianos/farmacología , Diferenciación Celular/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Silibina , Silimarina/farmacología , Staphylococcus aureus/efectos de los fármacos , Zinc/farmacología
20.
PLoS One ; 12(7): e0181535, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28727797

RESUMEN

Resveratrol (RSV), a polyphenolic compound and naturally occurring phytoalexin, has been reported to exert cardio-protective effects in several animal studies. However, the outcome of initial clinical trials with RSV was less effective compared to pre-clinical studies. Therefore, RSV treatment protocols need to be optimized. In this study we evaluated prophylactic versus therapeutic effect of resveratrol (RSV) in mitigating doxorubicin (Dox)-induced cardiac toxicity in rats. To investigate prophylactic effects, RSV was supplemented for 2 weeks along with Dox administration. After 2 weeks, Dox treatment was stopped and RSV was continued for another 4 weeks. To study therapeutic effects, RSV treatment was initiated after 2 weeks of Dox administration and continued for 4 weeks. Both prophylactic and therapeutic use of RSV mitigated Dox induced deterioration of cardiac function as assessed by echocardiography. Also RSV treatment (prophylactic and therapeutic) prevented Dox induced myocardial damage as measured by cardiac enzymes (LDH and CK-MB) in serum. Which was associated with decrease in Dox induced myocardial apoptosis and fibrosis. Interestingly our study also reveals that prophylactic use of RSV was more effective than its therapeutic use in mitigating Dox induced apoptosis and fibrosis in the myocardium. Therefore, prophylactic use of resveratrol may be projected as a possible future adjuvant therapy to minimize cardiotoxic side effects of doxorubicin in cancer patients.


Asunto(s)
Doxorrubicina/toxicidad , Corazón/efectos de los fármacos , Sustancias Protectoras/administración & dosificación , Estilbenos/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/sangre , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/prevención & control , Forma MB de la Creatina-Quinasa/sangre , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Ecocardiografía , Corazón/diagnóstico por imagen , Inmunohistoquímica , L-Lactato Deshidrogenasa/sangre , Masculino , Miocardio/metabolismo , Miocardio/patología , Factores de Transcripción NFATC/metabolismo , Distribución Aleatoria , Ratas Wistar , Resveratrol
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA