Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cardiovasc Drugs Ther ; 38(2): 223-236, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37421484

RESUMEN

Ischaemic heart disease is a global healthcare challenge with high morbidity and mortality. Early revascularisation in acute myocardial infarction has improved survival; however, limited regenerative capacity and microvascular dysfunction often lead to impaired function and the development of heart failure. New mechanistic insights are required to identify robust targets for the development of novel strategies to promote regeneration. Single-cell RNA sequencing (scRNA-seq) has enabled profiling and analysis of the transcriptomes of individual cells at high resolution. Applications of scRNA-seq have generated single-cell atlases for multiple species, revealed distinct cellular compositions for different regions of the heart, and defined multiple mechanisms involved in myocardial injury-induced regeneration. In this review, we summarise findings from studies of healthy and injured hearts in multiple species and spanning different developmental stages. Based on this transformative technology, we propose a multi-species, multi-omics, meta-analysis framework to drive the discovery of new targets to promote cardiovascular regeneration.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Isquemia Miocárdica , Humanos , Corazón , Infarto del Miocardio/genética , Regeneración
2.
Development ; 145(7)2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29592950

RESUMEN

The epicardium plays a key role during cardiac development, homeostasis and repair, and has thus emerged as a potential target in the treatment of cardiovascular disease. However, therapeutically manipulating the epicardium and epicardium-derived cells (EPDCs) requires insights into their developmental origin and the mechanisms driving their activation, recruitment and contribution to both the embryonic and adult injured heart. In recent years, studies of various model systems have provided us with a deeper understanding of the microenvironment in which EPDCs reside and emerge into, of the crosstalk between the multitude of cardiovascular cell types that influence the epicardium, and of the genetic programmes that orchestrate epicardial cell behaviour. Here, we review these discoveries and discuss how technological advances could further enhance our knowledge of epicardium-based repair mechanisms and ultimately influence potential therapeutic outcomes in cardiovascular regenerative medicine.


Asunto(s)
Corazón/embriología , Organogénesis/fisiología , Pericardio/citología , Regeneración/fisiología , Animales , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Corazón/fisiología , Humanos , Pericardio/metabolismo
3.
Cardiovasc Drugs Ther ; 34(6): 871-880, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32822006

RESUMEN

Despite various clinical modalities available for patients, heart disease remains among the leading causes of mortality and morbidity worldwide. Genetic medicine, particularly mRNA, has broad potential as a therapeutic. More specifically, mRNA-based protein delivery has been used in the fields of cancer and vaccination, but recent changes to the structural composition of mRNA have led the scientific community to swiftly embrace it as a new drug to deliver missing genes to injured myocardium and many other organs. Modified mRNA (modRNA)-based gene delivery features transient but potent protein translation and low immunogenicity, with minimal risk of insertional mutagenesis. In this review, we compared and listed the advantages of modRNA over traditional vectors for cardiac therapy, with particular focus on using modRNA therapy in cardiac repair. We present a comprehensive overview of modRNA's role in cardiomyocyte (CM) proliferation, cardiac vascularization, and prevention of cardiac apoptosis. We also emphasize recent advances in modRNA delivery strategies and discuss the challenges for its clinical translation.


Asunto(s)
Terapia Genética , Cardiopatías/terapia , ARN Mensajero/uso terapéutico , Animales , Proliferación Celular , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Cardiopatías/genética , Cardiopatías/metabolismo , Cardiopatías/fisiopatología , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Mensajero/efectos adversos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recuperación de la Función , Regeneración , Factores de Riesgo , Resultado del Tratamiento
4.
Int J Mol Sci ; 21(21)2020 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-33114756

RESUMEN

Direct cardiac reprogramming has emerged as a novel therapeutic approach to treat and regenerate injured hearts through the direct conversion of fibroblasts into cardiac cells. Most studies have focused on the reprogramming of fibroblasts into induced cardiomyocytes (iCMs). The first study in which this technology was described, showed that at least a combination of three transcription factors, GATA4, MEF2C and TBX5 (GMT cocktail), was required for the reprogramming into iCMs in vitro using mouse cells. However, this was later demonstrated to be insufficient for the reprogramming of human cells and additional factors were required. Thereafter, most studies have focused on implementing reprogramming efficiency and obtaining fully reprogrammed and functional iCMs, by the incorporation of other transcription factors, microRNAs or small molecules to the original GMT cocktail. In this respect, great advances have been made in recent years. However, there is still no consensus on which of these GMT-based varieties is best, and robust and highly reproducible protocols are still urgently required, especially in the case of human cells. On the other hand, apart from CMs, other cells such as endothelial and smooth muscle cells to form new blood vessels will be fundamental for the correct reconstruction of damaged cardiac tissue. With this aim, several studies have centered on the direct reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs) able to give rise to all myocardial cell lineages. Especially interesting are reports in which multipotent and highly expandable mouse iCPCs have been obtained, suggesting that clinically relevant amounts of these cells could be created. However, as of yet, this has not been achieved with human iCPCs, and exactly what stage of maturity is appropriate for a cell therapy product remains an open question. Nonetheless, the major concern in regenerative medicine is the poor retention, survival, and engraftment of transplanted cells in the cardiac tissue. To circumvent this issue, several cell pre-conditioning approaches are currently being explored. As an alternative to cell injection, in vivo reprogramming may face fewer barriers for its translation to the clinic. This approach has achieved better results in terms of efficiency and iCMs maturity in mouse models, indicating that the heart environment can favor this process. In this context, in recent years some studies have focused on the development of safer delivery systems such as Sendai virus, Adenovirus, chemical cocktails or nanoparticles. This article provides an in-depth review of the in vitro and in vivo cardiac reprograming technology used in mouse and human cells to obtain iCMs and iCPCs, and discusses what challenges still lie ahead and what hurdles are to be overcome before results from this field can be transferred to the clinical settings.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Factores de Transcripción/metabolismo , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Reprogramación Celular , Medios de Cultivo/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo
5.
Basic Res Cardiol ; 112(4): 41, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28540527

RESUMEN

Ischemic heart disease is still the leading cause of death even with the advancement of pharmaceutical therapies and surgical procedures. Early vascularization in the ischemic heart is critical for a better outcome. Although stem cell therapy has great potential for cardiovascular regeneration, the ideal cell type and delivery method of cells have not been resolved. We tested a new approach of stem cell therapy by delivery of induced vascular progenitor cells (iVPCs) grown on polymer micro-bundle scaffolds in a rat model of myocardial infarction. iVPCs partially reprogrammed from vascular endothelial cells (ECs) had potent angiogenic potential and were able to simultaneously differentiate into vascular smooth muscle cells (SMCs) and ECs in 2D culture. Under hypoxic conditions, iVPCs also secreted angiogenic cytokines such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) as measured by enzyme-linked immunosorbent assay (ELISA). A longitudinal micro-scaffold made from poly(lactic-co-glycolic acid) was sufficient for the growth and delivery of iVPCs. Co-cultured ECs and SMCs aligned well on the micro-bundle scaffold similarly as in the vessels. 3D cell/polymer micro-bundles formed by iVPCs and micro-scaffolds were transplanted into the ischemic myocardium in a rat model of myocardial infarction (MI) with ligation of the left anterior descending artery. Our in vivo data showed that iVPCs on the micro-bundle scaffold had higher survival, and better retention and engraftment in the myocardium than free iVPCs. iVPCs on the micro-bundles promoted better cardiomyocyte survival than free iVPCs. Moreover, iVPCs and iVPC/polymer micro-bundles treatment improved cardiac function (ejection fraction and fractional shortening, endocardial systolic volume) measured by echocardiography, increased vessel density, and decreased infarction size [endocardial and epicardial infarct (scar) length] better than untreated controls at 8 weeks after MI. We conclude that iVPCs grown on a polymer micro-bundle scaffold are new promising approach for cell-based therapy designed for cardiovascular regeneration in ischemic heart disease.


Asunto(s)
Células Progenitoras Endoteliales/trasplante , Ácido Láctico/química , Músculo Liso Vascular/trasplante , Infarto del Miocardio/cirugía , Miocardio/patología , Miocitos del Músculo Liso/trasplante , Neovascularización Fisiológica , Ácido Poliglicólico/química , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Músculo Liso Vascular/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocitos del Músculo Liso/metabolismo , Comunicación Paracrina , Fenotipo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas Sprague-Dawley , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Remodelación Ventricular
6.
Drug Discov Today ; 29(9): 104135, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39103145

RESUMEN

Oxygen-generating biomaterials are emerging as a groundbreaking solution for transforming cardiovascular engineering. These biomaterials generate and release oxygen within various biomedical applications, marking a new frontier in healthcare. Most cardiovascular treatments face a significant challenge, ensuring a consistent oxygen supply to nurture engineered tissues or even implanted devices. Traditional methods relying on passive oxygen diffusion often fall short, hindering functional cardiovascular tissue development. Oxygen-generating biomaterials, incorporating agents like calcium peroxide, provide a controlled oxygen source to the surrounding cells. This innovation potentially enhances cell viability, stimulates growth and boosts metabolic activity crucial for tissue health. Applications include repairing cardiac and vascular tissues, disease modeling, drug testing and personalized medicine, promising tailored treatments. Challenges like material toxicity and oxygen release control need consideration. As research progresses, the use of these innovative biomaterials in clinical translation could reshape cardiovascular healthcare, revolutionizing patient outcomes in heart disease treatment.


Asunto(s)
Materiales Biocompatibles , Oxígeno , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Animales , Oxígeno/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/terapia , Sistema Cardiovascular/efectos de los fármacos
7.
Methodist Debakey Cardiovasc J ; 20(4): 31-41, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39184159

RESUMEN

Myocardial recovery is characterized by a return toward normal structure and function of the heart after an injury. Mechanisms of myocardial recovery include restoration and/or adaptation of myocyte structure and function, mitochondrial activity and number, metabolic homeostasis, electrophysiological stability, extracellular matrix remodeling, and myocardial perfusion. Myocardial regeneration is an element of myocardial recovery that involves the generation of new myocardial tissue, a process which is limited in adult humans but may be therapeutically augmented. Understanding the mechanisms of myocardial recovery and myocardial regeneration will lead to novel therapies for heart failure.


Asunto(s)
Miocardio , Recuperación de la Función , Regeneración , Humanos , Miocardio/patología , Miocardio/metabolismo , Animales , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología
8.
Aging Cell ; 22(12): e14020, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37957823

RESUMEN

Cardiovascular aging presents a formidable challenge, as the aging process can lead to reduced cardiac function and heightened susceptibility to cardiovascular diseases. Consequently, there is an escalating, unmet medical need for innovative and effective cardiovascular regeneration strategies aimed at restoring and rejuvenating aging cardiovascular tissues. Altered redox homeostasis and the accumulation of oxidative damage play a pivotal role in detrimental changes to stem cell function and cellular senescence, hampering regenerative capacity in aged cardiovascular system. A mounting body of evidence underscores the significance of targeting redox machinery to restore stem cell self-renewal and enhance their differentiation potential into youthful cardiovascular lineages. Hence, the redox machinery holds promise as a target for optimizing cardiovascular regenerative therapies. In this context, we delve into the current understanding of redox homeostasis in regulating stem cell function and reprogramming processes that impact the regenerative potential of the cardiovascular system. Furthermore, we offer insights into the recent translational and clinical implications of redox-targeting compounds aimed at enhancing current regenerative therapies for aging cardiovascular tissues.


Asunto(s)
Sistema Cardiovascular , Senescencia Celular , Diferenciación Celular , Oxidación-Reducción
9.
Curr Stem Cell Res Ther ; 18(4): 440-444, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35927909

RESUMEN

Smoking habits represent a cardiovascular risk factor with a tremendous impact on health. Other than damaging differentiated and functional cells of the cardiovascular system, they also negatively affect reparative mechanisms, such as those involved in cardiac fibrosis and in endothelial progenitor cell (EPC) activation. In recent years, alternative smoking devices, dubbed modified tobacco risk products (MRPs), have been introduced, but their precise impact on human health is still under evaluation. Also, they have not been characterized yet about the possible negative effects on cardiovascular reparative and regenerative cells, such as EPCs or pluripotent stem cells. In this perspective, we critically review the still scarce available data on the effects of MRPs on molecular and cellular mechanisms of cardiovascular repair and regeneration.


Asunto(s)
Células Progenitoras Endoteliales , Productos de Tabaco , Humanos , Nicotiana , Humo , Fumar
10.
Curr Stem Cell Res Ther ; 18(2): 202-215, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35392790

RESUMEN

BACKGROUND: Cardiovascular disease (CVD) is one of the world's leading causes of increased morbidity and mortality. Current interventions for CVD, including percutaneous transluminal coronary angioplasty (PTCA) and coronary artery bypass grafting (CABG), carry certain risks and complications, which may also affect the patient's quality of life. It is important to minimize those risks and complications while speeding up the recovery. Insulin Growth Factor-1 (IGF-1) is a growth factor responsible for cellular migration, proliferation, differentiation, and angiogenesis, which supports cardiovascular regeneration. METHODS: In light of the current trend of regenerative medicine, the present review aims to pool data relating to the incorporation of IGF-1 in regenerative medicine and provide input on the current research gaps and concerns arising on translating this approach from benchwork into clinical settings. RESULTS: Using the keywords IGF-1 'OR' Insulin Growth Factor 1 'AND' Mesenchymal Stem Cells 'AND' Tissue Healing from 2009 to 2020, we identified 160 and 52 from Medline and PubMed, screening out 202 articles due to non-fulfilment of the inclusion criteria. CONCLUSION: Incorporating IGF-1 into regenerative and personalized medicine may be promising for treating CVD; however, the concerns include the role of IGF-1 in inducing cancer growth and its ability to migrate to the specific site of injury, especially for those who present with multiple pathologies should be addressed prior to its translation from bench work into clinical settings.


Asunto(s)
Enfermedades Cardiovasculares , Factor I del Crecimiento Similar a la Insulina , Humanos , Enfermedades Cardiovasculares/terapia , Insulina , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Medicina de Precisión , Calidad de Vida
11.
Front Cardiovasc Med ; 9: 802551, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35224044

RESUMEN

Cardiovascular diseases (CVDs) have a massive impact on human health. Due to the limited regeneration capacity of adult heart tissue, CVDs are the leading cause of death and disability worldwide. Even though there are surgical and pharmacological treatments for CVDs, regenerative strategies are the most promising approaches and have the potential to benefit millions of people. As in any other tissue engineering approach, the repair and regeneration of damaged cardiac tissues generally involve scaffolds made up of biodegradable and biocompatible materials, cellular components such as stem cells, and growth factors. This review provides an overview of biomaterial-based tissue engineering approaches for CVDs with a specific focus on the potential of 2D materials. It is essential to consider both physicochemical and immunomodulatory properties for evaluating the applicability of 2D materials in cardiac tissue repair and regeneration. As new members of the 2D materials will be explored, they will quickly become part of cardiac tissue engineering technologies.

12.
Methods Mol Biol ; 2191: 151-169, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32865744

RESUMEN

The delivery of cells into damaged myocardium induces limited cardiac regeneration due to extensive cell death. In an effort to limit cell death, our lab formulates three-dimensional matrices as a delivery system for cell therapy. Our primary work has been focused on the formation of engineered cardiac tissues (ECTs) from human-induced pluripotent stem cell-derived engineered cardiac cells. However, ECT immaturity hinders ability to fully recover damaged myocardium. Various conditioning regimens such as mechanical stretch and/or electric pacing have been used to activate maturation pathways. To improve ECT maturity, we use non-contacting chronic light stimulation using heterologously expressed light-sensitive channelrhodopsin ion channels. We transduce ECTs with an AAV packaged channelrhodopsin and chronically optically pace (C-OP) ECTs for 1 week above the intrinsic beat rate, resulting in increased ECT electrophysiological properties.


Asunto(s)
Channelrhodopsins/genética , Células Madre Pluripotentes Inducidas/citología , Optogenética/métodos , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular/genética , Fenómenos Electrofisiológicos/genética , Humanos , Células Madre Pluripotentes Inducidas/patología , Ratones , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Regeneración/genética
13.
Cells ; 10(1)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33477787

RESUMEN

mRNA has emerged as an important biomolecule in the global call for the development of therapies during the COVID-19 pandemic. Synthetic in vitro-transcribed (IVT) mRNA can be engineered to mimic naturally occurring mRNA and can be used as a tool to target "undruggable" diseases. Recent advancement in the field of RNA therapeutics have addressed the challenges inherent to this drug molecule and this approach is now being applied to several therapeutic modalities, from cancer immunotherapy to vaccine development. In this review, we discussed the use of mRNA for stem cell generation or enhancement for the purpose of cardiovascular regeneration.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , ARN Mensajero/uso terapéutico , Regeneración , Humanos , Células Madre
14.
Methods Mol Biol ; 2002: 51-59, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30159827

RESUMEN

Our laboratory has recently developed a novel three-dimensional in vitro model of the human heart, which we call the vascularized cardiac spheroid (VCS). These better recapitulate the human heart's cellular and extracellular microenvironment compared to the existing in vitro models. To achieve this, human-induced pluripotent stem cell (iPSC)-derived cardiomyocytes, cardiac fibroblasts, and human coronary artery endothelial cells are co-cultured in hanging drop culture in ratios similar to those found in the human heart in vivo. The resulting three-dimensional cellular organization, extracellular matrix, and microvascular network formation throughout the VCS has been shown to mimic the one present in the human heart tissue. Therefore, VCSs offer a promising platform to study cardiac physiology, disease, and pharmacology, as well as bioengineering constructs to regenerate heart tissue.


Asunto(s)
Matriz Extracelular/fisiología , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Miocitos Cardíacos/citología , Esferoides Celulares/citología , Bioingeniería , Técnicas de Cocultivo , Humanos , Imagenología Tridimensional , Técnicas In Vitro
15.
Adv Biochem Eng Biotechnol ; 163: 71-115, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29071403

RESUMEN

Many disorders are manifested by dysfunction of key cell types or their disturbed integration in complex organs. Thereby, adult organ systems often bear restricted self-renewal potential and are incapable of achieving functional regeneration. This underlies the need for novel strategies in the field of cell (re-)programming-based regenerative medicine as well as for drug development in vitro. The regenerative field has been hampered by restricted availability of adult stem cells and the potentially hazardous features of pluripotent embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Moreover, ethical concerns and legal restrictions regarding the generation and use of ESCs still exist. The establishment of direct reprogramming protocols for various therapeutically valuable somatic cell types has overcome some of these limitations. Meanwhile, new perspectives for safe and efficient generation of different specified somatic cell types have emerged from numerous approaches relying on exogenous expression of lineage-specific transcription factors, coding and noncoding RNAs, and chemical compounds.It should be of highest priority to develop protocols for the production of mature and physiologically functional cells with properties ideally matching those of their endogenous counterparts. Their availability can bring together basic research, drug screening, safety testing, and ultimately clinical trials. Here, we highlight the remarkable successes in cellular (re-)programming, which have greatly advanced the field of regenerative medicine in recent years. In particular, we review recent progress on the generation of cardiomyocyte subtypes, with a focus on cardiac pacemaker cells. Graphical Abstract.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Reprogramación Celular , Células Madre Embrionarias Humanas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Células Madre Embrionarias Humanas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología
16.
Front Cardiovasc Med ; 5: 52, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29942806

RESUMEN

Stem cell therapy has the potential to regenerate cardiac function after myocardial infarction. In this study, we sought to examine if fibrin microthread technology could be leveraged to develop a contractile fiber from human pluripotent stem cell derived cardiomyocytes (hPS-CM). hPS-CM seeded onto fibrin microthreads were able to adhere to the microthread and began to contract seven days after initial seeding. A digital speckle tracking algorithm was applied to high speed video data (>60 fps) to determine contraction behaviour including beat frequency, average and maximum contractile strain, and the principal angle of contraction of hPS-CM contracting on the microthreads over 21 days. At day 7, cells seeded on tissue culture plastic beat at 0.83 ± 0.25 beats/sec with an average contractile strain of 4.23±0.23%, which was significantly different from a beat frequency of 1.11 ± 0.45 beats/sec and an average contractile strain of 3.08±0.19% at day 21 (n = 18, p < 0.05). hPS-CM seeded on microthreads beat at 0.84 ± 0.15 beats/sec with an average contractile strain of 3.56±0.22%, which significantly increased to 1.03 ± 0.19 beats/sec and 4.47±0.29%, respectively, at 21 days (n = 18, p < 0.05). At day 7, 27% of the cells had a principle angle of contraction within 20 degrees of the microthread, whereas at day 21, 65% of hPS-CM were contracting within 20 degrees of the microthread (n = 17). Utilizing high speed calcium transient data (>300 fps) of Fluo-4AM loaded hPS-CM seeded microthreads, conduction velocities significantly increased from 3.69 ± 1.76 cm/s at day 7 to 24.26 ± 8.42 cm/s at day 21 (n = 5-6, p < 0.05). hPS-CM seeded microthreads exhibited positive expression for connexin 43, a gap junction protein, between cells. These data suggest that the fibrin microthread is a suitable scaffold for hPS-CM attachment and contraction. In addition, extended culture allows cells to contract in the direction of the thread, suggesting alignment of the cells in the microthread direction.

17.
Tissue Eng Regen Med ; 14(3): 253-265, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30603482

RESUMEN

In recent years, several kinds of cardiac progenitor cells have been identified and isolated from heart tissue. These cells showed differentiation potential into cardiomyocytes, smooth muscle cells, and endothelial cells in vitro and in vivo. Morphogenetic events are tightly regulated during development to determine cell destiny and reshape the embryonic lineage. In this study, we directly compared the characteristics of rat fetal cardiac progenitor cells (rFCPCs) isolated from the chamber formation stage at embryonic day 12 (E12) and at the septation stage of E15. Both kinds of rFCPCs expressed mesenchymal stem cell markers (CD105, CD73, and CD29) but not CD34 and CD45. The E12 rFCPCs expressed a high level of Oct4 compared to E15 until passage 5 and showed a steep decline of Nkx2.5 expression at passage 5. However, Nkx2.5 expression at E15 was maintained until passage 5 and Oct4 expression slightly increased at passage 5. We also detected an intense staining for Oct4 antibody in E12 heart tissue sections. The average doubling time of the E12 rFCPCs from passage 3 to passage 15 was about 5 hours longer than E15. These cells could also be induced into cardiomyocytes expressing α-MHC, cTnT, cTnC, and Cx43 under cardiomyogenic culture conditions and rFCPCs at E15 showed more intense staining of α-MHC than cells at E12 by immunocytochemistry. Taken together, our results show that developmental differences between E12 and E15 may influence their properties and differentiation. Furthermore those differences should be considered when deciding on the optimal cell source for cell replacement therapy in cardiovascular regeneration.

18.
Stem Cells Transl Med ; 6(10): 1859-1867, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28836732

RESUMEN

Experimental and early clinical data suggest that, due to several unique properties, mesenchymal stem cells (MSCs) may be more effective than other cell types for diseases that are difficult to treat or untreatable. Owing to their ease of isolation and culture as well as their secretory and immunomodulatory abilities, MSCs are the most promising option in the field of cell-based therapies. Although MSCs from various sources share several common characteristics, they also exhibit several important differences. These variations may reflect, in part, specific regional properties of the niches from which the cells originate. Moreover, morphological and functional features of MSCs are susceptible to variations across isolation protocols and cell culture conditions. These observations suggest that careful preparation of manufacturing protocols will be necessary for the most efficient use of MSCs in future clinical trials. A typical human myocardial infarct involves the loss of approximately 1 billion cardiomyocytes and 2-3 billion other (mostly endothelial) myocardial cells, leading (despite maximized medical therapy) to a significant negative impact on the length and quality of life. Despite more than a decade of intensive research, search for the "best" (safe and maximally effective) cell type to drive myocardial regeneration continues. In this review, we summarize information about the most important features of MSCs and recent discoveries in the field of MSCs research, and describe current data from preclinical and early clinical studies on the use of MSCs in cardiovascular regeneration. Stem Cells Translational Medicine 2017;6:1859-1867.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Medicina Regenerativa/métodos , Investigación Biomédica Traslacional/métodos , Animales , Humanos , Células Madre Mesenquimatosas/metabolismo
19.
Front Cardiovasc Med ; 4: 62, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29057227

RESUMEN

Global aging is a hallmark of our century. The natural multifactorial process resulting in aging involves structural and functional changes, affecting molecules, cells, and tissues. As the western population is getting older, we are witnessing an increase in the burden of cardiovascular events, some of which are known to be directly linked to cellular senescence and dysfunction. In this review, we will focus on the description of a few circulating molecules, which have been correlated to life span, aging, and cardiovascular homeostasis. We will review the current literature concerning the circulating levels and related signaling pathways of selected proteins (insulin-like growth factor 1, growth and differentiation factor-11, and PAI-1) and microRNAs of interest (miR-34a, miR-146a, miR-21), whose bloodstream levels have been associated to aging in different organisms. In particular, we will also discuss their potential role in the biology and senescence of cardiovascular regenerative cell types, such as endothelial progenitor cells, mesenchymal stromal cells, and cardiac progenitor cells.

20.
Biomaterials ; 135: 74-84, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28494265

RESUMEN

BACKGROUND: Magnetic activated cell sorting (MACS®) is routinely used to isolate stem cell subpopulations intended for the treatment of cardiovascular diseases. In strong contrast, studies examining the amount, effect and intramyocardial distribution of iron nanoparticles used for magnetic cell labelling are missing, although iron excess can cause functional disorders in the heart. METHODS AND RESULTS: CD133+ haematopoietic and CD271+ mesenchymal stem cells were purified from bone marrow using automatically and manually MACS® based systems. Flow cytometric measurements demonstrated a rapid loss of MACS® MicroBeads from cells under culture conditions, while storage under hypothermic conditions decelerated their detachment. Moreover, an average loading of ∼11 fg iron/cell caused by magnetic labelling was determined in magnetic particle spectroscopy. Importantly, hemodynamic measurements as well as histological examinations using a myocardial ischemia/reperfusion mouse model showed no influence of MACS® MicroBeads on cardiac regeneration, while the transplantation of stem cells caused a significant improvement. Furthermore, immunostainings demonstrated the clearance of co-injected iron nanoparticles from stem cells and the surrounding heart tissue within 48 h post transplantation. CONCLUSIONS: Our results indicate that iron amounts typically co-injected with MACS® purified stem cells do not harm cardiac functions and are cleared from heart tissue within a few hours. Therefore, we conclude that MACS® MicroBeads exhibit a good compatibility in the cardiac environment.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Hierro/metabolismo , Miocardio/metabolismo , Nanopartículas/metabolismo , Antígeno AC133/metabolismo , Adapaleno/metabolismo , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Humanos , Hierro/química , Leucocitos Mononucleares/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones SCID , Miocardio/citología , Nanopartículas/química
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda