Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
1.
Cells ; 10(11)2021 11 10.
Article in English | MEDLINE | ID: mdl-34831333

ABSTRACT

Personalized regenerative medicine and biomedical research have been galvanized and revolutionized by human pluripotent stem cells in combination with recent advances in genomics, artificial intelligence, and genome engineering. More recently, we have witnessed the unprecedented breakthrough life-saving translation of mRNA-based vaccines for COVID-19 to contain the global pandemic and the investment in billions of US dollars in space exploration projects and the blooming space-tourism industry fueled by the latest reusable space vessels. Now, it is time to examine where the translation of pluripotent stem cell research stands currently, which has been touted for more than the last two decades to cure and treat millions of patients with severe debilitating degenerative diseases and tissue injuries. This review attempts to highlight the accomplishments of pluripotent stem cell research together with cutting-edge genomics and genome editing tools and, also, the promises that have still not been transformed into clinical applications, with cardiovascular research as a case example. This review also brings to our attention the scientific and socioeconomic challenges that need to be effectively addressed to see the full potential of pluripotent stem cells at the clinical bedside.


Subject(s)
Cardiovascular Diseases/therapy , Genomics , Pluripotent Stem Cells/transplantation , Artificial Intelligence , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cardiovascular System/cytology , Cardiovascular System/growth & development , Cell Differentiation , Drug Discovery , Gene Editing , Humans , Models, Biological , Pluripotent Stem Cells/cytology , Precision Medicine , Regenerative Medicine , Safety , Translational Research, Biomedical
2.
Int J Mol Sci ; 22(17)2021 Aug 29.
Article in English | MEDLINE | ID: mdl-34502291

ABSTRACT

Early-stage mammalian embryos survive within a low oxygen tension environment and develop into fully functional, healthy organisms despite this hypoxic stress. This suggests that hypoxia plays a regulative role in fetal development that influences cell mobilization, differentiation, proliferation, and survival. The long-term hypoxic environment is sustained throughout gestation. Elucidation of the mechanisms by which cardiovascular stem cells survive and thrive under hypoxic conditions would benefit cell-based therapies where stem cell survival is limited in the hypoxic environment of the infarcted heart. The current study addressed the impact of long-term hypoxia on fetal Islet-1+ cardiovascular progenitor cell clones, which were isolated from sheep housed at high altitude. The cells were then cultured in vitro in 1% oxygen and compared with control Islet-1+ cardiovascular progenitor cells maintained at 21% oxygen. RT-PCR, western blotting, flow cytometry, and migration assays evaluated adaptation to long term hypoxia in terms of survival, proliferation, and signaling. Non-canonical Wnt, Notch, AKT, HIF-2α and Yap1 transcripts were induced by hypoxia. The hypoxic niche environment regulates these signaling pathways to sustain the dedifferentiation and survival of fetal cardiovascular progenitor cells.


Subject(s)
Cardiovascular System/embryology , Cell Hypoxia/physiology , Stem Cells/cytology , Animals , Cardiovascular System/cytology , Cell Cycle , Cell Differentiation , Cell Movement , Cell Survival , Female , Hypoxia/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Sheep , Stem Cells/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Development ; 148(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34080610

ABSTRACT

The lymphatic vasculature is an integral component of the cardiovascular system. It is essential to maintain tissue fluid homeostasis, direct immune cell trafficking and absorb dietary lipids from the digestive tract. Major advances in our understanding of the genetic and cellular events important for constructing the lymphatic vasculature during development have recently been made. These include the identification of novel sources of lymphatic endothelial progenitor cells, the recognition of lymphatic endothelial cell specialisation and heterogeneity, and discovery of novel genes and signalling pathways underpinning developmental lymphangiogenesis. Here, we review these advances and discuss how they inform our understanding of lymphatic network formation, function and dysfunction.


Subject(s)
Cardiovascular System/growth & development , Lymphangiogenesis/physiology , Lymphatic Vessels/physiology , Animals , Cardiovascular System/cytology , Cardiovascular System/embryology , Endothelial Cells/physiology , Homeostasis , Humans , Lymphatic Vessels/cytology , Lymphatic Vessels/embryology , Signal Transduction
4.
Int J Mol Sci ; 22(4)2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33669808

ABSTRACT

Recent technological advances have revolutionized the study of tissue biology and garnered a greater appreciation for tissue complexity. In order to understand cardiac development, heart tissue homeostasis, and the effects of stress and injury on the cardiovascular system, it is essential to characterize the heart at high cellular resolution. Single-cell profiling provides a more precise definition of tissue composition, cell differentiation trajectories, and intercellular communication, compared to classical bulk approaches. Here, we aim to review how recent single-cell multi-omic studies have changed our understanding of cell dynamics during cardiac development, and in the healthy and diseased adult myocardium.


Subject(s)
Cardiovascular System/cytology , Single-Cell Analysis , Transcriptome/genetics , Animals , COVID-19/genetics , COVID-19/pathology , Cellular Reprogramming/genetics , Embryonic Development/genetics , Humans
5.
Int J Mol Sci ; 22(5)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33652988

ABSTRACT

In this Review, we briefly describe the basic virology and pathogenesis of SARS-CoV-2, highlighting how stem cell technology and organoids can contribute to the understanding of SARS-CoV-2 cell tropisms and the mechanism of disease in the human host, supporting and clarifying findings from clinical studies in infected individuals. We summarize here the results of studies, which used these technologies to investigate SARS-CoV-2 pathogenesis in different organs. Studies with in vitro models of lung epithelia showed that alveolar epithelial type II cells, but not differentiated lung alveolar epithelial type I cells, are key targets of SARS-CoV-2, which triggers cell apoptosis and inflammation, while impairing surfactant production. Experiments with human small intestinal organoids and colonic organoids showed that the gastrointestinal tract is another relevant target for SARS-CoV-2. The virus can infect and replicate in enterocytes and cholangiocytes, inducing cell damage and inflammation. Direct viral damage was also demonstrated in in vitro models of human cardiomyocytes and choroid plexus epithelial cells. At variance, endothelial cells and neurons are poorly susceptible to viral infection, thus supporting the hypothesis that neurological symptoms and vascular damage result from the indirect effects of systemic inflammatory and immunological hyper-responses to SARS-CoV-2 infection.


Subject(s)
COVID-19/pathology , Organoids/virology , SARS-CoV-2/physiology , Stem Cells/virology , Animals , Apoptosis , COVID-19/virology , Cardiovascular System/cytology , Cardiovascular System/pathology , Cardiovascular System/virology , Central Nervous System/cytology , Central Nervous System/pathology , Central Nervous System/virology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/pathology , Gastrointestinal Tract/virology , Humans , Inflammation/pathology , Inflammation/virology , Lung/cytology , Lung/pathology , Lung/virology , Organoids/pathology , Stem Cells/pathology , Viral Tropism , Virus Internalization
6.
FASEB J ; 34(7): 8778-8786, 2020 07.
Article in English | MEDLINE | ID: mdl-32946179

ABSTRACT

Reporting the sex of biological material is critical for transparency and reproducibility in science. This study examined the reporting of the sex of cells used in cardiovascular studies. Articles from 16 cardiovascular journals that publish peer-reviewed studies in cardiovascular physiology and pharmacology in the year 2018 were systematically reviewed using terms "cultured" and "cells." Data were collected on the sex of cells, the species from which the cells were isolated, and the type of cells, and summarized as a systematic review. Sex was reported in 88 (38.6%) of the 228 studies meeting inclusion criteria. Reporting rates varied with Circulation, Cardiovascular Research and American Journal of Physiology: Heart and Circulatory Physiology having the highest rates of sex reporting (>50%). A majority of the studies used cells from male (54.5%) or both male and female animals (32.9%). Humans (31.8%), rats (20.4%), and mice (43.8%) were the most common sources for cells. Cardiac myocytes were the most commonly used cell type (37.0%). Overall reporting of sex of experimental material remains below 50% and is inconsistent among journals. Sex chromosomes in cells have the potential to affect protein expression and molecular signaling pathways and should be consistently reported.


Subject(s)
Biomedical Research , Cardiovascular System/physiopathology , Cardiovascular System/cytology , Cells, Cultured , Female , Humans , Male , Sex Factors
7.
Development ; 147(19)2020 10 12.
Article in English | MEDLINE | ID: mdl-32928907

ABSTRACT

Angiopoietin/TIE signalling plays a major role in blood and lymphatic vessel development. In mouse, Tek (previously known as Tie2) mutants die prenatally due to a severely underdeveloped cardiovascular system. In contrast, in zebrafish, previous studies have reported that although embryos injected with tek morpholinos (MOs) exhibit severe vascular defects, tek mutants display no obvious vascular malformations. To further investigate the function of zebrafish Tek, we generated a panel of loss-of-function tek mutants, including RNA-less alleles, an allele lacking the MO-binding site, an in-frame deletion allele and a premature termination codon-containing allele. Our data show that all these mutants survive to adulthood with no obvious cardiovascular defects. MO injections into tek mutants lacking the MO-binding site or the entire tek locus cause similar vascular defects to those observed in MO-injected +/+ siblings, indicating off-target effects of the MOs. Surprisingly, comprehensive phylogenetic profiling and synteny analyses reveal that Tek was lost in the largest teleost clade, suggesting a lineage-specific shift in the function of TEK during vertebrate evolution. Altogether, these data show that Tek is dispensable for zebrafish development, and probably dispensable in most teleost species.


Subject(s)
Cardiovascular System/metabolism , Zebrafish Proteins/metabolism , Animals , Cardiovascular System/cytology , Gene Editing , Organogenesis/genetics , Organogenesis/physiology , Phylogeny , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Zebrafish , Zebrafish Proteins/genetics
8.
Sci Rep ; 10(1): 12276, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32703995

ABSTRACT

The advantages of automatically recognition of fundamental tissues using computer vision techniques are well known, but one of its main limitations is that sometimes it is not possible to classify correctly an image because the visual information is insufficient or the descriptors extracted are not discriminative enough. An Ontology could solve in part this problem, because it gathers and makes possible to use the specific knowledge that allows detecting clear mistakes in the classification, occasionally simply by pointing out impossible configurations in that domain. One of the main contributions of this work is that we used a Histological Ontology to correct, and therefore improve the classification of histological images. First, we described small regions of images, denoted as blocks, using Local Binary Pattern (LBP) based descriptors and we determined which tissue of the cardiovascular system was present using a cascade Support Vector Machine (SVM). Later, we built Resource Description Framework (RDF) triples for the occurrences of each discriminant class. Based on that, we used a Histological Ontology to correct, among others, "not possible" situations, improving in this way the global accuracy in the blocks first and in tissues classification later. For the experimental validation, we used a set of 6000 blocks of [Formula: see text] pixels, obtaining F-Scores between 0.769 and 0.886. Thus, there is an improvement between 0.003 and [Formula: see text] when compared to the approach without the histological ontology. The methodology improves the automatic classification of histological images using a histological ontology. Another advantage of our proposal is that using the Ontology, we were capable of recognising the epithelial tissue, previously not detected by any of the computer vision methods used, including a CNN proposal called HistoResNet evaluated in the experiments. Finally, we also have created and made publicly available a dataset consisting of 6000 blocks of labelled histological tissues.


Subject(s)
Cardiovascular Physiological Phenomena , Cardiovascular System/cytology , Computational Biology , Gene Ontology , Histocytochemistry , Algorithms , Animals , Cardiovascular System/pathology , Computational Biology/methods , Histocytochemistry/methods , Humans , Image Processing, Computer-Assisted , Support Vector Machine
9.
J Am Coll Cardiol ; 75(17): 2169-2188, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32354384

ABSTRACT

The extracellular matrix (ECM) is the noncellular component of tissues in the cardiovascular system and other organs throughout the body. It is formed of filamentous proteins, proteoglycans, and glycosaminoglycans, which extensively interact and whose structure and dynamics are modified by cross-linking, bridging proteins, and cleavage by matrix degrading enzymes. The ECM serves important structural and regulatory roles in establishing tissue architecture and cellular function. The ECM of the developing heart has unique properties created by its emerging contractile nature; similarly, ECM lining blood vessels is highly elastic in order to sustain the basal and pulsatile forces imposed on their walls throughout life. In this part 1 of a 4-part JACC Focus Seminar, we focus on the role, function, and basic biology of the ECM in both heart development and in the adult.


Subject(s)
Cardiology/education , Cardiovascular System/cytology , Cardiovascular System/growth & development , Extracellular Matrix/physiology , Animals , Cardiovascular System/metabolism , Homeostasis/physiology , Humans , Proteoglycans/metabolism
10.
Cell ; 180(4): 764-779.e20, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32059779

ABSTRACT

The heterogeneity of endothelial cells (ECs) across tissues remains incompletely inventoried. We constructed an atlas of >32,000 single-EC transcriptomes from 11 mouse tissues and identified 78 EC subclusters, including Aqp7+ intestinal capillaries and angiogenic ECs in healthy tissues. ECs from brain/testis, liver/spleen, small intestine/colon, and skeletal muscle/heart pairwise expressed partially overlapping marker genes. Arterial, venous, and lymphatic ECs shared more markers in more tissues than did heterogeneous capillary ECs. ECs from different vascular beds (arteries, capillaries, veins, lymphatics) exhibited transcriptome similarity across tissues, but the tissue (rather than the vessel) type contributed to the EC heterogeneity. Metabolic transcriptome analysis revealed a similar tissue-grouping phenomenon of ECs and heterogeneous metabolic gene signatures in ECs between tissues and between vascular beds within a single tissue in a tissue-type-dependent pattern. The EC atlas taxonomy enabled identification of EC subclusters in public scRNA-seq datasets and provides a powerful discovery tool and resource value.


Subject(s)
Endothelial Cells/metabolism , Single-Cell Analysis , Transcriptome , Animals , Brain/cytology , Cardiovascular System/cytology , Endothelial Cells/classification , Endothelial Cells/cytology , Gastrointestinal Tract/cytology , Male , Mice , Mice, Inbred C57BL , Muscles/cytology , Organ Specificity , RNA-Seq , Testis/cytology
11.
Development ; 147(1)2020 01 08.
Article in English | MEDLINE | ID: mdl-31852685

ABSTRACT

In order to efficiently derive hematopoietic stem cells (HSCs) from pluripotent precursors, it is crucial to understand how mesodermal cells acquire hematopoietic and endothelial identities: two divergent, but closely related, cell fates. Although Npas4 has been recently identified as a conserved master regulator of hemato-vascular development, the molecular mechanisms underlying cell fate divergence between hematopoietic and vascular endothelial cells are still unclear. Here, we show in zebrafish that mesodermal cell differentiation into hematopoietic and vascular endothelial cells is regulated by Junctional adhesion molecule 3b (Jam3b) via two independent signaling pathways. Mutation of jam3b led to a reduction in npas4l expression in the posterior lateral plate mesoderm and defects in both hematopoietic and vascular development. Mechanistically, we show that Jam3b promotes endothelial specification by regulating npas4l expression through repression of the Rap1a-Erk signaling cascade. Jam3b subsequently promotes hematopoietic development, including HSCs, by regulating lrrc15 expression in endothelial precursors through the activation of an integrin-dependent signaling cascade. Our data provide insight into the divergent mechanisms for instructing hematopoietic or vascular fates from mesodermal cells.


Subject(s)
Cardiovascular System/embryology , Hematopoiesis , Receptors, Cell Surface/physiology , Zebrafish Proteins/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiovascular System/cytology , Endothelial Cells/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells , MAP Kinase Signaling System , Mesoderm/embryology , Receptors, Cell Surface/genetics , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Sci Rep ; 9(1): 16006, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31690816

ABSTRACT

Cardiovascular progenitor cells (CPCs) derived from human pluripotent stem cells (hPSCs) are proposed to be invaluable cell sources for experimental and clinical studies. This wide range of applications necessitates large-scale production of CPCs in an in vitro culture system, which enables both expansion and maintenance of these cells. In this study, we aimed to develop a defined and efficient culture medium that uses signaling factors for large-scale expansion of early CPCs, called cardiogenic mesodermal cells (CMCs), which were derived from hPSCs. Chemical screening resulted in a medium that contained a reproducible combination of three factors (A83-01, bFGF, and CHIR99021) that generated 1014 CMCs after 10 passages without the propensity for tumorigenicity. Expanded CMCs retained their gene expression pattern, chromosomal stability, and differentiation tendency through several passages and showed both the safety and possible cardio-protective potentials when transplanted into the infarcted rat myocardium. These CMCs were efficiently cryopreserved for an extended period of time. This culture medium could be used for both adherent and suspension culture conditions, for which the latter is required for large-scale CMC production. Taken together, hPSC-derived CMCs exhibited self-renewal capacity in our simple, reproducible, and defined medium. These cells might ultimately be potential, promising cell sources for cardiovascular studies.


Subject(s)
Cardiovascular System/cytology , Culture Media/metabolism , Pluripotent Stem Cells/cytology , Animals , Cardiovascular System/metabolism , Cell Differentiation , Cell Proliferation , Culture Media/chemistry , Fibroblast Growth Factor 2/metabolism , Humans , Male , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation , Pyrazoles/metabolism , Pyridines/metabolism , Pyrimidines/metabolism , Rats , Rats, Wistar , Thiosemicarbazones/metabolism
13.
Elife ; 82019 05 28.
Article in English | MEDLINE | ID: mdl-31135336

ABSTRACT

Differences in the response of cardiomyocytes to oxygen deprivation in humans and chimpanzees may explain why humans are more prone to certain heart diseases.


Subject(s)
Biological Evolution , Cardiovascular System/anatomy & histology , Cardiovascular Diseases/genetics , Cardiovascular System/cytology , Genetic Predisposition to Disease , Humans , Models, Biological , Stem Cells/cytology
14.
Front Immunol ; 10: 834, 2019.
Article in English | MEDLINE | ID: mdl-31040854

ABSTRACT

Monocytes are a subset of cells that are categorized together with dendritic cells (DCs) and macrophages in the mononuclear phagocyte system (MPS). Despite sharing several phenotypic and functional characteristics with MPS cells, monocytes are unique cells with the ability to function as both precursor and effector cells in their own right. Before the development of hematopoietic stem cells (HSCs) in utero, monocytes are derived from erythro-myeloid precursors (EMPs) in the fetal liver that are important for populating the majority of tissue resident macrophages. After birth, monocytes arise from bone marrow (BM)-derived HSCs and are released into the circulation upon their maturation, where they survey peripheral tissues and maintain endothelial integrity. Upon sensing of microbial breaches or inflammatory stimuli, monocytes migrate into tissues where their plasticity allows them to differentiate into cells that resemble macrophages or DCs according to the environmental niche. Alternatively, they may also migrate into tissues in the absence of inflammation and remain in an undifferentiated state where they perform homeostatic roles. As monocytes are typically on the move, the availability of intravital imaging approaches has provided further insights into their trafficking patterns in distinct tissue compartments. In this review, we outline the importance of understanding their functional behavior in the context of tissue compartments, and how these studies may contribute towards improved vaccine and future therapeutic strategies.


Subject(s)
Cell Movement , Monocytes/physiology , Animals , Cardiovascular System/cytology , Fetus/cytology , Humans , Leukopoiesis , Spatio-Temporal Analysis
15.
Can J Physiol Pharmacol ; 97(6): 515-527, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31063413

ABSTRACT

Stress responses are important to human physiology and pathology, and the inability to adapt to cellular stress leads to cell death. To mitigate cellular stress and re-establish homeostasis, cells, including those in the cardiovascular system, activate stress coping response mechanisms. The endoplasmic reticulum, a component of the cellular reticular network in cardiac cells, mobilizes so-called endoplasmic reticulum stress coping responses, such as the unfolded protein response. MicroRNAs play an important part in the maintenance of cellular and tissue homeostasis, perform a central role in the biology of the cardiac myocyte, and are involved in pathological cardiac function and remodeling. In this paper, we review a link between endoplasmic reticulum homeostasis and microRNA with an emphasis on the impact on stress responses in the cardiovascular system.


Subject(s)
Cardiovascular System/cytology , Cardiovascular System/metabolism , Endoplasmic Reticulum/metabolism , MicroRNAs/genetics , Animals , Base Sequence , Endoplasmic Reticulum Stress , Homeostasis , Humans , Up-Regulation
16.
Biomater Sci ; 7(7): 2686-2701, 2019 Jun 25.
Article in English | MEDLINE | ID: mdl-31099355

ABSTRACT

Functionalizing cardiovascular biomaterials with an extracellular matrix (ECM) via in vitro decellularization has been applied as an effective method to improve the biocompatibility of implants. However, the current ECM modified materials used for surface engineering implants have functional restrictions compared with the natural blood vessel due to distinguished cell phenotypes in vitro. Herein, we are inspired by the natural vascular basement membrane which is composed of an ECM secreted by physiological endothelial cells (EC) and smooth muscle cells (SMC), preparing a novel ECM coating by successive cell culture and decellularization: appropriately scaled hyaluronic acid (HA) micro-patterns are used to regulate the SMC phenotype to contraction and simulate the blood flow shear stress (BFSS) effect to control the EC physiological phenotype. The nature-inspired ECM coating significantly improves the material's hemocompatibility, cytocompatibility and tissue compatibility, and may be promising to break the function limitation of a single ECM and address more clinical complications.


Subject(s)
Biomimetics/methods , Cardiovascular System/cytology , Endothelial Cells/cytology , Extracellular Matrix/metabolism , Myocytes, Smooth Muscle/cytology , Tissue Engineering/methods , Animals , Cell Adhesion , Cell Proliferation/drug effects , Humans , Macrophages/cytology , Male , Phenotype , Rabbits , Rats
17.
Can J Physiol Pharmacol ; 97(9): 820-828, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30897335

ABSTRACT

Tumor necrosis factor alpha (TNFα) and its type 1 receptor (TNFR1) are implicated in several autoimmune diseases, including rheumatoid arthritis, and are associated with complications at the cardiovascular level. Using human cardiomyocytes, vascular smooth muscle, vascular endothelial, and endocardial endothelial cells coupled to indirect immunofluorescence, our results showed the presence of TNFR1 at the levels of the plasma membrane (including the cytosol) and mostly at the level of the nuclear membranes (including the nucleoplasm). The distribution of the receptor is different between cell types; however, the density is significantly higher at the nuclear level in all 4 cell types. The density of the receptor was the highest in contractile cells including the cardiomyocytes and vascular smooth muscle cells, compared with endothelial cells including endocardial endothelial and vascular endothelial cells. Using the Ca2+ probe Fluo-3 coupled to quantitative confocal microscopy, our results showed that the cytokine induced a sustained Ca2+ increase in both the cytosol and nucleoplasm of all 4 cell types. This increase was more significant at the nuclear level, mainly in endothelial cells. Our results demonstrated the presence of TNFR1 at both the cell and nuclear membranes of cardiovascular cells, and that its activation modulated both cytosolic and nuclear Ca2+.


Subject(s)
Calcium/metabolism , Cardiovascular System/cytology , Cell Nucleus/metabolism , Cytosol/metabolism , Extracellular Space/metabolism , Intracellular Space/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adolescent , Adult , Endocardium/cytology , Endothelial Cells/cytology , Female , Homeostasis , Humans , Male , Middle Aged , Young Adult
18.
Compr Physiol ; 9(2): 767-797, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30892694

ABSTRACT

Autologous stem/progenitor cell-based methods to restore blood flow and function to ischemic tissues are clinically appealing for the substantial proportion of the population with cardiovascular diseases. Early preclinical and case studies established the therapeutic potential of autologous cell therapies for neovascularization in ischemic tissues. However, trials over the past ∼15 years reveal the benefits of such therapies to be much smaller than originally estimated and a definitive clinical benefit is yet to be established. Recently, there has been an emphasis on improving the number and function of cells [herein generally referred to as circulating angiogenic cells (CACs)] used for autologous cell therapies. CACs include of several subsets of circulating cells, including endothelial progenitor cells, with proangiogenic potential that is largely exerted through paracrine functions. As exercise is known to improve CV outcomes such as angiogenesis and endothelial function, much attention is being given to exercise to improve the number and function of CACs. Accordingly, there is a growing body of evidence that acute, short-term, and chronic exercise have beneficial effects on the number and function of different subsets of CACs. In particular, recent studies show that aerobic exercise training can increase the number of CACs in circulation and enhance the function of isolated CACs as assessed in ex vivo assays. This review summarizes the roles of different subsets of CACs and the effects of acute and chronic exercise on CAC number and function, with a focus on the number and paracrine function of circulating CD34+ cells, CD31+ cells, and CD62E+ cells. © 2019 American Physiological Society. Compr Physiol 9:767-797, 2019.


Subject(s)
Exercise/physiology , Stem Cells/physiology , Animals , Antigens, CD , Cardiovascular System/cytology , Humans , Neovascularization, Physiologic
19.
Can J Physiol Pharmacol ; 97(7): 589-599, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30854877

ABSTRACT

The last 20 years witnessed the emergence of the thymosin ß4 (Tß4)-N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) pathway as a new source of future therapeutic tools to treat cardiovascular and renal diseases. In this review article, we attempted to shed light on the numerous experimental findings pertaining to the many promising cardiovascular therapeutic avenues for Tß4 and (or) its N-terminal derivative, Ac-SDKP. Specifically, Ac-SDKP is endogenously produced from the 43-amino acid Tß4 by 2 successive enzymes, meprin α and prolyl oligopeptidase. We also discussed the possible mechanisms involved in the Tß4-Ac-SDKP-associated cardiovascular biological effects. In infarcted myocardium, Tß4 and Ac-SDKP facilitate cardiac repair after infarction by promoting endothelial cell migration and myocyte survival. Additionally, Tß4 and Ac-SDKP have antifibrotic and anti-inflammatory properties in the arteries, heart, lungs, and kidneys, and stimulate both in vitro and in vivo angiogenesis. The effects of Tß4 can be mediated directly through a putative receptor (Ku80) or via its enzymatically released N-terminal derivative Ac-SDKP. Despite the localization and characterization of Ac-SDKP binding sites in myocardium, more studies are needed to fully identify and clone Ac-SDKP receptors. It remains promising that Ac-SDKP or its degradation-resistant analogs could serve as new therapeutic tools to treat cardiac, vascular, and renal injury and dysfunction to be used alone or in combination with the already established pharmacotherapy for cardiovascular diseases.


Subject(s)
Cardiovascular System/metabolism , Oligopeptides/metabolism , Thymosin/metabolism , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cardiovascular System/cytology , Cardiovascular System/drug effects , Cardiovascular System/pathology , Humans
20.
Exp Cell Res ; 377(1-2): 103-108, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30794804

ABSTRACT

Tissues are continuously exposed to forces in vivo, whether from fluid pressure in an artery from our blood or compressive forces on joints from our body weight. The forces that cells are exposed to arise almost immediately after conception; it is therefore important to understand how forces shape stem cell differentiation into lineage committed cells, how they help organize cells into tissues, and how forces can cause or exacerbate disease. No tissue is exempt, but cardiovascular tissues in particular are exposed to these forces. While animal models have been used extensively in the past, there is growing recognition of their limitations when modeling disease complexity or human genetics. In this mini review, we summarize current understanding of the mechanical influences on the differentiation of cardiovascular progeny, how the transduction of forces influence the onset of disease, and how engineering approaches applied to this problem have yielded systems that create mature-like human tissues in vitro in which to assess the impact of disease on cell function.


Subject(s)
Cardiovascular Diseases/therapy , Cardiovascular System/cytology , Cell Differentiation , Mechanotransduction, Cellular , Stem Cells/cytology , Stress, Mechanical , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...