Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Dev Cell ; 2024 May 20.
Article in English | MEDLINE | ID: mdl-38897197

ABSTRACT

Selective degradation of damaged mitochondria by autophagy (mitophagy) is proposed to play an important role in cellular homeostasis. However, the molecular mechanisms and the requirement of mitochondrial quality control by mitophagy for cellular physiology are poorly understood. Here, we demonstrated that primary human cells maintain highly active basal mitophagy initiated by mitochondrial superoxide signaling. Mitophagy was found to be mediated by PINK1/Parkin-dependent pathway involving p62 as a selective autophagy receptor (SAR). Importantly, this pathway was suppressed upon the induction of cellular senescence and in naturally aged cells, leading to a robust shutdown of mitophagy. Inhibition of mitophagy in proliferating cells was sufficient to trigger the senescence program, while reactivation of mitophagy was necessary for the anti-senescence effects of NAD precursors or rapamycin. Furthermore, reactivation of mitophagy by a p62-targeting small molecule rescued markers of cellular aging, which establishes mitochondrial quality control as a promising target for anti-aging interventions.

2.
Cells ; 13(4)2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38391966

ABSTRACT

Cardiovascular diseases (CVDs), a group of disorders affecting the heart or blood vessels, are the primary cause of death worldwide, with an immense impact on patient quality of life and disability. According to the World Health Organization, CVD takes an estimated 17.9 million lives each year, where more than four out of five CVD deaths are due to heart attacks and strokes. In the decades to come, an increased prevalence of age-related CVD, such as atherosclerosis, coronary artery stenosis, myocardial infarction (MI), valvular heart disease, and heart failure (HF) will contribute to an even greater health and economic burden as the global average life expectancy increases and consequently the world's population continues to age. Considering this, it is important to focus our research efforts on understanding the fundamental mechanisms underlying CVD. In this review, we focus on cellular senescence and mitochondrial dysfunction, which have long been established to contribute to CVD. We also assess the recent advances in targeting mitochondrial dysfunction including energy starvation and oxidative stress, mitochondria dynamics imbalance, cell apoptosis, mitophagy, and senescence with a focus on therapies that influence both and therefore perhaps represent strategies with the most clinical potential, range, and utility.


Subject(s)
Cardiovascular Diseases , Heart Failure , Mitochondrial Diseases , Myocardial Infarction , Humans , Quality of Life , Cellular Senescence
4.
Age Ageing ; 52(7)2023 07 01.
Article in English | MEDLINE | ID: mdl-37466640

ABSTRACT

Cellular senescence has emerged as a fundamental biological mechanism underpinning the ageing process and has been implicated in the pathogenesis of an increasing number of age-related conditions. Cellular senescence is a cell fate originally defined as an irreversible loss of replicative potential although it is now clear that it can be induced by a variety of mechanisms independent of replication and telomere attrition. The drivers include a persistent DNA damage response causing multiple alterations in cellular function. Senescent cells secrete a range of mediators that drive chronic inflammation and can convert other cells to the senescent state-the senescence-associated secretory phenotype. Much research to date has been conducted in animal models, but it is now clear that senescent cells accompany ageing in humans and their presence is an important driver of disease across systems. Proof-of-concept work suggests that preventing or reversing senescence may be a viable strategy to counteract human ageing and age-related disease. Possible interventions include exercise, nutrition and senolytics/senostatic drugs although there are a number of potential limitations to the use of senotherapeutics. These interventions are generally tested for single-organ conditions, but the real power of this approach is the potential to tackle multiple age-related conditions. The litmus test for this exciting new class of therapies, however, will be whether they can improve healthy life expectancy rather than merely extending lifespan. The outcomes measured in clinical studies need to reflect these aims if senotherapeutics are to gain the trust of clinicians, patients and the public.


Subject(s)
Cellular Senescence , Senotherapeutics , Animals , Humans , Cellular Senescence/physiology , Aging/physiology , Longevity , Inflammation
5.
NPJ Aging ; 9(1): 15, 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37316516

ABSTRACT

Myocardial infarction is a leading cause of morbidity and mortality. While reperfusion is now standard therapy, pathological remodelling leading to heart failure remains a clinical problem. Cellular senescence has been shown to contribute to disease pathophysiology and treatment with the senolytic navitoclax attenuates inflammation, reduces adverse myocardial remodelling and results in improved functional recovery. However, it remains unclear which senescent cell populations contribute to these processes. To identify whether senescent cardiomyocytes contribute to disease pathophysiology post-myocardial infarction, we established a transgenic model in which p16 (CDKN2A) expression was specifically knocked-out in the cardiomyocyte population. Following myocardial infarction, mice lacking cardiomyocyte p16 expression demonstrated no difference in cardiomyocyte hypertrophy but exhibited improved cardiac function and significantly reduced scar size in comparison to control animals. This data demonstrates that senescent cardiomyocytes participate in pathological myocardial remodelling. Importantly, inhibition of cardiomyocyte senescence led to reduced senescence-associated inflammation and decreased senescence-associated markers within other myocardial lineages, consistent with the hypothesis that cardiomyocytes promote pathological remodelling by spreading senescence to other cell-types. Collectively this study presents the demonstration that senescent cardiomyocytes are major contributors to myocardial remodelling and dysfunction following a myocardial infarction. Therefore, to maximise the potential for clinical translation, it is important to further understand the mechanisms underlying cardiomyocyte senescence and how to optimise senolytic strategies to target this cell lineage.

7.
Subcell Biochem ; 103: 45-78, 2023.
Article in English | MEDLINE | ID: mdl-37120464

ABSTRACT

During ageing molecular damage leads to the accumulation of several hallmarks of ageing including mitochondrial dysfunction, cellular senescence, genetic instability and chronic inflammation, which contribute to the development and progression of ageing-associated diseases including cardiovascular disease. Consequently, understanding how these hallmarks of biological ageing interact with the cardiovascular system and each other is fundamental to the pursuit of improving cardiovascular health globally. This review provides an overview of our current understanding of how candidate hallmarks contribute to cardiovascular diseases such as atherosclerosis, coronary artery disease and subsequent myocardial infarction, and age-related heart failure. Further, we consider the evidence that, even in the absence of chronological age, acute cellular stress leading to accelerated biological ageing expedites cardiovascular dysfunction and impacts on cardiovascular health. Finally, we consider the opportunities that modulating hallmarks of ageing offer for the development of novel cardiovascular therapeutics.


Subject(s)
Cardiovascular Diseases , Heart Diseases , Telomerase , Humans , Cardiovascular Diseases/genetics , Telomerase/genetics , Aging/genetics , Cellular Senescence , Mitochondria/genetics
8.
Front Aging ; 3: 1058435, 2022.
Article in English | MEDLINE | ID: mdl-36452034

ABSTRACT

Cancer continues to place a heavy burden on healthcare systems around the world. Although cancer survivorship continues to improve, cardiotoxicity leading to cardiomyopathy and heart failure as a consequence of cancer therapy is rising, and yesterday's cancer survivors are fast becoming today's heart failure patients. Although the mechanisms driving cardiotoxicity are complex, cellular senescence is gaining attention as a major contributor to chemotherapy-induced cardiotoxicity and, therefore, may also represent a novel therapeutic target to prevent this disease. Cellular senescence is a well-recognized response to clinical doses of chemotherapies, including anthracyclines, and is defined by cell cycle exit, phenotypic alterations which include mitochondrial dysfunction, and the expression of the pro-senescent, pro-fibrotic, and pro-inflammatory senescence-associated phenotype. Senescence has an established involvement in promoting myocardial remodeling during aging, and studies have demonstrated that the elimination of senescence can attenuate the pathophysiology of several cardiovascular diseases. Most recently, pharmacology-mediated elimination of senescence, using a class of drugs termed senolytics, has been demonstrated to prevent myocardial dysfunction in preclinical models of chemotherapy-induced cardiotoxicity. In this review, we will discuss the evidence that anthracycline-induced senescence causes the long-term cardiotoxicity of anticancer chemotherapies, consider how the senescent phenotype may promote myocardial dysfunction, and examine the exciting possibility that targeting senescence may prove a therapeutic strategy to prevent or even reverse chemotherapy-induced cardiac dysfunction.

9.
Cardiovasc Drugs Ther ; 36(1): 187-196, 2022 02.
Article in English | MEDLINE | ID: mdl-32979174

ABSTRACT

Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the leading cause of death in 40% of individuals over 65 years old. Ageing is associated with both an increased prevalence of cardiovascular disease including heart failure, coronary artery disease, and myocardial infarction. Furthermore, ageing is associated with a poorer prognosis to these diseases. Genetic models allowing the elimination of senescent cells revealed that an accumulation of senescence contributes to the pathophysiology of cardiovascular ageing and promotes the progression of cardiovascular disease through the expression of a proinflammatory and profibrotic senescence-associated secretory phenotype. These studies have resulted in an effort to identify pharmacological therapeutics that enable the specific elimination of senescent cells through apoptosis induction. These senescent cell apoptosis-inducing compounds are termed senolytics and their potential to ameliorate age-associated cardiovascular disease is the focus of this review.


Subject(s)
Cardiovascular Diseases/drug therapy , Cellular Senescence/drug effects , Senotherapeutics/pharmacology , Aged , Aging , Animals , Apoptosis/drug effects , Cardiovascular Diseases/physiopathology , Disease Progression , Humans , Prognosis , Risk Factors , Senescence-Associated Secretory Phenotype/physiology
10.
Mech Ageing Dev ; 198: 111540, 2021 09.
Article in English | MEDLINE | ID: mdl-34237321

ABSTRACT

Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the cause of death in 40 % of individuals over 65 years old. Ageing is associated with an increased prevalence of atherosclerosis, coronary artery stenosis and subsequent myocardial infarction, thoracic aortic aneurysm, valvular heart disease and heart failure. An accumulation of senescence and increased inflammation, caused by the senescence-associated secretory phenotype, have been implicated in the aetiology and progression of these age-associated diseases. Recently it has been demonstrated that compounds targeting components of anti-apoptotic pathways expressed by senescent cells can preferentially induce senescence cells to apoptosis and have been termed senolytics. In this review, we discuss the evidence demonstrating that senescence contributes to cardiovascular disease, with a particular focus on studies that indicate the promise of senotherapy. Based on these data we suggest novel indications for senolytics as a treatment of cardiovascular diseases which have yet to be studied in the context of senotherapy. Finally, while the potential benefits are encouraging, several complications may result from senolytic treatment. We, therefore, consider these challenges in the context of the cardiovascular system.


Subject(s)
Aging , Apoptosis Regulatory Proteins/metabolism , Cardiovascular Diseases , Cellular Senescence , Senotherapeutics/pharmacology , Aging/immunology , Aging/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/therapy , Cellular Senescence/drug effects , Cellular Senescence/physiology , Humans , Inflammation/metabolism , Senescence-Associated Secretory Phenotype , Signal Transduction/drug effects
11.
Aging Cell ; 19(10): e13249, 2020 10.
Article in English | MEDLINE | ID: mdl-32996233

ABSTRACT

A key component of cardiac ischemia-reperfusion injury (IRI) is the increased generation of reactive oxygen species, leading to enhanced inflammation and tissue dysfunction in patients following intervention for myocardial infarction. In this study, we hypothesized that oxidative stress, due to ischemia-reperfusion, induces senescence which contributes to the pathophysiology of cardiac IRI. We demonstrate that IRI induces cellular senescence in both cardiomyocytes and interstitial cell populations and treatment with the senolytic drug navitoclax after ischemia-reperfusion improves left ventricular function, increases myocardial vascularization, and decreases scar size. SWATH-MS-based proteomics revealed that biological processes associated with fibrosis and inflammation that were increased following ischemia-reperfusion were attenuated upon senescent cell clearance. Furthermore, navitoclax treatment reduced the expression of pro-inflammatory, profibrotic, and anti-angiogenic cytokines, including interferon gamma-induced protein-10, TGF-ß3, interleukin-11, interleukin-16, and fractalkine. Our study provides proof-of-concept evidence that cellular senescence contributes to impaired heart function and adverse remodeling following cardiac ischemia-reperfusion. We also establish that post-IRI the SASP plays a considerable role in the inflammatory response. Subsequently, senolytic treatment, at a clinically feasible time-point, attenuates multiple components of this response and improves clinically important parameters. Thus, cellular senescence represents a potential novel therapeutic avenue to improve patient outcomes following cardiac ischemia-reperfusion.


Subject(s)
Cellular Senescence/physiology , Reperfusion Injury/metabolism , Female , Humans , Male
12.
Aging Cell ; 18(3): e12945, 2019 06.
Article in English | MEDLINE | ID: mdl-30920115

ABSTRACT

Cardiovascular disease is the leading cause of death in individuals over 60 years old. Aging is associated with an increased prevalence of coronary artery disease and a poorer prognosis following acute myocardial infarction (MI). With age, senescent cells accumulate in tissues, including the heart, and contribute to age-related pathologies. However, the role of senescence in recovery following MI has not been investigated. In this study, we demonstrate that treatment of aged mice with the senolytic drug, navitoclax, eliminates senescent cardiomyocytes and attenuates profibrotic protein expression in aged mice. Importantly, clearance of senescent cells improved myocardial remodelling and diastolic function as well as overall survival following MI. These data provide proof-of-concept evidence that senescent cells are major contributors to impaired function and increased mortality following MI and that senolytics are a potential new therapeutic avenue for MI.


Subject(s)
Aging/drug effects , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Myocardial Infarction/drug therapy , Sulfonamides/pharmacology , Acute Disease , Aniline Compounds/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Survival/drug effects , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Sulfonamides/administration & dosage
13.
EMBO J ; 38(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30737259

ABSTRACT

Ageing is the biggest risk factor for cardiovascular disease. Cellular senescence, a process driven in part by telomere shortening, has been implicated in age-related tissue dysfunction. Here, we address the question of how senescence is induced in rarely dividing/post-mitotic cardiomyocytes and investigate whether clearance of senescent cells attenuates age-related cardiac dysfunction. During ageing, human and murine cardiomyocytes acquire a senescent-like phenotype characterised by persistent DNA damage at telomere regions that can be driven by mitochondrial dysfunction and crucially can occur independently of cell division and telomere length. Length-independent telomere damage in cardiomyocytes activates the classical senescence-inducing pathways, p21CIP and p16INK4a, and results in a non-canonical senescence-associated secretory phenotype, which is pro-fibrotic and pro-hypertrophic. Pharmacological or genetic clearance of senescent cells in mice alleviates detrimental features of cardiac ageing, including myocardial hypertrophy and fibrosis. Our data describe a mechanism by which senescence can occur and contribute to age-related myocardial dysfunction and in the wider setting to ageing in post-mitotic tissues.


Subject(s)
Cardiomegaly/pathology , Cellular Senescence , DNA Damage , Fibrosis/pathology , Mitosis , Myocytes, Cardiac/pathology , Telomere Shortening , Aging , Animals , Cardiomegaly/etiology , Female , Fibrosis/etiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monoamine Oxidase/physiology , Myocytes, Cardiac/metabolism , Phenotype , RNA/physiology , Rats, Sprague-Dawley , Telomerase/physiology
14.
Exp Gerontol ; 109: 5-15, 2018 08.
Article in English | MEDLINE | ID: mdl-29054534

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of death globally. One of the main risk factors for CVD is age, however the biological processes that occur in the heart during ageing are poorly understood. It is therefore important to understand the fundamental mechanisms driving heart ageing to enable the development of preventions and treatments targeting these processes. Cellular senescence is often described as the irreversible cell-cycle arrest which occurs in somatic cells. Emerging evidence suggests that cellular senescence plays a key role in heart ageing, however the cell-types involved and the underlying mechanisms are not yet elucidated. In this review we discuss the current understanding of how mechanisms known to contribute to senescence impact on heart ageing and CVD. Finally, we evaluate recent data suggesting that targeting senescent cells may be a viable therapy to counteract the ageing of the heart.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Heart/physiology , Animals , Cardiovascular Diseases/etiology , DNA Damage , DNA Repair , Humans , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Telomere
15.
J Vis Exp ; (111)2016 05 23.
Article in English | MEDLINE | ID: mdl-27285379

ABSTRACT

Although it is accepted that the heart has a limited potential to regenerate cardiomyocytes following injury and that low levels of cardiomyocyte turnover occur during normal ageing, quantification of these events remains challenging. This is in part due to the rarity of the process and the fact that multiple cellular sources contribute to myocardial maintenance. Furthermore, DNA duplication within cardiomyocytes often leads to a polyploid cardiomyocyte and only rarely leads to new cardiomyocytes by cellular division. In order to accurately quantify cardiomyocyte turnover discrimination between these processes is essential. The protocol described here employs long term nucleoside labeling in order to label all nuclei which have arisen as a result of DNA replication and cardiomyocyte nuclei identified by utilizing nuclei isolation and subsequent PCM1 immunolabeling. Together this allows the accurate and sensitive identification of the nucleoside labeling of the cardiomyocyte nuclei population. Furthermore, 4',6-diamidino-2-phenylindole labeling and analysis of nuclei ploidy, enables the discrimination of neo-cardiomyocyte nuclei from nuclei which have incorporated nucleoside during polyploidization. Although this method cannot control for cardiomyocyte binucleation, it allows a rapid and robust quantification of neo-cardiomyocyte nuclei while accounting for polyploidization. This method has a number of downstream applications including assessing the potential therapeutics to enhance cardiomyocyte regeneration or investigating the effects of cardiac disease on cardiomyocyte turnover and ploidy. This technique is also compatible with additional downstream immunohistological techniques, allowing quantification of nucleoside incorporation in all cardiac cell types.


Subject(s)
DNA/biosynthesis , Myocytes, Cardiac/physiology , Regeneration/physiology , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA/genetics , DNA Replication/physiology , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Ploidies
16.
Stem Cell Rev Rep ; 9(5): 731-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23619929

ABSTRACT

Cardiac side population (CSP) cells, defined by their ability to efflux the vital dye Hoechst 33342, have been identified as putative cardiac stem cells based on their potential to give rise to both cardiomyocytes and endothelial cells. The CSP phenotype relies on an active metabolic pathway and cell viability to identify a rare population of cells and therefore technical differences in the CSP staining protocol can lead to inconsistent results and discrepancies between studies. Here we describe an established protocol for CSP identification and have optimised a protocol for CSP analysis utilizing an automated cardiac digestion procedure using gentleMACs dissociation and Hoechst 33342 staining followed by dual wavelength flow cytometric analysis.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Myocardium/cytology , Side-Population Cells/cytology , Animals , Benzimidazoles/metabolism , Cell Separation/instrumentation , Cells, Cultured , Deoxyribonuclease I/metabolism , Flow Cytometry/instrumentation , Matrix Metalloproteinase 8/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Myocardium/metabolism , Pronase/metabolism , Reproducibility of Results , Side-Population Cells/metabolism
17.
FASEB J ; 26(12): 4832-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22919071

ABSTRACT

While the mammalian heart has low, but functionally significant, levels of telomerase expression, the cellular population responsible remains incompletely characterized. This study aimed to identify the cell types responsible for cardiac telomerase activity in neonatal, adult, and cryoinjured adult hearts using transgenic mice expressing green fluorescent protein (GFP), driven by the promoter for murine telomerase reverse transcriptase (mTert), which is a necessary and rate-limiting component of telomerase. A rare population of mTert-GFP-expressing cells was identified that possessed all detectable cardiac telomerase RNA and telomerase activity. It was heterogeneous and included cells coexpressing markers of cardiomyocytic, endothelial, and mesenchymal lineages, putative cardiac stem cell markers, and, interestingly, cardiomyocytes with a differentiated phenotype. Quantification using both flow cytometry and immunofluorescence identified a significant decline in mTert-GFP cells in adult animals compared to neonates (∼9- and ∼20-fold, respectively). Cardiac injury resulted in a ∼6.45-fold expansion of this population (P<0.005) compared with sham-operated controls. This study identifies the cells responsible for cardiac telomerase activity, demonstrates a significant diminution with age but a marked response to injury, and, given the relationship between telomerase activity and stem cell populations, suggests that they represent a potential target for further investigation of cardiac regenerative potential.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Myocardium/metabolism , Telomerase/genetics , Age Factors , Animals , Animals, Newborn , Antigens, Ly/genetics , Antigens, Ly/metabolism , Flow Cytometry , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Myocardium/cytology , Myocardium/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Stem Cells Dev ; 21(16): 3019-30, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22571645

ABSTRACT

In this study, we have demonstrated that cells of neural crest origin located in the dermal papilla (DP) exhibit endothelial marker expression and a functional activity. When grown in endothelial growth media, DP primary cultures upregulate expression of vascular endothelial growth factor receptor 1 (FLT1) mRNA and downregulate expression of the dermal stem cell marker α-smooth muscle actin. DP cells have demonstrated functional characteristics of endothelial cells, including the ability to form capillary-like structures on Matrigel, increase uptake of low-density lipoprotein and upregulate ICAM1 (CD54) in response to tumour necrosis factor alpha (TNF-α) stimulation. We confirmed that these observations were not due to contaminating endothelial cells, by using DP clones. We have also used the WNT1cre/ROSA26R and WNT1cre/YFP lineage-tracing mouse models to identify a population of neural crest-derived cells in DP cultures that express the endothelial marker PECAM (CD31); these cells also form capillary-like structures on Matrigel. Importantly, cells of neural crest origin that express markers of endothelial and mesenchymal lineages exist within the dermal sheath of the vibrissae follicle.


Subject(s)
Cell Differentiation , Cell Lineage , Dermis/cytology , Endothelial Cells/cytology , Stem Cells/cytology , Actins/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Clone Cells , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Flow Cytometry , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells , Humans , Integrases/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Neural Crest/cytology , Neural Crest/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats , Stem Cells/metabolism , von Willebrand Factor/metabolism
20.
Exp Dermatol ; 19(6): 546-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20456497

ABSTRACT

Human dermal papilla (DP) cells grown in two-dimensional (2D) culture have been studied extensively. However, key differences exist between DP cell activities in vivo and in vitro. Using a suspension method of cell culture to maintain DP cells, we created three-dimensional (3D) dermal spheres morphologically akin to intact (anagen) DPs. Analysis of these spheres using immunocytochemistry demonstrates that they have expression profiles different from papilla cells cultured in 2D but with many similarities to intact DPs. This method of DP cell culture may provide us with a tool to elucidate our understanding of signalling within the DP as it relates to induction, maintenance or even inhibition of hair growth.


Subject(s)
Cell Culture Techniques/methods , Dermis/cytology , Fibroblasts/cytology , Hair Follicle/cytology , Models, Biological , Spheroids, Cellular/cytology , Actins/metabolism , Adenomatous Polyposis Coli Protein/genetics , Alkaline Phosphatase/metabolism , Axin Protein , Cell Nucleus/metabolism , Connexin 43/metabolism , Cytoskeletal Proteins/genetics , Fibroblasts/metabolism , Gene Expression/genetics , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Heparan Sulfate Proteoglycans/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Neural Cell Adhesion Molecules/genetics , Proliferating Cell Nuclear Antigen/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Serine Endopeptidases/genetics , Spheroids, Cellular/metabolism , Versicans/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...