Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
BMC Res Notes ; 16(1): 341, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37974202

ABSTRACT

OBJECTIVE: Identification of patients at high risk of aggressive prostate cancer is a major clinical challenge. With the view of developing artificial intelligence-based methods for identification of these patients, we are constructing a comprehensive clinical database including 7448 prostate cancer (PCa) Danish patients. In this paper we provide an epidemiological description and patients' trajectories of this retrospective observational population, to contribute to the understanding of the characteristics and pathways of PCa patients in Denmark. RESULTS: Individuals receiving a PCa diagnosis during 2008-2014 in Region Southern Denmark were identified, and all diagnoses, operations, investigations, and biochemistry analyses, from 4 years prior, to 5 years after PCa diagnosis were obtained. About 85.1% were not diagnosed with metastatic PCa during the study period (unaggressive PCa); 9.2% were simultaneously diagnosed with PCa and metastasis (aggressive-advanced PCa), while 5.7% were not diagnosed with metastatic PCa at first, but they were diagnosed with metastasis at some point during the 5 years follow-up (aggressive-not advanced PCa). Patients with unaggressive PCa had more clinical investigations directly related to PCa detection (prostate ultrasounds and biopsies) during the 4 years prior to PCa diagnosis, compared to patients with aggressive PCa, which may have contributed to the early detection of PCa.


Subject(s)
Artificial Intelligence , Prostatic Neoplasms , Male , Humans , Retrospective Studies , Early Detection of Cancer , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/pathology , Denmark/epidemiology
2.
J Vis Exp ; (178)2021 12 28.
Article in English | MEDLINE | ID: mdl-35037661

ABSTRACT

Stem cells are used in many research areas within regenerative medicine in part because these treatments can be curative rather than symptomatic. Stem cells can be obtained from different tissues and several methods for isolation have been described. The presented method for the isolation of adipose-derived regenerative cells (ADRCs) can be used within many therapeutic areas because the method is a general procedure and, therefore, not limited to erectile dysfunction (ED) therapy. ED is a common and serious side effect to radical prostatectomy (RP) since ED often is not well treated with conventional therapy. Using ADRC's as treatment for ED has attracted great interest due to the initial positive results after a single injection of cells into the corpora cavernosum. The method used for the isolation of ADRC's is a simple, automated process, that is reproducible and ensures a uniform product. Furthermore, the sterility of the isolated product is ensured because the entire process takes place in a closed system. It is important to minimize the risk of contamination and infection since the stem cells are used for injection in humans. The whole procedure can be done within 2.5-3.5 hours and does not require a classified laboratory which eliminates the need for shipping tissue to an off-site. However, the procedure has some limitations since the minimum amount of drained lipoaspirate for the isolation device to function is 100 g.


Subject(s)
Erectile Dysfunction , Cell- and Tissue-Based Therapy , Erectile Dysfunction/etiology , Erectile Dysfunction/therapy , Humans , Male , Penis/surgery , Prostatectomy/adverse effects , Regenerative Medicine/methods
3.
Dev Biol ; 445(1): 1-7, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30389344

ABSTRACT

MESP1 is a key transcription factor in development of early cardiovascular tissue and it is required for induction of the cardiomyocyte (CM) gene expression program, but its role in vascular development is unclear. Here, we used inducible CRISPRi knock-down of MESP1 to analyze the molecular processes of the early differentiation stages of human induced pluripotent stem cells into mesoderm and subsequently vascular progenitor cells. We found that expression of the mesodermal marker, BRACHYURY (encoded by T) was unaffected in MESP1 knock-down cells as compared to wild type cells suggesting timely movement through the primitive streak whereas another mesodermal marker MIXL1 was slightly, but significantly decreased. In contrast, the expression of the vascular cell surface marker KDR was decreased and CD31 and CD34 expression were substantially reduced in MESP1 knock-down cells supporting inhibition or delay of vascular specification. In addition, mRNA microarray data revealed several other altered gene expressions including the EMT regulating transcription factors SNAI1 and TWIST1, which were both significantly decreased indicating that MESP1 knock-down cells are less likely to undergo EMT during vascular progenitor differentiation. Our study demonstrates that while leaving primitive streak markers unaffected, MESP1 expression is required for timely vascular progenitor specification. Thus, MESP1 expression is essential for the molecular features of early CM, EC and VSMC lineage specification.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Induced Pluripotent Stem Cells/metabolism , Primitive Streak/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/physiology , Cell Lineage , Embryonic Stem Cells/cytology , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Helix-Loop-Helix Motifs/physiology , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mesoderm/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Primitive Streak/cytology , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism
4.
Bone ; 110: 312-320, 2018 05.
Article in English | MEDLINE | ID: mdl-29499415

ABSTRACT

Soluble delta-like 1 homolog (DLK1) is a circulating protein that belongs to the Notch/Serrate/delta family, which regulates many differentiation processes including osteogenesis and adipogenesis. We have previously demonstrated an inhibitory effect of DLK1 on bone mass via stimulation of bone resorption and inhibition of bone formation. Further, serum DLK1 levels are elevated and positively correlated to bone turnover markers in estrogen (E)-deficient rodents and women. In this report, we examined whether inhibition of serum DLK1 activity using a neutralizing monoclonal antibody protects from E deficiency-associated bone loss in mice. Thus, we generated mouse monoclonal anti-mouse DLK1 antibodies (MAb DLK1) that enabled us to reduce and also quantitate the levels of bioavailable serum DLK1 in vivo. Ovariectomized (ovx) mice were injected intraperitoneally twice weekly with MAb DLK1 over a period of one month. DEXA-, microCT scanning, and bone histomorphometric analyses were performed. Compared to controls, MAb DLK1 treated ovx mice were protected against ovx-induced bone loss, as revealed by significantly increased total bone mass (BMD) due to increased trabecular bone volume fraction (BV/TV) and inhibition of bone resorption. No significant changes were observed in total fat mass or in the number of bone marrow adipocytes. These results support the potential use of anti-DLK1 antibody therapy as a novel intervention to protect from E deficiency associated bone loss.


Subject(s)
Antibodies/therapeutic use , Bone Resorption/prevention & control , Estrogens/deficiency , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Antibodies, Neutralizing/therapeutic use , Bone Density/drug effects , Calcium-Binding Proteins , Cell Line , Female , Flow Cytometry , Humans , Mice , NIH 3T3 Cells , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteoporosis/prevention & control , Ovariectomy , X-Ray Microtomography
5.
Cardiovasc Res ; 113(11): 1282-1293, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28859296

ABSTRACT

Cardiovascular diseases remain the leading cause of death worldwide and current treatment strategies have limited effect of disease progression. It would be desirable to have better models to study developmental and pathological processes and model vascular diseases in laboratory settings. To this end, human induced pluripotent stem cells (hiPSCs) have generated great enthusiasm, and have been a driving force for development of novel strategies in drug discovery and regenerative cell-therapy for the last decade. Hence, investigating the mechanisms underlying the differentiation of hiPSCs into specialized cell types such as cardiomyocytes, endothelial cells, and vascular smooth muscle cells (VSMCs) may lead to a better understanding of developmental cardiovascular processes and potentiate progress of safe autologous regenerative therapies in pathological conditions. In this review, we summarize the latest trends on differentiation protocols of hiPSC-derived VSMCs and their potential application in vascular research and regenerative therapy.


Subject(s)
Cell Lineage/physiology , Induced Pluripotent Stem Cells , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Animals , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/drug effects
6.
Sci Rep ; 7(1): 8362, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827644

ABSTRACT

After birth cardiomyocytes undergo terminal differentiation, characterized by binucleation and centrosome disassembly, rendering the heart unable to regenerate. Yet, it has been suggested that newborn mammals regenerate their hearts after apical resection by cardiomyocyte proliferation. Thus, we tested the hypothesis that apical resection either inhibits, delays, or reverses cardiomyocyte centrosome disassembly and binucleation. Our data show that apical resection rather transiently accelerates centrosome disassembly as well as the rate of binucleation. Consistent with the nearly 2-fold increased rate of binucleation there was a nearly 2-fold increase in the number of cardiomyocytes in mitosis indicating that the majority of injury-induced cardiomyocyte cell cycle activity results in binucleation, not proliferation. Concurrently, cardiomyocytes undergoing cytokinesis from embryonic hearts exhibited midbody formation consistent with successful abscission, whereas those from 3 day-old cardiomyocytes after apical resection exhibited midbody formation consistent with abscission failure. Lastly, injured hearts failed to fully regenerate as evidenced by persistent scarring and reduced wall motion. Collectively, these data suggest that should a regenerative program exist in the newborn mammalian heart, it is quickly curtailed by developmental mechanisms that render cardiomyocytes post-mitotic.


Subject(s)
Cell Differentiation , Heart Injuries , Myocytes, Cardiac/physiology , Animals , Animals, Newborn , Cell Proliferation , Rats, Sprague-Dawley , Regeneration
7.
Stem Cells Int ; 2016: 3846971, 2016.
Article in English | MEDLINE | ID: mdl-27994622

ABSTRACT

Background. Scaffolds for bone tissue engineering (BTE) can be loaded with stem and progenitor cells (SPC) from different sources to improve osteogenesis. SPC can be found in bone marrow, adipose tissue, and other tissues. Little is known about osteogenic potential of adipose-derived culture expanded, adherent cells (A-CEAC). This study compares in vivo osteogenic capacity between A-CEAC and bone marrow derived culture expanded, adherent cells (BM-CEAC). Method. A-CEAC and BM-CEAC were isolated from five female sheep and seeded on hydroxyapatite granules prior to subcutaneous implantation in immunodeficient mice. The doses of cells in the implants were 0.5 × 106, 1.0 × 106, or 1.5 × 106 A-CEAC and 0.5 × 106 BM-CEAC, respectively. After eight weeks, bone volume versus total tissue volume (BV/TV) was quantified using histomorphometry. Origin of new bone was assessed using human vimentin (HVIM) antibody staining. Results. BM-CEAC yielded significantly higher BV/TV than any A-CEAC group, and differences between A-CEAC groups were not statistically significant. HVIM antibody stain was successfully used to identify sheep cells in this model. Conclusion. A-CEAC and BM-CEAC were capable of forming bone, and BM-CEAC yielded significantly higher BV/TV than any A-CEAC group. In vitro treatment to enhance osteogenic capacity of A-CEAC is suggested for further research in ovine bone tissue engineering.

8.
Int J Cardiol ; 222: 448-456, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27505332

ABSTRACT

BACKGROUND/OBJECTIVES: Epicardium-derived progenitor cells (EPDCs) differentiate into all heart cell types in the embryonic heart, yet their differentiation into cardiomyocytes in the adult heart is limited and poorly described. This may be due to EPDCs lacking myogenic potential or the inert adult heart missing regenerative signals essential for directed differentiation of EPDCs. Herein, we aimed to evaluate the myogenic potential of neonatal EPDCs in adult and neonatal mouse myocardium, as well as in skeletal muscle. The two latter tissues have an intrinsic capability to develop and regenerate, in contrast to the adult heart. METHODS: Highly purified mouse EPDCs were transplanted into damaged neonatal and adult myocardium as well as regenerating skeletal muscle. Co-cultures with skeletal myoblasts were used to distinguish fusion independent myogenic conversion. RESULTS: No donor EPDC-derived cardiomyocytes were observed in hearts. In contrast, a remarkable contribution of EPDCs to skeletal muscle myofiber formation was evident in vivo. Furthermore, co-cultures of EPDCs with myoblasts showed that EPDCs became part of multinucleated fibers and appeared to acquire myogenic traits independent of a fusion event. Fluorescence activated cell sorting of EPDCs co-cultured with and without myoblasts and subsequent qRT-PCR of 64 transcripts established that the myogenic phenotype conversion was accomplished through induction of a transcriptional myogenic program. CONCLUSION: These results suggest that EPDCs may be more myogenic than previously anticipated. But, the heart may lack factors for induction of myogenesis of EPDCs, a scenario that should be taken into consideration when aiming for repair of damaged myocardium by stem cell transplantation.


Subject(s)
Muscle, Skeletal/cytology , Myocardium/cytology , Myocytes, Cardiac , Pericardium/cytology , Stem Cells , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques/methods , Mice , Mice, Inbred C57BL , Muscle, Skeletal/physiology , Myocytes, Cardiac/physiology , Pericardium/physiology , Stem Cells/physiology
9.
Cell Stem Cell ; 18(4): 541-53, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26971820

ABSTRACT

Developing technologies for efficient and scalable disruption of gene expression will provide powerful tools for studying gene function, developmental pathways, and disease mechanisms. Here, we develop clustered regularly interspaced short palindromic repeat interference (CRISPRi) to repress gene expression in human induced pluripotent stem cells (iPSCs). CRISPRi, in which a doxycycline-inducible deactivated Cas9 is fused to a KRAB repression domain, can specifically and reversibly inhibit gene expression in iPSCs and iPSC-derived cardiac progenitors, cardiomyocytes, and T lymphocytes. This gene repression system is tunable and has the potential to silence single alleles. Compared with CRISPR nuclease (CRISPRn), CRISPRi gene repression is more efficient and homogenous across cell populations. The CRISPRi system in iPSCs provides a powerful platform to perform genome-scale screens in a wide range of iPSC-derived cell types, dissect developmental pathways, and model disease.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Silencing , Induced Pluripotent Stem Cells/metabolism , Humans
10.
Cell Signal ; 28(4): 246-54, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26791579

ABSTRACT

Canonical NOTCH signaling, known to be essential for tissue development, requires the Delta-Serrate-LAG2 (DSL) domain for NOTCH to interact with its ligand. However, despite lacking DSL, Delta-like 1 homolog (DLK1), a protein that plays a significant role in mammalian development, has been suggested to interact with NOTCH1 and act as an antagonist. This non-canonical interaction is, however controversial, and evidence for a direct interaction, still lacking in mammals. In this study, we elucidated the putative DLK1-NOTCH1 interaction in a mammalian context. Taking a global approach and using Dlk1(+/+) and Dlk1(-/-) mouse tissues at E16.5, we demonstrated that several NOTCH signaling pathways indeed are affected by DLK1 during tissue development, and this was supported by a lower activation of NOTCH1 protein in Dlk1(+/+) embryos. Likewise, but using a distinct Dlk1-manipulated (siRNA) setup in a mammalian cell line, NOTCH signaling was substantially inhibited by DLK1. Using a mammalian two-hybrid system, we firmly established that the effect of DLK1 on NOTCH signaling was due to a direct interaction between DLK1 and NOTCH1. By careful dissection of this mechanism, we found this interaction to occur between EGF domains 5 and 6 of DLK1 and EGF domains 10-15 of NOTCH1. Thus, our data provide the first evidence for a direct interaction between DLK1 and NOTCH1 in mammals, and substantiate that non-canonical NOTCH ligands exist, adding to the complexity of NOTCH signaling.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology , 3T3-L1 Cells , Animals , Calcium-Binding Proteins , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Protein Structure, Tertiary , Receptor, Notch1/genetics
11.
J Renin Angiotensin Aldosterone Syst ; 16(4): 1288-97, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25031299

ABSTRACT

INTRODUCTION: MicroRNAs (miRNAs) are emerging as key regulators of cardiovascular development and disease; however, the cardiac miRNA target molecules are not well understood. We and others have described the Angiotensin II (AngII)-induced miR-132/212 family as novel regulators of cardiovascular function including regulation of cardiac hypertrophy, heart failure and blood pressure possibly through AT1R signalling. However, the miR-132/212 targets in the heart remain unknown. MATERIALS AND METHODS: To understand the role of these miRNAs in cardiac signalling networks, we undertook comprehensive in silico and in vitro experiments to identify miR-132/212 molecular targets in primary rat cardiac fibroblasts. RESULTS: MiR-132/212 overexpression increased fibroblast cell size and mRNA arrays detected several hundred genes that were differentially expressed, including a wide panel of receptors, signalling molecules and transcription factors. Subsequent comprehensive in silico analysis identified 24 target genes, of which 22 genes were qPCR validated. We identified seven genes involved in AngII signalling pathways. CONCLUSION: We here report novel insight of an extensive network of molecular pathways that fine-tuned by miR-132/212, suggesting a role for this miRNA family as master signalling switches in cardiac fibroblasts. Our data underscore the potential for miRNA tools to manipulate a large array of molecules and thereby control biological function.


Subject(s)
Angiotensin II/pharmacology , Fibroblasts/metabolism , MicroRNAs/metabolism , Myocardium/cytology , Signal Transduction/drug effects , Animals , Cell Size/drug effects , Cells, Cultured , Fibroblasts/drug effects , Male , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Reproducibility of Results , Signal Transduction/genetics , Software
12.
Circ Res ; 115(8): 696-708, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25085941

ABSTRACT

RATIONALE: Effective neovascularization is crucial for recovery after cardiovascular events. OBJECTIVE: Because microRNAs regulate expression of up to several hundred target genes, we set out to identify microRNAs that target genes in all pathways of the multifactorial neovascularization process. Using www.targetscan.org, we performed a reverse target prediction analysis on a set of 197 genes involved in neovascularization. We found enrichment of binding sites for 27 microRNAs in a single microRNA gene cluster. Microarray analyses showed upregulation of 14q32 microRNAs during neovascularization in mice after single femoral artery ligation. METHODS AND RESULTS: Gene silencing oligonucleotides (GSOs) were used to inhibit 4 14q32 microRNAs, miR-329, miR-487b, miR-494, and miR-495, 1 day before double femoral artery ligation. Blood flow recovery was followed by laser Doppler perfusion imaging. All 4 GSOs clearly improved blood flow recovery after ischemia. Mice treated with GSO-495 or GSO-329 showed increased perfusion already after 3 days (30% perfusion versus 15% in control), and those treated with GSO-329 showed a full recovery of perfusion after 7 days (versus 60% in control). Increased collateral artery diameters (arteriogenesis) were observed in adductor muscles of GSO-treated mice, as well as increased capillary densities (angiogenesis) in the ischemic soleus muscle. In vitro, treatment with GSOs led to increased sprout formation and increased arterial endothelial cell proliferation, as well as to increased arterial myofibroblast proliferation. CONCLUSIONS: The 14q32 microRNA gene cluster is highly involved in neovascularization. Inhibition of 14q32 microRNAs miR-329, miR-487b, miR-494, and miR-495 provides a promising tool for future therapeutic neovascularization.


Subject(s)
Blood Vessels/metabolism , MicroRNAs/genetics , Animals , Blood Flow Velocity/genetics , Blood Flow Velocity/physiology , Blood Vessels/physiopathology , Cell Proliferation , Cells, Cultured , Chromosomes, Human, Pair 14/genetics , Endothelial Cells/metabolism , Gene Expression Profiling , Gene Silencing , HeLa Cells , Hindlimb/blood supply , Humans , Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/blood supply , Myocytes, Smooth Muscle/metabolism , Oligonucleotide Array Sequence Analysis , Oligonucleotides/genetics
13.
Anal Bioanal Chem ; 405(29): 9585-91, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24196123

ABSTRACT

Stem cell therapy has opened up the possibility of treating numerous degenerating diseases. However, we are still merely at the stage of identifying appropriate sources of stem cells and exploring their full differentiation potential. Thus, tracking the stem cells upon in vivo engraftment and during in vitro co-culture is very important and is an area of research embracing many pitfalls. 5-Ethynyl-2'-deoxyuridine (EdU), a rather new thymidine analog incorporated into DNA, has recently been suggested to be a novel highly valid alternative to other dyes for labeling of stem cells and subsequent tracing of their proliferation and differentiation ability. However, our results herein do not at any stage support this recommendation, since EdU severely reduces the viability of stem cells. Accordingly, we found that transplanted EdU-labeled stem cells hardly survive upon in vivo transplantation into regenerating muscle, whereas stem cells labeled in parallel with another dye survived very well and also participated in myofiber formation. Similar data were obtained upon in vitro myogenic culture, and further analysis showed that EdU reduced cell numbers by up to 88 % and increased the cell volume of remaining cells by as much as 91 %. Even at low EdU concentrations, cell survival and phenotype were substantially compromised, and the myogenic differentiation potential was inhibited. Since we examined both primary derived cells and cell lines from several species with the same result, this appears to be a common trait of EdU. We therefore suggest that EdU labeling should be avoided (or used with precaution) for stem cell tracing purposes.


Subject(s)
Cell Proliferation , Cell Tracking/methods , Deoxyuridine/analogs & derivatives , Staining and Labeling/methods , Stem Cells/chemistry , Stem Cells/cytology , Animals , Bromodeoxyuridine/chemistry , Cell Survival , Cell Tracking/instrumentation , Deoxyuridine/chemistry , Humans , Rats , Staining and Labeling/instrumentation
14.
Ann Surg ; 258(5): 743-51; discussion 752-3, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24096771

ABSTRACT

OBJECTIVES: To study the role of microRNAs in hypertension-induced vascular pathology before the onset of symptoms of severe cardiovascular disease. BACKGROUND: MicroRNAs play a crucial role in cardiovascular disease. However, microRNAs are often studied in full-blown cardiovascular disease models, not during development of cardiovascular pathology. METHODS: Angiotensin II was infused into healthy adult rats, inducing chronic hypertension, and microRNA expression profiles were obtained. The most prominently regulated microRNA, miR-487b, was further investigated, using primary cultures of rat aortic and human umbilical cord arterial cells. RESULTS: MiR-487b is predicted to target insulin receptor substrate 1 (IRS1). IRS1 plays an important role in both insulin signaling and cell proliferation and survival. IRS1 mRNA and protein levels were downregulated in aortae of hypertensive rats. MiR-487b binds directly to both rat and human IRS1 3'UTR and inhibits reporter gene expression in vitro. In primary rat and human arterial adventitial fibroblasts, inhibition of miR-487b leads to upregulation of IRS1 expression. Upregulation of miR-487b had the opposite effect, confirming direct targeting of IRS1 by miR-487b.Immunohistochemistry of aortic cross sections and rt/qPCR analyses of the separate aortic wall layers showed that both IRS1 and miR-487b were present mainly in the adventitia and less or not at all in the intima and tunica media. IRS1 expression in adventitial fibroblasts was predominantly nuclear and nuclear IRS1 is known to have antiapoptotic effects. Indeed, inhibition of miR-487b protected adventitial fibroblasts, and also medial smooth muscle cells, against serum starvation-induced apoptosis and increased cell survival. CONCLUSIONS: Angiotensin II-induced hypertension leads to upregulation of miR-487b, which targets IRS1. Via downregulation of IRS1, miR-487b can contribute to cell death and loss of adventitial and medial integrity during hypertension-induced vascular pathology.


Subject(s)
Aorta/metabolism , Hypertension/metabolism , Insulin Receptor Substrate Proteins/metabolism , MicroRNAs/metabolism , Angiotensin II/pharmacology , Animals , Aorta/pathology , Apoptosis , Blotting, Western , Cell Proliferation , Cell Survival , Female , Fibroblasts/metabolism , Glucose/metabolism , Humans , Hypertension/pathology , Immunohistochemistry , Luciferases/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Umbilical Cord/blood supply
15.
Adipocyte ; 2(4): 272-5, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24052905

ABSTRACT

Obesity occurs when an excessive dietary fat intake leads to expansion of adipose tissue, which mainly consists of adipocytes that arise from proliferating and differentiating adipose stem cells, the preadipocytes. Obesity is a consequence of both adipocyte hypertrophy and hyperplasia. Knowledge about preadipocyte differentiation is relatively well established, whereas the mechanism responsible for preadipocyte proliferation is incompletely understood and only in the early stage of comprehension. In this regard, we have recently identified that Delta-like 1 homolog (Dlk1) (also known as Preadipocyte factor 1 [Pref-1]) inhibits preadipocyte proliferation by regulating their entry into G1/S-phase. This novel disclosure, adding to the previous published data on Dlk1 repression of preadipocyte differentiation, has given us the chance to firmly place Dlk1 as a master regulator of preadipocyte homeostasis and adipose tissue expansion. Dlk1 manipulation may, therefore, open new perspectives in obesity treatments.

17.
Int J Mol Sci ; 14(6): 11190-207, 2013 May 27.
Article in English | MEDLINE | ID: mdl-23712358

ABSTRACT

MicroRNAs (miRNAs), a group of small non-coding RNAs that fine tune translation of multiple target mRNAs, are emerging as key regulators in cardiovascular development and disease. MiRNAs are involved in cardiac hypertrophy, heart failure and remodeling following cardiac infarction; however, miRNAs involved in hypertension have not been thoroughly investigated. We have recently reported that specific miRNAs play an integral role in Angiotensin II receptor (AT1R) signaling, especially after activation of the Gαq signaling pathway. Since AT1R blockers are widely used to treat hypertension, we undertook a detailed analysis of potential miRNAs involved in Angiotensin II (AngII) mediated hypertension in rats and hypertensive patients, using miRNA microarray and qPCR analysis. The miR-132 and miR-212 are highly increased in the heart, aortic wall and kidney of rats with hypertension (159 ± 12 mm Hg) and cardiac hypertrophy following chronic AngII infusion. In addition, activation of the endothelin receptor, another Gαq coupled receptor, also increased miR-132 and miR-212. We sought to extend these observations using human samples by reasoning that AT1R blockers may decrease miR-132 and miR-212. We analyzed tissue samples of mammary artery obtained from surplus arterial tissue after coronary bypass operations. Indeed, we found a decrease in expression levels of miR-132 and miR-212 in human arteries from bypass-operated patients treated with AT1R blockers, whereas treatment with ß-blockers had no effect. Taken together, these data suggest that miR-132 and miR-212 are involved in AngII induced hypertension, providing a new perspective in hypertensive disease mechanisms.


Subject(s)
Angiotensin II/pharmacology , Hypertension/genetics , MicroRNAs/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure/drug effects , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Disease Models, Animal , Endothelin-1 , Female , Fibrosis , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Organ Specificity/drug effects , Organ Specificity/genetics , Rats, Sprague-Dawley , Reproducibility of Results , Vasoconstrictor Agents
18.
PLoS One ; 8(3): e58890, 2013.
Article in English | MEDLINE | ID: mdl-23516570

ABSTRACT

BACKGROUND: Functional cross-talk between seven transmembrane (7TM) receptors can dramatically alter their pharmacological properties, both in vitro and in vivo. This represents an opportunity for the development of novel therapeutics that potentially target more specific biological effects while causing fewer adverse events. Although several studies convincingly have established the existence of 7TM receptor cross-talk, little is known about the frequencey and biological significance of this phenomenon. METHODOLOGY/PRINCIPAL FINDINGS: To evaluate the extent of synergism in 7TM receptor signaling, we took a comprehensive approach and co-expressed 123 different 7TM receptors together with the angiotensin II type 1 receptor (AT1R) and analyzed how each receptor affected the angiotensin II (AngII) response. To monitor the effect we used integrative receptor activation/signaling assay called Receptor Selection and Amplification Technology (R-SAT). In this screen the thromboxane A2α receptor (TPαR) was the only receptor which significantly enhanced the AngII-mediated response. The TPαR-mediated enhancement of AngII signaling was significantly reduced when a signaling deficient receptor mutant (TPαR R130V) was co-expressed instead of the wild-type TPαR, and was completely blocked both by TPαR antagonists and COX inhibitors inhibiting formation of thromboxane A2 (TXA2). CONCLUSIONS/SIGNIFICANCE: We found a functional enhancement of AT1R only when co-expressed with TPαR, but not with 122 other 7TM receptors. In addition, the TPαR must be functionally active, indicating the AT1R enhancement is mediated by a paracrine mechanism. Since we only found one receptor enhancing AT1R potency, our results suggest that functional augmentation through 7TM receptor cross-talk is a rare event that may require specific conditions to occur.


Subject(s)
Receptor, Angiotensin, Type 1/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Angiotensin II/pharmacology , Animals , Cattle , Cell Line , Gene Expression , Humans , Paracrine Communication/drug effects , Receptor Cross-Talk/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Renal Artery/drug effects , Renal Artery/physiology , Signal Transduction/drug effects , Vasoconstriction/drug effects
19.
PLoS One ; 8(2): e56280, 2013.
Article in English | MEDLINE | ID: mdl-23441172

ABSTRACT

The lining of the adult heart contains epicardial mesothelial cells (EMCs) that have the potential to undergo fibrogenic Epithelial-to-Mesenchymal Transition (EMT) during cardiac injury. EMT of EMCs has therefore been suggested to contribute to the heterogeneous fibroblast pool that mediates cardiac fibrosis. However, the molecular basis of this process is poorly understood. Recently, microRNAs (miRNAs) have been shown to regulate a number of sub-cellular events in cardiac disease. Hence, we hypothesized that miRNAs regulate fibrogenic EMT in the adult heart. Indeed pro-fibrogenic stimuli, especially TGF-ß, promoted EMT progression in EMC cultures, which resulted in differential expression of numerous miRNAs, especially the pleiotropic miR-21. Accordingly, ectopic expression of miR-21 substantially promoted the fibroblast-like phenotype arising from fibrogenic EMT, whereas an antagonist that targeted miR-21 blocked this effect, as assessed on the E-cadherin/α-smooth muscle actin balance, cell viability, matrix activity, and cell motility, thus making miR-21 a relevant target of EMC-derived fibrosis. Several mRNA targets of miR-21 was differentially regulated during fibrogenic EMT of EMCs and miR-21-dependent targeting of Programmed Cell Death 4 (PDCD4) and Sprouty Homolog 1 (SPRY1) significantly contributed to the development of a fibroblastoid phenotype. However, PDCD4- and SPRY1-targeting was not entirely ascribable to all phenotypic effects from miR-21, underscoring the pleiotropic biological role of miR-21 and the increasing number of recognized miR-21 targets.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Pericardium/metabolism , Pericardium/pathology , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cluster Analysis , Fibrosis/genetics , Gene Expression Profiling , Gene Expression Regulation , Humans , Membrane Proteins/metabolism , MicroRNAs/metabolism , Myocardium , Phenotype , Phosphoproteins/metabolism , Primary Cell Culture , RNA-Binding Proteins/metabolism
20.
J Mol Cell Cardiol ; 57: 72-81, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23352991

ABSTRACT

Metastasis-associated protein, S100A4 is suggested as a marker for fibrosis in several organs. It also modulates DNA binding of p53 and affects its function. However, the functional role of S100A4 in the myocardium has remained unclear. Therefore, we investigated the role of S100A4 and its relationship with p53 in cardiac fibrosis. In Dahl-rat hypertensive heart disease model, S100A4 was upregulated in the hypertrophic myocardium and further activated during transition to heart failure (HF). It was expressed in various cells including fibroblasts. In in vitro cardiac fibroblasts, the knockdown of S100A4 significantly suppressed both cell proliferation and collagen expressions. S100A4 co-localized and interacted with p53 in the nucleus. S100A4 knockdown increased the expression of p53-downstream genes, p21 and mdm2, and concomitant knockdown of p53 recovered cell proliferation and collagen expression. Transverse aortic constriction (TAC) was performed in S100A4 knockout (KO) mice, which showed a similar baseline-phenotype to wild type (WT) mice. Although there was no difference in hypertrophic response, KO mice showed reduced interstitial fibrosis, decreased myofibroblasts, and suppressed expressions of collagens and profibrotic cytokines in the left ventricle. Also, DNA microarray analysis showed that S100A4 knockout in vivo had a significant impact on expressions of p53-associated genes. These findings suggest that S100A4 modulates p53 function in fibroblasts and thereby mediates myocardial interstitial fibrosis through two distinct mechanisms; cell proliferation and collagen expression. Blockade of S100A4 may have therapeutic potential in cardiac hypertrophy and HF by attenuating cardiac fibrosis.


Subject(s)
Heart Failure/metabolism , Heart Ventricles/pathology , Myofibroblasts/metabolism , S100 Proteins/physiology , Tumor Suppressor Protein p53/metabolism , Angiotensin II/physiology , Animals , Cell Proliferation , Collagen/genetics , Collagen/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Fibrosis , Gene Expression Regulation , Gene Knockout Techniques , Heart Failure/pathology , Heart Ventricles/metabolism , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Male , Mice , Mice, Knockout , Myofibroblasts/physiology , NIH 3T3 Cells , Natriuretic Peptide, Brain/blood , Rats , Rats, Inbred Dahl , S100 Calcium-Binding Protein A4 , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...