Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Int J Mol Sci ; 23(24)2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36555252

ABSTRACT

Rett syndrome (RTT) is a severe neurodevelopmental disorder caused by MeCP2 mutations. Nonetheless, the pathophysiological roles of MeCP2 mutations in the etiology of intrinsic cardiac abnormality and sudden death remain unclear. In this study, we performed a detailed functional studies (calcium and electrophysiological analysis) and RNA-sequencing-based transcriptome analysis of a pair of isogenic RTT female patient-specific induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs) that expressed either MeCP2wildtype or MeCP2mutant allele and iPSC-CMs from a non-affected female control. The observations were further confirmed by additional experiments, including Wnt signaling inhibitor treatment, siRNA-based gene silencing, and ion channel blockade. Compared with MeCP2wildtype and control iPSC-CMs, MeCP2mutant iPSC-CMs exhibited prolonged action potential and increased frequency of spontaneous early after polarization. RNA sequencing analysis revealed up-regulation of various Wnt family genes in MeCP2mutant iPSC-CMs. Treatment of MeCP2mutant iPSC-CMs with a Wnt inhibitor XAV939 significantly decreased the ß-catenin protein level and CACN1AC expression and ameliorated their abnormal electrophysiological properties. In summary, our data provide novel insight into the contribution of activation of the Wnt/ß-catenin signaling cascade to the cardiac abnormalities associated with MeCP2 mutations in RTT.


Subject(s)
Induced Pluripotent Stem Cells , Rett Syndrome , Humans , Female , Rett Syndrome/metabolism , Wnt Signaling Pathway , Myocytes, Cardiac/metabolism , Cell Line , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mutation
2.
J Environ Sci (China) ; 115: 443-452, 2022 May.
Article in English | MEDLINE | ID: mdl-34969472

ABSTRACT

The COVID-19 pandemic has raised awareness about various environmental issues, including PM2.5 pollution. Here, PM2.5 pollution during the COVID-19 lockdown was traced and analyzed to clarify the sources and factors influencing PM2.5 in Guangzhou, with an emphasis on heavy pollution. The lockdown led to large reductions in industrial and traffic emissions, which significantly reduced PM2.5 concentrations in Guangzhou. Interestingly, the trend of PM2.5 concentrations was not consistent with traffic and industrial emissions, as minimum concentrations were observed in the fourth period (3/01-3/31, 22.45 µg/m3) of the lockdown. However, the concentrations of other gaseous pollutants, e.g., SO2, NO2 and CO, were correlated with industrial and traffic emissions, and the lowest values were noticed in the second period (1/24-2/03) of the lockdown. Meteorological correlation analysis revealed that the decreased PM2.5 concentrations during COVID-19 can be mainly attributed to decreased industrial and traffic emissions rather than meteorological conditions. When meteorological factors were included in the PM2.5 composition and backward trajectory analyses, we found that long-distance transportation and secondary pollution offset the reduction of primary emissions in the second and third stages of the pandemic. Notably, industrial PM2.5 emissions from western, southern and southeastern Guangzhou play an important role in the formation of heavy pollution events. Our results not only verify the importance of controlling traffic and industrial emissions, but also provide targets for further improvements in PM2.5 pollution.


Subject(s)
Air Pollutants , Air Pollution , COVID-19 , Air Pollutants/analysis , Air Pollution/analysis , China/epidemiology , Communicable Disease Control , Environmental Monitoring , Humans , Pandemics , Particulate Matter/analysis , SARS-CoV-2
3.
Cells ; 10(9)2021 09 18.
Article in English | MEDLINE | ID: mdl-34572112

ABSTRACT

Pluripotent stem cells (PSCs) can undergo unlimited self-renewal and can differentiate into all the cell types present in our body, including cardiomyocytes. Therefore, PSCs can be an excellent source of cardiomyocytes for future regenerative medicine and medical research studies. However, cardiomyocytes obtained from PSC differentiation culture are regarded as immature structurally, electrophysiologically, metabolically, and functionally. Mitochondria are organelles responsible for various cellular functions such as energy metabolism, different catabolic and anabolic processes, calcium fluxes, and various signaling pathways. Cells can respond to cellular needs to increase the mitochondrial mass by mitochondrial biogenesis. On the other hand, cells can also degrade mitochondria through mitophagy. Mitochondria are also dynamic organelles that undergo continuous fusion and fission events. In this review, we aim to summarize previous findings on the changes of mitochondrial biogenesis, mitophagy, and mitochondrial dynamics during the maturation of cardiomyocytes. In addition, we intend to summarize whether changes in these processes would affect the maturation of cardiomyocytes. Lastly, we aim to discuss unanswered questions in the field and to provide insights for the possible strategies of enhancing the maturation of PSC-derived cardiomyocytes.


Subject(s)
Mitochondrial Dynamics , Mitophagy , Myocytes, Cardiac/pathology , Organelle Biogenesis , Animals , Humans
4.
Environ Sci Technol ; 54(15): 9519-9528, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32609501

ABSTRACT

Although the bioaccumulation of organophosphate flame retardants (OPFRs) in aquatic organisms has been investigated, little information is available about their bioaccumulation in mammals following chronic inhalation exposure. To address this knowledge gap, C57BL/6 mice were exposed to 7 PM2.5-associated OPFRs via the trachea to study their bioaccumulation, tissue distribution, and urinary metabolites. Low (corresponding to the real PM2.5 concentrations occurring during winter in Guangzhou), medium, and high dosages were examined. After 72 days' exposure, ∑OPFR concentrations in tissues from mice in the medium dosage group decreased in the order of intestine > heart > stomach > testis > kidney > spleen > brain > liver > lung > muscle. Of the OPFRs detected in all three exposure groups, chlorinated alkyl OPFRs were most heavily accumulated in mice. We found a significant positive correlation between the bioaccumulation ratio and octanol-air partition coefficient (KOA) in mice tissues for low log KOW OPFR congeners (log KOW ≤ 4, p < 0.05). Three urinary metabolites (di-p-cresyl phosphate: DCrP, diphenyl phosphate: DPhP, dibutyl phosphate: DnBP) were detected from the high dosage group. These results provide important insights into the bioaccumulation potential of OPFRs in mammals and emphasize the health risk of chlorinated alkyl OPFRs.


Subject(s)
Flame Retardants , Animals , Biomarkers , Environmental Exposure , Flame Retardants/analysis , Flame Retardants/toxicity , Male , Mice , Mice, Inbred C57BL , Organophosphates/analysis , Organophosphates/toxicity , Particulate Matter
5.
Cancers (Basel) ; 11(4)2019 Apr 18.
Article in English | MEDLINE | ID: mdl-31003514

ABSTRACT

Currently, there is no effective molecular-based therapy for triple-negative breast cancer (TNBC). Canonical transient receptor potential isoform 3 (TRPC3) was previously shown to be upregulated in breast cancer biopsy tissues when compared to normal breast tissues. However, the biological role of TRPC3 in breast cancer still remains to be elucidated. In this study, subcellular fractionation followed by Western blot and immunocytochemistry showed that TRPC3 was over-expressed on the plasma membrane of TNBC line MDA-MB-231 when compared to an estrogen receptor-positive cell line MCF-7. TRPC3 blocker Pyr3 and dominant negative of TRPC3 attenuated proliferation, induced apoptosis and sensitized cell death to chemotherapeutic agents in MDA-MB-231 as measured by proliferation assays. Interestingly, Ras GTPase-activating protein 4 (RASA4), a Ca2+-promoted Ras-MAPK pathway suppressor, was found to be located on the plasma membrane of MDA-MB-231. Blocking TRPC3 decreased the amount of RASA4 located on the plasma membrane, with concomitant activation of MAPK pathways. Our results suggest that, in TNBC MDA-MB-231 cells, Ca2+ influx through TRPC3 channel sustains the presence of RASA4 on the plasma membrane where it inhibits the Ras-MAPK pathway, leading to proliferation and apoptosis resistance. Our study reveals the novel TRPC3-RASA4-MAPK signaling cascade in TNBC cells and suggests that TRPC3 may be exploited as a potential therapeutic target for TNBC.

6.
J Vis Exp ; (143)2019 01 26.
Article in English | MEDLINE | ID: mdl-30741257

ABSTRACT

Cancer recurrence has long been studied by oncologists while the underlying mechanisms remain unclear. Recently, we and others found that a phenomenon named apoptosis reversal leads to increased tumorigenicity in various cell models under different stimuli. Previous studies have been focused on tracking this process in vitro and in vivo; however, the isolation of real reversed cells has yet to be achieved, which limits our understanding on the consequences of apoptosis reversal. Here, we take advantage of a Caspase-3/7 Green Detection dye to label cells with activated caspases after apoptotic induction. Cells with positive signals are further sorted out by fluorescence-activated cell sorting (FACS) for recovery. Morphological examination under confocal microscopy helps confirm the apoptotic status before FACS. An increase in tumorigenicity can often be attributed to the elevation in the percentage of cancer stem cell (CSC)-like cells. Also, given the heterogeneity of breast cancer, identifying the origin of these CSC-like cells would be critical to cancer treatment. Thus, we prepare breast non-stem cancer cells before triggering apoptosis, isolating caspase-activated cells and performing the apoptosis reversal procedure. Flow cytometry analysis reveals that breast CSC-like cells re-appear in the reversed group, indicating breast CSC-like cells are transited from breast non-stem cancer cells during apoptosis reversal. In summary, this protocol includes the isolation of apoptotic breast cancer cells and detection of changes in CSC percentage in reversed cells by flow cytometry.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Caspases/metabolism , Flow Cytometry/methods , Neoplastic Stem Cells/pathology , Breast Neoplasms/metabolism , Female , Humans , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured
7.
Ecotoxicol Environ Saf ; 168: 378-387, 2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30396134

ABSTRACT

A growing number of epidemiological surveys show that PM2.5 is an important promoter for the cardiovascular dysfunction induced by atmospheric pollution. PM2.5 is a complex mixture of solid and liquid airborne particles and its components determine the health risk of PM2.5to a great extent. However, the individual cardiotoxicities of different PM2.5 fractions are still unclear, especially in the cellular level. Here we used the neonatal rat cardiomyocytes (NRCMs) to evaluate the cardiac toxicity of PM2.5 exposure. The cytotoxicities of Total-PM2.5, water soluble components of PM2.5 (WS-PM2.5) and water insoluble components of PM2.5 (WIS-PM2.5), which include the cell viability, cell membrane damage, reactive oxygen species (ROS) generation, were examined with NRCMs in vitro. The results indicated that Total-PM2.5 or WIS-PM2.5 exposure significantly decreased the cell viability, induced the cell membrane damage and increased the ROS level in NRCMs at concentrations above 50 µg/mL. However, WS-PM2.5 exposure could induce the cytotoxicity on NRCMs until the concentration of WS-PM2.5 was raised to a higher concentration (75 µg/mL). Furthermore, the DNA damage was detected in NRCMs after 48 h of exposure with Total-PM2.5, WS-PM2.5 or WIS-PM2.5 (75 µg/mL) and the adverse effects on mitochondrial function and action potentials of NRCMs were detected only both in the Total-PM2.5 and WIS-PM2.5 treatment group. In summary, our project not only estimates the risk of PM2.5 on cardiac cells but also reveal that Total-PM2.5 and WIS-PM2.5 exposure were predominantly associated with the functional cardiotoxicities in NRCMs.


Subject(s)
Cardiotoxins/toxicity , Myocytes, Cardiac/drug effects , Particulate Matter/toxicity , Animals , Animals, Newborn , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Cells, Cultured , DNA Damage/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Rats , Reactive Oxygen Species/metabolism
8.
Neoplasia ; 20(3): 295-303, 2018 03.
Article in English | MEDLINE | ID: mdl-29476980

ABSTRACT

It has long been a puzzle in cancer treatment that despite the initial appearance of apoptosis, the process could be reversed in some cancer cells and often results in more aggressive tumors and metastasis. The mechanism for this recurrence is yet unknown. Here we report that human mammary carcinoma cells induced to undergo apoptosis could recover with increased tumorigenicity in vitro and in vivo, and induced lymph node metastasis. Specifically, the reversed cells underwent epithelial-to-mesenchymal transitions in the primary tumors in situ, and mesenchymal-to-epithelial transitions in the metastatic cells. Flow cytometry confirmed an elevated percentage of cells carrying cancer stem cells (CSCs) markers (CD44+/CD24-) in the reversed breast cancer cell population, with hypomethylated CD44 promoters and hypermethylated CD24 promoters. More importantly, CSCs were generated anew from non-stem cancer cells after apoptosis reversal possibly through epigenetic modifications. The results from this study can open doors to discovering more effective cancer treatments by suppressing apoptosis reversal.


Subject(s)
Apoptosis/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD24 Antigen/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Female , Humans , Hyaluronan Receptors/genetics , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology
9.
J Pharmacol Exp Ther ; 363(2): 211-220, 2017 11.
Article in English | MEDLINE | ID: mdl-28860353

ABSTRACT

Histone deacetylase (HDAC) inhibitors modulate acetylation/deacetylation of histone and nonhistone proteins. They have been widely used for cancer treatment. However, there have been only a few studies investigating the effect of HDAC inhibitors on vascular tone regulation, most of which employed chronic treatment with HDAC inhibitors. In the present study, we found that two hydroxamate-based pan-HDAC inhibitors, suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA), could partially but acutely relax high extracellular K+-contracted mouse aortas. SAHA and TSA also attenuated the high extracellular K+-induced cytosolic Ca2+ rise and inhibited L-type Ca2+ channel current in whole-cell patch-clamp. These data demonstrate that SAHA could inhibit L-type Ca2+ channels to cause vascular relaxation. In addition, SAHA and TSA dose dependently relaxed the arteries precontracted with phenylephrine. The relaxant effect of SAHA and TSA was greater in phenylephrine-precontracted arteries than in high K+-contracted arteries. Although part of the relaxant effect of SAHA and TSA on phenylephrine-precontracted arteries was related to L-type Ca2+ channels, both agents could also induce relaxation via a mechanism independent of L-type Ca2+ channels. Taken together, HDAC inhibitors SAHA and TSA can acutely relax blood vessels via their inhibitory action on L-type Ca2+ channels and via another L-type Ca2+ channel-independent mechanism.


Subject(s)
Aorta/drug effects , Aorta/physiology , Calcium Channels, L-Type/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Vasodilation/drug effects , Animals , Aorta/metabolism , Biological Transport/drug effects , Calcium/metabolism , Cytosol/drug effects , Cytosol/metabolism , Electrophysiological Phenomena/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Male , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Phenylephrine/pharmacology , Potassium/metabolism , Vorinostat
10.
PLoS One ; 10(7): e0133211, 2015.
Article in English | MEDLINE | ID: mdl-26208267

ABSTRACT

Cytosolic Ca2+ ([Ca2+]i) is an important signal that regulates cardiomyocyte differentiation during cardiogenesis. TRPV1 is a Ca2+-permeable channel that is expressed in cardiomyocytes. In the present study, we utilized mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) as a model to investigate the functional role of TRPV1 in cardiomyocyte differentiation. Induction of embryonic stem cells into cardiomyocytes was achieved using embryoid body (EB)-based differentiation method. Quantitative PCRs showed an increased TRPV1 expression during the differentiation process. In [Ca2+]i measurement study, application of TRPV1 agonists, capsaicin and camphor, elicited a [Ca2+]i rise in mESC-CMs, the effect of which was abolished by TRPV1-shRNA. In functional study, treatment of EBs with TRPV1 antagonists (capsazepine and SB366791) and TRPV1-shRNA reduced the size of the EBs and decreased the percentage of spontaneously beating EBs. TRPV1 antagonists and TRPV1-shRNA also suppressed the expression of cardiomyocyte marker genes, including cardiac actin, c-TnT, c-TnI, and α-MHC. Taken together, this study demonstrated an important functional role of TRPV1 channels in the differentiation of mESCs into cardiomyocytes.


Subject(s)
Cell Differentiation/genetics , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , TRPV Cation Channels/genetics , Animals , Biomarkers , Cells, Cultured , Gene Expression , Mice , RNA, Messenger/genetics , RNA, Small Interfering/genetics , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors
11.
J Cell Biochem ; 116(11): 2658-66, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25923408

ABSTRACT

In the past decade, miRNA emerges as a vital player in orchestrating gene regulation and maintaining cellular homeostasis. It is well documented that miRNA influences a variety of biological events, including embryogenesis, cell fate decision, and cellular differentiation. Adipogenesis is an organized process of cellular differentiation by which pre-adipocytes differentiate towards mature adipocytes. It has been shown that adipogenesis is tightly modulated by a number of transcription factors such as PPARγ, KLF4, and C/EBPα. However, the molecular mechanisms underlying the missing link between miRNA and adipogenesis-related transcription factors remain elusive. In this study, we unveiled that miR-25, a member of miR-106b-25 cluster, was remarkably downregulated during 3T3-L1 adipogenesis. Restored expression of miR-25 significantly impaired 3T3-L1 adipogenesis and downregulated the expression of serial adipogenesis-related genes. Further experiments presented that ectopic expression of miR-25 did not affect cell proliferation and cell cycle progression. Finally, KLF4 and C/EBPα, two key regulators of adipocyte differentiation, were experimentally identified as bona fide targets for miR-25. These data indicate that miR-25 is a novel negative regulator of adipocyte differentiation and it suppressed 3T3-L1 adipogenesis by targeting KLF4 and C/EBPα, which provides novel insights into the molecular mechanism of miRNA-mediated cellular differentiation.


Subject(s)
Adipogenesis , CCAAT-Enhancer-Binding Proteins/genetics , Kruppel-Like Transcription Factors/genetics , MicroRNAs/metabolism , 3T3-L1 Cells , Animals , Cell Differentiation , Gene Expression Regulation , Kruppel-Like Factor 4 , Mice , Promoter Regions, Genetic
12.
Proc Natl Acad Sci U S A ; 109(33): 13428-33, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22847428

ABSTRACT

The cell nucleus is a major site for polyglutamine (polyQ) toxicity, but the underlying mechanisms involved have yet been fully elucidated. Here, we report that mutant RNAs that carry an expanded CAG repeat (expanded CAG RNAs) induce apoptosis by activating the nucleolar stress pathway in both polyQ patients and transgenic animal disease models. We showed that expanded CAG RNAs interacted directly with nucleolin (NCL), a protein that regulates rRNA transcription. Such RNA-protein interaction deprived NCL of binding to upstream control element (UCE) of the rRNA promoter, which resulted in UCE DNA hypermethylation and subsequently perturbation of rRNA transcription. The down-regulation of rRNA transcription induced nucleolar stress and provoked apoptosis by promoting physical interaction between ribosomal proteins and MDM2. Consequently, p53 protein was found to be stabilized in cells and became concentrated in the mitochondria. Finally, we showed that mitochondrial p53 disrupted the interaction between the antiapoptotic protein, Bcl-xL, and the proapoptotic protein, Bak, which then caused cytochrome c release and caspase activation. Our work provides in vivo evidence that expanded CAG RNAs trigger nucleolar stress and induce apoptosis via p53 and describes a polyQ pathogenic mechanism that involves the nucleolus.


Subject(s)
Cell Nucleolus/genetics , Neurodegenerative Diseases/genetics , Peptides/metabolism , Stress, Physiological/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Caspases/metabolism , Cytochromes c/metabolism , DNA Methylation/genetics , Enzyme Activation , Humans , Mice , Mitochondria/genetics , Models, Biological , Phosphoproteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Polymerase I/metabolism , RNA Stability/genetics , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA-Binding Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Nucleolin
13.
Circ Res ; 111(3): e55-66, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22679141

ABSTRACT

RATIONALE: The expression of osteocalcin is augmented in human atherosclerotic lesions. How osteocalcin triggers vascular pathogenesis and remodeling is unclear. OBJECTIVE: To investigate whether osteocalcin promotes transformation of adventitial fibroblast to myofibroblasts and the molecular mechanism involved. METHODS AND RESULTS: Immunohistochemistry indicated that osteocalcin was expressed in the neointima of renal arteries from diabetic patients. Western blotting and wound-healing assay showed that osteocalcin induced fibroblast transformation and migration, which were attenuated by blockers of the renin-angiotensin system and protein kinase Cδ (PKCδ), toll-like receptor 4 (TLR4) neutralizing antibody, and antagonist and inhibitors of free radical production and cyclooxygenase-2. Small interfering RNA silencing of TLR4 and PKCδ abolished fibroblast transformation. Angiotensin II level in the conditioned medium from the osteocalcin-treated fibroblasts was found elevated using enzyme immunoassay. Culturing of fibroblasts in conditioned medium collected from differentiated osteoblasts promoted fibroblast transformation. The expression of fibronectin, TLR4, and cyclooxygenase-2 is augmented in human mesenteric arteries after 5-day in vitro exposure to osteocalcin. CONCLUSIONS: Osteocalcin transforms adventitial fibroblasts to myofibroblasts through stimulating angiotensin II release and subsequent activation of PKCδ/TLR4/reactive oxygen species/cyclooxygenase-2 signaling cascade. This study reveals that the skeletal hormone osteocalcin cross-talks with vascular system and contributes to vascular remodeling.


Subject(s)
Angiotensin II/metabolism , Cytoskeleton/metabolism , Fibroblasts/metabolism , Myofibroblasts/metabolism , Osteocalcin/physiology , Toll-Like Receptor 4/physiology , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Bone and Bones/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Cells, Cultured , Cyclooxygenase 2/physiology , Cytoskeleton/enzymology , Cytoskeleton/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Fibroblasts/cytology , Fibroblasts/enzymology , Humans , Myofibroblasts/cytology , Myofibroblasts/enzymology , Rats , Signal Transduction/physiology
14.
J Control Release ; 157(3): 321-30, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-21893122

ABSTRACT

Wound therapy remains a clinical challenge and much effort has been focused on the development of novel therapeutic approaches for wound management. New knowledge about the way in which signals control wound cellular and molecular behavior has promoted the topical application of multipotent stem cells and bioactive molecules to injured tissue, for skin regeneration with less scar formation. However, limited clinical success indicates that the effective delivery of polypeptides and therapeutic cells, with controlled releasing profile, is a major challenge which is yet to be overcome. Recently, a technique in which the genetically-manipulated stem cells were used both as the therapeutic agents and the vehicle for gene delivery for wound treatment - a method which serves to provide regenerative cells and bioactive genes within an optimal environment of regulatory molecular expression for wound sites - has emerged as a promising strategy for wound regenerative therapy. In this article, the roles of adult stem cells - as the therapeutics and the vehicles in these advanced biomimetic drug delivery systems for wound regeneration medicine - are scrutinized to indicate their mechanisms, characteristics, broad applicability and future lines of investigation.


Subject(s)
Adult Stem Cells , Genetic Therapy/methods , Regeneration , Wound Healing , Animals , Biomimetic Materials , Drug Delivery Systems , Gene Transfer Techniques , Humans
15.
J Cell Physiol ; 227(6): 2519-30, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21898397

ABSTRACT

Embryonic stem cells (ESCs) can self-renew indefinitely and differentiate into all cell lineages. Calcium is a universal second messenger which regulates a number of cellular pathways. Previous studies showed that store-operated calcium channels (SOCCs) but not voltage-operated calcium channels are present in mouse ESCs (mESCs). In this study, store-operated calcium entry (SOCE) was found to exist in mESCs using confocal microscopy. SOCC blockers lanthanum, 2-aminoethoxydiphenyl borate (2-APB) and SKF-96365 reduced mESC proliferation in a concentration-dependent manner, suggesting that SOCE is important for ESC proliferation. Pluripotent markers, Sox-2, Klf-4, and Nanog, were down-regulated by 2-APB, suggesting that self-renewal property of mESCs relies on SOCE. 17ß-estradiol (E2) enhanced mESC proliferation. This enhanced proliferation was associated with an increment of SOCE. Both stimulated proliferation and increased SOCE could be reversed by SOCC blockers suggesting that E2 mediates its stimulatory effect on proliferation via enhancing SOCE. Also, cyclosporin A and INCA-6, inhibitors of calcineurin [phosphatase that de-phosphorylates and activates nuclear factor of activated T-cells (NFAT)], reversed the proliferative effect of E2, indicating that NFAT is involved in E2-stimulated proliferation. Interestingly, E2 caused the nuclear translocation of NFATc4, and this could be reversed by 2-APB. These results suggested that NFATc4 is the downstream target of E2-induced SOCE. The present investigation provides the first line of evidence that SOCE and NFAT are crucial for ESCs to maintain their unique characteristics. In addition, the present investigation also provides novel information on the mechanisms of how E2, an important female sex hormone, affects ESC proliferation.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Cell Proliferation , Embryonic Stem Cells/metabolism , Estradiol/metabolism , NFATC Transcription Factors/metabolism , Pluripotent Stem Cells/metabolism , Animals , Biomarkers/metabolism , Calcineurin/metabolism , Calcineurin Inhibitors , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Signaling/drug effects , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/pharmacology , Homeodomain Proteins/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Microscopy, Confocal , NFATC Transcription Factors/genetics , Nanog Homeobox Protein , Pluripotent Stem Cells/drug effects , Recombinant Fusion Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Time Factors , Transfection
16.
J Pain ; 12(5): 600-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21296029

ABSTRACT

UNLABELLED: Irritable bowel syndrome (IBS), characterized mainly by abdominal pain, is a functional bowel disorder. The present study aimed to examine changes in the excitability and the activity of the voltage-gated K(+) channel in dorsal root ganglia (DRG) neurons innervating the colon of rats subjected to neonatal maternal separation (NMS). Colonic DRG neurons from NMS rats as identified by FAST DiI™ labeling showed an increased cell size compared with those from nonhandled (NH) rats. Whole cell current-clamp recordings showed that colonic DRG neurons from NMS rats displayed: 1) depolarized resting membrane potential; 2) increased input resistance; 3) a dramatic reduction in rheobase; and 4) a significant increase in the number of action potentials evoked at twice rheobase. Whole cell voltage-clamp recordings revealed that neurons from both groups exhibited transient A-type (I(A)) and delayed rectifier (I(K)) K(+) currents. Compared with NH rat neurons, the averaged density of I(K) was significantly reduced in NMS rat neurons. Furthermore, the Kv1.2 expression was significantly decreased in NMS rat colonic DRG neurons. These results suggest that NMS increases the excitability of colonic DRG neurons mainly by suppressing the I(K) current, which is likely accounted for by the downregulation of the Kv1.2 expression and somal hypertrophy. PERSPECTIVE: This study demonstrates the alteration of delayed rectifier K current and Kv1.2 expression in DRG neurons from IBS model rats, representing a molecular mechanism underlying visceral pain and sensitization in IBS, suggesting the potential of Kv1.2 as a therapeutic target for the treatment of IBS.


Subject(s)
Action Potentials/physiology , Colon/innervation , Ganglia, Spinal/physiology , Maternal Deprivation , Neurons/physiology , Potassium Channels, Voltage-Gated/metabolism , Animals , Colon/metabolism , Down-Regulation , Fluorescent Antibody Technique , Ganglia, Spinal/cytology , Male , Membrane Potentials/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
17.
Pflugers Arch ; 461(1): 191-202, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21110038

ABSTRACT

Embryonic stem cells (ESCs) can uniquely proliferate indefinitely and differentiate into all cell lineages. ESCs may therefore provide an unlimited supply of cells for cell-based therapies. Previous study reported the presence of hyperpolarization-activated inward currents in undifferentiated mouse (m) ESCs, but the functional role of this hyperpolarization-activated current in mESCs is unknown. In this study, the role of this current in maintaining the proliferative capacity and the cell cycle progression of ESCs was investigated. In D3 mESCs, this hyperpolarization-activated inward current can be blocked by HCN channel blocker ZD7288. Application of the HCN channel blockers, cesium (1-10 mM) or ZD7288 (0.1-30 µM), attenuated cell proliferation in a concentration-dependent manner. Both HCN blockers were found to be non-cytotoxic to mESCs as determined by cell viability test. Interestingly, ZD7288 at 10 and 30 µM was found to decrease the proportion of cells in G(0)/G(1) phase and increase the proportion of cells in S phase. This suggests that this hyperpolarization-activated current can affect the cell cycle progression in mESCs. In summary, the present investigation suggests that ESC proliferation and cell cycle progression can be regulated by this hyperpolarization-activated current.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Embryonic Stem Cells/cytology , Animals , Cell Survival/drug effects , Cesium/pharmacology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Cyclin B/biosynthesis , Embryonic Stem Cells/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membrane Potentials , Mice , Patch-Clamp Techniques , Pyrimidines/pharmacology
18.
J Cell Physiol ; 224(1): 165-77, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20333647

ABSTRACT

Embryonic stem cells (ESCs) possess two unique characteristics: self-renewal and pluripotency. In this study, roles of voltage-gated potassium channels (K(v)) in maintaining mouse (m) ESC characteristics were investigated. Tetraethylammonium (TEA(+)), a K(v) blocker, attenuated cell proliferation in a concentration-dependent manner. Possible reasons for this attenuation, including cytotoxicity, cell cycle arrest and differentiation, were examined. Blocking K(v) did not change the viability of mESCs. Interestingly, K(v) inhibition increased the proportion of cells in G(0)/G(1) phase and decreased that in S phase. This change in cell cycle distribution can be attributed to cell cycle arrest or differentiation. Loss of pluripotency as determined at both molecular and functional levels was detected in mESCs with K(v) blockade, indicating that K(v) inhibition in undifferentiated mESCs directs cells to differentiate instead of to self-renew and progress through the cell cycle. Membrane potential measurement revealed that K(v) blockade led to depolarization, consistent with the role of K(v) as the key determinant of membrane potential. The present results suggest that membrane potential changes may act as a "switch" for ESCs to decide whether to proliferate or to differentiate: hyperpolarization at G(1) phase would favor ESCs to enter S phase while depolarization would favor ESCs to differentiate. Consistent with this notion, S-phase-synchronized mESCs were found to be more hyperpolarized than G(0)/G(1)-phase-synchronized mESCs. Moreover, when mESCs differentiated, the differentiation derivatives depolarized at the initial stage of differentiation. This investigation is the first study to provide evidence that K(v) and membrane potential affect the fate determination of ESCs.


Subject(s)
Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Animals , Biomarkers/metabolism , Cell Cycle , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Cell Survival , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Gene Expression Regulation, Developmental , Membrane Potentials , Mice , Pluripotent Stem Cells/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Tetraethylammonium/pharmacology , Time Factors
19.
Biochem Biophys Res Commun ; 377(1): 46-51, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18823947

ABSTRACT

Gap junctions, encoded by the connexin (Cx) multi-gene family, couple adjacent cells and underlie cell-cell communications. Previous mouse studies suggest that Cxs play an important role in development but their role in human cardiogenesis is undefined. Human embryonic stem cells (hESC) provide a unique model for studying human differentiation. Lentivirus-mediated stable overexpression of Cx43 in hESC (Cx43-hESC) did not affect colony morphology, karyotype and expression of pluripotency genes such as Oct4 but completely suppressed the formation of spontaneously beating, cardiomyocyte-containing clusters in embryoid bodies (EBs). Unlike control hEBs, the transcripts of several mesodermal markers (kallikrein, delta-globin, and CMP), ventricular myosin light chain and cardiac troponin I were absent or delayed. Transcriptomic and pathway analyses showed that 194 genes crucial for movement, growth, differentiation and maintenance were differentially expressed in Cx43-hESC. We conclude that Cx43 mediates the expression of an array of genes involved in human cardiogenesis, in addition to intercellular communication.


Subject(s)
Connexin 43/metabolism , Embryonic Stem Cells/physiology , Gap Junctions/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Organogenesis/genetics , Pluripotent Stem Cells/physiology , Animals , Cell Communication/genetics , Cell Differentiation/genetics , Cell Line , Connexin 43/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Rats , Transcription, Genetic
20.
Mol Nutr Food Res ; 52(8): 950-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18496813

ABSTRACT

Previous reports demonstrated that hypocholesterolemic activity of apple was associated with its pectin and fiber. This report was to investigate the effect of apple polyphenols (AP) on blood cholesterol level and gene expression of cholesterol-regulating enzymes in Golden Syrian hamsters maintained on a 0.1% cholesterol diet. It was found that dietary supplementation of 0.3 or 0.6% of AP did not affect plasma total cholesterol (TC), but it increased HDL cholesterol (HDL-C) and decreased non-HDL-C, thus leading to a lower ratio of non-HDL-C to HDL-C. Plasma total triacylglycerol (TG) level was also significantly reduced when hamsters were fed a diet supplemented with 0.6% AP. Western blot analysis did not find any effect of AP on sterol regulatory element-binding protein 2 (SREBP-2), LDL receptor (LDLR), 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR), and cholesterol-7alpha-hydroxylase (CYP7A). Most interesting was that supplementation of AP had no effect on protein abundance of plasma cholesteryl ester transport protein (CETP), but it suppressed plasma CETP activity. A series of in vitro assays confirmed that AP inhibited CETP in a dose dependent manner. It was concluded that AP favorably improved distribution of cholesterol in lipoproteins, most likely, by its inhibition on CETP activity.


Subject(s)
Cholesterol Ester Transfer Proteins/blood , Cholesterol, HDL/blood , Cholesterol/blood , Flavonoids/pharmacology , Malus/chemistry , Phenols/pharmacology , Animals , Blotting, Western , Cricetinae , Eating/drug effects , Feces/chemistry , Hydroxymethylglutaryl CoA Reductases/analysis , Male , Mesocricetus , Organ Size/drug effects , Polyphenols , Receptors, LDL/analysis , Sterol Regulatory Element Binding Protein 2/analysis , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...