Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Oral Biosci ; 66(2): 288-299, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38403241

ABSTRACT

BACKGROUND: Regenerative dentistry aims to enhance the structure and function of oral tissues and organs. Modern tissue engineering harnesses cell and gene-based therapies to advance traditional treatment approaches. Studies have demonstrated the potential of mesenchymal stem cells (MSCs) in regenerative dentistry, with some progressing to clinical trials. This review comprehensively examines animal studies that have utilized MSCs for various therapeutic applications. Additionally, it seeks to bridge the gap between related findings and the practical implementation of MSC therapies, offering insights into the challenges and translational aspects involved in transitioning from preclinical research to clinical applications. HIGHLIGHTS: To achieve this objective, we have focused on the protocols and achievements related to pulp-dentin, alveolar bone, and periodontal regeneration using dental-derived MSCs in both animal and clinical studies. Various types of MSCs, including dental-derived cells, bone-marrow stem cells, and umbilical cord stem cells, have been employed in root canals, periodontal defects, socket preservation, and sinus lift procedures. Results of such include significant hard tissue reconstruction, functional pulp regeneration, root elongation, periodontal ligament formation, and cementum deposition. However, cell-based treatments for tooth and periodontium regeneration are still in early stages. The increasing demand for stem cell therapies in personalized medicine underscores the need for scientists and responsible organizations to develop standardized treatment protocols that adhere to good manufacturing practices, ensuring high reproducibility, safety, and cost-efficiency. CONCLUSION: Cell therapy in regenerative dentistry represents a growing industry with substantial benefits and unique challenges as it strives to establish sustainable, long-term, and effective oral tissue regeneration solutions.


Subject(s)
Tissue Engineering , Humans , Animals , Tissue Engineering/methods , Regeneration/physiology , Regenerative Medicine/methods , Regenerative Medicine/trends , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cell Transplantation/methods , Dental Pulp/cytology , Dental Pulp/physiology , Dentistry/trends , Dentistry/methods
2.
Exp Cell Res ; 436(1): 113961, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38341080

ABSTRACT

Non-coding RNAs, particularly small Cajal-body associated RNAs (scaRNAs), play a significant role in spliceosomal RNA modifications. While their involvement in ischemic myocardium regeneration is known, their role in cardiac development is unexplored. We investigated scaRNA20's role in iPSC differentiation into cardiomyocytes (iCMCs) via overexpression and knockdown assays. We measured scaRNA20-OE-iCMCs and scaRNA20-KD-iCMCs contractility using Particle Image Velocimetry (PIV), comparing them to control iCMCs. We explored scaRNA20's impact on alternative splicing via pseudouridylation (Ψ) of snRNA U12, analyzing its functional consequences in cardiac differentiation. scaRNA20-OE-iPSC differentiation increased beating colonies, upregulated cardiac-specific genes, activated TP53 and STAT3, and preserved contractility under hypoxia. Conversely, scaRNA20-KD-iCMCs exhibited poor differentiation and contractility. STAT3 inhibition in scaRNA20-OE-iPSCs hindered cardiac differentiation. RNA immunoprecipitation revealed increased Ψ at the 28th uridine of U12 RNA in scaRNA20-OE iCMCs. U12-KD iCMCs had reduced cardiac differentiation, which improved upon U12 RNA introduction. In summary, scaRNA20-OE in iPSCs enhances cardiomyogenesis, preserves iCMC function under hypoxia, and may have implications for ischemic myocardium regeneration.


Subject(s)
RNA, Small Nuclear , RNA , Humans , RNA, Small Nuclear/genetics , Alternative Splicing , Hypoxia , Myocytes, Cardiac
3.
Cell Tissue Res ; 394(1): 189-207, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37572165

ABSTRACT

Cardiovascular diseases, atherosclerosis, and strokes are the most common causes of death in patients with Hutchinson-Gilford progeria syndrome (HGPS). The LMNA variant c.1824C > T accounts for ~ 90% of HGPS cases. The detailed molecular mechanisms of Lamin A in the heart remain elusive due to the lack of appropriate in vitro models. We hypothesize that HGPS patient's induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iCMCs) will provide a model platform to study the cardio-pathologic mechanisms associated with HGPS. To elucidate the effects of progerin in cardiomyocytes, we first obtained skin fibroblasts (SFs) from a de-identified HGPS patient (hPGP1, proband) and both parents from the Progeria Research Foundation. Through Sanger sequencing and restriction fragment length polymorphism, with the enzyme EciI, targeting Lamin A, we characterized hPGP1-SFs as heterozygous mutants for the LMNA variant c.1824 C > T. Additionally, we performed LMNA exon 11 bisulfite sequencing to analyze the methylation status of the progeria cells. Furthermore, we reprogrammed the three SFs into iPSCs and differentiated them into iCMCs, which gained a beating on day 7. Through particle image velocimetry analysis, we found that hPGP1-iCMCs had an irregular contractile function and decreased cardiac-specific gene and protein expressions by qRT-PCR and Western blot. Our progeria-patient-derived iCMCs were found to be functionally and structurally defective when compared to normal iCMCs. This in vitro model will help in elucidating the role of Lamin A in cardiac diseases and the cardio-pathologic mechanisms associated with progeria. It provides a new platform for researchers to study novel treatment approaches for progeria-associated cardiac diseases.


Subject(s)
Heart Diseases , Progeria , Humans , Progeria/genetics , Progeria/metabolism , Progeria/pathology , Lamin Type A/genetics , Lamin Type A/metabolism , Myocytes, Cardiac/metabolism , Cell Differentiation
4.
Methods Mol Biol ; 2454: 197-212, 2022.
Article in English | MEDLINE | ID: mdl-33774811

ABSTRACT

Studies have shown that human-induced pluripotent stem cells (iPSCs) derived cardiomyocytes (iCMCs) would provide a limitless source of cells for regenerative therapy and drug discoveries. Similar to embryonic stem cells, iPSCs have the capability to differentiate into mature functional iCMCs. The objective of our study is to develop an animal-free and viral-free approach by using a highly efficient transfection method that utilizes a critical combination of DNAs and mRNAs of pluripotent genes to generate iPSCs from adult human skin fibroblasts (SF). Subsequently differentiated them into functional cardiomyocytes. We obtained 4% of SFs into iPSCs at Passage 0, which shows significantly higher efficiency of reprogramming when compared to the use of either DNA alone or mRNAs alone. These iPSCs cultured under cardiac culture conditions are capable of differentiating into iCMCs. Furthermore, >88% of iCMCs are positive for either cardiac troponin T (TNNT2) or GATA binding protein 4 (GATA4). The iCMCs produced from SFs have been used in our laboratory to demonstrate their in vitro and in vivo functional potentials. In this study, we present step-by-step procedures for the generation of iPSCs from SFs and further differentiate them toward functional iCMCs.


Subject(s)
Induced Pluripotent Stem Cells , Adult , Cell Differentiation/genetics , Cells, Cultured , Fibroblasts , Humans , Myocytes, Cardiac , Skin
5.
Exp Biol Med (Maywood) ; 247(5): 433-445, 2022 03.
Article in English | MEDLINE | ID: mdl-34648374

ABSTRACT

The process of bone repair has always been a natural mystery. Although bones do repair themselves, supplemental treatment is required for the initiation of the self-regeneration process. Predominantly, surgical procedures are employed for bone regeneration. Recently, cell-based therapy for bone regeneration has proven to be more effective than traditional methods, as it eliminates the immune risk and painful surgeries. In clinical trials, various stem cells, especially mesenchymal stem cells, have shown to be more efficient for the treatment of several bone-related diseases, such as non-union fracture, osteogenesis imperfecta, osteosarcoma, and osteoporosis. Furthermore, the stem cells grown in a suitable three-dimensional scaffold support were found to be more efficient for osteogenesis. It has been shown that the three-dimensional bioscaffolds support and simulate an in vivo environment, which helps in differentiation of stem cells into bone cells. Bone regeneration in patients with bone disorders can be improved through modification of stem cells with several osteogenic factors or using stem cells as carriers for osteogenic factors. In this review, we focused on the various types of stem cells and scaffolds that are being used for bone regeneration. In addition, the molecular mechanisms of various transcription factors, signaling pathways that support bone regeneration and the senescence of the stem cells, which limits bone regeneration, have been discussed.


Subject(s)
Osteogenesis , Tissue Scaffolds , Bone Regeneration , Cell Differentiation , Cells, Cultured , Humans , Stem Cells/metabolism , Technology
6.
J Cell Mol Med ; 25(18): 8904-8919, 2021 09.
Article in English | MEDLINE | ID: mdl-34390186

ABSTRACT

Generation of induced pluripotent stem cells (iPSCs) and their differentiation into mesenchymal stem/stromal cells (iMSCs) have created exciting source of cells for autologous therapy. In this study, we have compared the therapeutic potential of iMSCs generated from urinary epithelial (UE) cells with the available umbilical cord MSCs (UC-MSCs). For this, adult UE cells were treated with the mRNA of pluripotent genes (OCT4, NANOG, SOX2, KLF4, MYC and LIN28) and a cocktail of miRNAs under specific culture conditions for generating iPSCs. Our non-viral and mRNA-based treatment regimen demonstrated a high reprogramming efficiency to about 30% at passage 0. These UE-iPSCs were successfully differentiated further into ectoderm, endoderm and mesoderm lineage of cells. Moreover, these UE-iPSCs were subsequently differentiated into iMSCs and were compared with the UC-MSCs. These iMSCs were capable of differentiating into osteocytes, chondrocytes and adipocytes. Our qRT-PCR and Western blot data showed that the CD73, CD90 and CD105 gene transcripts and proteins were highly expressed in iMSCs and UC-MSCs but not in other cells. The comparative qRT-PCR data showed that the iMSCs maintained their MSC characteristics without any chromosomal abnormalities even at later passages (P15), during which the UC-MSCs started losing their MSC characteristics. Importantly, the wound-healing property demonstrated through migration assay was superior in iMSCs when compared to the UC-MSCs. In this study, we have demonstrated an excellent non-invasive and pain-free method of obtaining iMSCs for regenerative therapy. These homogeneous autologous highly proliferative iMSCs may provide an alternative source of cells to UC-MSCs for treating various diseases.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Regenerative Medicine/methods , Umbilical Cord/cytology , Cell Differentiation , Cells, Cultured , Humans , Male , Middle Aged
7.
Exp Cell Res ; 400(1): 112508, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33549576

ABSTRACT

Noonan syndrome (NS) is a dominant autosomal genetic disorder, associated with mutations in several genes that exhibit multisystem abnormal development including cardiac defects. NS associated with the Son of Sevenless homolog 1 (SOS1) gene mutation attributes to the development of cardiomyopathy and congenital heart defects. Since the treatment option for NS is very limited, an in vitro disease model with SOS1 gene mutation would be beneficial for exploring therapeutic possibilities for NS. We reprogrammed cardiac fibroblasts obtained from a NS patient and normal control skin fibroblasts (C-SF) into induced pluripotent stem cells (iPSCs). We identified NS-iPSCs carry a heterozygous single nucleotide variation in the SOS1 gene at the c.1654A > G. Furthermore, the control and NS-iPSCs were differentiated into induced cardiomyocytes (iCMCs), and the electron microscopic analysis showed that the sarcomeres of the NS-iCMCs were highly disorganized. FACS analysis showed that 47.5% of the NS-iCMCs co-expressed GATA4 and cardiac troponin T proteins, and the mRNA expression levels of many cardiac related genes, studied by qRT-PCR array, were significantly reduced when compared to the control C-iCMCs. We report for the first time that NS-iPSCs carry a single nucleotide variation in the SOS1 gene at the c.1654A>G were showing significantly reduced cardiac genes and proteins expression as well as structurally and functionally compromised when compared to C-iCMCs. These iPSCs and iCMCs can be used as a modeling platform to unravel the pathologic mechanisms and also the development of novel drug for the cardiomyopathy in patients with NS.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Mutation , Myocytes, Cardiac/pathology , Noonan Syndrome/pathology , SOS1 Protein/genetics , Case-Control Studies , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Noonan Syndrome/genetics
8.
Curr Genomics ; 22(7): 526-540, 2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35386434

ABSTRACT

Aims: Perform in-silico analysis of human SOS1 mutations to elucidate their pathogenic role in Noonan syndrome (NS). Background: NS is an autosomal dominant genetic disorder caused by single nucleotide mutation in PTPN11, SOS1, RAF1, and KRAS genes. NS is thought to affect approximately 1 in 1000. NS patients suffer different pathogenic effects depending on the mutations they carry. Analysis of the mutations would be a promising predictor in identifying the pathogenic effect of NS. Methods: We performed computational analysis of the SOS1 gene to identify the pathogenic nonsynonymous single nucleotide polymorphisms (nsSNPs) th a t cause NS. SOS1 variants were retrieved from the SNP database (dbSNP) and analyzed by in-silico tools I-Mutant, iPTREESTAB, and MutPred to elucidate their structural and functional characteristics. Results: We found that 11 nsSNPs of SOS1 that were linked to NS. 3D modeling of the wild-type and the 11 nsSNPs of SOS1 showed that SOS1 interacts with cardiac proteins GATA4, TNNT2, and ACTN2. We also found that GRB2 and HRAS act as intermediate molecules between SOS1 and cardiac proteins. Our in-silico analysis findings were further validated using induced cardiomyocytes (iCMCs) derived from NS patients carrying SOS1 gene variant c.1654A>G (NSiCMCs) and compared to control human skin fibroblast-derived iCMCs (C-iCMCs). Our in vitro data confirmed that the SOS1, GRB2 and HRAS gene expressions as well as the activated ERK protein, were significantly decreased in NS-iCMCs when compared to C-iCMCs. Conclusion: This is the first in-silico and in vitro study demonstrating that 11 nsSNPs of SOS1 play deleterious pathogenic roles in causing NS.

9.
Front Cell Dev Biol ; 8: 674, 2020.
Article in English | MEDLINE | ID: mdl-32984301

ABSTRACT

The material properties of tissues and their mechanical state is an important factor in development, disease, regenerative medicine and tissue engineering. Here we describe a microrheological measurement technique utilizing aggregates of microinjected ferromagnetic nickel particles to probe the viscoelastic properties of embryonic tissues. Quail embryos were cultured in a plastic incubator chamber located at the center of two pairs of crossed electromagnets. We found a pronounced viscoelastic behavior within the ECM-rich region separating the mesoderm and endoderm in Hamburger Hamilton stage 10 quail embryos, consistent with a Zener (standard generalized solid) model. The viscoelastic response is about 45% of the total response, with a characteristic relaxation time of 1.3 s.

10.
Differentiation ; 112: 58-66, 2020.
Article in English | MEDLINE | ID: mdl-31954271

ABSTRACT

Despite significant effort devoted to developing new treatments and procedures, cardiac disease is still one of the leading causes of death in the world. The loss of myocytes due to ischemic injury remains a major therapeutic challenge. However, cell-based therapy to repair the injured heart has shown significant promise in basic and translation research and in clinical trials. Embryonic stem cells have been successfully used to improve cardiac outcomes. Unfortunately, treatment with these cells is complicated by ethical and legal issues. Recent progress in developing induced pluripotent stem cells (iPSCs) using non-viral vectors has made it possible to derive cardiomyocytes for therapy. This review will focus on these non-integration-based approaches for reprogramming and their therapeutic advantages for cardiovascular medicine.


Subject(s)
Cell- and Tissue-Based Therapy , Heart Diseases/therapy , Induced Pluripotent Stem Cells/transplantation , Myocytes, Cardiac/transplantation , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Genetic Vectors/therapeutic use , Humans , Regenerative Medicine/trends
11.
Cells ; 10(1)2020 12 31.
Article in English | MEDLINE | ID: mdl-33396426

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent cells which can proliferate and replace dead cells in the body. MSCs also secrete immunomodulatory molecules, creating a regenerative microenvironment that has an excellent potential for tissue regeneration. MSCs can be easily isolated and grown in vitro for various applications. For the past two decades, MSCs have been used in research, and many assays and tests have been developed proving that MSCs are an excellent cell source for therapy. This review focusses on quality control parameters required for applications of MSCs including colony formation, surface markers, differentiation potentials, and telomere length. Further, the specific mechanisms of action of MSCs under various conditions such as trans-differentiation, cell fusion, mitochondrial transfer, and secretion of extracellular vesicles are discussed. This review aims to underline the applications and benefits of MSCs in regenerative medicine and tissue engineering.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Regenerative Medicine/methods , Tissue Engineering/methods , Extracellular Vesicles/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism
12.
Prog Mol Biol Transl Sci ; 160: 1-22, 2018.
Article in English | MEDLINE | ID: mdl-30470288

ABSTRACT

Cell therapy has been identified as an effective method to regenerate damaged tissue. Adult stem cells, also known as somatic stem cells or resident stem cells, are a rare population of undifferentiated cells, located within a differentiated organ, in a specialized structure, called a niche, which maintains the microenvironments that regulate the growth and development of adult stem cells. The adult stem cells are self-renewing, clonogenic, and multipotent in nature, and their main role is to maintain the tissue homeostasis. They can be activated to proliferate and differentiate into the required type of cells, upon the loss of cells or injury to the tissue. Adult stem cells have been identified in many tissues including blood, intestine, skin, muscle, brain, and heart. Extensive preclinical and clinical studies have demonstrated the structural and functional regeneration capabilities of these adult stem cells, such as bone marrow-derived mononuclear cells, hematopoietic stem cells, mesenchymal stromal/stem cells, resident adult stem cells, induced pluripotent stem cells, and umbilical cord stem cells. In this review, we focus on the human therapies, utilizing adult stem cells for their regenerative capabilities in the treatment of cardiac, brain, pancreatic, and eye disorders.


Subject(s)
Adult Stem Cells/cytology , Regenerative Medicine , Stem Cell Transplantation , Disease , Humans , Neurogenesis
13.
Int J Biochem Cell Biol ; 102: 117-127, 2018 09.
Article in English | MEDLINE | ID: mdl-30010012

ABSTRACT

Acute lung injury (ALI) is a common complication of sepsis that often leads to fatal lung disease without effective therapies. It is known that bone marrow derived macrophages are important in resolving the inflammation and maintaining tissue homeostasis. Here, we hypothesize that treatment in combination of DNA methyl transferase inhibitor (DNMTi) 5-Aza 2-deoxycytidine (Aza) and histone deacetylase inhibitor (HDACi) Trichostatin A (TSA) mitigates the inflammation induced pyroptosis and apoptosis during endotoxemia induced ALI. To test this hypothesis, the mice challenged with a sublethal dose of LPS followed by one-hour post-treatment with a single dose of Aza and TSA intraperitoneally showed a substantial attenuation of apoptosis and inflammation. Importantly, we observed significant changes in the mitochondrial membrane structure, and lower levels of DNA fragmentation, reduced expression of apoptotic and pyroptotic genes both transcriptionally and translationally in LPS induced BMDMs treated by a combination of Aza and TSA than in LPS-induced BMDMs treated with either drug alone. The protection was mediated by an inhibition of JNK-ERK and STAT3-JMJD3 activated pathways. Thus, targeting these important signaling pathways with the combination of Aza and TSA would be a good treatment modality for ALI.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Endotoxemia/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Jumonji Domain-Containing Histone Demethylases/metabolism , Macrophages/cytology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Decitabine/pharmacology , Decitabine/therapeutic use , Drug Interactions , Endotoxemia/immunology , Endotoxemia/pathology , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Lung/drug effects , Lung/pathology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondrial Membranes/drug effects , Pyroptosis/drug effects
14.
Acta Pharmacol Sin ; 39(10): 1590-1603, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29620051

ABSTRACT

Induced pluripotent stem cell (iPSC)-based cardiac regenerative medicine requires the efficient generation, structural soundness and proper functioning of mature cardiomyocytes, derived from the patient's somatic cells. The most important functional property of cardiomyocytes is the ability to contract. Currently available methods routinely used to test and quantify cardiomyocyte function involve techniques that are labor-intensive, invasive, require sophisticated instruments or can adversely affect cell vitality. We recently developed optical flow imaging method analyses and quantified cardiomyocyte contractile kinetics from video microscopic recordings without compromising cell quality. Specifically, our automated particle image velocimetry (PIV) analysis of phase-contrast video images captured at a high frame rate yields statistical measures characterizing the beating frequency, amplitude, average waveform and beat-to-beat variations. Thus, it can be a powerful assessment tool to monitor cardiomyocyte quality and maturity. Here we demonstrate the ability of our analysis to characterize the chronotropic responses of human iPSC-derived cardiomyocytes to a panel of ion channel modulators and also to doxorubicin, a chemotherapy agent with known cardiotoxic side effects. We conclude that the PIV-derived beat patterns can identify the elongation or shortening of specific phases in the contractility cycle, and the obtained chronotropic responses are in accord with known clinical outcomes. Hence, this system can serve as a powerful tool to screen the new and currently available pharmacological compounds for cardiotoxic effects.


Subject(s)
Cardiotoxicity/diagnosis , Cardiovascular Agents/pharmacology , Drug Evaluation, Preclinical/methods , Heart Rate/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Cell Shape/drug effects , Cells, Cultured , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Flow Cytometry/methods , Humans , Induced Pluripotent Stem Cells/physiology , Models, Biological , Myocytes, Cardiac/physiology
15.
Trends Cardiovasc Med ; 28(2): 81-90, 2018 02.
Article in English | MEDLINE | ID: mdl-28869095

ABSTRACT

Small nucleolar RNAs (snoRNAs) are a group of noncoding RNAs that perform various biological functions, including biochemical modifications of other RNAs, precursors of miRNA, splicing, and telomerase activity. The small Cajal body-associated RNAs (scaRNAs) are a subset of the snoRNA family and collect in the Cajal body where they perform their canonical function to biochemically modify spliceosomal RNAs prior to maturation. Failure of sno/scaRNAs have been implicated in pathology such as congenital heart anomalies, neuromuscular disorders, and various malignancies. Thus, understanding of sno/scaRNAs demonstrates the clinical value.


Subject(s)
Coiled Bodies/metabolism , RNA, Small Nucleolar/metabolism , Active Transport, Cell Nucleus , Animals , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Small Nucleolar/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism , Telomerase/metabolism
16.
Acta Pharmacol Sin ; 39(4): 501-513, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29219950

ABSTRACT

Extracellular vesicles (EVs) comprise apoptotic bodies, microvesicles and exosomes, and they perform as key regulators in cell-to-cell communication in normal as well as diseased states. EVs contain natural cargo molecules, such as miRNA, mRNA and proteins, and transfer these functional cargos to neighboring cells or more distant cells through circulation. These functionally active molecules then affect distinct signaling cascades. The message conveyed to the recipient cells is dependent upon the composition of the EV, which is determined by the parent cell and the EV biogenesis. Because of their properties such as increased stability in circulation, biocompatibility, low immunogenicity and toxicity, EVs have drawn attention as attractive delivery systems for therapeutics. This review focuses on the functional use of exosomes in therapy and the potential advantages and challenges in using exosomes for therapeutic purposes.


Subject(s)
Exosomes/drug effects , Exosomes/physiology , Animals , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/pharmacology , Enzyme Inhibitors/pharmacology , Exosomes/chemistry , Exosomes/metabolism , Gene Transfer Techniques , Heart Diseases/physiopathology , Humans , Inflammation/physiopathology , Liver Diseases/physiopathology , Neoplasms/physiopathology , Neurodegenerative Diseases/physiopathology
17.
Sci Rep ; 7(1): 10404, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871207

ABSTRACT

Characterization of cardiomyocyte beat patterns is needed for quality control of cells intended for surgical injection as well as to establish phenotypes in disease modeling or toxicity studies. Optical-flow based analysis of videomicroscopic recordings offer a manipulation-free and efficient characterization of contractile cycles, an important characteristics of cardiomyocyte phenotype. We demonstrate that by appropriate computational analysis of optical flow data one can identify distinct contractile centers and distinguish active cell contractility from passive elastic tissue deformations. Our proposed convergence measure correlates with myosin IIa immuno-localization and is capable to resolve contractile waves and their synchronization within maturing, unlabeled induced pluripotent stem cell-derived cardiomyocyte cultures.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Nonmuscle Myosin Type IIA/metabolism , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Computational Biology/methods , Humans , Induced Pluripotent Stem Cells/metabolism , Microscopy, Video , Myocardial Contraction , Myocytes, Cardiac/metabolism
18.
Adv Exp Med Biol ; 998: 285-307, 2017.
Article in English | MEDLINE | ID: mdl-28936747

ABSTRACT

Cardiovascular diseases are the number one cause of death globally with an estimated 7.4 million people dying from coronary heart disease. Studies have been conducted to identify the therapeutic utility of exosomes in many diseases, including cardiovascular diseases. It has been demonstrated that exosomes are immune modulators, can be used to treat cardiac ischemic injury, pulmonary hypertension and many other diseases, including cancers. Exosomes can be used as a biomarker for disease and cell-free drug delivery system for targeting the cells. Many studies suggest that exosomes can be used as a cell-free vaccine for many diseases. In this chapter, we explore the possibility of future therapeutic potential of exosomes in various cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/surgery , Cell-Free System/transplantation , Exosomes/transplantation , Myocardium/pathology , Regeneration , Regenerative Medicine/trends , Animals , Biomarkers/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cell-Free System/metabolism , Cell-Free System/pathology , Diffusion of Innovation , Drug Carriers , Drug Delivery Systems/trends , Exosomes/metabolism , Exosomes/pathology , Forecasting , Genetic Therapy/trends , Humans , Myocardium/metabolism , Recovery of Function
19.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 518-528, 2017 02.
Article in English | MEDLINE | ID: mdl-27919711

ABSTRACT

Even though the discovery of the term 'epigenetics' was in the 1940s, it has recently become one of the most promising and expanding fields to unravel the gene expression pattern in several diseases. The most well studied example is cancer, but other diseases like metabolic disorders, autism, or inflammation-associated diseases such as lung injury, autoimmune disease, asthma, and type-2 diabetes display aberrant gene expression and epigenetic regulation during their occurrence. The change in the epigenetic pattern of a gene may also alter gene function because of a change in the DNA status. Constant environmental pressure, lifestyle, as well as food habits are the other important parameters responsible for transgenerational inheritance of epigenetic traits. Discovery of epigenetic modifiers targeting DNA methylation and histone deacetylation enzymes could be an alternative source to treat or manipulate the pathogenesis of diseases. Particularly, the combination of epigenetic drugs such as 5-aza-2-deoxycytidine (Aza) and trichostatin A (TSA) are well studied to reduce inflammation in an acute lung injury model. It is important to understand the epigenetic machinery and the function of its components in specific diseases to develop targeted epigenetic therapy. Moreover, it is equally critical to know the specific inhibitors other than the widely used pan inhibitors in clinical trials and explore their roles in regulating specific genes in a more defined way during infection.


Subject(s)
Communicable Diseases/genetics , Epigenesis, Genetic , Inflammation/genetics , Animals , Communicable Diseases/drug therapy , DNA Methylation/drug effects , Drug Discovery , Epigenesis, Genetic/drug effects , Histone Code/drug effects , Humans , Inflammation/drug therapy , MicroRNAs/genetics
20.
Drug Discov Today ; 22(1): 186-193, 2017 01.
Article in English | MEDLINE | ID: mdl-27554801

ABSTRACT

Macrophages are a heterogeneous population of phagocytic cells present in all tissues. Recently, several drugs that target the epigenetic machinery have emerged as attractive molecules for treating infection and inflammation by modulating macrophages. Treatment of lipopolysaccharide (LPS)-challenged macrophages with epigenetic modifiers leads to phenotype switching. This could provide stimulatory/destructive (M1) or suppressive/protective (M2) therapeutic strategies, which are crucial in the cytokine milieu in which the macrophages reside. In this review, we provide an overview of macrophage functional diversity during various diseases, including infection, as well as the current status in the development and clinical utility of epigenetic modifiers.


Subject(s)
Cardiovascular Diseases/drug therapy , Epigenesis, Genetic/drug effects , Infections/drug therapy , Macrophages/drug effects , Wounds and Injuries/drug therapy , Animals , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Cardiovascular Diseases/immunology , Decitabine , Drug Therapy, Combination , Histone Deacetylase Inhibitors/therapeutic use , Humans , Infections/immunology , Macrophages/immunology , Wounds and Injuries/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...