Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 11991, 2024 05 25.
Article in English | MEDLINE | ID: mdl-38796487

ABSTRACT

Physiochemical tissue inducers and mechanical stimulation are both efficient variables in cartilage tissue fabrication and regeneration. In the presence of biomolecules, decellularized extracellular matrix (ECM) may trigger and enhance stem cell proliferation and differentiation. Here, we investigated the controlled release of transforming growth factor beta (TGF-ß1) as an active mediator of mesenchymal stromal cells (MSCs) in a biocompatible scaffold and mechanical stimulation for cartilage tissue engineering. ECM-derived hydrogel with TGF-ß1-loaded alginate-based microspheres (MSs) was created to promote human MSC chondrogenic development. Ex vivo explants and a complicated multiaxial loading bioreactor replicated the physiological conditions. Hydrogels with/without MSs and TGF-ß1 were highly cytocompatible. MSCs in ECM-derived hydrogel containing TGF-ß1/MSs showed comparable chondrogenic gene expression levels as those hydrogels with TGF-ß1 added in culture media or those without TGF-ß1. However, constructs with TGF-ß1 directly added within the hydrogel had inferior properties under unloaded conditions. The ECM-derived hydrogel group including TGF-ß1/MSs under loading circumstances formed better cartilage matrix in an ex vivo osteochondral defect than control settings. This study demonstrates that controlled local delivery of TGF-ß1 using MSs and mechanical loading is essential for neocartilage formation by MSCs and that further optimization is needed to prevent MSC differentiation towards hypertrophy.


Subject(s)
Alginates , Bioreactors , Chondrogenesis , Hydrogels , Mesenchymal Stem Cells , Microspheres , Tissue Engineering , Alginates/chemistry , Tissue Engineering/methods , Humans , Hydrogels/chemistry , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Animals , Cartilage/metabolism , Cartilage/cytology , Tissue Scaffolds/chemistry , Decellularized Extracellular Matrix/chemistry , Transforming Growth Factor beta1/metabolism , Cell Differentiation , Cells, Cultured , Transforming Growth Factor beta/metabolism , Extracellular Matrix/metabolism
2.
Int J Biol Macromol ; 242(Pt 1): 124602, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37141963

ABSTRACT

In this study, the effect of alumina nanowire on the physical and biological properties of polyhydroxybutyrate-keratin (PHB-K) electrospun scaffold was investigated. First, PHB-K/alumina nanowire nanocomposite scaffolds were made with an optimal concentration of 3 wt% alumina nanowire by using the electrospinning method. The samples were examined in terms of morphology, porosity, tensile strength, contact angle, biodegradability, bioactivity, cell viability, ALP activity, mineralization ability, and gene expression. The nanocomposite scaffold provided a porosity of >80 % and a tensile strength of about 6.72 MPa, which were noticeable for an electrospun scaffold. AFM images showed an increase in surface roughness with the presence of alumina nanowires. This led to an improvement in the degradation rate and bioactivity of PHB-K/alumina nanowire scaffolds. The viability of mesenchymal cells, alkaline phosphatase secretion, and mineralization significantly increased with the presence of alumina nanowire compared to PHB and PHB-K scaffolds. In addition, the expression level of collagen I, osteocalcin, and RUNX2 genes in nanocomposite scaffolds increased significantly compared to other groups. In general, this nanocomposite scaffold could be a novel and interesting construct for osteogenic induction in bone tissue engineering.


Subject(s)
Nanocomposites , Tissue Scaffolds , Osteogenesis , Tissue Engineering/methods , Bone Regeneration , Aluminum Oxide/pharmacology , Keratins/pharmacology , Polyesters/pharmacology , Cell Differentiation
3.
Differentiation ; 123: 9-17, 2022.
Article in English | MEDLINE | ID: mdl-34864442

ABSTRACT

Germ cell production from stem cells allows for studying the mechanisms involved in gamete development with the aim of helping infertile couples with the generation of healthy gametes. In this context, improving the protocols for in-vitro germ cell induction from stem cells is very important. Recently, SB4 small molecule has been introduced as a potent agonist for bone morphogenic protein 4 (BMP4). Herein, we investigated whether BMP4, is replaceable by SB4 for having affordable protocol for in vitro germ cell differentiation. We demonstrated that SB4 can induce Blimp1 (as the first gene induced germ line differentiation) expression significantly but at a lower level compared to BMP4. However, Tfap2c (a putative downstream target of Blimp1 during germ cell differentiation) expression level in SB4-induced aggregates was significantly higher than in BMP4-induced aggregates. Moreover, co-presence of both BMP4 and SB4 could increase the expression level of Prdm14, Nnose3 and Stella (Dppa3), and thereby improve establishment of the germ cell fate during in-vitro differentiation of embryonic stem cells. In summary, our data suggest that SB4 could improve germ line gene expression pattern induced by BMP4 during embryonic stem cells in-vitro differentiation.


Subject(s)
Embryonic Stem Cells , Germ Cells , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/genetics , Gene Expression
4.
Chem Biol Interact ; 351: 109687, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34653396

ABSTRACT

Because spermatogonia transmit genetic information across generations, their DNA must be protected from environmental damages, including exposure to zinc oxide nanoparticles (ZnO NPs), which are frequently used in modern technology. Here, we used an in vitro system enriched for spermatogonia and exposed them to 10 and 20 µg/ml ZnO NPs for one/seven days. We did not detect any significant cell death, chromosomal instability, or DNA fragmentation in the spermatogonia treated with the ZnO NPs following one-day treatment with 10 or 20 µg/ml ZnO NPs. However, ZnO NPs (both 10 and 20 µg/ml) induced chromosomal instability in the spermatogonia after seven days of treatment. Moreover, one-day exposure to these NPs induced reactive oxygen species (ROS) generation and upregulation of apoptotic pathway-related genes p53, Caspase3 and Il6, as an inflammatory factor. Taken together, our study provides preliminary evidence for possible damages induced by low concentrations of ZnO NPs in spermatogonia. We should pay increased attention when using these NPs because of the silent damages in spermatogonia that can be transmitted to the next generation and cause severe effects. However, more data and validation of these results are required to determine the extent of this concern.


Subject(s)
Metal Nanoparticles/toxicity , Spermatogonia/drug effects , Zinc Oxide/toxicity , Animals , CDC2 Protein Kinase/metabolism , Caspase 3/metabolism , Chromosomal Instability/drug effects , Interleukin-6/metabolism , Interleukin-8/metabolism , Male , Mice , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
5.
Basic Clin Neurosci ; 12(3): 315-323, 2021.
Article in English | MEDLINE | ID: mdl-34917291

ABSTRACT

INTRODUCTION: Cholinergic-associated diseases currently constitute a significant cause of neurological and neurodegenerative disabilities. As the drugs are not efficient in improving the suffered tissues, stem cell treatment is considered an effective strategy for substituting the lost cells. METHODS: In the current study, we set out to investigate the differentiation properties of human Adipose-Derived Mesenchymal Stem Cells (AD-MSCs) into cholinergic-like cells by two morphogens of Retinoic Acid (RA) and Sonic Hedgehog (Shh) using a three-step in vitro procedure. The results were evaluated using real-time PCR, flow cytometry, and immunocytochemistry for two weeks. RESULTS: Our data showed that the cells could express cholinergic specific markers, including Islet-1, Acetylcholinesterase (AChE), SMI-32, and Nestin, at mRNA and protein levels. We could also quantitatively evaluate the expression of Islet-1, AChE, and Nestin at 14 days post-induction using flow cytometry. CONCLUSION: Human AD-MSCs are potent cells to differentiate into cholinergic-like cells in the presence of RA and Shh through a three-step protocol. Thus, they could be a suitable cell candidate for the regeneration of cholinergic-associated diseases. However, more functional and electrophysiological analyses are needed in this regard.

6.
Cell Tissue Bank ; 22(2): 207-223, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33387152

ABSTRACT

Psychiatric disorders such as schizophrenia can generate distress and disability along with heavy costs on individuals and health care systems. Different genetic and environmental factors play a pivotal role in the appearance of the mentioned disorders. Since the conventional treatment options for psychiatric disorders are suboptimal, investigators are trying to find novel strategies. Herein, stem cell therapies have been recommended as novel choices. In this context, the preclinical examination of stem cell-based therapies specifically using appropriate models can facilitate passing strong filters and serious examination to ensure proper quality and safety of them as a novel treatment approach. Animal models cannot be adequately helpful to follow pathophysiological features. Nowadays, stem cell-based models, particularly induced pluripotent stem cells reflected as suitable alternative models in this field. Accordingly, the importance of stem cell-based models, especially to experiment with the regenerative medicine outcomes for schizophrenia as one of the severe typing of psychiatric disorders, is addressed here.


Subject(s)
Induced Pluripotent Stem Cells , Schizophrenia , Animals , Humans , Regenerative Medicine , Schizophrenia/therapy , Stem Cell Transplantation
7.
Adv Exp Med Biol ; 1326: 159-186, 2021.
Article in English | MEDLINE | ID: mdl-32926346

ABSTRACT

Rheumatoid arthritis as a common autoimmune inflammatory disorder with unknown etiology can affect 0.5-1% of adults in developed countries. It involves more than just the patient's joints and can be accompanied by several comorbidities and affect cardiovascular, pulmonary, and some other systems of the human body. Although cytokine-mediated pathways are mentioned to have a central role in RA pathogenesis, adaptive and innate immune systems and intracellular signaling pathways all have important roles in this process. Non-steroidal anti-inflammatory drugs, glucocorticoids, conventional disease-modifying anti-rheumatic drugs, and biological agents are some mentioned medications used for RA. They are accompanied by some adverse effects and treatment failures which elucidates the needing for novel and more powerful therapeutic approaches. Stem cell-based therapies and their beneficial effects on therapeutic processes of different diseases have been founded so far. They can be an alternative and promising therapeutic approach for RA, too; due to their effects on immune responses of the disease. This review, besides some explanations about RA characteristics, addresses the outcome of the stem cell-based therapies including mesenchymal stem cell transplantation and hematopoietic stem cell transplantation for RA and explains their effects on the disease improvement.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , Mesenchymal Stem Cell Transplantation , Arthritis, Rheumatoid/drug therapy , Humans , Immunity
8.
Hum Gene Ther ; 32(21-22): 1330-1345, 2021 11.
Article in English | MEDLINE | ID: mdl-33307949

ABSTRACT

Breast cancer is a heterogeneous disease, which is the consequence of several genetic and environmental factors. Also, it is one of the most common causes of cancer death and second leading cancer among women all around the world. Therefore, it is necessary to develop novel therapeutic approaches useful for the successful treatment of breast cancer. As conventional treatments had limited success, alternative approaches for the treatment of breast cancer have been applied in recent years. Hence, the molecular basis of breast cancer has provided the opportunity of using genetic materials for therapeutic uses. In this regard, gene therapy as one of the potentially efficient and beneficial treatments among various techniques became a popular treatment for different cancers, especially breast cancer. Accordingly, there are plenty of targets available for gene therapy of breast cancer. Gene therapy strategies have the potential to correct molecular defects that contributed to the cancer progression. These techniques should selectively target tumor cells without affecting normal cells. Moreover, data of clinical trials in gene therapy for breast cancer indicated that this approach has little toxicity compared to other therapeutic approaches. In this study, different aspects of breast neoplasm, gene therapy techniques, challenges, and recent developments will be mentioned.


Subject(s)
Breast Neoplasms , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Female , Genetic Therapy , Humans
9.
Cell Regen ; 9(1): 20, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33258056

ABSTRACT

Systemic sclerosis is a rare chronic autoimmune disease with extensive microvascular injury, damage of endothelial cells, activation of immune responses, and progression of tissue fibrosis in the skin and various internal organs. According to epidemiological data, women's populations are more susceptible to systemic sclerosis than men. Until now, various therapeutic options are employed to manage the symptoms of the disease. Since stem cell-based treatments have developed as a novel approach to rescue from several autoimmune diseases, it seems that stem cells, especially mesenchymal stem cells as a powerful regenerative tool can also be advantageous for systemic sclerosis treatment via their remarkable properties including immunomodulatory and anti-fibrotic effects. Accordingly, we discuss the contemporary status and future perspectives of mesenchymal stem cell transplantation for systemic sclerosis.

10.
Int J Biochem Cell Biol ; 127: 105822, 2020 10.
Article in English | MEDLINE | ID: mdl-32771442

ABSTRACT

Extensive application of zinc oxide (ZnO) nanoparticles (NPs) in everyday life results in increased exposure to these NPs. Spermatogonial stem cells (SSCs) guarantee sperm production throughout the male reproductive life by providing a balance between self-renewal and differentiation. We used an in vitro platform to investigate the ZnO NPs effects on SSCs. We successfully synthesized ZnO NPs. In order to investigate these NPs, we isolated SSCs from mouse testes and cultured them in vitro. Our results confirmed the uptake of ZnO NPs by the cultured SSCs. We observed a dose- and time-dependent decrease in SSC viability. Both spherical and nanosheet ZnO NPs had the same cytotoxic effects on the SSCs, irrespective of their shapes. Moreover, we have shown that short time (one day) exposure of SSCs to a low concentration of ZnO NPs (10 µg/mL) promoted expressions of specific genes (Plzf, Gfr α1 and Bcl6b) for SSC self-renewal and differentiation genes (Vasa, Dazl, C-kit and Sycp3) expressed by spermatogonia during spermatogenesis. Our study provides the first insight into ZnO NPs function in SSCs and suggests a new function for ZnO NPs in the male reproductive system. We demonstrated that ZnO NPs might promote spermatogenesis via upregulation of gene expression related to SSC self-renewal and differentiation at low concentrations. Additional research should clarify the possible effect of ZnO NPs on the SSC genome and its effects on human SSCs.


Subject(s)
Nanoparticles/administration & dosage , Spermatogenesis/drug effects , Spermatogenesis/genetics , Spermatogonia/drug effects , Stem Cells/drug effects , Zinc Oxide/administration & dosage , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cells, Cultured , Gene Expression/drug effects , Male , Mice , Nanoparticles/chemistry , Spermatogonia/cytology , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Time Factors , Zinc Oxide/chemistry
11.
Cell Tissue Bank ; 21(3): 405-425, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32588163

ABSTRACT

Acute respiratory infections as one of the most common problems of healthcare systems also can be considered as an important reason for worldwide morbidity and mortality from infectious diseases. Coronaviruses are a group of well-known respiratory viruses that can cause acute respiratory infections. At the current state, the 2019 novel coronavirus is cited as the most worldwide problematic agent for the respiratory system. According to investigations, people with old age and underlying diseases are at higher risk of 2019 novel coronavirus infection. Indeed, they may show a severe form of the disease (with severe acute respiratory infections). Based on the promising role of cell therapy and regenerative medicine approaches in the treatment of several life-threatening diseases, it seems that applying cell-based approaches can also be a hopeful strategy for improving subjects with severe acute respiratory infections caused by the 2019 novel coronavirus. Herein, due to the amazing effects of mesenchymal stem cells in the treatment of various diseases, this review focuses on the auxiliary role of mesenchymal stem cells to reduce inflammatory processes of acute respiratory infections caused by the 2019 novel coronavirus.


Subject(s)
Coronavirus Infections/therapy , Inflammation/therapy , Mesenchymal Stem Cells , Pneumonia, Viral/therapy , Regeneration , COVID-19 , Coronavirus Infections/complications , Humans , Inflammation/etiology , Pandemics , Pneumonia, Viral/complications , Regenerative Medicine/methods
12.
Mater Sci Eng C Mater Biol Appl ; 109: 110603, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32228891

ABSTRACT

Cardiovascular diseases (CVD) are a major cause of mortality worldwide. Accessibility to heart tissue is limited due to sampling issues and lack of appropriate culture conditions. In addition, animal models are not an ideal choice for physiological, pharmacological, and fundamental evaluations in the cardiovascular field due to interspecies differences. Hence, there is an inevitable need for functional in vitro cardiac models. In this study, we have synthesized a novel electroconductive scaffold comprised of cardiac extracellular matrix (ECM) derived pre-cardiogel (pCG) blended with polypyrrole (Ppy). Our data revealed that 2.5% (w/v) pyrrole (Py) had the highest possible Py ratio that provided pCG-Ppy gel formation. The prepared mixture was fabricated into a scaffold by using the freeze-dried method. The scaffolds had open interconnected pores that ranged from 55 ± 24 µm for the cardiogel (CG)-Ppy to 74 ± 26 µm for the CG scaffolds, with no alterations in vital ECM components of collagen, polysaccharides, and glycosaminoglycans (GAGs). Incorporation of Ppy increased the CG stiffness with a final complex modulus from 80 pa to 140 pa. The CG-Ppy group had significantly greater electrical conductivity than the CG group. Scaffolds supported neonatal mouse cardiomyocyte (NMCM) adhesion, viability, cardiac-specific gene expression, and spontaneous beating up to 14 days after seeding. Among the fabricated hydrogels, the CG-Ppy group resulted in the synchronous beating of cardiomyocyte clusters and upregulation of cardiac genes involved in cardiac muscle contraction (cardiac troponin T [cTNT]) and cardiomyocyte electrical coupling (connexin 43 [Cx43]). Thus, this ECM-based electro-conductive scaffold might provide a promising substrate for constructing in vitro cardiac models for drug testing, disease modeling, developmental studies, and cardiac regenerative approaches.


Subject(s)
Electric Conductivity , Extracellular Matrix/chemistry , Myocardial Contraction , Myocardium/chemistry , Myocytes, Cardiac/metabolism , Tissue Scaffolds/chemistry , Animals , Cell Survival , Mice , Myocytes, Cardiac/cytology , Sheep
13.
J Biomed Mater Res A ; 108(4): 938-946, 2020 04.
Article in English | MEDLINE | ID: mdl-31894891

ABSTRACT

Cartilage tissue engineering is the interdisciplinary science that will help to improve cartilage afflictions, such as arthrosis, arthritis, or following joints traumatic injuries. In the present work, we developed an injectable hydrogel which derived from decellularized extracellular matrix of sheep cartilage. Successful decellularization was evaluated by measuring the DNA, glycosaminoglycans (GAG), collagen contents, and histological analyses. There was a minor difference in GAG and collagen contents among natural cartilage and decellularized tissue as well as ultimate hydrogel. Rheological analysis showed that the temperature and gelation time of prepared hydrogel were 37°C and between 5 and 7 min, respectively. Mechanical properties evaluation indicated a storage modulus of 20 kPa. The results show that prepared hydrogel possessed cell-friendly microenvironment as confirmed via calcein staining and MTT assay. Also, cells were able to proliferate which observed by H&E and alcian blue staining. Cell attachment and proliferation at the surface of the decellularized hydrogel was apparent by Scanning Electron Microscope (SEM) images and microphotographs. Furthermore, the cells embedded within the hydrogel were able to differentiate into chondrocyte with limited evidence of hypertrophy and osteogenesis in utilized cells which proved by SOX9, CoL2, ACAN, and also CoL1 and CoL10 gene expression levels. In summary, the results suggest that developed novel injectable hydrogel from decellularized cartilage could be utilized as a promising substrate for cartilage tissue engineering applications.


Subject(s)
Cartilage, Articular/physiology , Extracellular Matrix/metabolism , Hydrogels/pharmacology , Knee Joint/physiology , Regeneration/drug effects , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/ultrastructure , Rabbits , Sheep
14.
Hum Reprod ; 34(12): 2430-2442, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31886487

ABSTRACT

STUDY QUESTION: Could small molecules (SM) which target (or modify) signaling pathways lead to increased proliferation of undifferentiated spermatogonia following chemotherapy? SUMMARY ANSWER: Inhibition of transforming growth factor-beta (TGFb) signaling by SM can enhance the proliferation of undifferentiated spermatogonia and spermatogenesis recovery following chemotherapy. WHAT IS KNOWN ALREADY: Spermatogonial stem cells (SSCs) hold great promise for fertility preservation in prepubertal boys diagnosed with cancer. However, the low number of SSCs limits their clinical applications. SM are chemically synthesized molecules that diffuse across the cell membrane to specifically target proteins involved in signaling pathways, and studies have reported their ability to increase the proliferation or differentiation of germ cells. STUDY DESIGN, SIZE, DURATION: In our experimental study, spermatogonia were collected from four brain-dead individuals and used for SM screening in vitro. For in vivo assessments, busulfan-treated mice were treated with the selected SM (or vehicle, the control) and assayed after 2 (three mice per group) and 5 weeks (two mice per group). PARTICIPANTS/MATERIALS, SETTING, METHODS: We investigated the effect of six SM on the proliferation of human undifferentiated spermatogonia in vitro using a top-bottom approach for screening. We used histological, hormonal and gene-expression analyses to assess the effect of selected SM on mouse spermatogenesis. All experiments were performed at least in triplicate and were statistically evaluated by Student's t-test and/or one-way ANOVA followed by Scheffe's or Tukey's post-hoc. MAIN RESULTS AND THE ROLE OF CHANCE: We found that administration of SB431542, as a specific inhibitor of the TGFb1 receptor (TGFbR1), leads to a two-fold increase in mouse and human undifferentiated spermatogonia proliferation. Furthermore, injection of SB to busulfan-treated mice accelerated spermatogenesis recovery as revealed by increased testicular size, weight and serum level of inhibin B. Moreover, SB administration accelerated both the onset and completion of spermatogenesis. We demonstrated that SB promotes proliferation in testicular tissue by regulating the cyclin-dependent kinase (CDK) inhibitors 4Ebp1 and P57 (proliferation inhibitor genes) and up-regulating Cdc25a and Cdk4 (cell cycle promoting genes). LIMITATIONS, REASONS FOR CAUTION: The availability of human testis was the main limitation in this study. WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to report acceleration of spermatogenesis recovery following chemotherapy by administration of a single SM. Our findings suggest that SB is a promising SM and should be assessed in future clinical trials for preservation of fertility in men diagnosed with cancer or in certain infertility cases (e.g. oligospermia). STUDY FUNDING/COMPETING INTEREST(S): This study was supported by Royan Institute and National Institute for Medical Research Development (NIMAD, grant no 963337) granted to H.B. The authors have no conflict of interest to report.


Subject(s)
Benzamides/pharmacology , Dioxoles/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Spermatogenesis/drug effects , Spermatogonia/drug effects , Adolescent , Adult , Animals , Female , Fertility Preservation , Humans , Male , Mice , Primary Cell Culture , Spermatogonia/cytology
15.
Syst Biol Reprod Med ; 65(6): 409-419, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31675245

ABSTRACT

Interest in the role of male factor in infertility continues to mount with defects related to sperm movement considered as one of the more severe forms of subfertility. The peroxisome proliferator-activated receptor gamma (PPARγ) primarily regulates the expression of target genes involved in energy control as well as lipid and glucose metabolism. Although the pivotal roles of these receptors on female fertility have been reported, there are limited studies addressing PPARs role(s) in the male. This study was designed to determine and compare PPARα, PPARß and PPARγ mRNA expression in sperm cells of normozoospermic and asthenozoospermic men. In addition, flow cytometric analyses, immunofluorescence and western blot were used to evaluate PPARγ protein levels in spermatozoa. We have compared the sperm PPARs mRNA relative expression in 27 normozoospermic and 28 asthenozoospermic samples and monitored sperm PPARγ protein levels in 39 normozoospermic and 40 asthenozoospermic samples using flow cytometry. We have also assessed in a sub-group of seven normozoospermic and eight asthenozoospermic samples, PPARγ protein levels by western blotting. Relative expression of PPARγ mRNA in normozoospermic men was found to be significantly higher (P = 0.004) than in asthenozoospermic men while PPARα and PPARß relative expression was similar in the two groups. Likewise, PPARγ showed a positive correlation with motility (r = 0.34; P < 0.05), sperm concentration (r = 0.33) and the percentage of progressive motile spermatozoa (r = 0.31). In agreement with the mRNA behavior, sperm PPARγ protein levels as measured by flow cytometry (P = 0.066) and western blot (P = 0.089) showed a tendency to be higher in normozoospermic than asthenozoospermic men. The present study proposes a link between PPARγ gene expression level and motility in human sperm.Abbreviations: PPARs: Peroxisome Proliferator-Activated Receptors; CASA: Computer Assisted Semen Analysis; TFA: Trans Fatty Acids; HTF: Human Tubal Fluid; PBS: Phosphate-Buffered Saline; PPP: Pentose Phosphate Pathway; PI3K: Phosphoinositide 3-Kinase; G6PDH: Glucose 6-Phosphate Dehydrogenase.


Subject(s)
Asthenozoospermia/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Spermatozoa/metabolism , Adult , Humans , Male
16.
Biofabrication ; 12(1): 015021, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31658444

ABSTRACT

Mesenchymal stem cells (MSCs) are considered primary candidates for treating complex bone defects in cell-based therapy and tissue engineering. Compared with monolayer cultures, spheroid cultures of MSCs (mesenspheres) are favorable due to their increased potential for differentiation, extracellular matrix (ECM) synthesis, paracrine activity, and in vivo engraftment. Here, we present a strategy for the incorporation of microparticles for the fabrication of osteogenic micro-tissues from mesenspheres in a cost-effective and scalable manner. A facile method was developed to synthesize mineral microparticles with cell-sized spherical shape, biphasic calcium phosphate composition (hydroxyapatite and ß-tricalcium phosphate), and a microporous structure. Calcium phosphate microparticles (CMPs) were incorporated within the mesenspheres through mixing with the single cells during cell aggregation. Interestingly, the osteogenic genes were upregulated significantly (collagen type 1 (Col 1) 30-fold, osteopontin (OPN) 10-fold, and osteocalcin (OCN) 3-fold) after 14 days of culture with the incorporated CMPs, while no significant upregulation was observed with the incorporation of gelatin microparticles. The porous structure of the CMPs was exploited for loading and sustained release of an angiogenic small molecule. Dimethyloxaloylglycine (DMOG) was loaded efficiently onto the CMPs (loading efficiency: 65.32 ± 6%) and showed a sustained release profile over 12 days. Upon incorporation of the DMOG-loaded CMPs (DCMPs) within the mesenspheres, a similar osteogenic differentiation and an upregulation in angiogenic genes (VEGF 5-fold and kinase insert domain (KDR) 2-fold) were observed after 14 days of culture. These trends were also observed in immunostaining analysis. To evaluate scalable production of the osteogenic micro-tissues, the incorporation of microparticles was performed during cell aggregation in a spinner flask. The DCMPs were efficiently incorporated and directed the mesenspheres toward osteogenesis and angiogenesis. Finally, the DCMP mesenspheres were loaded within a three-dimensional printed cell trapper and transplanted into a critical-sized defect in a rat model. Computed tomography and histological analysis showed significant bone formation with blood vessel reconstruction after 8 weeks in this group. Taken together, we provide a scalable and cost-effective approach for fabrication of osteogenic micro-tissues, as building blocks of macro-tissues, that can address the large amounts of cells required for cell-based therapies.


Subject(s)
Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Animals , Bioprinting/economics , Cell Proliferation , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/metabolism , Osteocalcin/metabolism , Osteogenesis , Rats , Rats, Wistar , Tissue Engineering/economics , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry , Tissue Scaffolds/economics
17.
J Biomed Mater Res A ; 107(9): 2040-2052, 2019 09.
Article in English | MEDLINE | ID: mdl-31077544

ABSTRACT

Large bone defects treatment is one of the challenges in current bone tissue engineering approaches. Various strategies have been proposed to address this issue, among which, prevascularization by coculturing of angiogenic and osteogenic cells on the scaffolds can alleviate this problem. In the present study, modified fibrous scaffolds were prepared by electrospinning and subsequent ultrasonication of polycaprolactone (PCL) containing nano-hydroxyapatite (n-HA), with/without nano-zinc oxide (n-ZnO), and polyethylene oxide [PEO] as a sacrificial agent. The physical, mechanical, and chemical characteristics of the scaffolds were evaluated. The results showed the presence of n-ZnO, which in turn increased Young's module of the scaffolds from 5.5 ± 0.67 to 6.7 ± 1.77 MPa. Moreover, MTT, SEM, alkaline phosphatase (ALP) activity, chicken embryo chorioallantoic membrane (CAM) assay, and real-time RT-PCR were utilized to investigate the biocompatibility, cell adhesion and infiltration, osteoconductivity, angiogenic properties, and expression of osteogenic and angiogenic related genes. ALP assay showed that the highest enzyme activity was noted when the modified scaffolds containing n-ZnO were seeded with HUVEC:hBMSC at the cell ratio of 1:5. CAM assay showed induction of angiogenesis for the scaffolds containing n-ZnO. Real-time RT-PCR results showed significant upregulation of angiogenic related genes. Thus, the scaffolds containing n-ZnO may have great potential for osteogenesis and angiogenesis in tissue engineering applications.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Materials Testing , Mesenchymal Stem Cells/metabolism , Nanoparticles/chemistry , Neovascularization, Physiologic , Osteogenesis , Polyesters/chemistry , Tissue Scaffolds/chemistry , Zinc Oxide/chemistry , Animals , Chick Embryo , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology
18.
J Biomed Mater Res B Appl Biomater ; 107(1): 50-64, 2019 01.
Article in English | MEDLINE | ID: mdl-29468802

ABSTRACT

Critical-sized bone defects constitute a major health issue in orthopedics and usually cause mal-unions due to an inadequate number of migrated progenitor cells into the defect site or their incomplete differentiation into osteogenic precursor cells. The current study aimed to develop an optimized osteoinductive and angiogenic scaffold by incorporation of strontium (Sr) and bioglass (BG) into gelatin/nano-hydroxyapatite (G/nHAp) seeded with bone marrow mesenchymal stem cells to enhance bone regeneration. The scaffolds were fabricated by a freeze-drying technique and characterized in terms of morphology, structure, porosity and degradation rate. The effect of fabricated scaffolds on cell viability, attachment and differentiation into osteoblastic lineages was evaluated under in vitro condition. Micro computed tomography scan, histological and histomorphometric analysis were performed after implantation of scaffolds into the radial bone defects in rat. RT-PCR analysis showed that G/nHAp/BG/Sr scaffold significantly increased the expression level of osteogenic and angiogenic markers in comparison to other groups (P < 0.05). Moreover, the defects treated with the BMSCs-seeded scaffolds showed superior bone formation and mechanical properties compared to the cell-free scaffolds 4 and 12 weeks post-implantation. Finally, the BMSCs-seeded G/nHAp/BG/Sr scaffold showed the greatest bone regenerative capacity which was more similar to autograft. It is concluded that combination of Sr, BG, and nHAp can synergistically enhance the bone regeneration process. In addition, our results demonstrated that the BMSCs have the potential to considerably increase the bone regeneration ability of osteoinductive scaffolds. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 107B: 50-64, 2019.


Subject(s)
Bone Marrow Cells/metabolism , Bone Regeneration , Cells, Immobilized , Durapatite/chemistry , Glass/chemistry , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Nanostructures/chemistry , Radius , Strontium/chemistry , Allografts , Animals , Bone Marrow Cells/pathology , Cells, Immobilized/metabolism , Cells, Immobilized/pathology , Cells, Immobilized/transplantation , Male , Mesenchymal Stem Cells/pathology , Radius/injuries , Radius/metabolism , Radius/pathology , Rats , Rats, Wistar
19.
J Mater Sci Mater Med ; 30(1): 1, 2018 Dec 18.
Article in English | MEDLINE | ID: mdl-30564959

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α), a well-studied angiogenesis pathway, plays an essential role in angiogenesis-osteogenesis coupling. Targeting the HIF-1a pathway frequently leads to successful reconstruction of large-sized bone defects through promotion of angiogenesis. Dimethyloxalylglycine (DMOG) small molecule regulates the stability of HIF-1α at normal oxygen tension by mimicking hypoxia, which subsequently accelerates angiogenesis. The current study aims to develop a novel construct by seeding adipose derived mesenchymal stem cells (ADMSCs) onto a scaffold that contains DMOG to induce angiogenesis and regeneration of a critical size calvarial defect in a rat model. The spongy scaffolds have been synthesized in the presence and absence of DMOG and analyzed in terms of morphology, porosity, pore size, mechanical properties and DMOG release profile. The effect of DMOG delivery on cellular behaviors of adhesion, viability, osteogenic differentiation, and angiogenesis were subsequently evaluated under in vitro conditions. Histological analysis of cell-scaffold constructs were also performed following transplantation into the calvarial defect. Physical characteristics of fabricated scaffolds confirmed higher mechanical strength and surface roughness of DMOG-loaded scaffolds. Scanning electron microscopy (SEM) images and MTT assay demonstrated the attachment and viability of ADMSCs in the presence of DMOG, respectively. Osteogenic activity of ADMSCs that included alkaline phosphatase (ALP) activity and calcium deposition significantly increased in the DMOG-loaded scaffold. Computed tomography (CT) imaging combined with histomorphometry and immunohistochemistry analysis showed enhanced bone formation and angiogenesis in the DMOG-loaded scaffolds. Therefore, spongy scaffolds that contained DMOG and had angiogenesis ability could be utilized to enhance bone regeneration of large-sized bone defects.


Subject(s)
Alginic Acid/chemistry , Amino Acids, Dicarboxylic/pharmacology , Bone Development , Calcium Phosphates/chemistry , Gelatin/chemistry , Tissue Scaffolds , Amino Acids, Dicarboxylic/administration & dosage , Animals , Biocompatible Materials , Bone and Bones/injuries , Cell Adhesion/drug effects , Cell Survival , Drug Liberation , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Male , Mesenchymal Stem Cells , Microscopy, Electron, Scanning , Neovascularization, Physiologic , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
Biomaterials ; 154: 99-112, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29127860

ABSTRACT

Bioengineering of whole hearts using human embryonic stem cells (hESCs)-derived cardiovascular progenitor cells (CPCs) and natural matrices is a promising approach to overcome organ donor shortage threatening millions of patients awaiting for heart transplantation. Here, we developed a novel strategy for generation of heart constructs by repopulating engineered decellularized rat hearts using hESCs-derived CPCs. Careful expansion of CPCs in a scalable stirred-suspension bioreactor combined with step-wise seeding (60 million cells in 3 steps of 20 million per 1.5 h) onto decellularized hearts containing immobilized basic fibroblast growth factor (bFGF) resulted in improved retention of CPCs and differentiation to cardiomyocytes, smooth muscle cells and endothelial cells as evaluated by immunohistochemistry and qRT-PCR. We observed spontaneous and synchronous contractions of humanized hearts after 12 days of perfusion as well as advanced alignment of myofilaments. Our study provides a robust platform for generation of artificial human hearts and resolves major bottlenecks hindering further development of this technology.


Subject(s)
Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/pharmacology , Heart/physiology , Human Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cattle , Cell Differentiation/drug effects , Cell Lineage/drug effects , Colony-Forming Units Assay , Extracellular Matrix/drug effects , Gene Expression Regulation/drug effects , Heart/drug effects , Human Embryonic Stem Cells/drug effects , Humans , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...