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










Publication year range
1.
Stem Cell Res Ther ; 14(1): 127, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37170113

ABSTRACT

BACKGROUND: Childhood cancer treatment-induced gonadotoxicity causes permanent infertility/sub-infertility in nearly half of males. The current clinical and experimental approaches are limited to cryopreservation of prepubertal testicular strips and in vitro spermatogenesis which are inadequate to achieve the expanded spermatogonial stem/progenitor cells and spermatogenesis in vitro. Recently, we reported the supportive effect of bone marrow-derived mesenchymal cell co-culture which is inadequate after 14 days of culture in static conditions in prepubertal mouse testis due to lack of microvascular flow and diffusion. Therefore, we generated a novel, pumpless, single polydimethylsiloxane-layered testis-on-chip platform providing a continuous and stabilized microfluidic flow and real-time cellular paracrine contribution of allogeneic bone marrow-derived mesenchymal stem cells. METHODS: We aimed to evaluate the efficacy of this new setup in terms of self-renewal of stem/progenitor cells, spermatogenesis and structural and functional maturation of seminiferous tubules in vitro by measuring the number of undifferentiated and differentiating spermatogonia, spermatocytes, spermatids and tubular growth by histochemical, immunohistochemical, flow cytometric and chromatographic techniques. RESULTS: Bone marrow-derived mesenchymal stem cell-based testis-on-chip platform supported the maintenance of SALL4(+) and PLZF(+) spermatogonial stem/progenitor cells, for 42 days. The new setup improved in vitro spermatogenesis in terms of c-Kit(+) differentiating spermatogonia, VASA(+) total germ cells, the meiotic cells including spermatocytes and spermatids and testicular maturation by increasing testosterone concentration and improved tubular growth for 42 days in comparison with hanging drop and non-mesenchymal stem cell control. CONCLUSIONS: Future fertility preservation for male pediatric cancer survivors depends on the protection/expansion of spermatogonial stem/progenitor cell pool and induction of in vitro spermatogenesis. Our findings demonstrate that a novel bone marrow-derived mesenchymal stem cell-based microfluidic testis-on-chip device supporting the maintenance of stem cells and spermatogenesis in prepubertal mice in vitro. This new, cell therapy-based microfluidic platform may contribute to a safe, precision-based cell and tissue banking protocols for prepubertal fertility restoration in future.


Subject(s)
Sertoli Cells , Spermatogenesis , Male , Mice , Animals , Animals, Newborn , Culture Media, Conditioned , Spermatogenesis/physiology , Testis , Spermatogonia , Stem Cells
2.
Sci Rep ; 12(1): 11494, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35798781

ABSTRACT

Prepubertal cancer treatment leads to irreversible infertility in half of the male patients. Current in vitro spermatogenesis protocols and cryopreservation techniques are inadequate to expand spermatogonial stem/progenitor cells (SSPC) from testicles. Bone marrow derived mesenchymal stem cells (BM-MSC) bearing a close resemblance to Sertoli cells, improved spermatogenesis in animal models. We asked if a co-culture setup supported by syngeneic BM-MSC that contributes to the air-liquid interphase (ALI) could lead to survival, expansion and differentiation of SSPCs in vitro. We generated an ALI platform able to provide a real-time cellular paracrine contribution consisting of syngeneic BM-MSCs to neonatal C57BL/6 mice testes. We aimed to evaluate the efficacy of this culture system on SSPC pool expansion and spermatogenesis throughout a complete spermatogenic cycle by measuring the number of total germ cells (GC), the undifferentiated and differentiating spermatogonia, the spermatocytes and the spermatids. Furthermore, we evaluated the testicular cell cycle phases, the tubular and luminal areas using histochemical, immunohistochemical and flow cytometric techniques. Cultures in present of BM-MSCs displayed survival of ID4(+) spermatogonial stem cells (SSC), expansion of SALL4(+) and OCT4(+) SSPCs, VASA(+) total GCs and Ki67(+) proliferative cells at 42 days and an increased number of SCP3(+) spermatocytes and Acrosin(+) spermatids at 28 days. BM-MSCs increased the percentage of mitotic cells within the G2-M phase of the total testicular cell cycle increased for 7 days, preserved the cell viability for 42 days and induced testicular maturation by enlargement of the tubular and luminal area for 42 days in comparison to the control. The percentage of PLZF(+) SSPCs increased within the first 28 days of culture, after which the pool started to get smaller while the number of spermatocytes and spermatids increased simultaneously. Our findings established the efficacy of syngeneic BM-MSCs on the survival and expansion of the SSPC pool and differentiation of spermatogonia to round spermatids during in vitro culture of prepubertal mice testes for 42 days. This method may be helpful in providing alternative cures for male fertility by supporting in vitro differentiated spermatids that can be used for round spermatid injection (ROSI) to female oocyte in animal models. These findings can be further exploited for personalized cellular therapy strategies to cure male infertility of prepubertal cancer survivors in clinics.


Subject(s)
Adult Germline Stem Cells , Mesenchymal Stem Cells , Animals , Animals, Newborn , Cell Differentiation , Female , Male , Mice , Mice, Inbred C57BL , Spermatogenesis , Spermatogonia/metabolism , Testis/metabolism
3.
J Neurosurg Sci ; 66(1): 40-48, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33056946

ABSTRACT

BACKGROUND: Syringomyelia is a pathological cavitation of the spinal cord. In this study, we examined whether a syrinx cavity would limit itself with axonal regeneration and stem cell activity in the cavity, and we evaluated subjects on a functional basis. METHODS: Groups were designated as kaolin, trauma, kaolin-trauma, and saline groups. Also divided out of the syringomyelia treated groups were those given human mesenchymal stem cells (hMSCs). All groups were evaluated with immunohistochemistry, electron microscopy, confocal microscopy and functionally. RESULTS: The kaolin-trauma group had a significant correction of BBB score with hMSCs therapy. The syrinx cavity measurements showed significant improvement in groups treated with hMSCs. The tissue surrounding the syrinx cavity, however, appeared to be better organized in groups treated with hMSCs. The process of repair and regeneration of damaged axons in the lesion were more improved in groups treated with hMSCs. Using confocal microscopy, fluorescence of hMSCs was observed in the central canal, in the ependymal tissue, and around the lesion. CONCLUSIONS: It was concluded that axonal repair accelerated in groups receiving stem cells, and thus, stem cells may be effective in recovery of neural tissue and myelin damage in syringomyelia.


Subject(s)
Mesenchymal Stem Cells , Spinal Cord Injuries , Syringomyelia , Humans , Kaolin/pharmacology , Spinal Cord/pathology , Syringomyelia/pathology , Syringomyelia/therapy
4.
Cell Tissue Bank ; 23(1): 93-100, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33797678

ABSTRACT

The level of the major endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG) are altered in several types of carcinomas, and are known to regulate tumor growth. Thusly, this study hypothesized that the HEp-2 human laryngeal squamous cell carcinoma (LSCC) cell line releases AEA and 2-AG, and aimed to determine if their exogenous supplementation has an anti-proliferative effect in vitro. In this in vitro observational study a commercial human LSCC cell line (HEp-2) was used to test for endogenous AEA and 2-AG release via liquid chromatography-tandem mass spectrometry (LC-MS/MS). The anti-proliferative effect of AEA and 2-AG supplementation was evaluated via WST-1 proliferation assay. It was observed that the HEp-2 LSCC cell line released AEA and 2-AG; the median quantity of AEA released was 15.69 ng mL-1 (range: 14.55-15.95 ng mL-1) and the median quantity of 2-AG released was 2.72 ng -1 (range: 2.67-2.74 ng mL-1). Additionally, both AEA and 2-AG exhibited an anti-proliferative effect. The anti-proliferative effect of 2-AG was stronger than that of AEA. These findings suggest that AEA might function via a CB1 receptor-independent pathway and that 2-AG might function via a CB2-dependent pathway. The present findings show that the HEp-2 LSCC cell line releases the major endocannabinoids AEA and 2-AG, and that their supplementation inhibits tumor cell proliferation in vitro. Thus, cannabinoid ligands might represent novel drug candidates for laryngeal cancers, although functional in vivo studies are required in order to validate their potency.


Subject(s)
Endocannabinoids , Head and Neck Neoplasms , Arachidonic Acids , Cell Line , Chromatography, Liquid , Dietary Supplements , Endocannabinoids/metabolism , Endocannabinoids/pharmacology , Glycerides , Humans , Polyunsaturated Alkamides , Squamous Cell Carcinoma of Head and Neck , Tandem Mass Spectrometry
5.
Adv Exp Med Biol ; 1347: 135-162, 2021.
Article in English | MEDLINE | ID: mdl-33977438

ABSTRACT

Lysosomal storage disorders (LSDs) are rare inborn errors of metabolism caused by defects in lysosomal function. These diseases are characterized by accumulation of completely or partially degraded substrates in the lysosomes leading to cellular dysfunction of the affected cells. Currently, enzyme replacement therapies (ERTs), treatments directed at substrate reduction (SRT), and hematopoietic stem cell (HSC) transplantation are the only treatment options for LSDs, and the effects of these treatments depend strongly on the type of LSD and the time of initiation of treatment. However, some of the LSDs still lack a durable and curative treatment. Therefore, a variety of novel treatments for LSD patients has been developed in the past few years. However, despite significant progress, the efficacy of some of these treatments remains limited because these therapies are often initiated after irreversible organ damage has occurred.Here, we provide an overview of the known effects of LSDs on stem cell function, as well as a synopsis of available stem cell-based cell and gene therapies that have been/are being developed for the treatment of LSDs. We discuss the advantages and disadvantages of use of hematopoietic stem cell (HSC), mesenchymal stem cell (MSC), and induced pluripotent stem cell (iPSC)-related (gene) therapies. An overview of current research data indicates that when stem cell and/or gene therapy applications are used in combination with existing therapies such as ERT, SRT, and chaperone therapies, promising results can be achieved, showing that these treatments may result in alleviation of existing symptoms and/or prevention of progression of the disease. All together, these studies offer some insight in LSD stem cell biology and provide a hopeful perspective for the use of stem cells. Further development and improvement of these stem cell (gene) combination therapies may greatly improve the current treatment options and outcomes of patients with a LSD.


Subject(s)
Lysosomal Storage Diseases , Enzyme Replacement Therapy , Genetic Therapy , Hematopoietic Stem Cells , Humans , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/therapy , Lysosomes
6.
Cytokine ; 139: 155407, 2021 03.
Article in English | MEDLINE | ID: mdl-33383380

ABSTRACT

Although G-CSF mobilized peripheral blood stem cell (PBSC) transplantation is commonly used in adults, bone marrow (BM) is still the preferred stem cell source in pediatric stem cell transplantation. Despite the fact that G-CSF is increasingly being used to enhance the hematopoietic stem/progenitor cell (HSPC) yield in BM transplantation (G-BM), the direct effects of G-CSF on the pediatric BM microenvironment have never been investigated. The BM hematopoietic niche provides the physical space where the HSPCs reside. This BM niche regulates HSPC quiescence and proliferation through direct interactions with other niche cells, including Mesenchymal Stromal Cells (MSCs). These cells have been shown to secrete a wide range of hematopoietic cytokines (CKs) and growth factors (GFs) involved in differentiation, retention and homing of hematopoietic cells. Here, we assessed changes in the BM microenvironment by measuring levels of 48 different CKs and GFs in G-BM and control BM (C-BM) plasma from pediatric donors. In addition, the effect of G-CSF on cell numbers and characteristics of HSPCs and MSCs was assessed. IL-16, SCGF-b, MIP-1b (all >1000 pg/mL) and RANTES (>10.000 pg/mL) were highly expressed in healthy donor pediatric BM plasma. Levels of IL-3, IL-18, GROa, MCP-3 (p<0.05) were increased in G-BM, whereas levels of RANTES (p<0.001) decreased after G-CSF treatment. We found a negative correlation with increasing age for IL2-Ra and LIF (p<0.05). In addition, a concomitant increase in the number of both hematopoietic and fibroblast colony forming units was observed, indicating that G-CSF affects both HSPC and MSC numbers. In conclusion, G-CSF treatment of healthy pediatric donors affects the hematopoietic BM microenvironment by expansion of HSPC and MSC numbers and modifying local CK and GF levels.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow/drug effects , Cytokines/metabolism , Granulocyte Colony-Stimulating Factor/therapeutic use , Mesenchymal Stem Cells/drug effects , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Child , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Tissue Donors
7.
J Assist Reprod Genet ; 37(11): 2825-2838, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32840762

ABSTRACT

PURPOSE: To keep and increase spermatogonial stem cell number (SSC) is the only available option for pediatric cancer survivors to maintain fertility. Leptin is secreted by the epididymal white adipose tissue and has receptors on stem/progenitor spermatogonia. The purpose of this study is to demonstrate dose- and time-dependent proliferative effect of leptin on stem/progenitor spermatogonia cultures from prepubertal mice testes. METHODS: CD90.2 (+) stem/progenitor spermatogonia were isolated from the C57BL/6 mouse testis on postnatal day 6 and placed in culture. The proliferative effect of leptin supplementation was assessed by colony formation (diameter and number), WST proliferation assays, and xCELLigence real-time cell analysis (RTCA) on days 3, 5, and 7 of culture. Expressions of p-ERK1/2, p-STAT3, total STAT3, and p-SHP2 levels were determined by western blot analysis. RESULTS: Leptin supplementation of 100 ng/ml increased the diameter (p = 0.001) and number (p = 0.01) of colonies in stem/progenitor spermatogonial cultures and caused higher proliferation by WST-1 (p = 0.009) compared with the control on day 7. The EC50 was calculated as 114 ng/ml for leptin by RTCA. Proliferative dose of leptin induced increased expression of p-ERK1/2 (p = 0.009) and p-STAT3 (p = 0.023) on stem/progenitor spermatogonia when compared with the untreated group. CONCLUSION: The results indicated that leptin supplementation exhibited a dose- and time-dependent proliferative effect on stem/progenitor spermatogonia that was associated with increased expression of ERK1/2 and STAT3 pathways while maintaining their undifferentiated state. This output presents a new agent that may help to expand the stem/progenitor spermatogonia pool from the neonatal testis in order to autotransplant after cancer treatment.


Subject(s)
Adult Germline Stem Cells/cytology , Cell Proliferation/genetics , Leptin/genetics , Stem Cells/cytology , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Cell Differentiation/genetics , Humans , Mice
8.
Adv Exp Med Biol ; 1212: 127-153, 2020.
Article in English | MEDLINE | ID: mdl-31342461

ABSTRACT

The bone marrow (BM) hematopoietic niche is the microenvironment where in the adult hematopoietic stem and progenitor cells (HSPCs) are maintained and regulated. This regulation is tightly controlled through direct cell-cell interactions with mesenchymal stromal stem (MSCs) and reticular cells, adipocytes, osteoblasts and endothelial cells, through binding to extracellular matrix molecules and through signaling by cytokines and hematopoietic growth factors. These interactions provide a healthy environment and secure the maintenance of the HSPC pool, their proliferation, differentiation and migration. Recent studies have shown that innervation of the BM and interactions with the peripheral sympathetic neural system are important for maintenance of the hematopoietic niche, through direct interactions with HSCPs or via interactions with other cells of the HSPC microenvironment. Signaling through adrenergic receptors (ARs), opioid receptors (ORs), endocannabinoid receptors (CRs) on HSPCs and MSCs has been shown to play an important role in HSPC homeostasis and mobilization. In addition, a wide range of neuropeptides and neurotransmitters, such as Neuropeptide Y (NPY), Substance P (SP) and Tachykinins, as well as neurotrophins and neuropoietic growth factors have been shown to be involved in regulation of the hematopoietic niche. Here, a comprehensive overview is given of their role and interactions with important cells in the hematopoietic niche, including HSPCs and MSCs, and their effect on HSPC maintenance, regulation and mobilization.


Subject(s)
Bone Marrow Cells/cytology , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Stem Cell Niche/physiology , Cell Differentiation , Hematopoietic Stem Cells/cytology , Humans , Mesenchymal Stem Cells/cytology
9.
Biol Trace Elem Res ; 193(2): 364-376, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31069715

ABSTRACT

Metabolic diseases or injuries damage bone structure and self-renewal capacity. Trace elements and hydroxyapatite crystals are important in the development of biomaterials to support the renewal of bone extracellular matrix. In this study, it was assumed that the boron-loaded nanometer-sized hydroxyapatite composite supports the construction of extracellular matrix by controlled boron release in order to prevent its toxic effect. In this context, boron release from nanometer-sized hydroxyapatite was calculated by ICP-MS as in large proportion within 1 h and continuing release was provided at a constant low dose. The effect of the boron-containing nanometer-sized hydroxyapatite composite on the proliferation of SaOS-2 osteoblasts and human bone marrow-derived mesenchymal stem cells was evaluated by WST-1 and compared with the effects of nano-hydroxyapatite and boric acid. Boron increased proliferation of mesenchymal stem cells at high doses and exhibited different effects on osteoblastic cell proliferation. Boron-containing nano-hydroxyapatite composites increased osteogenic differentiation of mesenchymal stem cells by increasing alkaline phosphatase activity, when compared to nano-hydroxyapatite composite and boric acid. The molecular mechanism of effective dose of boron-containing hydroxyapatite has been assessed by transcriptomic analysis and shown to affect genes involved in Wnt, TGF-ß, and response to stress signaling pathways when compared to nano-hydroxyapatite composite and boric acid. Finally, a safe osteoconductive dose range of boron-containing nano-hydroxyapatite composites for local repair of bone injuries and the molecular effect profile in the effective dose should be determined by further studies to validation of the regenerative therapeutic effect window.


Subject(s)
Boron/pharmacology , Durapatite/pharmacology , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Alkaline Phosphatase/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Biocompatible Materials/pharmacology , Bone Regeneration/drug effects , Boron/chemistry , Boron/pharmacokinetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Drug Liberation , Durapatite/chemistry , Durapatite/pharmacokinetics , Humans , Mesenchymal Stem Cells/metabolism , Nanocomposites/chemistry , Osteoblasts/metabolism
10.
Turk J Biol ; 43(3): 171-178, 2019.
Article in English | MEDLINE | ID: mdl-31320815

ABSTRACT

Mucolipidosis type II (ML-II, I-cell disease) is a fatal inherited lysosomal storage disease caused by a deficiency of the enzyme N-acetylglucosamine-1-phosphotransferase. A characteristic skeletal phenotype is one of the many clinical manifestations of ML-II. Since the mechanisms underlying these skeletal defects in ML-II are not completely understood, we hypothesized that a defect in osteogenic differentiation of ML-II bone marrow mesenchymal stem cells (BM-MSCs) might be responsible for this skeletal phenotype. Here, we assessed and characterized the cellular phenotype of BM-MSCs from a ML-II patient before (BBMT) and after BM transplantation (ABMT), and we compared the results with BM-MSCs from a carrier and a healthy donor. Morphologically, we did not observe differences in ML-II BBMT and ABMT or carrier MSCs in terms of size or granularity. Osteogenic differentiation was not markedly affected by disease or carrier status. Adipogenic differentiation was increased in BBMT ML-II MSCs, but chondrogenic differentiation was decreased in both BBMT and ABMT ML-II MSCs. Immunophenotypically no significant differences were observed between the samples. Interestingly, the proliferative capacity of BBMT and ABMT ML-II MSCs was increased in comparison to MSCs from age-matched healthy donors. These data suggest that MSCs are not likely to cause the skeletal phenotype observed in ML-II, but they may contribute to the pathogenesis of ML-II as a result of lysosomal storage-induced pathology.

11.
Methods Mol Biol ; 1879: 153-163, 2019.
Article in English | MEDLINE | ID: mdl-30306535

ABSTRACT

Magnetic-activated cell sorting (MACS) is the technology that is recently used as a magnetic-based cell isolation/purification technique. This technique enables the isolation and selection of germ, hematopoietic, and somatic stem cells including skin stem cells (SkSCs). Here, we have tried to describe the isolation of stem cells by MACS using CD34 antigen for SkSCs, again CD34 for hematopoietic stem cells (HSCs) and Thy-1 for spermatogonial stem cells (SpSCs). MACS allowed the isolation of CD34+, CD34+, and Thy-1+ human SkSCs, HSCs, and SpSCs with minimum 98% purity.


Subject(s)
Cell Separation/methods , Stem Cells/cytology , Animals , Antigens, CD34/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunomagnetic Separation/methods , Magnetics/methods , Mice , Mice, Inbred C57BL , Stem Cells/metabolism , Thy-1 Antigens/metabolism
12.
Adv Exp Med Biol ; 1077: 317-342, 2018.
Article in English | MEDLINE | ID: mdl-30357696

ABSTRACT

Bioceramics are type of biomaterials generally used for orthopaedic applications due to their similar structure with bone. Especially regarding to their osteoinductivity and osteoconductivity, they are used as biodegradable scaffolds for bone regeneration along with mesenchymal stem cells. Since chemical properties of bioceramics are important for regeneration of tissue, physical properties are also important for cell proliferation. In this respect, several different manufacturing methods are used for manufacturing nano scale bioceramics. These nano scale bioceramics are used for regeneration of bone and cartilage both alone or with other types of biomaterials. They can also act as carrier for the delivery of drugs in musculoskeletal infections without causing any systemic toxicity.


Subject(s)
Biocompatible Materials , Bone Regeneration , Ceramics , Nanostructures , Osteogenesis , Cell Proliferation , Humans , Mesenchymal Stem Cells , Tissue Scaffolds
13.
Adv Exp Med Biol ; 1107: 15-40, 2018.
Article in English | MEDLINE | ID: mdl-29882209

ABSTRACT

Recent advances require a dual evaluation of germ and somatic stem cell niches with a regenerative medicine perspective. For a better point of view of the niche concept, it is needed to compare the microenvironments of those niches in respect to several components. The cellular environment of spermatogonial stem cells' niche consists of Sertoli cells, Leydig cells, vascular endothelial cells, epididymal fat cells, peritubular myoid cells while hematopoietic stem cells have mesenchymal stem cells, osteoblasts, osteoclasts, megacaryocytes, macrophages, vascular endothelial cells, pericytes and adipocytes in their microenvironment. Not only those cells', but also the effect of the other factors such as hormones, growth factors, chemokines, cytokines, extracellular matrix components, biomechanical forces (like shear stress, tension or compression) and physical environmental elements such as temperature, oxygen level and pH will be clarified during the chapter. Because it is known that the microenvironment has an important role in the stem cell homeostasis and disease conditions, it is crucial to understand the details of the microenvironment and to be able to compare the niche concepts of the different types of stem cells from each other, for the regenerative interventions. Indeed, the purpose of this chapter is to point out the usage of niche engineering within the further studies in the regenerative medicine field. Decellularized, synthetic or non-synthetic scaffolds may help to mimic the stem cell niche. However, the shared or different characteristics of germ and somatic stem cell microenvironments are necessary to constitute a proper niche model. When considered from this aspect, it is possible to produce some strategies on the personalized medicine by using those artificial models of stem cell microenvironment.


Subject(s)
Hematopoietic Stem Cells/cytology , Regenerative Medicine/trends , Spermatogonia/cytology , Stem Cell Niche , Humans , Male
14.
Exp Hematol ; 57: 30-41.e1, 2018 01.
Article in English | MEDLINE | ID: mdl-29030083

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is a well-known hematopoietic stem cell (HSC)-mobilizing agent used in both allogeneic and autologous transplantation. However, a proportion of patients or healthy donors fail to mobilize a sufficient number of cells. New mobilization agents are therefore needed. Endocannabinoids (eCBs) are endogenous lipid mediators generated in the brain and peripheral tissues and activate the cannabinoid receptors CB1 and CB2. We suggest that eCBs may act as mobilizers of HSCs from the bone marrow (BM) under stress conditions as beta-adrenergic receptors (Adrß). This study demonstrates that BM mesenchymal stem cells (MSCs) secrete anandamide (AEA) and 2-arachidonylglycerol (2-AG) and the peripheral blood (PB) and BM microenvironment contain AEA and 2-AG. 2-AG levels are significantly higher in PB of the G-CSF-treated group compared with BM plasma. BM mononuclear cells (MNCs) and CD34+ HSCs express CB1, CB2, and Adrß subtypes. CD34+ HSCs had higher CB1 and CB2 receptor expression in G-CSF-untreated and G-CSF-treated groups compared with MSCs. MNCs but not MSCs expressed CB1 and CB2 receptors based on qRT-PCR and flow cytometry. AEA- and 2-AG-stimulated HSC migration was blocked by eCB receptor antagonists in an in vitro migration assay. In conclusion, components of the eCB system and their interaction with Adrß subtypes were demonstrated on HSCs and MSCs of G-CSF-treated and G-CSF-untreated healthy donors in vitro, revealing that eCBs might be potential candidates to enhance or facilitate G-CSF-mediated HSC migration under stress conditions in a clinical setting.


Subject(s)
Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Glycerides/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Adolescent , Adrenergic beta-Antagonists/pharmacology , Adult , Arachidonic Acids/analysis , Arachidonic Acids/pharmacology , Bone Marrow/chemistry , Cell Movement/drug effects , Cells, Cultured , Cellular Microenvironment , Endocannabinoids/analysis , Endocannabinoids/pharmacology , Gene Expression Regulation/drug effects , Glycerides/analysis , Glycerides/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Humans , Plasma , Polyunsaturated Alkamides/analysis , Polyunsaturated Alkamides/pharmacology , Receptor, Cannabinoid, CB1/biosynthesis , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/biosynthesis , Receptor, Cannabinoid, CB2/drug effects , Receptor, Cannabinoid, CB2/genetics , Receptors, Adrenergic, beta/biosynthesis , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/genetics , Stress, Physiological/genetics , Young Adult
15.
Leuk Res ; 63: 62-71, 2017 12.
Article in English | MEDLINE | ID: mdl-29102598

ABSTRACT

Multipotent mesenchymal stem cells (MSC) are key components of the bone marrow (BM) microenvironment. The contribution of this microenvironment to the pathophysiology of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) is not well defined. A recent study in mice demonstrated that DICER1 gene deletion in osteoprogenitor cells from the BM microenvironment suppressed osteogenic differentiation and induced MDS and AML-like haematological findings. The present study evaluated the expression profiles of microRNAs (miRNAs) and DICER1 gene in BM-derived MSC of patients with AML (n=12), MDS (n=10) and healthy controls (HC) (n=8).miRNA expression profiles were analyzed by microarray and confirmations were performed using quantitative real-time PCR (qRT-PCR). Patient MSC displayed impaired proliferative and differentiation potential compared to HC. DICER1 gene expression was lower in MSC from MDS and AML patients than HC and some differentially expressed miRNAs indicated the potential involvement of DICER1 in the pathogenesis of MDS and AML. qRT-PCR confirmation revealed down-regulated miRNAs (hsa-miR-30d-5p, hsa-miR-222-3p and hsa-miR-30a-3p in MDS; hsa-miR-1275, hsa-miR-4725-5p and hsa-miR-143-3p in AML) and over-expressed miRNAs (hsa-miR-4462 in MDS; hsa-miR-134-5p and hsa-miR-874-3p in AML) in MDS and AML. Thus, our findings validate the results of the aforementioned animal study and demonstrate downregulation of DICER1 gene and abnormal miRNA profile in MDS and AML, which may have implications for understanding MDS and AML pathogenesis and contribute to developing targeted treatment strategies.


Subject(s)
Biomarkers, Tumor/genetics , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Myelodysplastic Syndromes/genetics , Ribonuclease III/genetics , Adult , Aged , Case-Control Studies , Cell Differentiation , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mesenchymal Stem Cells/pathology , Middle Aged , Myelodysplastic Syndromes/pathology , Prognosis , Young Adult
16.
Acta Histochem ; 119(5): 523-532, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28549792

ABSTRACT

Adenomyosis that is a form of endometriosis is the growth of ectopic endometrial tissue within the muscular wall of the uterus (myometrium), which may cause dysmenorrhea and infertility. Endocannabinoid mediated apoptotic mechanisms of endometriosis and adenomyosis are not known. We hypothesized that the down regulation of endocannabinoid receptors and/or alteration in their regulatory enzymes may have a direct role in the pathogenesis of endometriosis and adenomyosis through apoptosis. Endocannabinoid receptors CB1 and CB2, their synthesizing and catabolizing enzymes (FAAH, NAPE-PLD, DAGL, MAGL) and the apoptotic indexes were immunohistochemically assessed in endometriotic and adenomyotic tissues. Findings were compared to normal endometrium and myometrium. Endometrial adenocarcinoma (Ishikawa) and ovarian endometriosis cyst wall stromal (CRL-7566) cell lines were furthermore cultured with or without cannabinoid receptor agonists. The IC50 value for CB1 and CB2 receptor agonists was quantified. Cannabinoid agonists on cell death were investigated by Annexin-V/Propidium iodide labeling with flow cytometry. CB1 and CB2 receptor levels decreased in endometriotic and adenomyotic tissues compared to the control group (p=0,001 and p=0,001). FAAH, NAPE-PLD, MAGL and DAGL enzyme levels decreased in endometriotic and adenomyotic tissues compared to control (p=0,001, p=0,001, p=0,001 and p=0,002 respectively). Apoptotic cell indexes both in endometriotic and adenomyotic tissues also decreased significantly, compared to the control group (p=0,001 and p=0,001). CB1 and CB2 receptor agonist mediated dose dependent fast anti-proliferative and pro-apoptotic effects were detected in Ishikawa and ovarian endometriosis cyst wall stromal cell lines (CRL-7566). Endocannabinoids are suggested to increase apoptosis mechanisms in endometriosis and adenomyosis. CB1 and CB2 antagonists can be considered as potential medical therapeutic agents for endometriosis and adenomyosis.


Subject(s)
Adenomyosis/physiopathology , Apoptosis/physiology , Endocannabinoids/metabolism , Endometriosis/physiopathology , Apoptosis/drug effects , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Cell Line , Female , Flow Cytometry , Humans , Inhibitory Concentration 50 , Protein Binding/drug effects , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors
17.
J Appl Oral Sci ; 24(1): 67-75, 2016.
Article in English | MEDLINE | ID: mdl-27008259

ABSTRACT

UNLABELLED: An increasing body of evidence suggests that the use of probiotic bacteria is a promising intervention approach for the treatment of inflammatory diseases with a polymicrobial etiology. P. gingivalis has been noted to have a different way of interacting with the innate immune response of the host compared to other pathogenic bacteria, which is a recognized feature that inhibits CXCL8 expression. OBJECTIVE: The aim of the study was to determine if P. gingivalis infection modulates the inflammatory response of gingival stromal stem cells (G-MSSCs), including the release of CXCL8, and the expression of TLRs and if immunomodulatory L. rhamnosus ATCC9595 could prevent CXCL8 inhibition in experimental inflammation. MATERIAL AND METHODS: G-MSSCs were pretreated with L. rhamnosus ATCC9595 and then stimulated with P. gingivalis ATCC33277. CXCL8 and IL-10 levels were investigated with ELISA and the TLR-4 and 2 were determined through flow cytometer analysis. RESULTS: CXCL8 was suppressed by P. gingivalis and L. rhamnosus ATCC9595, whereas incubation with both strains did not abolish CXCL8. L. rhamnosus ATCC9595 scaled down the expression of TLR4 and induced TLR2 expression when exposed to P. gingivalis stimulation (p<0.01). CONCLUSIONS: These findings provide evidence that L. rhamnosus ATCC9595 can modulate the inflammatory signals and could introduce P. gingivalis to immune systems by inducing CXCL8 secretion.


Subject(s)
Interleukin-8/analysis , Lacticaseibacillus rhamnosus/physiology , Mesenchymal Stem Cells/microbiology , Porphyromonas gingivalis/immunology , Probiotics/pharmacology , Bacterial Adhesion/immunology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunity, Innate , Interferon-gamma/analysis , Interferon-gamma/immunology , Interleukin-10 , Interleukin-8/immunology , Periodontitis/microbiology , Statistics, Nonparametric , Toll-Like Receptor 4/analysis , Toll-Like Receptor 4/immunology , Young Adult
18.
J. appl. oral sci ; 24(1): 67-75, Jan.-Feb. 2016. tab, graf
Article in English | LILACS, BBO - Dentistry | ID: lil-777353

ABSTRACT

ABSTRACT An increasing body of evidence suggests that the use of probiotic bacteria is a promising intervention approach for the treatment of inflammatory diseases with a polymicrobial etiology. P. gingivalis has been noted to have a different way of interacting with the innate immune response of the host compared to other pathogenic bacteria, which is a recognized feature that inhibits CXCL8 expression. Objective The aim of the study was to determine if P. gingivalis infection modulates the inflammatory response of gingival stromal stem cells (G-MSSCs), including the release of CXCL8, and the expression of TLRs and if immunomodulatory L. rhamnosus ATCC9595 could prevent CXCL8 inhibition in experimental inflammation. Material and Methods G-MSSCs were pretreated with L. rhamnosus ATCC9595 and then stimulated with P. gingivalis ATCC33277. CXCL8 and IL-10 levels were investigated with ELISA and the TLR-4 and 2 were determined through flow cytometer analysis. Results CXCL8 was suppressed by P. gingivalis and L. rhamnosus ATCC9595, whereas incubation with both strains did not abolish CXCL8. L. rhamnosus ATCC9595 scaled down the expression of TLR4 and induced TLR2 expression when exposed to P. gingivalis stimulation (p<0.01). Conclusions These findings provide evidence that L. rhamnosus ATCC9595 can modulate the inflammatory signals and could introduce P. gingivalis to immune systems by inducing CXCL8 secretion.


Subject(s)
Humans , Young Adult , Interleukin-8/analysis , Porphyromonas gingivalis/immunology , Probiotics/pharmacology , Lacticaseibacillus rhamnosus/physiology , Mesenchymal Stem Cells/microbiology , Periodontitis/microbiology , Bacterial Adhesion/immunology , Enzyme-Linked Immunosorbent Assay , Cells, Cultured , Interleukin-8/immunology , Interferon-gamma/analysis , Interferon-gamma/immunology , Interleukin-10 , Statistics, Nonparametric , Toll-Like Receptor 4/analysis , Toll-Like Receptor 4/immunology , Flow Cytometry , Immunity, Innate
19.
Curr Stem Cell Res Ther ; 11(6): 475-486, 2016.
Article in English | MEDLINE | ID: mdl-25747695

ABSTRACT

Advancements in biomaterials and stem cell technology have lead current medical technology to tissue engineering and regenerative medicine. Human engineered cartilage, bone, fascia, tendon, nerve and skin tissues have been used for the treatment of tissue injuries and degenerative diseases in combination with embryonic, fetal or adult stem and progenitor cells. Mesenchymal stem cells are one of the most extensively studied adult stem cell population and are widely utilized in cell therapies. Regeneration and 3-dimensional reconstruction of specialized connective tissues by combining differently originated micro and nanoscaled, natural or synthetic scaffolds with stem or progenitor cells are highly expected to guarantee patients to maintain acceptable life quality. In this review we discuss the important issues in biomaterial and stem cell interactions based on histological biocompatibility, updating recent basic research in this field and addressing possible future perspectives.

20.
Curr Stem Cell Res Ther ; 11(6): 487-493, 2016.
Article in English | MEDLINE | ID: mdl-25643122

ABSTRACT

Orthopedic disorders and trauma usually result in bone loss. Bone grafts are widely used to replace this tissue. Bone grafts excluding autografts unfortunately have disadvantages like evoking immune response, contamination and rejection. Autografts are of limited sources and optimum biomaterials that can replace bone have been searched for several decades. Bioceramics, which have the similar inorganic structure of natural bone, are widely used to regenerate bone or coat metallic implants. As people continuously look for a higher life quality, there are developments in technology almost everyday to meet their expectations. Nanotechnology is one of such technologies and it attracts everyone's attention in biomaterial science. Nano scale biomaterials have many advantages like larger surface area and higher biocompatibility and these properties make them more preferable than micro scale. Also, stem cells are used for bone regeneration besides nano-bioceramics due to their differentiation characteristics. This review covers current research on nano-bioceramics and mesenchymal stem cells and their role in bone regeneration.

SELECTION OF CITATIONS
SEARCH DETAIL
...