Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Int J Mol Sci ; 24(24)2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38139018

RESUMO

Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episode (MELAS) syndrome, caused by a single base substitution in mitochondrial DNA (m.3243A>G), is one of the most common maternally inherited mitochondrial diseases accompanied by neuronal damage due to defects in the oxidative phosphorylation system. There is no established treatment. Our previous study reported a superior restoration of mitochondrial function and bioenergetics in mitochondria-deficient cells using highly purified mesenchymal stem cells (RECs). However, whether such exogenous mitochondrial donation occurs in mitochondrial disease models and whether it plays a role in the recovery of pathological neuronal functions is unknown. Here, utilizing induced pluripotent stem cells (iPSC), we differentiated neurons with impaired mitochondrial function from patients with MELAS. MELAS neurons and RECs/mesenchymal stem cells (MSCs) were cultured under contact or non-contact conditions. Both RECs and MSCs can donate mitochondria to MELAS neurons, but RECs are more excellent than MSCs for mitochondrial transfer in both systems. In addition, REC-mediated mitochondrial transfer significantly restored mitochondrial function, including mitochondrial membrane potential, ATP/ROS production, intracellular calcium storage, and oxygen consumption rate. Moreover, mitochondrial function was maintained for at least three weeks. Thus, REC-donated exogenous mitochondria might offer a potential therapeutic strategy for treating neurological dysfunction in MELAS.


Assuntos
Acidose Láctica , Síndrome MELAS , Células-Tronco Mesenquimais , Doenças Mitocondriais , Humanos , Síndrome MELAS/genética , Síndrome MELAS/terapia , Mitocôndrias/genética , Acidose Láctica/metabolismo , Acidose Láctica/patologia , DNA Mitocondrial/metabolismo , Doenças Mitocondriais/metabolismo , Neurônios/patologia , Células-Tronco Mesenquimais/metabolismo
2.
Int J Mol Sci ; 24(12)2023 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-37373439

RESUMO

Mitochondria are essential organelles for maintaining intracellular homeostasis. Their dysfunction can directly or indirectly affect cell functioning and is linked to multiple diseases. Donation of exogenous mitochondria is potentially a viable therapeutic strategy. For this, selecting appropriate donors of exogenous mitochondria is critical. We previously demonstrated that ultra-purified bone marrow-derived mesenchymal stem cells (RECs) have better stem cell properties and homogeneity than conventionally cultured bone marrow-derived mesenchymal stem cells. Here, we explored the effect of contact and noncontact systems on three possible mitochondrial transfer mechanisms involving tunneling nanotubes, connexin 43 (Cx43)-mediated gap junction channels (GJCs), and extracellular vesicles (Evs). We show that Evs and Cx43-GJCs provide the main mechanism for mitochondrial transfer from RECs. Through these two critical mitochondrial transfer pathways, RECs could transfer a greater number of mitochondria into mitochondria-deficient (ρ0) cells and could significantly restore mitochondrial functional parameters. Furthermore, we analyzed the effect of exosomes (EXO) on the rate of mitochondrial transfer from RECs and recovery of mitochondrial function. REC-derived EXO appeared to promote mitochondrial transfer and slightly improve the recovery of mtDNA content and oxidative phosphorylation in ρ0 cells. Thus, ultrapure, homogenous, and safe stem cell RECs could provide a potential therapeutic tool for diseases associated with mitochondrial dysfunction.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Conexina 43/genética , Conexina 43/metabolismo , Vesículas Extracelulares/metabolismo , Mitocôndrias/metabolismo , Canais Iônicos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Junções Comunicantes/metabolismo
3.
Stem Cell Res Ther ; 14(1): 40, 2023 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-36927781

RESUMO

BACKGROUND: Mitochondrial dysfunction caused by mutations in mitochondrial DNA (mtDNA) or nuclear DNA, which codes for mitochondrial components, are known to be associated with various genetic and congenital disorders. These mitochondrial disorders not only impair energy production but also affect mitochondrial functions and have no effective treatment. Mesenchymal stem cells (MSCs) are known to migrate to damaged sites and carry out mitochondrial transfer. MSCs grown using conventional culture methods exhibit heterogeneous cellular characteristics. In contrast, highly purified MSCs, namely the rapidly expanding clones (RECs) isolated by single-cell sorting, display uniform MSCs functionality. Therefore, we examined the differences between RECs and MSCs to assess the efficacy of mitochondrial transfer. METHODS: We established mitochondria-deficient cell lines (ρ0 A549 and ρ0 HeLa cell lines) using ethidium bromide. Mitochondrial transfer from RECs/MSCs to ρ0 cells was confirmed by PCR and flow cytometry analysis. We examined several mitochondrial functions including ATP, reactive oxygen species, mitochondrial membrane potential, and oxygen consumption rate (OCR). The route of mitochondrial transfer was identified using inhibition assays for microtubules/tunneling nanotubes, gap junctions, or microvesicles using transwell assay and molecular inhibitors. RESULTS: Co-culture of ρ0 cells with MSCs or RECs led to restoration of the mtDNA content. RECs transferred more mitochondria to ρ0 cells compared to that by MSCs. The recovery of mitochondrial function, including ATP, OCR, mitochondrial membrane potential, and mitochondrial swelling in ρ0 cells co-cultured with RECs was superior than that in cells co-cultured with MSCs. Inhibition assays for each pathway revealed that RECs were sensitive to endocytosis inhibitor, dynasore. CONCLUSIONS: RECs might serve as a potential therapeutic strategy for diseases linked to mitochondrial dysfunction by donating healthy mitochondria.


Assuntos
DNA Mitocondrial , Mitocôndrias , Humanos , Células HeLa , Mitocôndrias/metabolismo , DNA Mitocondrial/genética , Células Clonais , Trifosfato de Adenosina/metabolismo
4.
Cells ; 12(3)2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36766847

RESUMO

Intervertebral disc (IVD) degeneration is a major cause of low back pain. However, treatments directly approaching the etiology of IVD degeneration and discogenic pain are not yet established. We previously demonstrated that intradiscal implantation of cell-free bioresorbable ultra-purified alginate (UPAL) gel promotes tissue repair and reduces discogenic pain, and a combination of ultra-purified, Good Manufacturing Practice (GMP)-compliant, human bone marrow mesenchymal stem cells (rapidly expanding clones; RECs), and the UPAL gel increasingly enhanced IVD regeneration in animal models. This study investigated the therapeutic efficacy of injecting a mixture of REC and UPAL non-gelling solution for discogenic pain and IVD regeneration in a rat caudal nucleus pulposus punch model. REC and UPAL mixture and UPAL alone suppressed not only the expression of TNF-α, IL-6, and TrkA (p < 0.01, respectively), but also IVD degeneration and nociceptive behavior compared to punching alone (p < 0.01, respectively). Furthermore, REC and UPAL mixture suppressed these expression levels and nociceptive behavior compared to UPAL alone (p < 0.01, respectively). These results suggest that this minimally invasive treatment strategy with a single injection may be applied to treat discogenic pain and as a regenerative therapy.


Assuntos
Degeneração do Disco Intervertebral , Células-Tronco Mesenquimais , Núcleo Pulposo , Ratos , Humanos , Animais , Alginatos/farmacologia , Degeneração do Disco Intervertebral/tratamento farmacológico , Degeneração do Disco Intervertebral/metabolismo , Células-Tronco Mesenquimais/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo
5.
Stem Cells Transl Med ; 11(4): 434-449, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35267026

RESUMO

Mesenchymal stem cells (MSCs) exhibit self-renewal, multi-lineage differentiation potential and immunomodulatory properties, and are promising candidates for cellular therapy of various tissues. Despite the effective function of MSCs, the gradual loss of stem cell characteristics that occurs with repeated passages may significantly limit their therapeutic potential. A novel 3D shaking method was previously established to generate MSC spheroids in growth medium (GM-spheroids) and successfully maintain the multipotency of expanded MSCs, yet the expression of MSC-related genes was still low. In this study, we used a neurosphere culture technique to optimize the shaking culture method using human bone marrow-derived MSCs (BM-MSCs). MSC spheroids generated in neurosphere medium (NM-spheroids) maintained high expression of MSC-related genes during 3 weeks of prolonged shaking culture. Moreover, NM-spheroids generated from expanded MSCs showed high viability, upregulation of MSC-related and immune-related genes, and recovery of differentiation potential in vitro. Expanded adherent MSCs, GM-spheroids, and NM-spheroids were transplanted into a rat femur bone defect model to investigate their therapeutic potential in bone repair. Adherent MSCs and GM-spheroids showed delayed bone healing. In contrast, NM-spheroids showed high transplantation efficiency and enhanced bone regeneration. These data suggest that NM-spheroids generated using modified neurosphere culture conditions under continuous shaking recovered their stem cell characteristics in vitro and enhanced bone regeneration in vivo. Therefore, NM-spheroids should have great clinical potential for bone and tissue regenerative therapies as a stem cell-based biomaterial therapy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Regeneração Óssea , Diferenciação Celular , Osteogênese/fisiologia , Ratos , Esferoides Celulares
6.
Nanomaterials (Basel) ; 11(2)2021 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-33503931

RESUMO

This study was performed to examine the applicability of the newly developed nano-biocomposite, ß-tricalcium phosphate (ß-TCP)/u-HA/poly-d/l-lactide (PDLLA), to bone defects in the oral and maxillofacial area. This novel nano-biocomposite showed several advantages, including biocompatibility, biodegradability, and osteoconductivity. In addition, its optimal plasticity also allowed its utilization in irregular critical bone defect reconstructive surgery. Here, three different nano-biomaterials, i.e., ß-TCP/PDLLA, ß-TCP, and PDLLA, were implanted into critical bone defects in the right lateral mandible of 10-week-old Sprague-Dawley (SD) rats as bone graft substitutes. Micro-computed tomography (Micro-CT) and immunohistochemical staining for the osteogenesis biomarkers, Runx2, osteocalcin, and the leptin receptor, were performed to investigate and compare bone regeneration between the groups. Although the micro-CT results showed the highest bone mineral density (BMD) and bone volume to total volume (BV/TV) with ß-TCP, immunohistochemical analysis indicated better osteogenesis-promoting ability of ß-TCP/PDLLA, especially at an early stage of the bone healing process. These results confirmed that the novel nano-biocomposite, ß-TCP/PDLLA, which has excellent biocompatibility, bioresorbability and bioactive/osteoconductivity, has the potential to become a next-generation biomaterial for use as a bone graft substitute in maxillofacial reconstructive surgery.

7.
Stem Cells ; 39(3): 318-330, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33338299

RESUMO

Human mesenchymal stem/stromal cells (hMSCs) have garnered enormous interest as a potential resource for cell-based therapies. However, the molecular mechanisms regulating senescence in hMSCs remain unclear. To elucidate these mechanisms, we performed gene expression profiling to compare clonal immature MSCs exhibiting multipotency with less potent MSCs. We found that the transcription factor Frizzled 5 (FZD5) is expressed specifically in immature hMSCs. The FZD5 cell surface antigen was also highly expressed in the primary MSC fraction (LNGFR+ THY-1+ ) and cultured MSCs. Treatment of cells with the FZD5 ligand WNT5A promoted their proliferation. Upon FZD5 knockdown, hMSCs exhibited markedly attenuated proliferation and differentiation ability. The observed increase in the levels of senescence markers suggested that FZD5 knockdown promotes cellular senescence by regulating the noncanonical Wnt pathway. Conversely, FZD5 overexpression delayed cell cycle arrest during the continued culture of hMSCs. These results indicated that the intrinsic activation of FZD5 plays an essential role in negatively regulating senescence in hMSCs and suggested that controlling FZD5 signaling offers the potential to regulate hMSC quality and improve the efficacy of cell-replacement therapies using hMSCs.


Assuntos
Diferenciação Celular/fisiologia , Senescência Celular/fisiologia , Receptores Frizzled/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proliferação de Células/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Humanos , Transplante de Células-Tronco Mesenquimais/métodos
8.
J Invest Surg ; 34(3): 243-256, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31122080

RESUMO

This study compared the in vivo applicability of three-dimensional uncalcined and unsintered hydroxyapatite/poly-d/l-lactide (3D-HA/PDLLA) with beta-tricalcium phosphate (ß-TCP). 3D-HA/PDLLA is a newly developed bioactive, osteoconductive, bioresorbable bone regenerative composite. We performed critical-defect surgery on the mandible body of rats; the defects were filled with one of two bone graft substitutes. After a 4-week follow-up period, the mandibular specimens were examined using hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) staining and micro-computed tomography (micro-CT). The H&E staining showed an increase in newly formed bone in both groups from week 1 to 4. The difference in the Runx2 IHC optical density (OD) scores of 3D-HA/PDLLA and ß-TCP was not statistically significant (p > 0.05); however, the osteocalcin IHC OD scores of the groups differed significantly (p < 0.05). Micro-CT demonstrated a similar trabecular thickness, trabecular spacing, and bone volume per total volume in the two groups (p > 0.05), indicating that bone formation in the two groups was nearly the same from a macro-perspective of bone regeneration. These results demonstrated that a different bone regeneration pattern and earlier osteoblast differentiation occurred in 3D-HA/PDLLA compared with ß-TCP. In conclusion, our study demonstrates that 3D-HA/PDLLA is feasible for clinical application as a new bioactive, osteoconductive/bioresorbable bone graft substitute for maxillofacial surgery.


Assuntos
Substitutos Ósseos , Animais , Regeneração Óssea , Fosfatos de Cálcio , Dioxanos , Durapatita , Ratos , Microtomografia por Raio-X
9.
Front Bioeng Biotechnol ; 8: 590332, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33195156

RESUMO

Mesenchymal stromal/stem cells (MSCs), which generally expand into adherent monolayers, readily lose their proliferative and multilineage potential following repeated passages. Floating culture systems can be used to generate MSC spheroids, which are expected to overcome limitations associated with conventional adherent cultures while facilitating scaffold-free cell transplantation. However, the phenotypic characteristics of spheroids after long-term culture are unknown. In addition, regenerative therapies require new culture systems to maintain their undifferentiated state. In this study, we established a novel culture method employing three-dimensional (3D) "shaking" to generate MSC spheroids using bone marrow derived MSCs. Floating 3D cultures of mouse or human MSCs formed spheroids after shaking (85-95 rpm), within 1 month. These spheroids maintained their osteogenic-, adipogenic-, and chondrogenic-differentiation capacity. The adipogenic-differentiation capacity of adherent cultured mouse and human MSCs, which is lost following several passages, was remarkedly restored by shaking-culture. Notably, human MSC spheroids exhibited a renewable "undifferentiated MSC-pool" property, wherein undifferentiated MSCs grew from spheroids seeded repeatedly on a plastic culture dish. These data suggest that the shaking-culture method maintains and restores multipotency that is lost following monolayer expansion and thereby shows potential as a promising strategy for regenerative therapies with mesenchymal tissues.

10.
J Immunother Cancer ; 8(2)2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32675195

RESUMO

BACKGROUND: Mesenchymal stem/stromal cells (MSC) accumulate and reside in tumor sites. METHODS: Taking advantage of this feature in anticancer therapy, immortalized murine MSC (iMSC) were genetically altered to produce chemokine (C-C motif) ligand 19 (iMSC/CCL19), which attracts dendritic cells (DC) and T lymphocytes. Thereafter, iMSC/CCL19 were examined for their therapeutic efficacy using a syngeneic CT26 colon carcinoma cell line. RESULTS: Co-injection of iMSC/CCL19 into mice significantly suppressed the in vivo growth of CT26 cells compared with that of CCL19-expressing immortalized fibroblasts (iFib/CCL19). This anticancer effect was not observed when injected in CT26-bearing nude mice. Co-injected iMSC/CCL19 survived longer than iFib/CCL19 in the tumor sites. In a therapeutic model, local injection of iMSC/CCL19 suppressed the tumor growth, and increased IFN (interferon)-γ+ CD8+ T cells and CCR7+ DC infiltration in tumor site was observed when treated with iMSC/CCL19, but not with iMSC. This antitumor effect was completely negated by depletion of CD4+ cells and partially negated by depletion of CD8+ cells. Furthermore, the antitumor effects induced by local injection of iMSC/CCL19 were augmented by additional therapy with anti-programmed death (PD)-ligand 1 (PD-L1) antibody, but not with anti-PD-1 antibody. This combination therapy cured most of the tumors in CT26-bearing mice. CONCLUSION: These results suggest that local therapy with iMSC/CCL19 can suppress tumor growth via effective recruitment of CCR7+ DC into tumor sites and increase IFN-γ+ CD8+ T cells, and that combination with anti-PD-L1 antibody therapy can be a powerful anticancer therapy.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Quimiocina CCL19/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos
11.
Exp Hematol ; 76: 24-37, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31408689

RESUMO

The polycomb group protein Bmi1 maintains hematopoietic stem cell (HSC) functions. We previously reported that Bmi1-deficient mice exhibited progressive fatty changes in bone marrow (BM). A large portion of HSCs reside in the perivascular niche created partly by endothelial cells and leptin receptor+ (LepR+) BM stromal cells. To clarify how Bmi1 regulates the HSC niche, we specifically deleted Bmi1 in LepR+ cells in mice. The Bmi1 deletion promoted the adipogenic differentiation of LepR+ stromal cells and caused progressive fatty changes in the BM of limb bones with age, resulting in reductions in the numbers of HSCs and progenitors in BM and enhanced extramedullary hematopoiesis. This adipogenic change was also evident during BM regeneration after irradiation. Several adipogenic regulator genes appeared to be regulated by Bmi1. Our results indicate that Bmi1 keeps the adipogenic differentiation program repressed in BM stromal cells to maintain the integrity of the HSC niche.


Assuntos
Adipogenia/fisiologia , Células-Tronco Hematopoéticas/citologia , Complexo Repressor Polycomb 1/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Nicho de Células-Tronco , Animais , Medula Óssea/patologia , Medula Óssea/fisiologia , Linhagem Celular , Autorrenovação Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo Repressor Polycomb 1/deficiência , Proteínas Proto-Oncogênicas/deficiência , Receptores para Leptina/análise , Regeneração , Células Estromais/química , Células Estromais/patologia
12.
J Oral Sci ; 61(2): 284-293, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31217377

RESUMO

Bone marrow-derived mesenchymal stem cells (BMMSCs) remain the most widely used source of osteogenic cells in bone tissue engineering research. A cell-based treatment for alveolar ridge augmentation has received attention as an alternative to bone grafting. In the present study, BMMSC transplantation into tooth extraction sockets of C57BL/6J mice was evaluated for alveolar ridge regeneration. The first right maxillary molars were extracted, and then BMMSCs (PDGFRα+ Sca-1+ CD45- TER119- cells) isolated from femoral and tibial bone marrow were immediately transplanted into the extraction sockets. A control group underwent the same procedure except for BMMSC transplantation. Bone formation in the sockets was evaluated using micro-computed tomography and histological and immunohistochemical analyses. At 3 weeks, bone formation in the sockets was more advanced in the experimental group than in the control group. Histological analysis at 6 weeks after transplantation showed that the sockets in the experimental group also contained a greater quantity of bone marrow. Interestingly, socket bone mineral density was lower in the experimental group than in the control group at 6 weeks. These findings suggest that BMMSC transplantation accelerates bone healing and augments bone marrow formation in tooth extraction sockets.


Assuntos
Células-Tronco Mesenquimais , Animais , Medula Óssea , Regeneração Óssea , Camundongos , Camundongos Endogâmicos C57BL , Extração Dentária , Alvéolo Dental , Microtomografia por Raio-X
13.
Blood ; 133(23): 2495-2506, 2019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-30917958

RESUMO

Recurrent inactivating mutations have been identified in the X-linked plant homeodomain finger protein 6 (PHF6) gene, encoding a chromatin-binding transcriptional regulator protein, in various hematological malignancies. However, the role of PHF6 in normal hematopoiesis and its tumor-suppressor function remain largely unknown. We herein generated mice carrying a floxed Phf6 allele and inactivated Phf6 in hematopoietic cells at various developmental stages. The Phf6 deletion in embryos augmented the capacity of hematopoietic stem cells (HSCs) to proliferate in cultures and reconstitute hematopoiesis in recipient mice. The Phf6 deletion in neonates and adults revealed that cycling HSCs readily acquired an advantage in competitive repopulation upon the Phf6 deletion, whereas dormant HSCs only did so after serial transplantations. Phf6-deficient HSCs maintained an enhanced repopulating capacity during serial transplantations; however, they did not induce any hematological malignancies. Mechanistically, Phf6 directly and indirectly activated downstream effectors in tumor necrosis factor α (TNFα) signaling. The Phf6 deletion repressed the expression of a set of genes associated with TNFα signaling, thereby conferring resistance against the TNFα-mediated growth inhibition on HSCs. Collectively, these results not only define Phf6 as a novel negative regulator of HSC self-renewal, implicating inactivating PHF6 mutations in the pathogenesis of hematological malignancies, but also indicate that a Phf6 deficiency alone is not sufficient to induce hematopoietic transformation.


Assuntos
Autorrenovação Celular , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Proliferação de Células/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Materials (Basel) ; 12(5)2019 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-30818862

RESUMO

A novel three-dimensional (3D) porous uncalcined and unsintered hydroxyapatite/poly-d/l-lactide (3D-HA/PDLLA) composite demonstrated superior biocompatibility, osteoconductivity, biodegradability, and plasticity, thereby enabling complex maxillofacial defect reconstruction. Mesenchymal stem cells (MSCs)-a type of adult stem cell-have a multipotent ability to differentiate into chondrocytes, adipocytes, and osteocytes. In a previous study, we found that CD90 (Thy-1, cluster of differentiation 90) and CD271 (low-affinity nerve growth factor receptor) double-positive cell populations from human bone marrow had high proliferative ability and differentiation capacity in vitro. In the present study, we investigated the utility of bone regeneration therapy using implantation of 3D-HA/PDLLA loaded with human MSCs (hMSCs) in mandibular critical defect rats. Microcomputed tomography (Micro-CT) indicated that implantation of a 3D-HA/PDLLA-hMSC composite scaffold improved the ability to achieve bone regeneration compared with 3D-HA/PDLLA alone. Compared to the sufficient blood supply in the mandibular defection superior side, a lack of blood supply in the inferior side caused delayed healing. The use of Villanueva Goldner staining (VG staining) revealed the gradual progression of the nucleated cells and new bone from the scaffold border into the central pores, indicating that 3D-HA/PDLLA loaded with hMSCs had good osteoconductivity and an adequate blood supply. These results further demonstrated that the 3D-HA/PDLLA-hMSC composite scaffold was an effective bone regenerative method for maxillofacial boney defect reconstruction.

15.
Sci Rep ; 9(1): 20408, 2019 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-31892733

RESUMO

The spleen is comprised of spatially distinct compartments whose functions, such as immune responses and removal of aged red blood cells, are tightly controlled by the non-hematopoietic stromal cells that provide regionally-restricted signals to properly activate hematopoietic cells residing in each area. However, information regarding the ontogeny and relationships of the different stromal cell types remains limited. Here we have used in vivo lineage tracing analysis and in vitro mesenchymal stromal cell assays and found that Tlx1, a transcription factor essential for embryonic spleen organogenesis, marks neonatal stromal cells that are selectively localized in the spleen and retain mesenchymal progenitor potential to differentiate into mature follicular dendritic cells, fibroblastic reticular cells and marginal reticular cells. Furthermore, by establishing a novel three-dimensional cell culture system that enables maintenance of Tlx1-expressing cells in vitro, we discovered that signals from the lymphotoxin ß receptor and TNF receptor promote differentiation of these cells to express MAdCAM-1, CCL19 and CXCL13, representative functional molecules expressed by different subsets of mature stromal cells in the spleen. Taken together, these findings indicate that mesenchymal progenitor cells expressing Tlx1 are a subset of lymphoid tissue organizer-like cells selectively found in the neonatal spleen.


Assuntos
Proteínas de Homeodomínio/metabolismo , Tecido Linfoide/metabolismo , Células-Tronco Mesenquimais/metabolismo , Baço/metabolismo , Células Estromais/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Regulação da Expressão Gênica , Camundongos
16.
Tissue Eng Part A ; 25(5-6): 499-510, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30234439

RESUMO

IMPACT STATEMENT: Construction of capillary networks is a fundamental challenge for the development of three-dimensional (3D) tissue engineering. However, it is not well understood how to construct stable capillary networks that maintain a luminal size similar to that of capillary structures in vivo (i.e., <10 µm diameter). In this study, we demonstrated the construction of stable capillary networks covered by pericyte-like perivascular cells using an in vitro 3D angiogenesis model by optimizing interactions between endothelial cells and perivascular cells. Our 3D angiogenesis model can be combined with 3D culture of epithelial cells in the context of vascularization of 3D tissue-engineered constructs.


Assuntos
Capilares/citologia , Pericitos/citologia , Engenharia Tecidual/métodos , Membrana Basal/metabolismo , Proliferação de Células , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Imageamento Tridimensional , Células-Tronco Mesenquimais/citologia , Microfluídica , Neovascularização Fisiológica
17.
Materials (Basel) ; 11(10)2018 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-30347816

RESUMO

This study evaluated the feasibility of a novel three-dimensional (3D) porous composite of uncalcined and unsintered hydroxyapatite (u-HA) and poly-d/l-lactide (PDLLA) (3D-HA/PDLLA) for the bony regenerative biomaterial in maxillofacial surgery, focusing on cellular activities and osteoconductivity properties in vitro and in vivo. In the in vitro study, we assessed the proliferation and ingrowth of preosteoblastic cells (MC3T3-E1 cells) in 3D-HA/PDLLA biomaterials using 3D cell culture, and the results indicated enhanced bioactive proliferation. After osteogenic differentiation of those cells on 3D-HA/PDLLA, the osteogenesis marker genes runt-related transcription factor-2 (Runx2), and Sp7 (Osterix) were upregulated. For the in vivo study, we evaluated the utility of 3D-HA/PDLLA biomaterials compared to the conventional bone substitute of beta-tricalcium phosphate (ß-TCP) in rats with critical mandibular bony defects. The implantation of 3D-HA/PDLLA biomaterials resulted in enhanced bone regeneration, by inducing high osteoconductivity as well as higher ß-TCP levels. Our study thus showed that the novel composite, 3D-HA/PDLLA, is an excellent bioactive/bioresorbable biomaterial for use as a cellular scaffold, both in vitro and in vivo, and has utility in bone regenerative therapy, such as for patients with irregular maxillofacial bone defects.

18.
Inflamm Regen ; 37: 8, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29259707

RESUMO

Dental pulp stem cells/progenitor cells (DPSCs) can be easily obtained and can have excellent proliferative and mineralization potentials. Therefore, many studies have investigated the isolation and bone formation of DPSCs. In most previous reports, human DPSCs were traditionally isolated by exploiting their ability to adhere to plastic tissue culture dishes. DPSCs isolated by plastic adherence are frequently contaminated by other cells, which limits the ability to investigate their basic biology and regenerative properties. Additionally, the proliferative and osteogenic potentials vary depending on the isolated cells. It is very difficult to obtain cells of a sufficient quality to elicit the required effect upon transplantation. Considering clinical applications, stem cells used for regenerative medicine need to be purified in order to increase the efficiency of bone regeneration, and a stable supply of these cells must be generated. Here, we review the purification of DPSCs and studies of cranio-maxillofacial bone regeneration using these cells. Additionally, we introduce the prospective isolation of DPSCs using specific cell surface markers: low-affinity nerve growth factor and thymocyte antigen 1.

19.
Sci Rep ; 7: 42777, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28220862

RESUMO

The dermal papilla (DP) is a specialised mesenchymal component of the hair follicle (HF) that plays key roles in HF morphogenesis and regeneration. Current technical difficulties in preparing trichogenic human DP cells could be overcome by the use of highly proliferative and plastic human induced pluripotent stem cells (hiPSCs). In this study, hiPSCs were differentiated into induced mesenchymal cells (iMCs) with a bone marrow stromal cell phenotype. A highly proliferative and plastic LNGFR(+)THY-1(+) subset of iMCs was subsequently programmed using retinoic acid and DP cell activating culture medium to acquire DP properties. The resultant cells (induced DP-substituting cells [iDPSCs]) exhibited up-regulated DP markers, interacted with human keratinocytes to up-regulate HF related genes, and when co-grafted with human keratinocytes in vivo gave rise to fibre structures with a hair cuticle-like coat resembling the hair shaft, as confirmed by scanning electron microscope analysis. Furthermore, iDPSCs responded to the clinically used hair growth reagent, minoxidil sulfate, to up-regulate DP genes, further supporting that they were capable of, at least in part, reproducing DP properties. Thus, LNGFR(+)THY-1(+) iMCs may provide material for HF bioengineering and drug screening for hair diseases.


Assuntos
Diferenciação Celular , Folículo Piloso/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Regeneração , Animais , Linhagem Celular , Células Cultivadas , Feminino , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Queratinócitos/citologia , Queratinócitos/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores de Fator de Crescimento Neural/genética , Receptores de Fator de Crescimento Neural/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Tretinoína/farmacologia
20.
PLoS One ; 11(11): e0165946, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27855169

RESUMO

Mesenchymal stem/stromal cells (MSCs) reside in the bone marrow and maintain their stemness under hypoxic conditions. However, the mechanism underlying the effects of hypoxia on MSCs remains to be elucidated. This study attempted to uncover the signaling pathway of MSC proliferation. Under low-oxygen culture conditions, MSCs maintained their proliferation and differentiation abilities for a long term. The Notch2 receptor was up-regulated in MSCs under hypoxic conditions. Notch2-knockdown (Notch2-KD) MSCs lost their cellular proliferation ability and showed reduced gene expression of hypoxia-inducible transcription factor (HIF)-1α, HIF-2α, and c-Myc. Overexpression of the c-Myc gene in Notch2-KD MSCs allowed the cells to regain their proliferation capacity. These results suggested that Notch2 signaling is linked to c-Myc expression and plays a key role in the regulation of MSC proliferation. Our findings provide important knowledge for elucidating the self-replication competence of MSCs in the bone marrow microenvironment.


Assuntos
Regulação da Expressão Gênica , Genes myc , Células-Tronco Mesenquimais/metabolismo , Receptor Notch2/metabolismo , Transdução de Sinais , Animais , Medula Óssea/metabolismo , Diferenciação Celular , Hipóxia Celular , Proliferação de Células , Células Cultivadas , Feminino , Células-Tronco Mesenquimais/citologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA