Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Cell Physiol ; 237(9): 3627-3639, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35766589

RESUMO

The adipose-derived stromal vascular fraction (SVF) is composed of a heterogeneous mix of adipose-derived stem cells (ADSCs), macrophages, pericytes, fibroblasts, blood, and other cells. Previous studies have found that the paracrine effects of SVF cells may be therapeutic, but their role in osteoarthritis treatment remains unclear. This study aimed to investigate the therapeutic effect of SVF cells on chondrocytes. Chondrocytes were seeded on culture plates alone (control) or cocultured with SVF or ADSCs on cell culture inserts. After 48 h of coculture, chondrocyte collagen II, tissue inhibitors of metalloproteinases-3 (TIMP-3), and matrix metalloproteinases-13 (MMP-13) messenger RNA (mRNA) expression levels were evaluated using reverse-transcription polymerase chain reaction, and the transforming growth factor-ß (TGF-ß) levels in the supernatant were measured using ELISA. Immunohistochemical staining and flow cytometry were used to evaluate the macrophages in the SVF. These macrophages were characterized according to phenotype using the F4/80, CD86, and CD163 markers. To determine whether the Smad2/3 signaling pathways were involved, the chondrocytes were pre-treated with a Smad2/3 phosphorylation inhibitor and stimulated with the SVF, and then Smad2/3 phosphorylation levels were analyzed using western blot. The mRNA expression levels of various paracrine factors and chondrocyte pellet size were also assessed. Collagen II and TIMP-3 expression were higher in the SVF group than in the ADSC group and controls, while MMP-13 expression was the highest in the ADSC group and the lowest in the controls. TGF-ß levels in the SVF group were also elevated. Immunohistochemical staining and flow cytometry revealed that the macrophages in the SVF were of the anti-inflammatory phenotype. Western blot analysis showed that the SVF increased Smad2/3 phosphorylation, while Smad2/3 inhibitors decreased phosphorylation. Smad2/3 inhibitors also reduced the expression of various other paracrine factors and decreased chondrocyte pellet size. These findings suggested that the paracrine effect of heterogeneous cells, such as anti-inflammatory macrophages, in the SVF partly supports chondrocyte regeneration through TGF-ß-induced Smad2/3 phosphorylation.


Assuntos
Condrócitos , Inibidor Tecidual de Metaloproteinase-3 , Condrócitos/metabolismo , Colágeno/metabolismo , Humanos , Macrófagos/metabolismo , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Fração Vascular Estromal , Inibidor Tecidual de Metaloproteinase-3/genética , Fator de Crescimento Transformador beta/metabolismo
2.
BMC Musculoskelet Disord ; 21(1): 207, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-32252731

RESUMO

BACKGROUND: Adipose-derived stromal vascular fraction (SVF) cells are a mixed cell population that includes cells with multilineage potential, similar to bone marrow-derived mesenchymal stem cells. Our purpose is to investigate the influence of SVF cells in patients with knee osteoarthritis (OA) and the short-term treatment effects. METHODS: Fifty-seven patients were enrolled and treated with intra-articular injection of 2.5 × 107 SVF cells into the knee joint between September 2017 and March 2018. All patients were followed up for 12 months or longer. Mean age at treatment and follow-up period were 69.4 ± 6.9 years and 13.7 ± 2.0 months, respectively. The mean preoperative hip-knee-ankle angle was 6.7 ± 3.6°. SVF cells were prepared using the Celution®800/CRS system from the patients' abdominal or breech subcutaneous fat. The mean SVF cell viability was 90.6 ± 2.7%. Clinical evaluations were performed for range of motion, Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), visual analog scale (VAS) for pain, and the Knee injury and Osteoarthritis Score (KOOS). Imaging evaluations, which included the hip-knee-ankle angle assessed via radiography, and T2 mapping value using a 1.5-T magnetic resonance imaging unit were also assessed. Both clinical and imaging evaluations were performed preoperatively, 1, 3, 6, and 12 months postoperatively, and compared among all timepoints (p < 0.05). RESULTS: Knee extension angle at 6 and 12 months postoperatively was significantly better than the preoperative angle. Total WOMAC, VAS, and KOOS scores at 1, 3, 6 and 12 months postoperatively were significantly better than preoperative scores. There was no significant difference in hip-knee-ankle angle among the five time periods. T2 mapping values of lateral femur and tibia were significantly higher 12 months postoperatively than preoperatively. CONCLUSIONS: The short-term clinical effects of intra-articular SVF cell injection on knee OA were excellent. Intra-articular SVF cell injection is a novel and innovative approach for treating patients with knee OA.


Assuntos
Tecido Adiposo/citologia , Transplante de Células-Tronco Mesenquimais , Osteoartrite do Joelho/terapia , Idoso , Feminino , Humanos , Injeções Intra-Articulares , Articulação do Joelho/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade , Osteoartrite do Joelho/diagnóstico por imagem , Medição da Dor , Radiografia , Amplitude de Movimento Articular , Resultado do Tratamento
3.
Surg Today ; 47(12): 1500-1511, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28555267

RESUMO

PURPOSE: More effective methods are needed for breast reconstruction after breast-conserving surgery for breast cancer. The aim of this clinical study was to assess the perioperative and long-term outcomes of adipose-derived regenerative cell (ADRC)-enriched autologous fat grafting. METHODS: Ten female patients who had undergone breast-conserving surgery and adjuvant radiotherapy for breast cancer were enrolled. An ADRC-enriched fat graft prepared from the patient's adipose tissue was implanted at the time of adipose tissue harvest. The perioperative and long-term outcomes of the grafts, which included safety, efficacy, and questionnaire-based patient satisfaction, were investigated. RESULTS: The mean operation time was 188 ± 30 min, and the mean duration of postoperative hospitalization was 1.2 ± 0.4 days. No serious postoperative complications were associated with the procedure. Neither recurrence nor metastatic disease was observed during the follow-up period (7.8 ± 1.5 years) after transplantation. Of 9 available patients, "more than or equal to average" satisfaction with breast appearance and overall satisfaction were reported by 6 (66.7%) and 5 (55.6%) patients, respectively. CONCLUSIONS: ADRC-enriched autologous fat transplantation is thus considered to be safe perioperatively, with no long-term recurrence, for patients with breast cancer treated by breast-conserving surgery, and it may be an option for breast reconstruction, even after adjuvant radiotherapy.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/transplante , Neoplasias da Mama/cirurgia , Mama/cirurgia , Mastectomia Segmentar/métodos , Procedimentos de Cirurgia Plástica/métodos , Transplante Autólogo/métodos , Adulto , Idoso , Povo Asiático , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Duração da Cirurgia , Satisfação do Paciente , Qualidade de Vida , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
4.
Cell Transplant ; 32: 9636897231190175, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37551027

RESUMO

The aim of the article is to compare the clinical and radiological outcomes between single and double stromal vascular fraction (SVF) cell injections in patients with knee osteoarthritis (OA). We included 54 patients treated for varus knee OA with intra-articular SVF cell injection. They were divided into two groups: those who received one injection and those who received two. The Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score, knee range of motion, and knee muscle force were assessed at baseline and 3, 6, 12, and 24 months after the first injection. The preoperative hip-knee-ankle (HKA) angle was evaluated using plain radiographs, and T2 mapping values were assessed. The total WOMAC score improved significantly in the single injection group from 3 to 24 months, but the total WOMAC score in the double injection group improved significantly at 24 months. The T2 mapping values in both the groups improved, with a significant difference at 12 months. The preoperative mean HKA angle and the correlation coefficients between the HKA angle and the total WOMAC score and between the HKA angle and the T2 mapping value of the medial femur were significant. In conclusion, double injections may provide more satisfactory treatment outcomes in patients with severe varus knee alignment. This clinical trial is registered in the Japanese Ministry of Health, Labour and Welfare (URL: https://saiseiiryo.mhlw.go.jp/published_plan/index/2) with the registration name "Cell transplantation therapy for osteoarthritis using autologous subcutaneous adipose tissue-derived regenerative (stem) cells (ADRCs)," and the registration number was "PB5160012."


Assuntos
Osteoartrite do Joelho , Humanos , Tecido Adiposo , Injeções Intra-Articulares , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/cirurgia , Osteoartrite do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/terapia , Fração Vascular Estromal , Resultado do Tratamento
5.
Regen Ther ; 24: 94-102, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37363753

RESUMO

Introduction: Currently, studies on adipose-derived stromal vascular fraction (SVF) cells are attracting increasing attention because they have the potential to differentiate into a subset of cell types, such as bone marrow-derived mesenchymal stromal cells (MSCs), and are easier to harvest than MSCs, thus making them easier to apply clinically. This study evaluated the short-term clinical outcomes of SVF cell therapy for hip osteoarthritis (OA). Methods: Forty-two patients were enrolled in this study; these patients received a single injection comprising an average of 3.8 (standard deviation [SD], ±1.3) × 107 SVF cells into the hip joint. All patients were followed-up for at least 6 months. The mean age of the patients was 60.2 years (SD, ±9.4 years). Kellgren-Lawrence (KL) grades II, III, and IV based on radiography were 13, 13, and 16 patients, respectively. SVF cells were obtained from the subcutaneous fat of the abdomen or breech using a Celution® 800/CRS system. The average cell viability of SVF cells was 90.8% (SD, ±2.8%). Clinical assessments were performed using the Harris Hip Score (HHS), Japanese Orthopaedic Association Hip Disease Evaluation Questionnaire (JHEQ) score, and visual analog scale (VAS) score to evaluate pain. Images were evaluated using radiography, and T2 mapping values were obtained using a 1.5-T magnetic resonance imaging system. These clinical and imaging assessments were followed from preoperatively to 6 months postoperatively. Results: The HHS, JHEQ score, and VAS score improved significantly from 22.5 (SD, ±16.6), 26.6 (SD, ±11.3), and 75.5 (SD, ±15.8) preoperatively to 46.8 (SD, ±27.2), 39.4 (SD, ±19.7), and 46.5 (SD, ±27.9), respectively, at 6 months postoperatively. KL grade II showed significant improvement in clinical outcome from preoperative to postoperative, while KL grade IV showed slight or little improvement. The center edge angle, acetabular head index on the radiographs, and T2 mapping values did not change significantly from preoperatively to 6 months postoperatively. Conclusions: SVF cell injection in the hip joint showed good short-term clinical efficacy for reducing hip OA symptoms. SVF cell therapy is thus an innovative and effective treatment for hip OA.

6.
Int J Urol ; 19(7): 652-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22435469

RESUMO

OBJECTIVES: To report a novel cell therapy using autologous adipose tissue-derived regenerative cells for male stress urinary incontinence caused by urethral sphincteric deficiency, and the outcomes in the initial cases undergoing periurethral injection of adipose tissue-derived regenerative cells. METHODS: Three patients with moderate stress incontinence after radical prostatectomy and holmium laser enucleation of the prostate were enrolled. Adipose tissue-derived regenerative cells were isolated from the abdominal adipose tissue by using the Celution system. Subsequently, the isolated adipose tissue-derived regenerative cells, and a mixture of adipose tissue-derived regenerative cells and adipose tissue were transurethrally injected into the rhabdosphincter and submucosal space of the urethra, respectively. Short-term outcomes during a 6-month follow up were assessed by a 24-h pad test, a validated patient questionnaire, urethral pressure profile, transrectal ultrasonography and magnetic resonance imaging. RESULTS: Urinary incontinence progressively improved after 2 weeks of injection up to 6 months in terms of decreased leakage volume, decreased frequency and amount of incontinence, and improved quality of life. Both maximum urethral closing pressure and functional profile length increased. Magnetic resonance imaging suggested a sustained presence of the injected adipose tissue. Enhanced ultrasonography showed a progressive increase in the blood flow to the injected area. No significant adverse events were observed peri- and postoperatively. CONCLUSION: These preliminary findings suggest that periurethral injection of the autologous adipose tissue-derived regenerative cells is a safe and feasible treatment modality for male stress urinary incontinence.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/transplante , Células-Tronco Multipotentes/transplante , Incontinência Urinária por Estresse/terapia , Idoso , Humanos , Injeções , Imageamento por Ressonância Magnética , Masculino , Qualidade de Vida , Resultado do Tratamento , Ultrassonografia , Uretra/diagnóstico por imagem , Incontinência Urinária por Estresse/diagnóstico por imagem , Urodinâmica
7.
Surg Today ; 41(1): 18-23, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21191687

RESUMO

Regenerative medicine is emerging as a rapidly evolving field of research and therapeutics. Stem cells hold great promise for future translational research and clinical applications in many fields. Much research has focused on mesenchymal stem cells isolated from bone marrow in vitro and in vivo; however, bone marrow procurement causes considerable discomfort to the patient and yields a relatively small number of harvested cells. By contrast, adipose tissue represents an abundant and easily accessible source of adult stem cells, termed adipose-derived stem cells (ADSCs), with the ability to equally differentiate along multiple lineage pathways. These stem cells have angiogenic properties, possibly because of their secretion of cytokines. They may also play a role in healing acute and chronic tissue damage. Subsequently, they have a wide range of potential clinical implications. This article reviews the potential preclinical and clinical applications of mesenchymal stem cells, especially ADSCs, in surgery.


Assuntos
Adipócitos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Medicina Regenerativa/métodos , Humanos , Transplante de Células-Tronco/métodos , Coleta de Tecidos e Órgãos/métodos
8.
J Clin Med ; 10(19)2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34640324

RESUMO

Adipose-derived regenerative cells (ADRCs) are non-cultured heterogeneous or mixed populations of cells obtained from adipose tissue by collagenase digestion. The injection of ADRCs have been tried clinically for the treatment of osteoarthritis (OA). The purpose of this study was to evaluate the effect of intra-articular transplantation of human ADRCs on OA progression in mice and the effect of ADRCs on macrophage polarization. In in vivo experiments, BALB/c-nu mice with knee OA received intra-articular transplantation of either phosphate buffered-saline or human ADRCs. OA progression was evaluated histologically and significantly attenuated in the ADRC group at both four and eight weeks postoperatively. The expression of OA-related proteins in the cartilage and macrophage-associated markers in the synovium were examined by immunohistochemistry. The numbers of MMP-13-, ADAMTS-5-, IL-1ß-, IL-6- and iNOS-positive cells significantly decreased, and type II collagen- and CD206-positive cells were more frequently detected in the ADRC group compared with that in the control group. In vitro co-culture experiments showed that ADRCs induced macrophage polarization toward M2. The results of this study suggest that the intra-articular transplantation of human ADRCs could attenuate OA progression possibly by reducing catabolic factors in chondrocytes and modulating macrophage polarization.

9.
Cell Transplant ; 30: 9636897211067454, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35392685

RESUMO

Favorable clinical outcomes of intra-articular injection of adipose-derived stromal vascular fraction (SVF) cells for knee osteoarthritis (OA) have been reported, but the effects of different doses of SVF cells have not been examined. This study aimed to compare the short-term clinical and imaging outcomes of different doses of SVF cells for knee OA treatment. This study included 60 patients with knee OA who underwent intra-articular injection of SVF cells. The follow-up period was at least 12 months. Thirty patients received an intra-articular injection of 2.5×107 SVF cells (low-dose group), and the remaining 30 patients received an intra-articular injection of 5.0×107 SVF cells (high-dose group). Clinical evaluations were performed for the Knee injury and Osteoarthritis Outcome Score (KOOS). Imaging evaluations, including the magnetic resonance imaging Osteoarthritis Knee Score (MOAKS) features (bone marrow lesions, cartilage defects, osteophytes, Hoffa's synovitis, and effusion synovitis), were also performed. All clinical and imaging evaluations were performed preoperatively and 12 months postoperatively and compared between the groups. In demographic data, no significant differences were found between the two groups. The total score of KOOS at 12 months postoperatively was significantly more favorable than the preoperative score in the high-dose groups. Pain and symptoms subscale scores of KOOS at 12 months postoperatively were significantly better in the high-dose group than in the low-dose group. The bone marrow lesions, Hoffa's synovitis, and effusion synovitis improved approximately 30-40% at 12 months postoperatively compared to baseline in both groups. However, there were no significant differences in imaging evaluations between the two groups. In conclusion, the pain and symptoms subscale scores of KOOS from baseline to 12 months postoperatively improved better in the high-dose group than in the low-dose group. Our findings suggest that intra-articular injection of SVF cells for knee OA is an innovative approach.


Assuntos
Transplante de Células-Tronco Mesenquimais , Osteoartrite do Joelho , Sinovite , Tecido Adiposo/patologia , Humanos , Injeções Intra-Articulares/métodos , Imageamento por Ressonância Magnética , Transplante de Células-Tronco Mesenquimais/métodos , Osteoartrite do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/terapia , Dor , Fração Vascular Estromal , Resultado do Tratamento
10.
Int J Urol ; 17(1): 75-82, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20002225

RESUMO

OBJECTIVES: To report a novel cell therapy using autologous adipose tissue-derived stem cells (ADSC) for stress urinary incontinence caused by urethral sphincteric deficiency and the outcomes in two initial cases undergoing periurethral injection of stem cells for the treatment of urinary incontinence after radical prostatectomy. METHODS: Two patients with moderate stress incontinence after radical prostatectomy were enrolled. After liposuction of 250 mL of adipose tissue from the abdomen, we isolated ADSC from this tissue by using the Celution system. Subsequently, the isolated ADSC and a mixture of stem cells and adipose tissue were transurethrally injected into the rhabdosphincter and submucosal space of the urethra, respectively. Short-term outcomes during a 12-week follow-up were assessed by a 24-h pad test, a validated patient questionnaire, urethral pressure profile, transrectal ultrasonography, and magnetic resonance imaging. RESULTS: Urinary incontinence progressively improved after 2 weeks of injection up to 12 weeks in terms of decreased leakage volume in a 24-h pad test, decreased frequency and amount of incontinence, and improved quality of life as per the questionnaire. In urethral pressure profile, both maximum urethral closing pressure and functional profile length increased. Ultrasonography and magnetic resonance imaging showed sustained presence of the injected adipose tissue. Enhanced ultrasonography showed a progressive increase in the blood flow to the injected area. No significant adverse events were observed peri- and postoperatively. CONCLUSION: This preliminary study showed that periurethral injection of the autologous ADSC is a safe and feasible treatment modality for stress urinary incontinence.


Assuntos
Tecido Adiposo/citologia , Prostatectomia/efeitos adversos , Transplante de Células-Tronco , Incontinência Urinária por Estresse/etiologia , Incontinência Urinária por Estresse/cirurgia , Idoso , Idoso de 80 Anos ou mais , Humanos , Injeções , Masculino , Transplante de Células-Tronco/métodos , Uretra
11.
Regen Med ; 15(7): 1891-1904, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32698666

RESUMO

Aim: Embryo implantation and subsequent pregnancy depends on endometrial thickness. To investigate potential fertility strategies for women with thin endometrium, we explored the efficacy of adipose tissue-derived regenerative cells (ADRCs) on thin endometrium and embryo implantation in a mouse model. Materials & methods: ADRCs isolated from mouse subcutaneous fat were characterized by flow cytometry. Endometrium thickness, endometrial fibrosis, embryo implantation and angiogenesis factors were evaluated in uterine cavities of ethanol-induced thin endometrium mice with ADRC transplantation. Results: ADRCs included adipose-derived stem cells and some blood vessel component cells. ADRCs improved endometrial thickness, endometrial fibrosis and embryo implantation and augmented vascular endothelial growth factor expression in the mouse uterine. Conclusion: ADRCs may be a useful therapeutic strategy to improve fertility of women with thin endometrium.


Assuntos
Tecido Adiposo/transplante , Implantação do Embrião , Endométrio/citologia , Infertilidade Feminina/terapia , Regeneração , Doenças Uterinas/terapia , Zigoto/fisiologia , Tecido Adiposo/citologia , Animais , Terapia Baseada em Transplante de Células e Tecidos , Endométrio/patologia , Feminino , Camundongos , Camundongos Endogâmicos ICR , Gravidez
12.
Sci Rep ; 10(1): 3076, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32080313

RESUMO

Adipose-derived stem cells (ADSCs) have anti-inflammatory and regenerative properties. The purpose of this study was to investigate the effect of locally administered ADSCs in a rheumatoid arthritis (RA) mouse model. In an in vivo experiment, single-cell ADSCs and three dimensionally-cultured ADSC spheroids were injected intra-articularly into the knees of RA model mice and histologically assessed. Marked improvement of synovial inflammation and articular cartilage regeneration was found in ADSC-treated mice. Proliferation, migration, and apoptosis assays of synovial fibroblasts incubated with single-cell and spheroid ADSCs were performed. The expression levels of total cytokine RNA in ADSC single cells, spheroids, and ADSC-treated inflammatory synovial fibroblasts were also evaluated by quantitative reverse transcription PCR. ADSCs suppressed the proliferation and migration of activated inflammatory cells and downregulated inflammatory cytokines. TSG-6 and TGFß1 were significantly upregulated in ADSCs compared to controls and TGFß1 was significantly upregulated in ADSC spheroids compared to single cells. The apoptosis rate of ADSC spheroids was significantly lower than that of single-cell ADSCs. These results indicated that intra-articular administration of ADSC single cells and spheroids was effective in an RA mouse model, offering a novel approach for the development of effective localized treatments for patients with RA.


Assuntos
Tecido Adiposo/patologia , Artrite Reumatoide/patologia , Artrite Reumatoide/terapia , Transplante de Células-Tronco , Animais , Cartilagem Articular/patologia , Moléculas de Adesão Celular/metabolismo , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Glucanos , Inflamação/patologia , Macrófagos/metabolismo , Camundongos Endogâmicos BALB C , Regeneração , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Membrana Sinovial/patologia , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima
13.
Circulation ; 118(2): 157-65, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18591437

RESUMO

BACKGROUND: Despite accumulating evidence that proves the pivotal role of endothelial progenitor cells (EPCs) in ischemic neovascularization, the key signaling cascade that regulates functional EPC kinetics remains unclear. METHODS AND RESULTS: In this report, we show that inactivation of specific Jagged-1 (Jag-1)-mediated Notch signals leads to inhibition of postnatal vasculogenesis in hindlimb ischemia via impairment of proliferation, survival, differentiation, and mobilization of bone marrow-derived EPCs. Bone marrow-derived EPCs obtained from Jag-1-/- mice, but not Delta-like (Dll)-1-/- mice, demonstrated less therapeutic potential for ischemic neovascularization than EPCs from the wild type. In contrast, a gain-of-function study using 3T3 stromal cells overexpressing Notch ligand revealed that Jag-1-mediated Notch signals promoted EPC commitment, which resulted in enhanced neovascularization. The impaired neovascularization in Jag-1-/- mice was profoundly rescued by transplantation of Jag-1-stimulated EPCs. CONCLUSIONS: These data indicate that specific Jag-1-derived Notch signals from the bone marrow microenvironment are critical for EPC-mediated vasculogenesis, thus providing an important clue for modulation of strategies for therapeutic neovascularization.


Assuntos
Medula Óssea/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Células Endoteliais/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Proteínas de Membrana/fisiologia , Neovascularização Fisiológica , Células-Tronco/citologia , Animais , Proteínas de Ligação ao Cálcio/deficiência , Células Endoteliais/citologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Proteína Jagged-1 , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Receptores Notch , Proteínas Serrate-Jagged , Transdução de Sinais
14.
Circ Res ; 101(6): 598-606, 2007 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-17656679

RESUMO

Estrogen has been demonstrated to promote therapeutic reendothelialization after vascular injury by bone marrow (BM)-derived endothelial progenitor cell (EPC) mobilization and phenotypic modulation. We investigated the primary hypothesis that estrogen regulates physiological postnatal vasculogenesis by modulating bioactivity of BM-derived EPCs through the estrogen receptor (ER), in cyclic hormonally regulated endometrial neovascularization. Cultured human EPCs from peripheral blood mononuclear cells (PB-MNCs) disclosed consistent gene expression of ER alpha as well as downregulated gene expressions of ER beta. Under the physiological concentrations of estrogen (17beta-estradiol, E2), proliferation and migration were stimulated, whereas apoptosis was inhibited on day 7 cultured EPCs. These estrogen-induced activities were blocked by the receptor antagonist, ICI182,780 (ICI). In BM transplanted (BMT) mice with ovariectomy (OVX) from transgenic mice overexpressing beta-galactosidase (lacZ) regulated by an endothelial specific Tie-2 promoter (Tie-2/lacZ/BM), the uterus demonstrated a significant increase in BM-derived EPCs (lacZ expressing cells) incorporated into neovasculatures detected by CD31 immunohistochemistry after E2 administration. The BM-derived EPCs that were incorporated into the uterus dominantly expressed ER alpha, rather than ER beta in BMT mice from BM of transgenic mice overexpressing EGFP regulated by Tie-2 promoter with OVX (Tie-2/EGFP/BMT/OVX) by ERs fluorescence immunohistochemistry. An in vitro assay for colony forming activity as well as flow cytometry for CD133, CD34, KDR, and VE-cadherin, using human PB-MNCs at 5 stages of the female menstrual-cycle (early-proliferative, pre-ovulatory, post-ovulatory, mid-luteal, late-luteal), revealed cycle-specific regulation of EPC kinetics. These findings demonstrate that physiological postnatal vasculogenesis involves cyclic, E2-regulated bioactivity of BM-derived EPCs, predominantly through the ER alpha.


Assuntos
Proliferação de Células , Endométrio/irrigação sanguínea , Células Endoteliais/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Neovascularização Fisiológica , Células-Tronco/metabolismo , Adulto , Animais , Apoptose , Transplante de Medula Óssea , Movimento Celular , Forma Celular , Células Cultivadas , Neovascularização da Córnea/metabolismo , Endométrio/metabolismo , Células Endoteliais/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Fulvestranto , Humanos , Cinética , Ciclo Menstrual/metabolismo , Camundongos , Camundongos Nus , Camundongos SCID , Camundongos Transgênicos , Neovascularização Fisiológica/efeitos dos fármacos , Ovariectomia , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Células-Tronco/efeitos dos fármacos
15.
Arterioscler Thromb Vasc Biol ; 27(3): 512-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17170379

RESUMO

BACKGROUND: Although potential participation of bone marrow-derived circulating endothelial progenitor cells (EPCs) to neoangiogenesis has been proposed, the precise molecular mechanisms of EPC recruitment to vascular endothelium has not been fully elucidated. METHODS AND RESULTS: Peripheral blood mononuclear cells were isolated from healthy volunteers and cultured for 7 days to obtain EPCs. Tumor necrosis factor-alpha-activated human umbilical vein endothelial cells (HUVECs) supported significantly more rolling and adhesion of EPCs compared with inactivated HUVEC monolayer. Pretreatment of activated HUVEC with an adhesion-blocking mAb to E-selectin significantly reduced EPCs adhesion to HUVECs. When HUVECs were transduced with a recombinant adenovirus of E-selectin (AdRSVE-sel) or that of beta-galactosidase (AdRSVLacZ), E-selectin-transduced but not LacZ-transduced HUVECs exhibited significantly more EPC rolling as well as adhesion. Further, effect of AdRSVE-sel or AdRSVLacZ was examined in mouse hind limb ischemic model. AdRSVE-sel-transduced mice showed significantly less limb necrosis and higher laser Doppler ratio when compared with AdRSVLacZ-transduced mice. Interestingly, blood flow recovery of ischemic limb observed in AdRSVE-sel-transduced mice was more prominent when combined with EPC administration compared with that of AdRSVLacZ-transduced mice. CONCLUSIONS: Endothelial E-selectin plays a crucial role in EPC-endothelial interaction in vitro. The importance of E-selectin was also confirmed in vivo even in the absence of exogenous EPC. These data provide molecular background for novel cell-based therapy for ischemic atherosclerosis.


Assuntos
Selectina E/farmacologia , Células Endoteliais/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Análise de Variância , Animais , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/fisiologia , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Modelos Animais , Neovascularização Fisiológica/fisiologia , Probabilidade , Sensibilidade e Especificidade , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Fator de Necrose Tumoral alfa/farmacologia
16.
Stem Cells Dev ; 16(4): 649-56, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17784838

RESUMO

Nicotine has been demonstrated to stimulate postnatal angiogenesis, having an antiapoptotic effect on endothelial cells. Given the extent of this angiogenesis-promoting effect, we hypothesized that nicotine may also stimulate postnatal vasculogenesis on endothelial progenitor cells (EPCs). Our analyses reveal some intriguing results using an in vitro assay with 2 x 10(-6) M of nicotine (smoker's average nicotine concentration and the dose of nicotine replacement therapy). The proliferation and migration activities of human EPCs cultured from peripheral blood mononuclear cells of non-smoking healthy volunteers were not affected by nicotine. The effect of nicotine on EPC survival was significantly enhanced under serum starvation on the ratio of Hoechest 33342-stained pyknotic nuclear cells as well as Annexin-V-stained cells to total cells. Furthermore, the antiapoptotic effect of nicotine was blocked completely by nicotinic acetylcholine receptor (nAChR) antagonist hexamethonium. Next, we verified how nicotine acts in vivo. Nicotine (100 ng/ml) was administered orally for 7 days before and 4 weeks after injection of cultured EPCs (1 x 10(5) /mouse) into the tail veins of 8-week-old athymic nude mice with ischemic hindlimbs. Laser doppler imaging analysis indicated that blood perfusion in the ischemic hindlimb was significantly enhanced in EPCs plus nicotine, as compared with EPCs alone. These findings suggest nicotine improves blood flow following EPC transplantation in patients with ischemic diseases.


Assuntos
Apoptose/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo/fisiologia , Células Endoteliais/transplante , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Leucócitos Mononucleares/transplante , Nicotina/farmacologia , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Técnicas de Cultura de Células , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Humanos , Fluxometria por Laser-Doppler , Camundongos
17.
Arterioscler Thromb Vasc Biol ; 26(9): 1998-2004, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16794220

RESUMO

OBJECTIVE: Tissue regeneration requires both growth factor and extracellular matrix such as collagen, serving as a scaffold for cell growth. We established FNCBD-VEGF121, consisting of the fibronectin collagen-binding domain (FNCBD) and vascular endothelial growth factor (VEGF) 121, and investigated its properties. METHODS AND RESULTS: FNCBD-VEGF121 specifically bound to gelatin and type I, II, III, IV, and V collagen. This collagen-bound FNCBD-VEGF121 captured soluble VEGF receptor 2 (VEGFR-2)/Fc chimeric protein. Cell growth-promoting activity of FNCBD-VEGF121 was almost identical to that of VEGF121. The VEGF fusion protein significantly enhanced the expression of VEGFR-2 (71.6+/-0.8%) on endothelial progenitor cells (EPCs) derived from umbilical cord blood. Expectably, the collagen-bound VEGF fusion protein not only promoted the growth of endothelial cells (ECs) but also induced the expression of VEGFR-2 (63.7+/-0.8%) on non-adherent cells expanded from bone marrow CD34+ cells. Moreover, the VEGF fusion protein enhanced sprout formation of ECs in a matrigel model. In vivo experiments revealed that FNCBD-VEGF121 had local effects but not systemic effect on EPC mobilization. CONCLUSIONS: These results suggest that FNCBD-VEGF121 stably maintains an optimally high and local concentration of VEGF with collagen matrix and stimulates both ECs and EPCs in situ, supplying a vascular regeneration niche.


Assuntos
Colágeno/metabolismo , Fibronectinas/genética , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Antígenos CD34/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular , Células Cultivadas , Tecido Conjuntivo/metabolismo , Células Endoteliais/citologia , Gelatina/metabolismo , Humanos , Monócitos/citologia , Proteínas Recombinantes de Fusão/farmacologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
PLoS One ; 11(11): e0166660, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27846321

RESUMO

Notch signaling is involved in cell fate decisions during murine vascular development and hematopoiesis in the microenvironment of bone marrow. To investigate the close relationship between hematopoietic stem cells and human endothelial progenitor cells (EPCs) in the bone marrow niche, we examined the effects of Notch signals [Jagged-1 and Delta-like ligand (Dll)-1] on the proliferation and differentiation of human CD133+ cell-derived EPCs. We established stromal systems using HESS-5 murine bone marrow cells transfected with human Jagged-1 (hJagged-1) or human Dll-1 (hDll-1). CD133+ cord blood cells were co-cultured with the stromal cells for 7 days, and then their proliferation, differentiation, and EPC colony formation was evaluated. We found that hJagged-1 induced the proliferation and differentiation of CD133+ cord blood EPCs. In contrast, hDll-1 had little effect. CD133+ cells stimulated by hJagged-1 differentiated into CD31+/KDR+ cells, expressed vascular endothelial growth factor-A, and showed enhanced EPC colony formation compared with CD133+ cells stimulated by hDll-1. To evaluate the angiogenic properties of hJagged-1- and hDll-1-stimulated EPCs in vivo, we transplanted these cells into the ischemic hindlimbs of nude mice. Transplantation of EPCs stimulated by hJagged-1, but not hDll-1, increased regional blood flow and capillary density in ischemic hindlimb muscles. This is the first study to show that human Notch signaling influences EPC proliferation and differentiation in the bone marrow microenvironment. Human Jagged-1 induced the proliferation and differentiation of CD133+ cord blood progenitors compared with hDll-1. Thus, hJagged-1 signaling in the bone marrow niche may be used to expand EPCs for therapeutic angiogenesis.


Assuntos
Antígeno AC133/genética , Diferenciação Celular/genética , Proteína Jagged-1/genética , Neovascularização Fisiológica/genética , Animais , Células da Medula Óssea/metabolismo , Proteínas de Ligação ao Cálcio , Proliferação de Células/genética , Células Progenitoras Endoteliais/metabolismo , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Membro Posterior/crescimento & desenvolvimento , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1/metabolismo , Camundongos , Nicho de Células-Tronco/genética , Transplante de Células-Tronco , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
20.
Atherosclerosis ; 249: 132-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27100923

RESUMO

BACKGROUND AND AIMS: Heparin/protamine micro/nanoparticles (LH/P-MPs) were recently developed as low-molecular weight, biodegradable carriers for adipose-derived stromal cells (ADSCs). These particles can be used for a locally delivered stem cell therapy that promotes angiogenesis. LH/P-MPs bind to the cell surface of ADSCs and promote cell-to-cell interaction and aggregation of ADSCs. Cultured ADSC/LH/P-MP aggregates remain viable. Here, we examined the ability of these aggregates to rescue limb loss in a mouse model of hindlimb ischemia. METHODS: Unilateral hindlimb ischemia was induced in adult male BALB/c mice by ligation of the iliac artery and hindlimb vein. For allotransplantation of ADSCs from the same inbred strain, we injected ADSC alone or ADSC/LH/P-MP aggregates or control medium (sham-treated) directly into the ischemic muscles. Ischemic limb blood perfusion, vessel density, and vessel area were recorded. The extent of ischemic limb necrosis or limb loss was assessed on postoperative days 2, 7, and 14. RESULTS: Compared with the sham-treatment control, treatment with ADSCs alone showed modest effects on blood perfusion recovery and increased the number of α-SMA-positive vessels. Response to ADSC/LH/P-MP aggregates was significantly greater than ADSCs alone for every endpoint. ADSC/LH/P-MP aggregates more effectively prevented the loss of ischemic hindlimbs than ADSCs alone or the sham-treatment. CONCLUSION: The LH/P-MPs augmented the effects of ADSCs on angiogenesis and reversal of limb ischemia. Use of ADSC/LH/P-MP aggregates offers a novel and convenient treatment method and potentially represents a promising new therapeutic approach to inducing angiogenesis in ischemic diseases.


Assuntos
Adipócitos/citologia , Heparina de Baixo Peso Molecular/uso terapêutico , Isquemia/terapia , Protaminas/uso terapêutico , Transplante de Células-Tronco , Células Estromais/citologia , Tecido Adiposo/citologia , Adsorção , Animais , Comunicação Celular , Sobrevivência Celular , Membro Posterior/patologia , Isquemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Fisiológica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA