Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell Transplant ; 32: 9636897231190175, 2023.
Article in English | MEDLINE | ID: mdl-37551027

ABSTRACT

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."


Subject(s)
Osteoarthritis, Knee , Humans , Adipose Tissue , Injections, Intra-Articular , Knee Joint/diagnostic imaging , Knee Joint/surgery , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/therapy , Stromal Vascular Fraction , Treatment Outcome
2.
Regen Ther ; 24: 94-102, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37363753

ABSTRACT

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.

3.
J Cell Physiol ; 237(9): 3627-3639, 2022 09.
Article in English | MEDLINE | ID: mdl-35766589

ABSTRACT

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.


Subject(s)
Chondrocytes , Tissue Inhibitor of Metalloproteinase-3 , Chondrocytes/metabolism , Collagen/metabolism , Humans , Macrophages/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Smad2 Protein/metabolism , Stromal Vascular Fraction , Tissue Inhibitor of Metalloproteinase-3/genetics , Transforming Growth Factor beta/metabolism
5.
J Clin Med ; 10(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34640324

ABSTRACT

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.

6.
Cell Transplant ; 30: 9636897211067454, 2021.
Article in English | MEDLINE | ID: mdl-35392685

ABSTRACT

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.


Subject(s)
Mesenchymal Stem Cell Transplantation , Osteoarthritis, Knee , Synovitis , Adipose Tissue/pathology , Humans , Injections, Intra-Articular/methods , Magnetic Resonance Imaging , Mesenchymal Stem Cell Transplantation/methods , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/therapy , Pain , Stromal Vascular Fraction , Treatment Outcome
7.
Regen Med ; 15(7): 1891-1904, 2020 07.
Article in English | MEDLINE | ID: mdl-32698666

ABSTRACT

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.


Subject(s)
Adipose Tissue/transplantation , Embryo Implantation , Endometrium/cytology , Infertility, Female/therapy , Regeneration , Uterine Diseases/therapy , Zygote/physiology , Adipose Tissue/cytology , Animals , Cell- and Tissue-Based Therapy , Endometrium/pathology , Female , Mice , Mice, Inbred ICR , Pregnancy
8.
BMC Musculoskelet Disord ; 21(1): 207, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32252731

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Mesenchymal Stem Cell Transplantation , Osteoarthritis, Knee/therapy , Aged , Female , Humans , Injections, Intra-Articular , Knee Joint/diagnostic imaging , Magnetic Resonance Imaging , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Pain Measurement , Radiography , Range of Motion, Articular , Treatment Outcome
9.
Sci Rep ; 10(1): 3076, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32080313

ABSTRACT

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.


Subject(s)
Adipose Tissue/pathology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/therapy , Stem Cell Transplantation , Animals , Cartilage, Articular/pathology , Cell Adhesion Molecules/metabolism , Cell Movement , Cell Proliferation , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Glucans , Inflammation/pathology , Macrophages/metabolism , Mice, Inbred BALB C , Regeneration , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Synovial Membrane/pathology , Transforming Growth Factor beta1/metabolism , Up-Regulation
10.
Surg Today ; 47(12): 1500-1511, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28555267

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/transplantation , Breast Neoplasms/surgery , Breast/surgery , Mastectomy, Segmental/methods , Plastic Surgery Procedures/methods , Transplantation, Autologous/methods , Adult , Aged , Asian People , Female , Follow-Up Studies , Humans , Middle Aged , Operative Time , Patient Satisfaction , Quality of Life , Time Factors , Treatment Outcome , Young Adult
11.
PLoS One ; 11(11): e0166660, 2016.
Article in English | MEDLINE | ID: mdl-27846321

ABSTRACT

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.


Subject(s)
AC133 Antigen/genetics , Cell Differentiation/genetics , Jagged-1 Protein/genetics , Neovascularization, Physiologic/genetics , Animals , Bone Marrow Cells/metabolism , Calcium-Binding Proteins , Cell Proliferation/genetics , Endothelial Progenitor Cells/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Hematopoietic Stem Cells/metabolism , Hindlimb/growth & development , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/metabolism , Mice , Stem Cell Niche/genetics , Stem Cell Transplantation , Vascular Endothelial Growth Factor Receptor-2/genetics
12.
Atherosclerosis ; 249: 132-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27100923

ABSTRACT

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.


Subject(s)
Adipocytes/cytology , Heparin, Low-Molecular-Weight/therapeutic use , Ischemia/therapy , Protamines/therapeutic use , Stem Cell Transplantation , Stromal Cells/cytology , Adipose Tissue/cytology , Adsorption , Animals , Cell Communication , Cell Survival , Hindlimb/pathology , Ischemia/pathology , Male , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic/drug effects
13.
Biomed Res ; 35(6): 397-405, 2014.
Article in English | MEDLINE | ID: mdl-25743346

ABSTRACT

Transplantation of cultured adipose-derived regenerative cells (ADRCs) into ischemic tissues promotes neovascularization and blood perfusion recovery. These effects are attenuated in diabetes patients. We examined the effects of hyperglycemia on the angiogenic capacity of ADRCs derived from Wistar rats both in vivo and in vitro. Cultured ADRCs were predominantly composed of CD90 positive cells; prevalence of CD90 positive cells was not affected by hyperglycemia. mRNA and protein levels of vascular endothelial growth factor (VEGF) were significantly decreased in ADRCs under hyperglycemic conditions independent of osmolarity, whereas mRNA levels of hepatocyte growth factor and fibroblast growth factor were unaffected. Since ADRCs express glucose transporter proteins GLUT1, 3 and 4, we examined the effects of the glucose transporter inhibitor phloretin on reactive oxygen species (ROS) and angiogenic factors. Phloretin decreased the glucose uptake rate, reduced ROS, and increased VEGF mRNA in ADRCs exposed to a hyperglycemic condition. In vivo transplantation of ADRCs cultured under hyperglycemic conditions into mouse ischemic limbs resulted in significantly decreased blood perfusion and capillary density in ischemic regions compared with transplantation of ADRCs cultured under normoglycemic conditions. These results suggest that hyperglycemia impaired VEGF production in ADRCs via an increase of ROS, impairing the angiogenic capacity of ADRCs transplanted into ischemic limbs.


Subject(s)
Adipose Tissue/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Hyperglycemia/metabolism , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cells, Cultured , Flow Cytometry , Mice , Neovascularization, Physiologic/physiology , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Regeneration/physiology , Vascular Endothelial Growth Factor A/genetics
14.
Cell Transplant ; 21(2-3): 387-99, 2012.
Article in English | MEDLINE | ID: mdl-22793046

ABSTRACT

Human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells have an enormous potential; however, their potential clinical application is being arrested due to various limitations such as teratoma formation followed by tumorigenesis, emergent usage, and the quality control of cells, as well as safety issues regarding long-term culture are also delaying their clinical application. In addition, human ES cells have two crucial issues: immunogenicity and ethical issues associated with their clinical application. The efficient generation of human iPS cells requires gene transfer, yet the mechanism underlying pluripotent stem cell induction has not yet been fully elucidated. Otherwise, although human adult regenerative cells including mesenchymal stem cells have a limited capacity for differentiation, they are nevertheless promising candidates for tissue regeneration in a clinical setting. This review highlights the use of regenerative cells for transplantation in hepatic failure.


Subject(s)
Embryonic Stem Cells/cytology , Liver Failure/therapy , Mesenchymal Stem Cell Transplantation , Regenerative Medicine , Adipose Tissue/cytology , Bone Marrow Cells/cytology , Cell Differentiation , Embryonic Stem Cells/immunology , Embryonic Stem Cells/transplantation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Tissue Engineering
15.
Int J Urol ; 19(7): 652-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22435469

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/transplantation , Multipotent Stem Cells/transplantation , Urinary Incontinence, Stress/therapy , Aged , Humans , Injections , Magnetic Resonance Imaging , Male , Quality of Life , Treatment Outcome , Ultrasonography , Urethra/diagnostic imaging , Urinary Incontinence, Stress/diagnostic imaging , Urodynamics
16.
Surg Today ; 41(1): 18-23, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21191687

ABSTRACT

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.


Subject(s)
Adipocytes/physiology , Mesenchymal Stem Cells/physiology , Regenerative Medicine/methods , Humans , Stem Cell Transplantation/methods , Tissue and Organ Harvesting/methods
17.
Curr Stem Cell Res Ther ; 5(2): 182-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19941447

ABSTRACT

Severe hepatic dysfunctions including hepatic cirrhosis and hepatocarcinoma are life-threatening conditions for which effective medical treatments are needed. With the only effective treatment to date being orthotropic liver transplantation, alternative approaches are needed because of the limited number of donors and the possibility of immune-rejection. One alternative is regenerative medicine, which holds promise for the development of a cell-based therapy enabling hepatic regeneration through transplantation of adipose tissue-derived mesenchymal stem cells (AT-MSCs) or hepatocyte-like cells generated from AT-MSCs. When compared with embryonic stem (ES) cells and induced pluripotent stem (iPS) cells, the use of AT-MSCs as regenerative cells would be advantageous in regard to ethical and safety issues since AT-MSCs are somatic cells and have the potential to be used without in vitro culture. These autologous cells are immuno-compatible and exhibit controlled differentiation and multi-functional abilities and do not undergo post-transplantation rejection or unwanted differentiation such as formation of teratomas. AT-MSC-based therapies may provide a novel approach for hepatic regeneration and hepatocyte differentiation and thereby support hepatic function in diseased individuals.


Subject(s)
Adipose Tissue/pathology , Carcinoma, Hepatocellular/therapy , Liver Cirrhosis/therapy , Liver Neoplasms/therapy , Mesenchymal Stem Cells/metabolism , Stem Cell Niche/pathology , Stem Cell Transplantation , Animals , Carcinoma, Hepatocellular/pathology , Cell Differentiation , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Immune Tolerance , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Liver Regeneration , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/pathology , Regenerative Medicine
18.
Int J Urol ; 17(1): 75-82, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20002225

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Prostatectomy/adverse effects , Stem Cell Transplantation , Urinary Incontinence, Stress/etiology , Urinary Incontinence, Stress/surgery , Aged , Aged, 80 and over , Humans , Injections , Male , Stem Cell Transplantation/methods , Urethra
19.
Circulation ; 118(2): 157-65, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18591437

ABSTRACT

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.


Subject(s)
Bone Marrow/physiology , Calcium-Binding Proteins/physiology , Endothelial Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Neovascularization, Physiologic , Stem Cells/cytology , Animals , Calcium-Binding Proteins/deficiency , Endothelial Cells/cytology , Intercellular Signaling Peptides and Proteins/deficiency , Jagged-1 Protein , Membrane Proteins/deficiency , Mice , Mice, Knockout , Receptors, Notch , Serrate-Jagged Proteins , Signal Transduction
20.
PLoS One ; 3(3): e1789, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18335059

ABSTRACT

BACKGROUND: Cellular cardiomyoplasty for myocardial infarction has been developed using various cell types. However, complete differentiation and/or trans-differentiation into cardiomyocytes have never occurred in these transplant studies, whereas functional contributions were reported. METHODS AND RESULTS: Skeletal muscle interstitium-derived CD34(+)/CD45(-) (Sk-34) cells were purified from green fluorescent protein transgenic mice by flowcytometory. Cardiac differentiation of Sk-34 cells was examined by in vitro clonal culture and co-culture with embryonic cardiomyocytes, and in vivo transplantation into a nude rat myocardial infarction (MI) model (left ventricle). Lower relative expression of cardiomyogenic transcription factors, such as GATA-4, Nkx2-5, Isl-1, Mef2 and Hand2, was seen in clonal cell culture. However, vigorous expression of these factors was seen on co-culture with embryonic cardiomyocytes, together with formation of gap-junctions and synchronous contraction following sphere-like colony formation. At 4 weeks after transplantation of freshly isolated Sk-34 cells, donor cells exhibited typical cardiomyocyte structure with formation of gap-junctions, as well as intercalated discs and desmosomes, between donor and recipient and/or donor and donor cells. Fluorescence in situ hybridization (FISH) analysis detecting the rat and mouse genomic DNA and immunoelectron microscopy using anti-GFP revealed donor-derived cells. Transplanted Sk-34 cells were incorporated into infarcted portions of recipient muscles and contributed to cardiac reconstitution. Significant improvement in left ventricular function, as evaluated by transthoracic echocardiography and micro-tip conductance catheter, was also observed. CONCLUSIONS AND SIGNIFICANCE: Skeletal muscle-derived multipotent Sk-34 cells that can give rise to skeletal and smooth muscle cells as reported previously, also give rise to cardiac muscle cells as multi-myogenic stem cells, and thus are a potential source for practical cellular cardiomyoplasty.


Subject(s)
Cell Transplantation , Muscle, Skeletal/cytology , Myocardium/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Coculture Techniques , Green Fluorescent Proteins/genetics , Immunophenotyping , In Situ Hybridization, Fluorescence , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...