Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 471
Filter
1.
N Engl J Med ; 385(24): 2264-2270, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34881838

ABSTRACT

Inherited junctional epidermolysis bullosa is a severe genetic skin disease that leads to epidermal loss caused by structural and mechanical fragility of the integuments. There is no established cure for junctional epidermolysis bullosa. We previously reported that genetically corrected autologous epidermal cultures regenerated almost an entire, fully functional epidermis on a child who had a devastating form of junctional epidermolysis bullosa. We now report long-term clinical outcomes in this patient. (Funded by POR FESR 2014-2020 - Regione Emilia-Romagna and others.).


Subject(s)
Epidermis/transplantation , Epidermolysis Bullosa, Junctional/therapy , Keratinocytes/transplantation , Transduction, Genetic , Transgenes , Cell Self Renewal , Cells, Cultured/transplantation , Child , Clone Cells , Epidermis/pathology , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/pathology , Follow-Up Studies , Genetic Diseases, Inborn/pathology , Genetic Diseases, Inborn/therapy , Genetic Therapy , Genetic Vectors , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Male , Regeneration , Stem Cells/physiology , Transplantation, Autologous
2.
J Ocul Pharmacol Ther ; 37(4): 200-208, 2021 05.
Article in English | MEDLINE | ID: mdl-33481657

ABSTRACT

Purpose: The aim of this study was to investigate the microRNA (miRNA) expressions of the corneal tissue after an alkaline burn and to compare the efficiency of adipose- and bone marrow-derived mesenchymal stem cells (MSCs) on expressions. Methods: Thirty-two rats were divided into 4 groups. No intervention was made in the control group. A chemical burn was created by applying 4 µL NaOH soaked in 6 mm filter paper to the right eye of each animal in the other groups. Whereas only subconjunctival 0.1 mL phosphate-buffered saline (PBS) was injected to in the group 1, 2 × 106 adipose- or bone marrow-derived MSC in 0.1 mL PBS was injected subconjunctivally to the animals in the remaining groups (groups 2 and 3, respectively). Tissue samples were collected for miRNA analysis on the third day after the burn. Results: When group 1 was compared with the control group, the expression of 3 of 93 miRNAs increased significantly, whereas the expression of 50 miRNAs decreased significantly. Significant changes in miRNA expressions were observed when group 1 was compared with groups 2 and 3. Although a significant change was observed in the expression of 6 miRNAs in the adipose-derived MSC group, it was found that the expression of 65 miRNAs significantly changed in the bone marrow-derived MSC group. Conclusion: This study shows that there are significant changes in some miRNA expressions after corneal alkaline burn and these changes can be reversed with the subconjunctival injection of MSCs.


Subject(s)
Burns/metabolism , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Animals , Bone Marrow Cells/metabolism , Burns/therapy , Case-Control Studies , Cells, Cultured/transplantation , Cornea/metabolism , Corneal Injuries/chemically induced , Corneal Injuries/pathology , Disease Models, Animal , Male , Microscopy, Fluorescence/methods , Rats , Rats, Sprague-Dawley
3.
Acta Ophthalmol ; 99(3): e302-e309, 2021 May.
Article in English | MEDLINE | ID: mdl-32914585

ABSTRACT

Anterior lens capsule, as the thickest basement membrane in the body, has its unique physiology characteristics. In ophthalmology, many attempts have been made to culture different kinds of cells including iris pigment epithelial cells, retinal pigment epithelial cells, corneal epithelium and endothelium cells, trabecular meshwork cells etc and anterior lens capsule has been confirmed to be served as an excellent scaffold for the growth and expansion of different ocular cells. Furthermore, anterior lens capsule also has unique potential in gestation evaluation and the treatment of various ocular diseases, including corneal ulcer, glaucoma, age-related macular degeneration and macular hole, etc. Here, we provide an overview of the biomechanical properties and biomedical engineering perspectives of anterior lens capsule.


Subject(s)
Anterior Capsule of the Lens/cytology , Biomedical Engineering/methods , Animals , Cells, Cultured/transplantation , Eye Diseases/therapy , Humans
5.
Expert Rev Gastroenterol Hepatol ; 14(3): 185-196, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32098516

ABSTRACT

Introduction: Hepatocyte transplantation (HT) is a promising alternative to liver transplantation for the treatment of liver-based metabolic diseases and acute liver failure (ALF). However, shortage of good-quality liver tissues, early cell loss post-infusion, reduced cell engraftment and function restricts clinical application.Areas covered: A comprehensive literature search was performed to cover pre-clinical and clinical HT studies. The review discusses the latest developments to address HT limitations: cell sources from marginal/suboptimal donors to neonatal livers, differentiating pluripotent stem cells into hepatocyte-like cells, in vitro expansion, prevention of immune response to transplanted cells by encapsulation or using innate immunity-inhibiting agents, and enhancing engraftment through partial hepatectomy or irradiation.Expert opinion: To date, published data are highly encouraging specially the alginate-encapsulated hepatocyte treatment of children with ALF. Hepatocyte functions can be further improved through co-culturing with mesenchymal stromal cells. Moreover, ex-vivo genetic correction will enable the use of autologous cells in future personalized medicine.


Subject(s)
Cell Transplantation/standards , Hepatocytes/transplantation , Liver Diseases/therapy , Cell Transplantation/methods , Cells, Cultured/transplantation , Humans , Liver Diseases/metabolism , Liver Diseases/surgery , Liver Transplantation
6.
Cancer Immunol Immunother ; 69(5): 825-834, 2020 May.
Article in English | MEDLINE | ID: mdl-32060687

ABSTRACT

Adjuvant cytokine-induced killer (CIK) cell immunotherapy has shown potential in improving the prognosis of hepatocellular carcinoma (HCC) patients after curative resection. However, whether an individual could obtain survival benefit from CIK cell treatment remains unknown. In the present study, we focused on the characteristics of CIK cells and aimed to identify the best predictive biomarker for adjuvant CIK cell treatment in patients with HCC after surgery. This study included 48 patients with HCC treated with postoperative adjuvant CIK cell immunotherapy. The phenotype activity and cytotoxic activity of CIK cells were determined by flow cytometry and xCELLigence™ Real-Time Cell Analysis (RTCA) system, respectively. Correlation analysis revealed that the cytotoxic activity of CIK cells was significantly negative correlated with the percentage of CD3+ CD4+ cell subsets, but significantly positive correlated with CD3-CD56+ and CD3+ CD56+ cell subsets. Survival analysis showed that there were no significant associations between patients' prognosis and the phenotype of CIK cells. By contrast, there was statistically significant improvement in recurrence-free survival (RFS) and overall survival (OS) for patients with high cytotoxic activity of CIK cells as compared with those with low cytotoxic activity of CIK cells. Univariate and multivariate analyses indicated that CIK cell cytotoxicity was an independent prognostic factor for RFS and OS. In conclusion, a high cytotoxic activity of CIK cells can serve as a valuable biomarker for adjuvant CIK cell immunotherapy of HCC patients after surgery.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cytokine-Induced Killer Cells/transplantation , Cytotoxicity, Immunologic , Immunotherapy/methods , Liver Neoplasms/therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Cell Culture Techniques , Cells, Cultured/immunology , Cells, Cultured/transplantation , Combined Modality Therapy/methods , Cytokine-Induced Killer Cells/immunology , Cytotoxicity Tests, Immunologic , Female , Flow Cytometry , Hepatectomy , Humans , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Male , Middle Aged , Postoperative Period , Prognosis , Survival Analysis , Transplantation, Autologous/methods
7.
Transplantation ; 104(1): 190-196, 2020 01.
Article in English | MEDLINE | ID: mdl-31365472

ABSTRACT

BACKGROUND: Europe is currently the most active region in the field of pancreatic islet transplantation, and many of the leading groups are actually achieving similar good outcomes. Further collaborative advances in the field require the standardization of islet cell product isolation processes, and this work aimed to identify differences in the human pancreatic islet isolation processes within European countries. METHODS: A web-based questionnaire about critical steps, including donor selection, pancreas processing, pancreas perfusion and digestion, islet counting and culture, islet quality evaluation, microbiological evaluation, and release criteria of the product, was completed by isolation facilities participating at the Ninth International European Pancreas and Islet Transplant Association (EPITA) Workshop on Islet-Beta Cell Replacement in Milan. RESULTS: Eleven islet isolation facilities completed the questionnaire. The facilities reported 445 and 53 islet isolations per year over the last 3 years from deceased organ donors and pancreatectomized patients, respectively. This activity resulted in 120 and 40 infusions per year in allograft and autograft recipients, respectively. Differences among facilities emerged in donor selection (age, cold ischemia time, intensive care unit length, amylase concentration), pancreas procurement, isolation procedures (brand and concentration of collagenase, additive, maximum acceptable digestion time), quality evaluation, and release criteria for transplantation (glucose-stimulated insulin secretion tests, islet numbers, and purity). Moreover, even when a high concordance about the relevance of one parameter was evident, thresholds for the acceptance were different among facilities. CONCLUSIONS: The result highlighted the presence of a heterogeneity in the islet cell product process and product release criteria.


Subject(s)
Cell Separation/methods , Donor Selection/methods , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Tissue and Organ Harvesting/methods , Adolescent , Adult , Age Factors , Aged , Cell Count/standards , Cell Count/statistics & numerical data , Cell Separation/statistics & numerical data , Cells, Cultured/transplantation , Child , Child, Preschool , Cold Ischemia/standards , Cold Ischemia/statistics & numerical data , Donor Selection/standards , Donor Selection/statistics & numerical data , Europe , Humans , Infant , Infant, Newborn , Islets of Langerhans Transplantation/standards , Middle Aged , Perfusion/methods , Perfusion/statistics & numerical data , Practice Guidelines as Topic , Primary Cell Culture/methods , Primary Cell Culture/standards , Primary Cell Culture/statistics & numerical data , Surveys and Questionnaires/statistics & numerical data , Time Factors , Tissue and Organ Harvesting/standards , Tissue and Organ Harvesting/statistics & numerical data , Young Adult
8.
Lancet Haematol ; 7(2): e134-e145, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31704264

ABSTRACT

BACKGROUND: Benefits of cord blood transplantation include low rates of relapse and chronic graft-versus-host disease (GVHD). However, the use of cord blood is rapidly declining because of the high incidence of infections, severe acute GVHD, and transplant-related mortality. UM171, a haematopoietic stem cell self-renewal agonist, has been shown to expand cord blood stem cells and enhance multilineage blood cell reconstitution in mice. We aimed to investigate the safety and feasibility of single UM171-expanded cord blood transplantation in patients with haematological malignancies who do not have a suitable HLA-matched donor. METHODS: This single-arm, open-label, phase 1-2 safety and feasibility study was done at two hospitals in Canada. The study had two parts. In part 1, patients received two cord blood units (one expanded with UM171 and one unmanipulated cord blood) until UM171-expanded cord blood demonstrated engraftment. Once engraftment was documented we initiated part 2, reported here, in which patients received a single UM171-expanded cord blood unit with a dose de-escalation design to determine the minimal cord blood unit cell dose that achieved prompt engraftment. Eligible patients were aged 3-64 years, weighed 12 kg or more, had a haematological malignancy with an indication for allogeneic hematopoietic stem cell transplant and did not have a suitable HLA-matched donor, and a had a Karnofsky performance status score of 70% or more. Five clinical sites were planned to participate in the study; however, only two study sites opened, both of which only treated adult patients, thus no paediatric patients (aged <18 years) were recruited. Patients aged younger than 50 years without comorbidities received a myeloablative conditioning regimen (cyclophosphamide 120 mg/kg, fludarabine 75 mg/m2, and 12 Gy total body irradiation) and patients aged older than 50 years and those with comorbidities received a less myeloablative conditioning regimen (cyclophosphamide 50 mg/kg, thiotepa 10 mg/kg, fludarabine 150 mg/m2, and 4 Gy total body irradiation). Patients were infused with the 7-day UM171-expanded CD34-positive cells and the lymphocyte-containing CD34-negative fraction. The primary endpoints were feasibility of UM171 expansion, safety of the transplant, kinetics of hematopoietic reconstitution (time to neutrophil and platelet engraftment) of UM171-expanded cord blood, and minimal pre-expansion cord blood unit cell dose that achieved prompt engraftment. We analysed feasibility in all enrolled patients and all other primary outcomes were analysed per protocol, in all patients who received single UM171-expanded cord blood transplantation. This trial has been completed and was registered with ClinicalTrials.gov, NCT02668315. FINDINGS: Between Feb 17, 2016, and Nov 11, 2018, we enrolled 27 patients, four of whom received two cord blood units for safety purposes in part 1 of the study. 23 patients were subsequently enrolled in part 2 to receive a single UM171-expanded cord blood transplant and 22 patients received a single UM171-expanded cord blood transplantation. At data cutoff (Dec 31, 2018), median follow-up was 18 months (IQR 12-22). The minimal cord blood unit cell dose at thaw that achieved prompt engraftment as a single cord transplant after UM171 expansion was 0·52 × 105 CD34-positive cells. We successfully expanded 26 (96%) of 27 cord blood units with UM171. Among the 22 patients who received single UM171-expanded cord blood transplantation, median time to engraftment of 100 neutrophils per µL was 9·5 days (IQR 8-12), median time to engraftment of 500 neutrophils per µL was 18 days (12·5-20·0), and no graft failure occurred. Median time to platelet recovery was 42 days (IQR 35-47). The most common non-haematological adverse events were grade 3 febrile neutropenia (16 [73%] of 22 patients) and bacteraemia (nine [41%]). No unexpected adverse events were observed. One (5%) of 22 patients died due to treatment-related diffuse alveolar haemorrhage. INTERPRETATION: Our preliminary findings suggest that UM171 cord blood stem cell expansion is feasible, safe, and allows for the use of small single cords without compromising engraftment. UM171-expanded cord blood might have the potential to overcome the disadvantages of other cord blood transplants while maintaining the benefits of low risk of chronic GVHD and relapse, and warrants further investigation in randomised trials. FUNDING: Canadian Institutes of Health Research, Canadian Cancer Society and Stem Cell Network.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/drug effects , Indoles/pharmacology , Pyrimidines/pharmacology , Adolescent , Adult , Cell Self Renewal/drug effects , Cells, Cultured/drug effects , Cells, Cultured/transplantation , Cord Blood Stem Cell Transplantation/adverse effects , Disease-Free Survival , Feasibility Studies , Febrile Neutropenia/etiology , Female , Graft Survival , Graft vs Host Disease/etiology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/cytology , Humans , Infant, Newborn , Male , Middle Aged , Proportional Hazards Models , Treatment Outcome , Young Adult
9.
Immunol Lett ; 216: 97-105, 2019 12.
Article in English | MEDLINE | ID: mdl-31622634

ABSTRACT

Mesenchymal stem cell (MSCs) therapy are among new strategies that are used to combat infections through immunomodulation. Cell number, route and frequency of injection and the duration of exposure to the infectious agent are of the main factors to determine the effectiveness of cell therapy. The current study was aimed to assess the effect of multiple intravenous (i.v.) injection of adipose tissue derived (AD)-MSCs on immune response of Leishmania (L.) major-infected BALB/c mice. Therefore, infected mice received AD-MSCs four times during the early phase of infection through i.v. route. They were then monitored weekly for footpad swelling and lesion development. Parasite burden, nitric oxide (NO) and cytokine production were measured in the spleen and lymph node 90 days post-infection. Delayed lesion development, significant reduction in footpad swelling and lower parasite burden in the spleen of AD-MSCs-treated mice showed the relative effect of AD-MSCs therapy in the control of L. major dissemination. In addition, MSCs were able to manage direct cytokine responses toward T-helper 1 (Th1). Although the level of interleukin (IL)-10 was still higher than the associated level of tumor necrosis factor (TNF)-α, a shift towards higher level of TNF-α was also observed.


Subject(s)
Abdominal Fat/cytology , Leishmania major/immunology , Leishmaniasis, Cutaneous/therapy , Mesenchymal Stem Cell Transplantation/methods , Th1 Cells/immunology , Animals , Cells, Cultured/transplantation , Disease Models, Animal , Female , Humans , Injections, Intravenous , Leishmania major/isolation & purification , Leishmaniasis, Cutaneous/blood , Leishmaniasis, Cutaneous/immunology , Lymph Nodes/immunology , Lymph Nodes/parasitology , Mice , Mice, Inbred BALB C , Parasite Load , Primary Cell Culture , Skin/immunology , Skin/parasitology , Spleen/immunology , Spleen/parasitology
10.
Am J Physiol Gastrointest Liver Physiol ; 317(6): G845-G852, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31589468

ABSTRACT

Intestinal organoid cultures provide an in vitro model system for studying pathways and mechanisms involved in epithelial damage and repair. Derived from either embryonic or induced pluripotent stem cells or adult intestinal stem cells or tissues, these self-organizing, multicellular structures contain polarized mature cells that recapitulate both the physiology and heterogeneity of the intestinal epithelium. These cultures provide a cutting-edge technology for defining regenerative pathways that are induced following radiation or chemical damage, which directly target the cycling intestinal stem cell, or damage resulting from viral, bacterial, or parasitic infection of the epithelium. Novel signaling pathways or biological mechanisms identified from organoid studies that mediate regeneration of the epithelium following damage are likely to be important targets of preventive or therapeutic modalities to mitigate intestinal injury. The evolution of these cultures to include more components of the intestinal wall and the ability to genetically modify them are key components for defining the mechanisms that modulate epithelial regeneration.


Subject(s)
Adult Stem Cells , Intestinal Diseases , Intestines , Organoids , Regeneration/physiology , Animals , Cells, Cultured/physiology , Cells, Cultured/transplantation , Humans , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Intestinal Diseases/therapy , Intestines/drug effects , Intestines/radiation effects , Models, Biological , Organoids/physiology , Organoids/transplantation , Tissue Engineering/methods
11.
Cells ; 8(9)2019 09 11.
Article in English | MEDLINE | ID: mdl-31514443

ABSTRACT

Muscular dystrophies (MDs) are a group of heterogeneous genetic disorders caused by mutations in the genes encoding the structural components of myofibres. The current state-of-the-art treatment is oligonucleotide-based gene therapy that restores disease-related protein. However, this therapeutic approach has limited efficacy and is unlikely to be curative. While the number of studies focused on cell transplantation therapy has increased in the recent years, this approach remains challenging due to multiple issues related to the efficacy of engrafted cells, source of myogenic cells, and systemic injections. Technical innovation has contributed to overcoming cell source challenges, and in recent studies, a combination of muscle resident stem cells and gene editing has shown promise as a novel approach. Furthermore, improvement of the muscular environment both in cultured donor cells and in recipient MD muscles may potentially facilitate cell engraftment. Artificial skeletal muscle generated by myogenic cells and muscle resident cells is an alternate approach that may enable the replacement of damaged tissues. Here, we review the current status of myogenic stem cell transplantation therapy, describe recent advances, and discuss the remaining obstacles that exist in the search for a cure for MD patients.


Subject(s)
Cell Engineering/methods , Cells, Cultured/transplantation , Muscle, Skeletal , Muscular Dystrophies/therapy , Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Humans , Mice , Muscle Cells/cytology , Muscle Development/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/pathology
12.
Stem Cell Res Ther ; 10(1): 177, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31200769

ABSTRACT

BACKGROUND: Human decidual stromal cells (DSCs) are involved in the maintenance and development of pregnancy, in which they play a key role in the induction of immunological maternal-fetal tolerance. Precursors of DSCs (preDSCs) are located around the vessels, and based on their antigen phenotype, previous studies suggested a relationship between preDSCs and mesenchymal stromal/stem cells (MSCs). This work aimed to further elucidate the MSC characteristics of preDSCs. METHODS: We established 15 human preDSC lines and 3 preDSC clones. Physiological differentiation (decidualization) of these cell lines and clones was carried out by in vitro culture with progesterone (P4) and cAMP. Decidualization was confirmed by the change in cellular morphology and prolactin (PRL) secretion, which was determined by enzyme immunoassay of the culture supernatants. We also studied MSC characteristics: (1) In mesenchymal differentiation, under appropriate culture conditions, these preDSC lines and clones differentiated into adipocytes, osteoblasts, and chondrocytes, and differentiation was confirmed by cytochemical assays and RT-PCR. (2) The expression of stem cell markers was determined by RT-PCR. (3) Cloning efficiency was evaluated by limited dilution. (4) Immunoregulatory activity in vivo was estimated in DBA/2-mated CBA/J female mice, a murine model of immune-based recurrent abortion. (5) Survival of preDSC in immunocompetent mice was analyzed by RT-PCR and flow cytometry. RESULTS: Under the effect of P4 and cAMP, the preDSC lines and clones decidualized in vitro: the cells became rounder and secreted PRL, a marker of physiological decidualization. PreDSC lines and clones also exhibited MSC characteristics. They differentiated into adipocytes, osteoblasts, and chondrocytes, and preDSC lines expressed stem cell markers OCT-4, NANOG, and ABCG2; exhibited a cloning efficiency of 4 to 15%; significantly reduced the embryo resorption rate (P < 0.001) in the mouse model of abortion; and survived for prolonged periods in immunocompetent mice. The fact that 3 preDSC clones underwent both decidualization and mesenchymal differentiation shows that the same type of cell exhibited both DSC and MSC characteristics. CONCLUSIONS: Together, our results confirm that preDSCs are decidual MSCs and suggest that these cells are involved in the mechanisms of maternal-fetal immune tolerance.


Subject(s)
Abortion, Habitual/therapy , Abortion, Spontaneous/therapy , Decidua/transplantation , Mesenchymal Stem Cell Transplantation , Abortion, Habitual/pathology , Abortion, Spontaneous/pathology , Animals , Cell Differentiation , Cells, Cultured/transplantation , Decidua/cytology , Disease Models, Animal , Endometrium/cytology , Endometrium/transplantation , Female , Humans , Mesenchymal Stem Cells/cytology , Mice , Pregnancy
13.
J Invest Dermatol ; 139(10): 2115-2124.e11, 2019 10.
Article in English | MEDLINE | ID: mdl-31054844

ABSTRACT

Inherited skin disorders have been reported recently to have sporadic normal-looking areas, where a portion of the keratinocytes have recovered from causative gene mutations (revertant mosaicism). We observed a case of recessive dystrophic epidermolysis bullosa treated with cultured epidermal autografts (CEAs), whose CEA-grafted site remained epithelized for 16 years. We proved that the CEA product and the grafted area included cells with revertant mosaicism. Based on these findings, we conducted an investigator-initiated clinical trial of CEAs from clinically revertant skin for recessive dystrophic epidermolysis bullosa. The donor sites were analyzed by genetic analysis, immunofluorescence, electron microscopy, and quantification of the reverted mRNA with deep sequencing. The primary endpoint was the ulcer epithelization rate per patient at 4 weeks after the last CEA application. Three patients with recessive dystrophic epidermolysis bullosa with 8 ulcers were enrolled, and the epithelization rate for each patient at the primary endpoint was 87.7%, 100%, and 57.0%, respectively. The clinical effects were found to persist for at least 76 weeks after CEA transplantation. One of the three patients had apparent revertant mosaicism in the donor skin and in the post-transplanted area. CEAs from clinically normal skin are a potentially well-tolerated treatment for recessive dystrophic epidermolysis bullosa.


Subject(s)
Epidermal Cells/transplantation , Epidermis/transplantation , Epidermolysis Bullosa Dystrophica/pathology , Epidermolysis Bullosa Dystrophica/surgery , Skin Transplantation/methods , Wound Healing/physiology , Adult , Autografts/transplantation , Biopsy, Needle , Cells, Cultured/transplantation , Child , Epidermolysis Bullosa Dystrophica/genetics , Female , Follow-Up Studies , Humans , Immunohistochemistry , Japan , Male , Middle Aged , Pilot Projects , Risk Assessment , Severity of Illness Index , Time Factors , Treatment Outcome
14.
Medicine (Baltimore) ; 97(48): e13313, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30508922

ABSTRACT

RATIONALE: This study reviewed the use of a combination of meshed dermis graft and cultured epithelial autografts (CEA) made in Japan "JACE" (JACE; Japan Tissue Engineering Co., Ltd. Japan) for the treatment of massively burns. JACE is a Green-type CEA. We recently described a method in which we prepare the wound bed for burned patients by using artificial dermis and graft with JACE on a meshed 6:1 split-thickness autograft. In this report, we used a meshed 3:1 split-thickness dermis graft without epithelial cells. There are several reports of combination of using CEA on meshed split-thickness autograft, however this is the first report of using CEA on meshed split-thickness dermis graft. PATIENT CONCERNS AND DIAGNOSIS: Between March 2015 and August 2017, 3 burn patients were enrolled in this study. The patients ranged in age from 51 to 66 years. All 3 patients suffered severe burn injury that caused by flame. % Total Body Surface Area (TBSA) burned were ranged from 37.5% to 69%. INTERVENTIONS: All patients received surgical treatment with tangential excision within a week from admission. We implanted artificial dermis immediately after debridement. Basically, we applied meshed 6:1 split-thickness autografts to the wound bed and covered with JACE. However, in the absence of split-thickness autografts, we used a meshed 3:1 split-thickness dermis graft instead of a meshed 6:1 split-thickness autograft. OUTCOMES: At 3 weeks after the transplantation of JACE, the take rate for JACE sheets was >60% on the meshed 3:1 split-thickness dermis graft. Furthermore, almost all of the burn wounds had healed at 6 weeks after surgery. LESSONS: We observed good results by grafting JACE on meshed 3:1 dermis graft. With this new method, it is possible to cover a large burn wound by harvesting tissue from only a small site.


Subject(s)
Burns/surgery , Skin Transplantation/methods , Aged , Burns/pathology , Cells, Cultured/transplantation , Dermatologic Surgical Procedures/methods , Epithelium/transplantation , Humans , Injury Severity Score , Male , Middle Aged , Skin, Artificial , Tissue Culture Techniques/methods , Transplantation, Autologous/methods , Treatment Outcome
15.
Sci Rep ; 8(1): 12060, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30104715

ABSTRACT

Regenerative therapeutic approaches for myocardial diseases often involve delivery of stem cells expanded ex vivo. Prior studies indicate that cell culture conditions affect functional and phenotypic characteristics, but relationship(s) of cultured cells derived from freshly isolated populations and the heterogeneity of the cultured population remain poorly defined. Functional and phenotypic characteristics of ex vivo expanded cells will determine outcomes of interventional treatment for disease, necessitating characterization of the impact that ex vivo expansion has upon isolated stem cell populations. Single-cell RNA-Seq profiling (scRNA-Seq) was performed to determine consequences of culture expansion upon adult cardiac progenitor cells (CPCs) as well as relationships with other cell populations. Bioinformatic analyses demonstrate that identity marker genes expressed in freshly isolated cells become undetectable in cultured CPCs while low level expression emerges for thousands of other genes. Transcriptional profile of CPCs exhibited greater degree of similarity throughout the cultured population relative to freshly isolated cells. Findings were validated by comparative analyses using scRNA-Seq datasets of various cell types generated by multiple scRNA-Seq technology. Increased transcriptome diversity and decreased population heterogeneity in the cultured cell population may help account for reported outcomes associated with experimental and clinical use of CPCs for treatment of myocardial injury.


Subject(s)
Adult Stem Cells/physiology , Cells, Cultured/physiology , Myocytes, Cardiac/physiology , Stem Cell Transplantation/methods , Adult , Adult Stem Cells/transplantation , Animals , Cell Differentiation/genetics , Cells, Cultured/transplantation , Computational Biology , Datasets as Topic , Female , Gene Expression Profiling/methods , Heart Failure/pathology , Heart Failure/therapy , Humans , Mice , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/transplantation , Primary Cell Culture/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome/genetics , Treatment Outcome
16.
Sci Rep ; 8(1): 10502, 2018 Jul 12.
Article in English | MEDLINE | ID: mdl-30002380

ABSTRACT

Limbal stem cell deficiency can be treated with transplantation of cultured human limbal epithelial cells (LEC). It can be advantageous to produce LEC in centralized labs and thereafter ship them to eye clinics. The present study used transport simulations of LEC to determine if vigorous shaking during transport altered the viability, morphology and phenotype during a 4 day-long storage of LEC with a previously described serum-free storage method. Inserts with LEC cultured on amniotic membranes were sutured to caps inside air-tight containers with generous amounts of 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES)-buffered minimal essential medium (MEM). The containers were distributed among the following testing conditions: 6 hours with full containers, 36 hours with full containers, 36 hours with container three quarters full of medium, and 36 hours with container full of medium containing a shear-protecting agent (Pluronic-F68). Compared to stored, but non-transported controls, no statistically significant changes in viability and immunohistochemical staining were observed. The epithelial sheets remained intact. However, an air-liquid interface in the containers reduced the number of desmosomes and hemi-desmosomes compared to the controls. In conclusion, cultured LEC sheets appear to endure vigorous shaking for at least 36 hours if the container is full.


Subject(s)
Corneal Diseases/surgery , Epithelium, Corneal/transplantation , Limbus Corneae/pathology , Stem Cell Transplantation/methods , Transportation , Aged , Aged, 80 and over , Cell Adhesion , Cell Survival , Cells, Cultured/transplantation , Cells, Cultured/ultrastructure , Corneal Diseases/pathology , Epithelium, Corneal/cytology , Humans , Limbus Corneae/cytology , Male , Microscopy, Electron, Transmission , Stem Cells/pathology , Stem Cells/ultrastructure
17.
Adv Exp Med Biol ; 1059: 219-240, 2018.
Article in English | MEDLINE | ID: mdl-29736576

ABSTRACT

Stem cell research plays a central role in the future of medicine, which is mainly dependent on the advances on regenerative medicine (RM), specifically in the disciplines of tissue engineering (TE) and cellular therapeutics. All RM strategies depend upon the harnessing, stimulation, or guidance of endogenous developmental or repair processes in which cells have an important role. Among the most clinically challenging disorders, cartilage degeneration, which also affects subchondral bone becoming an osteochondral (OC) defect, is one of the most demanding. Although primary cells have been clinically applied, stem cells are currently seen as the promising tool of RM-related research because of its availability, in vitro proliferation ability, pluri- or multipotency, and immunosuppressive features. Being the OC unit, a transition from the bone to cartilage, mesenchymal stem cells (MSCs) are the main focus for OC regeneration. Promising alternatives, which can also be obtained from the patient or at banks and have great differentiation potential toward a wide range of specific cell types, have been reported. Still, ethical concerns and tumorigenic risk are currently under discussion and assessment. In this book chapter, we revise the existing stem cell-based approaches for engineering bone and cartilage, focusing on cell therapy and TE. Furthermore, 3D OC composites based on cell co-cultures are described. Finally, future directions and challenges still to be faced are critically discussed.


Subject(s)
Bone Diseases/therapy , Cartilage Diseases/therapy , Regenerative Medicine/methods , Stem Cell Transplantation/methods , Adult Stem Cells/transplantation , Bone Marrow Transplantation , Cells, Cultured/transplantation , Chondrocytes/transplantation , Chondrogenesis , Embryonic Stem Cells/cytology , Forecasting , Humans , Induced Pluripotent Stem Cells/transplantation , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Osteogenesis , Regenerative Medicine/trends , Tissue Engineering/methods
18.
Contrib Nephrol ; 195: 74-80, 2018.
Article in English | MEDLINE | ID: mdl-29734152

ABSTRACT

Treatment using the cell sheet technology has been applied to various organs, including the cornea, heart, esophagus, periodontium, cartilage, middle ear, and lungs. It has been shown that the therapeutic efficacy of cell sheet transplantation involves 2 aspects, supplementation of cells and provision of cytokines to the affected organ. In addition, cell sheet transplantation also promotes repair of damage through the paracrine effects of cytokines derived from the transplanted cells. It is known that in cases of cell transplantation by injection, the transplanted cells are less likely to differentiate into renal tissue to supply cells, but repair is promoted by the actions of the transplanted cell-derived renotropic factors. Renal function requires functional conjugation of various tissues, including blood vessels, glomeruli, renal tubules, and collecting ducts. It is difficult to supply the necessary cells directly to the affected site of the renal tissue composed of complex structures. On the contrary, the 2-dimensional cell sheet can produce proteins such as erythropoietin, and is thus suitable for transplantation into the living body. It would be desirable to develop cell sheet therapy for the suppression of kidney damage in the future, taking advantage of the beneficial characteristics of cell sheets.


Subject(s)
Cells, Cultured/transplantation , Renal Insufficiency/therapy , Stem Cell Transplantation/methods , Cell Transplantation/methods , Cells, Cultured/metabolism , Cholecalciferol/metabolism , Culture Techniques/methods , Cytokines/metabolism , Erythropoietin/biosynthesis , Humans , Kidney/physiology , Kidney Diseases/therapy , Regeneration , Tissue Engineering/methods
19.
Eur J Vasc Endovasc Surg ; 55(2): 257-265, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29208350

ABSTRACT

OBJECTIVE: Endothelial cells (ECs) play an important role in neovascularisation, but are too limited in number for adequate therapeutic applications. Mesenchymal stem cells (MSCs) have the potential to differentiate into endothelial lineage cells, which makes them attractive candidates for therapeutic angiogenesis. The aim of this study was to investigate efficient differentiation of MSCs into ECs by inducing medium in vitro. METHODS: MSCs were isolated from bone marrow by density gradient centrifugation. The characterisation of the MSCs was determined by their cluster of differentiation (CD) marker profile. Inducing medium containing vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), insulin like growth factor (IGF), epidermal growth factor (EGF), ascorbic acid, and heparin was applied to differentiate the MSCs into ECs. Endothelial differentiation was quantitatively evaluated using flow cytometry. Real time quantitative PCR (qRT-PCR) was used to analyse mRNA expression of endothelial markers. Tube formation assay was further performed to examine the functional status of the differentiated MSCs. RESULTS: Flow cytometry analysis demonstrated that CD31+ and CD34+ cells increased steadily from 12% at 3 days, to 40% at 7 days, and to 60% at 14 days. Immunofluorescence staining further confirmed the expression of CD31 and CD34. qRT-PCR showed that expression of von Willebrand factor (vWF), vascular endothelial cadherin (VE-cadherin) and vascular endothelial growth factor receptor-2 (VEGFR-2) were significantly higher in the induced MSCs group compared with the uninduced MSCs group. The functional behavior of the differentiated cells was tested by tube formation assay in vitro on matrigel. Induced MSCs were capable of developing capillary networks, and progressive formation of vessel like structures was associated with increased EC population. CONCLUSIONS: These results provide a method to efficiently promote differentiation of MSCs into ECs in vitro for potential application in the treatment of peripheral arterial disease.


Subject(s)
Cell Differentiation/physiology , Cytokines/metabolism , Endothelial Cells/physiology , Mesenchymal Stem Cells/physiology , Peripheral Arterial Disease/therapy , Biomarkers/metabolism , Cell Separation/methods , Cells, Cultured/physiology , Cells, Cultured/transplantation , Centrifugation, Density Gradient/methods , Culture Media/metabolism , Endothelial Cells/transplantation , Flow Cytometry , Humans , Neovascularization, Physiologic/physiology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A
SELECTION OF CITATIONS
SEARCH DETAIL
...