Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Methods Mol Biol ; 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38329617

ABSTRACT

In oncological research, the function of tumor-infiltrating natural killer (NK) cells in skin carcinoma presents a viable avenue for novel therapeutic methods. NK cells are essential to the body's defense against malignancies, including skin cancer, and are especially important in more sophisticated cancer immunotherapies such as vaccinations containing dendritic cells. The deadliest type of skin cancer, malignant melanoma, still has a poor prognosis even with advancements in early-stage therapies, which emphasizes the need for novel therapeutic strategies. NK cells from human melanoma metastases were subjected to single-cell RNA-seq analysis, which demonstrated notable variations in the transcriptional programs of tumor-infiltrating and circulating NK cells. Different transcriptional states are displayed by NK cells that have invaded tumors, indicating that they are functionally specialized in areas like chemokine production and cytotoxicity. These results emphasize the functions of NK cells in recruiting other significant immune cell types, such as cross-presenting dendritic cells, and in direct cytotoxicity against malignant cells. Investigating NK cells that infiltrate tumors in skin carcinomas presents a viable approach to comprehending and may be modifying the immune environment surrounding these cancers. It is essential to comprehend the distinct characteristics and roles of NK cells inside the tumor microenvironment in order to create more potent immunotherapeutic approaches to treat skin cancer. In order to perhaps open the door for new directions in cancer immunotherapy, the project intends to establish a thorough technique for the isolation and thorough phenotypic characterization of tumor-infiltrating NK cells in skin carcinoma.

2.
Methods Mol Biol ; 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38095836

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC), a non-melanoma skin cancer that is frequently diagnosed, is distinguished by its propensity for aggressive behavior, frequent poor response to standard therapy, and capacity to metastasize to distant areas. Utilizing the body's natural immune defense mechanisms, particularly through the use of chimeric antigen receptor (CAR) technology, is receiving increasing attention in the dynamic field of oncological therapies. Although T cells have received most of the attention, this strategy has proven to be highly effective in battling some blood-related malignancies. However, there are considerable challenges when using this method in the context of solid tumors. The innate immune system's natural killer (NK) cells are essential parts because they have the ability to detect and destroy cancer cells. CAR-NK cells are a very promising approach because they combine the natural cytotoxic properties of natural killer (NK) cells with the precise targeting skills of chimeric antigen receptor (CAR) technology. With the use of this integrated strategy, the intrinsic diversity of cutaneous squamous cell carcinoma (cSCC) tumors may be successfully targeted, increasing treatment effectiveness and lowering the risk of tumor recurrence. This tactic is improved by the development of dual-specificity chimeric antigen receptors (CARs), which fully resolve the antigen presentation heterogeneity among tumor cells. In conclusion, the use of CAR-NK cells that precisely target cSCC-specific antigens has the potential to drastically transform therapy approaches for cSCC as well as other difficult solid tumors as oncological research advances. In order to create chimeric antigen receptor (CAR)-natural killer (NK) cells that particularly target antigens linked to cutaneous squamous cell carcinoma (cSCC), the goal of this protocol is to present a detailed method. The ultimate objective is to lay the groundwork for the development of precision immunotherapy.

3.
Adv Exp Med Biol ; 1409: 83-110, 2023.
Article in English | MEDLINE | ID: mdl-35999347

ABSTRACT

Tissue engineering as an important field in regenerative medicine is a promising therapeutic approach to replace or regenerate injured tissues. It consists of three vital steps including the selection of suitable cells, formation of 3d scaffolds, and adding growth factors. Mesenchymal stem cells (MSCs) and embryonic stem cells (ESCs) are mentioned as two main sources for this approach that have been used for the treatment of various types of disorders. However, the main focus of literature in the field of dental tissue engineering is on utilizing MSCs. On the other hand, biocompatible scaffolds play a notable role in this regenerative process which is mentioned to be harmless with acceptable osteoinductivity. Their ability in inhibiting inflammatory responses also makes them powerful tools. Indeed, stem cell functions should be supported by biomaterials acting as scaffolds incorporated with biological signals. Naturally derived polymeric scaffolds and synthetically engineered polymeric/ceramic scaffolds are two main types of scaffolds regarding their materials that are defined further in this review. Various strategies of tissue bioengineering can affect the regeneration of dentin-pulp complex, periodontium regeneration, and whole teeth bioengineering. In this regard, in vivo/ex vivo experimental models have been developed recently in order to perform preclinical studies of dental tissue engineering which make it more transferable to be used for clinic uses. This review summarizes dental tissue engineering through its different components. Also, strategies of tissue bioengineering and experimental models are introduced in order to provide a perspective of the potential roles of dental tissue engineering to be used for clinical aims.


Subject(s)
Mesenchymal Stem Cells , Tissue Engineering , Biocompatible Materials/therapeutic use , Regenerative Medicine , Mesenchymal Stem Cells/metabolism , Embryonic Stem Cells , Tissue Scaffolds , Dental Pulp
4.
Med J Islam Repub Iran ; 36: 34, 2022.
Article in English | MEDLINE | ID: mdl-36128298

ABSTRACT

Background: In patients with diabetes, transplantation of stem cells increases C-peptide levels and induces insulin independence for some period. Today, this positive therapeutic outcome is widely attributed to the well-documented immunomodulatory properties of stem cells. The aim of this study was to report alternations (the trend of increase or decrease) in different lymphocyte populations in a stem cell clinical trial performed in our institute. Methods: Recorded data of a clinical trial conducted on 72 patients with type 1 diabetes who had received fetal stem cell transplantation several years ago and were regularly monitored before and after the procedure in 1, 3, 6, 12, 24 months were analyzed. In these regular follow-up visits, insulin demand, HbA1c, C-peptide, and alternation to B cell and T cell populations were analyzed and recorded. For the purpose of the current study, patients were retrospectively divided into 2 groups, namely, those with the positive response to treatment and patients without such response. Temporary positive therapeutic response was defined by 2 different indicators, namely, plasma C-peptide levels and insulin dose-adjusted A1C (IDAA1c), which was calculated as A1C (percent) + (4 × insulin dose (units per kilogram per 24 h). Data analysis was performed by means of SPSS Version 18. Results: Besides the short-term therapeutic effect, we observed remarkably significant alternations to the populations of B and T lymphocytes in the recipients. When patients were retrospectively assigned to 2 different groups of patients with a positive therapeutic response (based on C-peptide changes) and those without it, it was observed that alternations to different populations of B-cells and T-cells were significantly different in these 2 groups. Conclusion: Our results demonstrated that transplantation of stem cells leads to significant positive therapeutic outcomes in one group of patients who showed totally distinct patterns of alternation to different groups of lymphocytes.

5.
Cell Tissue Bank ; 23(4): 653-668, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34545506

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent cells which are popular in human regenerative medicine. These cells can renew themselves and differentiate into several specialized cell types including osteoblasts, adipocytes, and chondrocytes under physiological and experimental conditions. MSCs can secret a lot of components including proteins and metabolites. These components have significant effects on their surrounding cells and also can be used to characterize them. This characterization of multipotent MSCs plays a critical role in their therapeutic potential. The metabolic profile of culture media verified by applying matrix-assisted laser desorption and ionization time-of-flight mass spectrometry (MALDI-TOF-MS) techniques. Also, the differentiation and development of MSCs have monitored through culture media metabolome or secretome (secreted metabolites). Totally, 24 potential metabolites were identified. Between them 12 metabolites are unique to BM-MSCs and 5 metabolites are unique to AD-MSCs. Trilineage differentiation including chondrocytes, osteoblasts, and adipocytes, as well as metabolites that are being differentiated, have been shown in different weeks. In the present study, the therapeutic effects of MSCs analyzed by decoding the metabolome for MSCs secretome via metabolic profiling using MALDI-TOF-MS techniques.


Subject(s)
Mesenchymal Stem Cells , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Culture Media/metabolism , Cell Differentiation , Adipocytes
6.
Article in English | MEDLINE | ID: mdl-33538679

ABSTRACT

Type 1 Diabetes (T1D) is a complex autoimmune disorder which occurs as a result of an intricate series of pathologic interactions between pancreatic ß-cells and a wide range of components of both the innate and the adaptive immune systems. Stem-cell therapy, a recently-emerged potentially therapeutic option for curative treatment of diabetes, is demonstrated to cause significant alternations to both different immune cells such as macrophages, natural killer (NK) cells, dendritic cells, T cells, and B cells and non-cellular elements, including serum cytokines and different components of the complement system. Although there exists overwhelming evidence indicating that the documented therapeutic effects of stem cells on patients with T1D are primarily due to their potential for immune regulation rather than pancreatic tissue regeneration, to date, the precise underlying mechanisms remain obscure. On the other hand, immune-mediated rejection of stem cells remains one of the main obstacles to regenerative medicine. Moreover, the consequences of efferocytosis of stem-cells by the recipients' lung-resident macrophages have recently emerged as a mechanism responsible for some immune-mediated therapeutic effects of stem-cells. This review focuses on the nature of the interactions amongst different compartments of the immune systems which are involved in the pathogenesis of T1D and provides an explanation as to how stem cell- based interventions can influence immune system and maintain the physiologic equilibrium.


Subject(s)
Autoimmune Diseases , Diabetes Mellitus, Type 1 , Humans , Immunomodulation , Killer Cells, Natural , Stem Cells
7.
J Diabetes Metab Disord ; 20(1): 1067-1073, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34222099

ABSTRACT

INTRODUCTION: Stem-cell therapy, which has recently emerged as a potentially therapeutic option for diabetes, is demonstrated to significantly alter both cellular and non-cellular elements of the immune system. In addition, it is demonstrated that allogenic stem-cells, once considered immune-privileged, can be rejected by the host immune system almost similar to any other somatic cell. To date, nonetheless, details of these intricate interactions remain obscure. The current study is designed to illuminate both aforementioned favorable and unfavorable stem cell-mediated immune reactions. Findings of this study may shed some light on how stem cells may exert their therapeutic effect in type 1 diabetes through immune system-mediated mechanisms and illuminate the partially-obscure immune-caused rejection of these cells. METHODS AND ANALYSIS: For the purpose of this study, frozen whole blood samples obtained from patients with type 1 diabetes who received stem cells at the Endocrinology and Metabolism Research Institute of Tehran University of Medical Sciences in two different clinical trials will be thawed and analyzed. These clinical trials were carried out using two different sources of stem cells, namely allogenic fetal and autologous mesenchymal cells. The samples we aim to analyze were obtained from the patients before the procedure and regularly after it, one, three, six, 12, and 24 months later. For the purpose of this study, the following parameters will be measured: C-peptide levels, IDAA1c (a surrogate marker of beta cell function which is calculated as HbA1c (%) + [4 × insulin dose (units per kilogram per day)]), frequencies of islet-specific autoreactive CD8+ T cells (CTL), different lymphocyte subsets, thymic function indicators, T cell repertoire diversity (including Treg/Tconv ratios), plasma levels of several pro- and anti-inflammatory cytokines, diabetes autoantibodies, and HLA typing. ETHICS AND DISSEMINATION: The stem cell transplantation clinical trials which provided the primary source of our samples were carried out at the Endocrinology and Metabolism Research Institute of Tehran University of Medical Sciences between 2008 and 2012. These series of clinical trials have secured approval of the ethics committee of Tehran University of Medical Sciences (ethical code number: E-0089) and registered on the national clinical trial registry of Islamic Republic of Iran (IRCT) with the identifier codes: IRCT138810271414N8 (for autologous mesenchymal cells) and IRCT201103171414N23 (for allogenic fetal cells). Our findings are to be presented at international scientific events, published in peer-reviewed journals, and disseminated both electronically and in print. Besides, results of the current study will be used for design and implementation of future laboratory investigations and clinical trials at the Endocrinology and Metabolism Research Institute of Tehran University of Medical Sciences.

8.
Stem Cell Res Ther ; 11(1): 396, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32928295

ABSTRACT

BACKGROUND: Regenerative medicine plays a major role in biomedicine, and given the ever-expanding boundaries of this knowledge, numerous ethical considerations have been raised. MAIN TEXT: Rapid advancement of regenerative medicine science and technology in Iran, emerged the Iranian National Committee for Ethics in Biomedical Research to develop a comprehensive national ethical guideline. Therefore, the present ethical guideline which comprises eleven chapters was developed in 2019 and approved in early 2020. The titles of these chapters were selected based on the ethical considerations of various aspects of the field of regenerative medicine: (1) ethical principles of research on stem cells and regenerative medicine; (2) ethical considerations for research on stem cells (embryonic stem cells, epiblast stem cells, tissue-specific stem cells, stem cells derived from transdifferentiation, induced pluripotent stem cells [iPSCs], germline pluripotent stem cells, germline stem cells, and somatic cell nuclear transfer [SCNT] stem cells); (3) ethical considerations for research on somatic cells in regenerative medicine (adult somatic cells, fetal tissue somatic cells, and somatic cells derived from pregnancy products [other than fetus]); (4) ethical considerations for research on gametes in regenerative medicine; (5) ethical considerations for research related to genetic manipulation (human and animal) in regenerative medicine; (6) ethical considerations for research on tissue engineering in regenerative medicine; (7) ethical considerations for pre-clinical studies in regenerative medicine; (8) ethical considerations for clinical trials in regenerative medicine; (9) ethical considerations for stem cells and regenerative medicine bio-banks; (10) ethical considerations for privacy and confidentiality; and (11) ethical considerations for obtaining informed consent. CONCLUSION: This article discusses the process of developing the present ethical guidelines and its practical points. We hope that it can play an important worldwide role in advancing ethics of research on stem cells and regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Embryonic Stem Cells , Female , Humans , Iran , Pregnancy , Regenerative Medicine
9.
J Immunol Methods ; 477: 112690, 2020 02.
Article in English | MEDLINE | ID: mdl-31678265

ABSTRACT

Adoptive cell therapy (ACT) using tumor antigen-independent killer cells has been widely used in clinical trials of cancer treatment. Circumventing the need for identification of a particular tumor-associated antigen on tumor cells, the approach has opened possibilities for the extension of ACT immunotherapy to patients with a wide variety of cancer types. Namely, Natural Killer (NK), Lymphokine-activated Killer (LAK) cells and Cytokine-induced killer (CIK) cells are the most commonly used cell types in antigen-independent adoptive immunotherapy of cancer. They all originate from peripheral blood mononuclear cells and share several common features in their killing mechanisms. However, despite broad application in clinical settings, the boundaries between these cell types are not very clearly defined. The current study aims to review different aspects of these cell populations in terms of phenotypical characteristic and preparation media, to clarify how the boundaries are set.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Primary Cell Culture/methods , Clinical Trials as Topic , Culture Media , Cytokine-Induced Killer Cells/immunology , History, 20th Century , History, 21st Century , Humans , Immunotherapy, Adoptive/history , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Treatment Outcome
10.
Arch Iran Med ; 22(1): 7-10, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30821155

ABSTRACT

BACKGROUND: Current advancements in the field of chimeric antigen receptor (CAR) therapy, particularly U.S. FDA approval of Kymriah and Yescarta, heralds a new era of cancer treatment. This rapid progress in technology has urged more countries and institutions to keep pace with the fast-growing and developing technology of producing CAR T cell-based therapies in the race to develop new cancer-targeting drugs. Hence, for stepping in line with global advances and to pave the way for subsequent preclinical and clinical studies, we have established a development protocol for a cancer-targeting CAR T cell; we have chosen CD19 CAR T cell as a well-defined model to set-up T cell expansion, activation, and viral transduction as the prerequisites for diverse CAR T cell therapies. METHODS: T cells from peripheral blood mononuclear cells (PBMCs) were activated and expanded. CD19 CAR lentiviral particles were produced in the Lenti-X™ 293T Cell Line using PolyFect Transfection Reagent. RESULTS: Activation protocol resulted in (65 ± 4%; P = 0.046) increase in the rate of activated T cells 24 hours after the initiation of the procedure. The expansion methodology resulted in a high purity of the T cell population (96 ± 3%) in the pool of PBMCs within 14 days of the procedure. Finally, 35 ± 6% of T cells were transduced with CD19 lentivirus with MOI of 3. CONCLUSION: Collectively, the results of this study prove that we have successfully overcome the first hurdle on the road to reach CAR T cell technology which is the prerequisite for developing preclinical and clinical phases of CAR therapy in settings with basic resources.


Subject(s)
Antigens, CD19/metabolism , Biomarkers, Tumor/metabolism , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/therapy , Lymphoma, B-Cell/therapy , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen/therapeutic use , Cells, Cultured , Humans , Leukemia, B-Cell/immunology , Lymphoma, B-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism
11.
Cell Tissue Bank ; 20(1): 11-24, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30535614

ABSTRACT

Regenerative medicine as a background of stem cell research and therapy has a long history. A wide variety of diseases including Parkinson's disease, heart diseases, multiple sclerosis, spinal cord injury, diabetes mellitus and etc. are candidate to be treated using different types of stem cells. There are several sources of stem cells such as bone marrow, umbilical cord, peripheral blood, germ cells and the embryo/fetus tissues. Fetal stem cells (FSCs) and embryonic stem cells (ESCs) have been described as the most potent stem cell source. Although their pluri- or multipotent properties leads to promising reports for their clinical applications, owning to some ethical and legal obstacles in different communities such as Muslim countries, care should be taken for therapeutic applications of FSCs and ESCs. Derivation of these cell types needs termination of pregnancy and embryo or fetus life that is prohibited according to almost all rules and teaches in Muslim communities. Abortion and termination of pregnancy under a normal condition for the procurement of stem cell materials is forbidden by nearly all the major world religions such as Islam. Legislated laws in the most of Muslim countries permit termination of pregnancy and abortion only when the life of the mother is severely threatened or when continuing pregnancy may lead to the birth of a mentally retarded, genetically or anatomically malformed child. Based on the rules and conditions in Islamic countries, finding an alternative and biologically normal source for embryonic or fetal stem cell isolation will be too difficult. On the one hand, Muslim scientists have the feasibility for finding of genetically and anatomically normal embryonic or fetal stem cell sources for research or therapy, but on the other hand they should adhere to the law and related regional and local rules in all parts of their investigation. The authors suggest that the utilization of ectopic pregnancy (EP) conceptus, extra-embryonic tissues, and therapeutic abortion materials as a valuable source of stem cells for research and medical purposes can overcome limitations associated with finding the appropriate stem cell source. Pregnancy termination because of the mentioned subjects is accepted by almost all Islamic laws because of maternal lifesaving. Also, there are no ethical or legal obstacles in the use of extra-embryonic or EP derived tissues which lead to candidate FSCs as a valuable source for stem cell researches and therapeutic applications.


Subject(s)
Abortion, Therapeutic , Fetal Stem Cells/transplantation , Pregnancy, Ectopic/therapy , Stem Cell Research , Cell Differentiation , Clinical Trials as Topic , Female , Humans , Iran , Pregnancy , Stem Cell Research/ethics , Stem Cell Research/legislation & jurisprudence
12.
Adv Exp Med Biol ; 1119: 133-149, 2018.
Article in English | MEDLINE | ID: mdl-29858972

ABSTRACT

Skin as the outer layer covers the body. Wounds can affect this vital organ negatively and disrupt its functions. Wound healing as a biological process is initiated immediately after an injury. This process consists of three stages: inflammation, proliferation, remodeling. Generally, these three stages occur continuously and timely. However, some factors such as infection, obesity and diabetes mellitus can interfere with these stages and impede the normal healing process which results in chronic wounds. Financial burden on both patients and health care systems, negative biologic effect on the patient's general health status and reduction in quality of life are a number of issues which make chronic wounds as a considerable challenge. During recent years, along with advances in the biomedical sciences, various surgical and non-surgical therapeutic methods have been suggested. All of these suggested treatments have their own advantages and disadvantages. Recently, cell-based therapies and regenerative medicine represent promising approaches to wound healing. Accordingly, several types of mesenchymal stem cells have been used in both preclinical and clinical settings for the treatment of wounds. Adipose-derived stromal cells are a cost-effective source of mesenchymal stem cells in wound management which can be easily harvest from adipose tissues through the less invasive processes with high yield rates. In addition, their ability to secrete multiple cytokines and growth factors, and differentiation into skin cells make them an ideal cell type to use in wound treatment. This is a concise overview on the application of adipose-derived stromal cells in wound healing and their role in the treatment of chronic wounds.


Subject(s)
Mesenchymal Stem Cells , Quality of Life , Adipose Tissue , Cell Differentiation , Humans , Wound Healing
13.
Acta Med Iran ; 55(6): 368-374, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28843237

ABSTRACT

Stress urinary incontinence is a common medical problem among women. The urethral closure complex and/or the supportive mechanisms are responsible for incontinence in the majority of patients. Several surgical procedures with different degrees of invasiveness and outcomes have been reported to treat the problem. Although most of these procedures are reasonably effective, a general trend towards the study of natural and biocompatible tissues is emerging over popular synthetic materials. Here we report our experience of autologous adipose-derived stem cells transplantation into the periurethral region as a new method of stress urinary incontinence treatment. Ten women with symptoms of stress urinary incontinence were treated by injections of autologous adipose-derived stem cells into the periurethral region via transurethral and transvaginal approach under urethroscopic observation. This report presents the short-term outcome of the patients. The outcome measured by pad test results, ICIQ-SF scores, and Qmax. The mean age of the participants was 45.8±8.7 years. Urinary incontinence significantly decreased through the first two, 6 and 24 weeks after the injection therapy. The difference was significant in pad test results (P<0.001) and ICIQ-SF scores (P<0.001), especially comparing results between 2 and 6 weeks and among 6 and 24 weeks, but not for 2 and 6 weeks compared to each other. Surprisingly, Qmax showed improvement after the study period (means 32.6 vs. 35.7; P=0.002). This study showed that injection of the autologous adipose-derived stem cells to the periurethral region is a safe, yet short-term effective treatment option for stress urinary incontinence. Further studies with longer follow up are needed to confirm its long term efficacy.


Subject(s)
Adipose Tissue/cytology , Stem Cell Transplantation/methods , Urinary Incontinence, Stress/therapy , Adult , Female , Humans , Injections , Middle Aged , Transplantation, Autologous , Treatment Outcome , Urethra
14.
Acta Med Iran ; 53(9): 540-6, 2015.
Article in English | MEDLINE | ID: mdl-26553081

ABSTRACT

In this study, our aim was to produce a generation of GMP-grade adipose tissue-derived mesenchymal stem cells for clinical applications. According to our results, we fulfill to establish consistent and also reproducible current good manufacturing practice (cGMP) compliant adipose tissue-derived mesenchymal stem cells from five female donors. The isolated cells were cultured in DMEM supplemented with 10% fetal bovine serum and characterized by standard methods. Moreover, karyotyping was performed to evaluate chromosomal stability. Mean of donors' age was 47.6 ± 8.29 year, mean of cell viability was 95.6 ± 1.51%, and cell count was between 9×106 and 14×106 per microliter with the mean of 12.2×106 ± 2863564.21 per microliter. The main aim of this project was demonstrating the feasibility of cGMP-compliant and clinical grade adipose tissue-derived mesenchymal stem cells preparation and banking for clinical cell transplantation trials.


Subject(s)
Adipose Tissue/cytology , Biological Specimen Banks , Cell Survival , Mesenchymal Stem Cells/physiology , Adult , Cell Differentiation , Cell Proliferation , Cells, Cultured , Culture Media , Feasibility Studies , Female , Humans
15.
Methods Mol Biol ; 1283: 123-36, 2015.
Article in English | MEDLINE | ID: mdl-25092054

ABSTRACT

Stem cell therapy seems a promising avenue in regenerative medicine. Within various stem cells, mesenchymal stem cells have progressively used for cellular therapy. Because of the age-related decreasing in the frequency and differentiating capacity of adult MSCs, fetal tissues such as fetal liver, lung, pancreas, spleen, etc. have been introduced as an alternative source of MSCs for cellular therapy. On the other hand, using stem cells as advanced therapy medicinal products, must be performed in compliance with cGMP as a quality assurance system to ensure the safety, quality, and identity of cell products during translation from the basic stem cell sciences into clinical cell transplantation. In this chapter the authors have demonstrated the manufacturing of GMP-grade human fetal liver-derived mesenchymal stem cells.


Subject(s)
Fetus/cytology , Guideline Adherence , Liver/cytology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cell Transplantation/standards , Mesenchymal Stem Cells/cytology , Cell Culture Techniques/methods , Cell Culture Techniques/standards , Cell Separation/methods , Cell Separation/standards , Cryopreservation/methods , Cryopreservation/standards , Humans , Mesenchymal Stem Cells/metabolism , Specimen Handling/methods , Specimen Handling/standards
16.
Methods Mol Biol ; 1283: 93-107, 2015.
Article in English | MEDLINE | ID: mdl-25138723

ABSTRACT

Stem cells, which can be derived from different sources, demonstrate promising therapeutic evidences for cellular therapies. Among various types of stem cell, mesenchymal stem cells are one of the most common stem cells that are used in cellular therapy. Human subcutaneous adipose tissue provides an easy accessible source of mesenchymal stem cells with some considerable advantages. Accordingly, various preclinical and clinical investigations have shown enormous potential of adipose-derived stromal cells in regenerative medicine. Consequently, increasing clinical applications of these cells has elucidated the importance of safety concerns regarding clinical transplantation. Therefore, clinical-grade preparation of adipose-derived stromal cells in accordance with current good manufacturing practice guidelines is an essential part of their clinical applications to ensure the safety, quality, characteristics, and identity of cell products. Additionally, GMP-compliant cell manufacturing involves several issues to provide a quality assurance system during translation from the basic stem cell sciences into clinical investigations and applications. On the other hand, advanced cellular therapy requires extensive validation, process control, and documentation. It also evidently elucidates the critical importance of production methods and probable risks. Therefore, implementation of a quality management and assurance system in accordance with GMP guidelines can greatly reduce these risks particularly in the higher-risk category or "more than minimally manipulated" products.


Subject(s)
Adipose Tissue/cytology , Cell Culture Techniques/methods , Cell Culture Techniques/standards , Cell Differentiation , Guideline Adherence , Mesenchymal Stem Cells/cytology , Cell Separation/methods , Cell Separation/standards , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/standards , Cryopreservation/methods , Cryopreservation/standards , Humans , Mesenchymal Stem Cells/metabolism
17.
Acta Med Iran ; 50(8): 541-6, 2012.
Article in English | MEDLINE | ID: mdl-23109026

ABSTRACT

Stem cell-based therapies have recently opened up new horizons for treatment of various types of diseases including diabetes mellitus. However, long-term efficacy and safety of these novel modalities still remain a serious question. Hereby, we aim to report the one-year follow-up results in the diabetic patients who underwent fetal liver-derived hematopoietic stem cell allotransplantation. Fifty six patients with type one (n=30) and type two (n=26) diabetes, aged 10-58 years old (32.8 ± 16.3) were divided into the intervention and placebo group. The patients in the intervention group underwent fetal liver-derived hematopoietic stem cell transplantation while the patients in the placebo group received 5 ml of normal saline both via an intravenous route. The patients were visited at regular intervals to evaluate the efficacy of transplantation in glycemic control as well as possible complications. In the 6th month of the follow-up, there was a significant decrease in HbA1c levels in all groups without any rise in the fasting c-peptide. However, none of the precipitants transiently or continuously became insulin free in the first year after transplantation. It can be concluded that, in this study, fetal liver-derived hematopoietic stem cell transplantation had no significant effects on glycemic control. The heterogeneity of our patients might account for the negative results. Hence, longer follow-up results will be reported in the near future.


Subject(s)
Cell Transplantation , Diabetes Mellitus, Type 1/surgery , Diabetes Mellitus, Type 2/surgery , Liver/cytology , Adolescent , Adult , Blood Glucose/analysis , Child , Follow-Up Studies , Humans , Insulin/administration & dosage , Liver/embryology , Middle Aged , Placebos , Transplantation, Homologous , Young Adult
18.
Cell Tissue Bank ; 13(2): 281-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21484231

ABSTRACT

Clinical grade cultivation of human schwann cell by the utilization of human autologous serum instead of fetal bovine serum, and also avoiding any growth factors, can increase safety level of this procedure in cases of clinical cell transplantation. The aim of this study was demonstration of the feasibility of clinical grade schwann cell cultivation. In this experimental study after obtaining consent from close relatives we harvested 10 sural nerves from brain death donors and then cultured in 10 seperated culture media plus autologous serum. We also prepared autologous serum from donor's whole blood. Then cultured cells were evaluated by S100 antibody staining for both morphology and purity. Cell purity range was from 97% to 99% (mean=98.11 ± 0.782%). Mean of the cell count was 14,055.56 ± 2,480.479 per micro liter. There was not significant correlation between cell purity and either the culture period or the age of donors (P>0.05). The spearman correlation coefficient for the cell purity with the period or the age of donors was 0.21 and 0.09, respectively. We demonstrated the feasibility of clinical grade schwann cell cultivation by the using of human autologous serum instead of fetal bovine serum and also without the using of growth factors. We also recommended all cell preparation facilities to adhere to the GMP and other similar quality disciplines especially in the preparation of clinically-used cell products.


Subject(s)
Cell Culture Techniques/methods , Intercellular Signaling Peptides and Proteins/pharmacology , Schwann Cells/cytology , Schwann Cells/drug effects , Serum/metabolism , Adult , Animals , Cattle , Cells, Cultured , Demography , Female , Humans , Male , Middle Aged , S100 Proteins/metabolism , Schwann Cells/metabolism , Staining and Labeling , Sural Nerve/drug effects , Sural Nerve/transplantation , Tissue Donors
19.
Neurosci Lett ; 443(1): 46-50, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18662744

ABSTRACT

Several experimental studies have introduced Schwann cell transplantation as a means of recovery in animal models of spinal cord injury (SCI). The reported promising results together with the availability of autologous sources for Schwann cells indicate Schwann cell transplantation as a possible treatment for SCI. To address the safety and feasibility concerns we report 1-year follow-up of four patients aged between 22 and 43 years who had stable chronic (28-80 months) spinal cord injury at mid-thoracic level and treated with autologous Schwann cell transplantation. Purified Schwann cells used for transplantation were acquired from autologous sural nerve and cultured without the use of any specific mitogenic or growth factors. The patients were evaluated by means of American Spinal Injury Association (ASIA) criteria, sphincter, sexual function and Magnetic Resonance Imaging assessments for 1 year after transplantation. None of the patients were found to have any adverse effects indicating transfer of infection, neurological deterioration or other related clinical problems. Of the four patients, only one patient with incomplete SCI showed motor and sensory improvement 1 year after transplantation with extensive and continuous rehabilitation. All the four patients experienced transient paresthesia or increased muscle spasm after transplantation. Magnetic Resonance (MR) images of the patients did not show any visible changes or pathological findings after 1 year. This preliminary report shows that autologous Schwann cell transplantation is generally safe for the selected number of SCI patients but it does not prove beneficial effects. Further safety and outcome studies are recommended.


Subject(s)
Cell Transplantation/methods , Schwann Cells/transplantation , Spinal Cord Injuries/surgery , Adult , Chronic Disease , Female , Follow-Up Studies , Humans , Male , Recovery of Function , Schwann Cells/physiology , Thoracic Vertebrae , Transplantation, Autologous/methods , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...