1
|
Petrella F, Cassina EM, Libretti L, Pirondini E, Raveglia F, Tuoro A. Mesenchymal Stromal Cell Therapy for Thoracic Surgeons: An Update. J Pers Med 2023; 13:1632. [PMID: 38138859 PMCID: PMC10744666 DOI: 10.3390/jpm13121632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Stem cells are undifferentiated cells presenting extensive self-renewal features and the ability to differentiate "in vitro" and "in vivo" into a range of lineage cells, like chondrogenic, osteogenic and adipogenic lineages when cultured in specific inducing media. Two major domains of clinical applications of stem cells in thoracic surgery have been investigated: regenerative medicine, which is a section of translational research in tissue engineering focusing on the replacement, renewal or regeneration of cells, tissues and organs to re-establish damaged physiologic functions; drug loading and delivery, representing a new branch proposing stem cells as carriers to provide selected districts with anti-cancer agents for targeted treatments.
Collapse
Affiliation(s)
- Francesco Petrella
- Department of Thoracic Surgery, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (E.M.C.); (L.L.); (E.P.); (F.R.); (A.T.)
| | | | | | | | | | | |
Collapse
|
2
|
Oh MS, Lee SG, Lee GH, Kim CY, Song JH, Yu BY, Chung HM. Verification of Therapeutic Effect through Accelerator Mass Spectrometry-Based Single Cell Level Quantification of hESC-Endothelial Cells Distributed into an Ischemic Model. Adv Healthc Mater 2023; 12:e2300476. [PMID: 37068221 DOI: 10.1002/adhm.202300476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/01/2023] [Indexed: 04/19/2023]
Abstract
As the potential of pluripotent stem cell-derived differentiated cells has been demonstrated in regenerative medicine, differentiated vascular endothelial cells (ECs) are emerging as a therapeutic agent for the cardiovascular system. To verify the therapeutic efficacy of differentiated ECs in an ischemic model, human embryonic stem cells (hESCs) are induced as EC lineage and produce high-purity ECs through fluorescence-activated cell sorting (FACS). When hESC-ECs are transplanted into a hindlimb ischemic model, it is confirmed that blood flow and muscle regeneration are further improved by creating new blood vessels together with autologous ECs than the primary cell as cord blood endothelial progenitor cells (CB-EPCs). In addition, previously reported studies show the detection of transplanted cells engrafted in blood vessels through various tracking methods, but fail to provide accurate quantitative values over time. In this study, it is demonstrated that hESC-ECs are engrafted approximately sevenfold more than CB-EPCs by using an accelerator mass spectrometry (AMS)-based cell tracking technology that can perform quantification at the single cell level. An accurate quantification index is suggested. It has never been reported in in vivo kinetics of hESC-ECs that can act as therapeutic agents.
Collapse
Affiliation(s)
- Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Gwan-Ho Lee
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Jong Han Song
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Byung-Yong Yu
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Mirae Cell Bio Co. Ltd, Seoul, 04795, Republic of Korea
| |
Collapse
|
3
|
Oh MS, Lee SG, Lee GH, Kim CY, Kim EY, Song JH, Yu BY, Chung HM. In vivo tracking of 14C thymidine labeled mesenchymal stem cells using ultra-sensitive accelerator mass spectrometry. Sci Rep 2021; 11:1360. [PMID: 33446731 PMCID: PMC7809063 DOI: 10.1038/s41598-020-80416-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
Despite the tremendous advancements made in cell tracking, in vivo imaging and volumetric analysis, it remains difficult to accurately quantify the number of infused cells following stem cell therapy, especially at the single cell level, mainly due to the sensitivity of cells. In this study, we demonstrate the utility of both liquid scintillator counter (LSC) and accelerator mass spectrometry (AMS) in investigating the distribution and quantification of radioisotope labeled adipocyte derived mesenchymal stem cells (AD-MSCs) at the single cell level after intravenous (IV) transplantation. We first show the incorporation of 14C-thymidine (5 nCi/ml, 24.2 ng/ml) into AD-MSCs without affecting key biological characteristics. These cells were then utilized to track and quantify the distribution of AD-MSCs delivered through the tail vein by AMS, revealing the number of AD-MSCs existing within different organs per mg and per organ at different time points. Notably, the results show that this highly sensitive approach can quantify one cell per mg which effectively means that AD-MSCs can be detected in various tissues at the single cell level. While the significance of these cells is yet to be elucidated, we show that it is possible to accurately depict the pattern of distribution and quantify AD-MSCs in living tissue. This approach can serve to incrementally build profiles of biodistribution for stem cells such as MSCs which is essential for both research and therapeutic purposes.
Collapse
Affiliation(s)
- Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, 05029, Republic of Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, 05029, Republic of Korea
| | - Gwan-Ho Lee
- Advanced Analysis Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, 05029, Republic of Korea
| | - Eun-Young Kim
- Mirae Cell Bio Co. Ltd, Seoul, 04795, Republic of Korea
| | - Jong Han Song
- Advanced Analysis Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Byung-Yong Yu
- Advanced Analysis Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, 05029, Republic of Korea. .,Mirae Cell Bio Co. Ltd, Seoul, 04795, Republic of Korea.
| |
Collapse
|
4
|
Shimizu H, Kuze Y, Higuchi T, Matsumoto SI, Yamamoto S, Goto A, Moriya Y, Hirabayashi H. Development of a bioanalytical method for circulating human T cells in animals using Arthrobacter luteus-based quantitative polymerase chain reaction and its application in preclinical biodistribution studies. Regen Ther 2020; 15:251-257. [PMID: 33426226 PMCID: PMC7770409 DOI: 10.1016/j.reth.2020.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/22/2020] [Accepted: 10/19/2020] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION In the development of cell therapy products for human use, studies on the biodistribution of transplanted cells in animals are important for assessing the safety and efficacy of these products. Although a few reports have described the biodistribution of human cells in animals using Arthrobacter luteus-based-polymerase chain reaction (Alu-PCR), most have used genomic DNA or synthetic oligonucleotide as calibrators, as opposed to actual cells. In addition, bioanalytical variability in the quantification of cells with respect to specificity, selectivity, accuracy, and precision, has not been evaluated. Accordingly, in this study, we validated the utility of this bioanalytical method for human T cells in mice to establish assay performance using cells as a calibrator. METHODS A standard curve was constructed for the addition of cell lysates to mouse tissues and blood, and DNA was extracted. Alu-PCR was applied for the quantification of human peripheral blood CD8+ T cells in mice. To determine assay performance, we evaluated accuracy, precision, selectivity, specificity, and stability. In vivo cell kinetics and biodistribution were investigated based on intravenous administration of human T cells to mice. RESULTS Alu-PCR enabled us to specifically detect human T cells in mouse blood and tissues. The lower detection limit of Alu-PCR was 10 cells/15 mg tissue (7.5 mg for spleen and lung) or cells/50 μL blood. Given that PCR threshold cycle (Cq) values among mouse samples (blood, liver spleen, lung, heart, and kidney) show slight variation, calibration curves should be generated using the same tissue as used for the assay. Most coefficients of variation in the assay were within 30%. The cell kinetics of administered human T cells in mice were successfully evaluated using the established Alu-qPCR. CONCLUSIONS The Alu-PCR technique developed in this study showed sufficient specificity and sensitivity in detecting human peripheral blood CD8+ T cells in mice. This technique, which targets the primate-specific Alu gene, is applicable for quantifying transplanted human cells in animals without the necessity of cell labeling. The data presented herein will be useful for standardizing bioanalytical approaches in biodistribution studies of cell therapy products.
Collapse
Affiliation(s)
- Hisao Shimizu
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Yoji Kuze
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Tomoaki Higuchi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Shin-ichi Matsumoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Syunsuke Yamamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Akihiko Goto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Yuu Moriya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| |
Collapse
|
5
|
Bukulmez H, Horkayne-Szakaly I, Bilgin A, Baker TP, Caplan AI, Jones OY. Intrarenal injection of mesenchymal stem cell for treatment of lupus nephritis in mice - a pilot study. Lupus 2020; 30:52-60. [PMID: 33135563 DOI: 10.1177/0961203320968897] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The current project is to explore feasibility of direct intra-renal injection of human bone marrow derived mesenchymal stem cells (hMSC) for treatment of lupus nephritis in mice. The treatment protocol involved aged male BXSB (20 weeks) injected with 1 × 106 hMSC unilaterally under the renal capsule. Mice were harvested after 10 weeks follow-up for postmortem exam. Controls included untreated age matched male BXSB and healthy C57Bl/6. At the end of follow-up period, the survival of treated BXSB was 10 folds higher at 62.5% compared to survival of untreated control at 6.3%. The survival of C57Bl/6 remained at 100% with or without similar treatment. The renal pathology review was most significant for decreased tissue inflammation in treated BXSB compared to untreated controls. Renal tissue expression of IL-1b, IL17 were decreased and CTLA-4 was increased by RT PCR among treated compared to untreated BXSB. Thus, direct delivery of hMSC by intrarenal injection is a promising route for treatment of lupus nephritis as shown in this xenogeneic model. Further studies -using expanded numbers of mice to include other lupus strains- are warranted to investigate the mechanisms involved and to optimize treatment protocol for safety and efficacy.
Collapse
Affiliation(s)
- Hulya Bukulmez
- Department of Pediatrics, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | | | - Asuman Bilgin
- Skeletal Research Center, Case Western Reserve University, Cleveland, OH, USA
| | - Thomas P Baker
- Joint Pathology Center, Defense Health Agency, Silver Spring, MD, USA
| | - Arnold I Caplan
- Skeletal Research Center, Case Western Reserve University, Cleveland, OH, USA
| | - Olcay Y Jones
- Division Pediatric Rheumatology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| |
Collapse
|
6
|
Marzorati S, Messina M, Ghiglieri A. Autoradioluminography, a powerful and reliable tool for drug development: Accelera's experience. J Labelled Comp Radiopharm 2019; 62:776-784. [PMID: 31379030 DOI: 10.1002/jlcr.3790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 11/08/2022]
Abstract
A deeper understanding of the pharmacokinetic and pharmacodynamic properties of a drug candidate is a pivotal component of drug discovery and development. Autoradiography is an excellent technique allowing exploiting the advantages of the use of radioisotopes in the drug disscovery field. The introduction of phosphor imaging technology has revolutionized the handling of drug distribution studies providing high-resolution images. Specifically, quantitative whole-body autoradioluminography is employed for preclinical study where the aim is to obtain information about the route of elimination and tissue distribution of a drug candidate. Autoradioluminography is also the technique of choice pursued to deal with all the issue that it is possible to encounter in all stage of drug development (ie, site-specific drug localization and retention, drug-drug interactions, penetration into specific target, specific tissue binding, crossing of brain blood barrier, and placental transfer). The purpose of this review is to give a picture of how autoradiography is employed in our laboratory as a key tool for advances in the assessment of the drug disposition and to validate new experimental models.
Collapse
|
7
|
Anti-aging effects exerted by Tetramethylpyrazine enhances self-renewal and neuronal differentiation of rat bMSCs by suppressing NF-kB signaling. Biosci Rep 2019; 39:BSR20190761. [PMID: 31171713 PMCID: PMC6591573 DOI: 10.1042/bsr20190761] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/20/2019] [Accepted: 05/31/2019] [Indexed: 12/13/2022] Open
Abstract
In order to improve the therapeutic effects of mesenchymal stem cell (MSC)-based therapies for a number of intractable neurological disorders, a more favorable strategy to regulate the outcome of bone marrow MSCs (bMSCs) was examined in the present study. In view of the wide range of neurotrophic and neuroprotective effects, Tetramethylpyrazine (TMP), a biologically active alkaloid isolated from the herbal medicine Ligusticum wallichii, was used. It was revealed that treatment with 30–50 mg/l TMP for 4 days significantly increased cell viability, alleviated senescence by suppressing NF-κB signaling, and promoted bMSC proliferation by regulating the cell cycle. In addition, 40–50 mg/l TMP treatment may facilitate the neuronal differentiation of bMSCs, verified in the present study by presentation of neuronal morphology and expression of neuronal markers: microtubule-associated protein 2 (MAP-2) and neuron-specific enolase (NSE). The quantitative real-time polymerase chain reaction (qRT-PCR) revealed that TMP treatment may promote the expression of neurogenin 1 (Ngn1), neuronal differentiation 1 (NeuroD) and mammalian achaete–scute homolog 1 (Mash1). In conclusion, 4 days of 40–50 mg/l TMP treatment may significantly delay bMSC senescence by suppressing NF-κB signaling, and enhancing the self-renewal ability of bMSCs, and their potential for neuronal differentiation.
Collapse
|
8
|
Comparison of the Proliferation and Differentiation Potential of Human Urine-, Placenta Decidua Basalis-, and Bone Marrow-Derived Stem Cells. Stem Cells Int 2018; 2018:7131532. [PMID: 30651734 PMCID: PMC6311712 DOI: 10.1155/2018/7131532] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/05/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023] Open
Abstract
Human multipotent stem cell-based therapies have shown remarkable potential in regenerative medicine and tissue engineering applications due to their abilities of self-renewal and differentiation into multiple adult cell types under appropriate conditions. Presently, human multipotent stem cells can be isolated from different sources, but variation among their basic biology can result in suboptimal selection of seed cells in preclinical and clinical research. Thus, the goal of this study was to compare the biological characteristics of multipotent stem cells isolated from human bone marrow, placental decidua basalis, and urine, respectively. First, we found that urine-derived stem cells (USCs) displayed different morphologies compared with other stem cell types. USCs and placenta decidua basalis-derived mesenchymal stem cells (PDB-MSCs) had superior proliferation ability in contrast to bone marrow-derived mesenchymal stem cells (BMSCs); these cells grew to have the highest colony-forming unit (CFU) counts. In phenotypic analysis using flow cytometry, similarity among all stem cell marker expression was found, excluding CD29 and CD105. Regarding stem cell differentiation capability, USCs were observed to have better adipogenic and endothelial abilities as well as vascularization potential compared to BMSCs and PDB-MSCs. As for osteogenic and chondrogenic induction, BMSCs were superior to all three stem cell types. Future therapeutic indications and clinical applications of BMSCs, PDB-MSCs, and USCs should be based on their characteristics, such as growth kinetics and differentiation capabilities.
Collapse
|
9
|
Svitina H, Skrypkina I, Areshkov P, Kyryk V, Bukreieva T, Klymenko P, Garmanchuk L, Lobintseva G, Shablii V. Transplantation of placenta-derived multipotent cells in rats with dimethylhydrazine-induced colon cancer decreases survival rate. Oncol Lett 2018; 15:5034-5042. [PMID: 29552139 PMCID: PMC5840552 DOI: 10.3892/ol.2018.7996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 01/22/2018] [Indexed: 12/19/2022] Open
Abstract
Transplantation of placenta-derived multipotent cells (PDMCs) is a promising treatment method for many diseases. However, the impact of PDMCs on colon cancer has not yet been studied. PDMCs were obtained from rat placentas by culturing tissue explants. Colon cancer was experimentally induced in male albino Wistar rats by administering 20 mg/kg dimethylhydrazine (DMH) once a week for 20 consecutive weeks. The administration of the PDMCs was performed at the 20th week after the first DMH injection. The number and size of each tumour lesion were calculated in the 5th week after transplantation. The tumour type was determined by standard histological methods. To study the engraftment of PDMCs in the body of rats, the cells were transduced with enhanced green fluorescent protein. Cell engraftment was determined by assessing the presence of EGFP by PCR and immunohistochemistry. Survival of all rats was monitored daily. Allogeneic transplantation of PDMCs to rats at middle phase of DMH-induced colon carcinogenesis did not significantly influence the number of neoplasms and the parameters of mean and total tumour area, but led to an increase in size of the most invasiveness tumours. Intravenous allogeneic transplantation of PDMCs reduced the survival rate of rats with colon cancer by 17 days. PDMCs from rats engrafted into tissues of the normal intestine, tumours, lungs, liver, and spleen of rats for five weeks after intravenous transplantation. These results suggest that intravenous allogeneic transplantation of PDMCs promotes colon cancer progression and has a negative impact on survival of rats.
Collapse
Affiliation(s)
- Hanna Svitina
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine.,Department of Biochemistry, Educational and Scientific Centre 'Institute of Biology and Medicine', Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Inessa Skrypkina
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine.,Department of Functional Genomics, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, Kyiv 03680, Ukraine
| | - Pavlo Areshkov
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine.,Department of Functional Genomics, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, Kyiv 03680, Ukraine
| | - Vitaliy Kyryk
- Department of Cell and Tissue Technologies, State Institute of Genetic and Regenerative Medicine of National Academy of Medicine of Ukraine, Kyiv 04114, Ukraine
| | - Tetiana Bukreieva
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine
| | - Pavlo Klymenko
- Department of Cell and Tissue Technologies, State Institute of Genetic and Regenerative Medicine of National Academy of Medicine of Ukraine, Kyiv 04114, Ukraine
| | - Liudmyla Garmanchuk
- Department of Biochemistry, Educational and Scientific Centre 'Institute of Biology and Medicine', Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Galyna Lobintseva
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine
| | - Volodymyr Shablii
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine.,Department of Biochemistry, Educational and Scientific Centre 'Institute of Biology and Medicine', Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| |
Collapse
|
10
|
Eve DJ, Sanberg PR, Buzanska L, Sarnowska A, Domanska-Janik K. Human Somatic Stem Cell Neural Differentiation Potential. Results Probl Cell Differ 2018; 66:21-87. [DOI: 10.1007/978-3-319-93485-3_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
11
|
Svitina H, Kyryk V, Skrypkina I, Kuchma M, Bukreieva T, Areshkov P, Shablii Y, Denis Y, Klymenko P, Garmanchuk L, Ostapchenko L, Lobintseva G, Shablii V. Placenta-derived multipotent cells have no effect on the size and number of DMH-induced colon tumors in rats. Exp Ther Med 2017; 14:2135-2147. [PMID: 28962134 PMCID: PMC5609206 DOI: 10.3892/etm.2017.4792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
Transplantation of placenta-derived multipotent cells (PDMCs) is a promising approach for cell therapy to treat inflammation-associated colon diseases. However, the effect of PDMCs on colon cancer cells remains unknown. The aim of the present study was to characterize PDMCs obtained from human (hPDMCs) and rat (rPDMCs) placentas and to evaluate their impact on colon cancer progression in rats. PDMCs were obtained from human and rat placentas by tissue explant culturing. Stemness- and trophoblast-related gene expression was studied using reverse transcription-polymerase chain reaction (RT-PCR), and surface markers and intracellular proteins were detected using flow cytometry and immunofluorescence, respectively. Experimental colon carcinogenesis was induced in male albino Wistar rats by injecting 20 mg/kg dimethylhydrazine (DMH) once a week for 20 consecutive weeks. The administration of rPDMCs and hPDMC was performed at week 22 after the initial DMH-injection. All animals were sacrificed through carbon dioxide asphyxiation at week 5 after cell transplantation. The number and size of each tumor lesion was calculated. The type of tumor was determined by standard histological methods. Cell engraftment was determined by PCR and immunofluorescence. Results demonstrated that rPDMCs possessed the immunophenotype and differentiation potential inherent in MSCs; however, hPDMCs exhibited a lower expression of cluster of differentiation 44 and did not express trophoblast-associated genes. The data of the present study indicated that PDMCs may engraft in different tissues but do not significantly affect DMH-induced tumor growth during short-term observations.
Collapse
Affiliation(s)
- Hanna Svitina
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, 03680 Kyiv, Ukraine.,Department of Biochemistry, Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine
| | - Vitaliy Kyryk
- Department of Cell and Tissue Technologies, State Institute of Genetics and Regenerative Medicine of Academy of Medicine of Ukraine, 04114 Kyiv, Ukraine
| | - Inessa Skrypkina
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, 03680 Kyiv, Ukraine.,Department of Functional Genomics, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, 03680 Kyiv, Ukraine
| | - Maria Kuchma
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, 03680 Kyiv, Ukraine.,Department of Functional Genomics, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, 03680 Kyiv, Ukraine
| | - Tetiana Bukreieva
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, 03680 Kyiv, Ukraine
| | - Pavlo Areshkov
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, 03680 Kyiv, Ukraine.,Department of Functional Genomics, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, 03680 Kyiv, Ukraine
| | - Yulia Shablii
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, 03680 Kyiv, Ukraine
| | - Yevheniy Denis
- Department of Biochemistry, Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine
| | - Pavlo Klymenko
- Department of Cell and Tissue Technologies, State Institute of Genetics and Regenerative Medicine of Academy of Medicine of Ukraine, 04114 Kyiv, Ukraine
| | - Liudmyla Garmanchuk
- Department of Biochemistry, Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine
| | - Liudmyla Ostapchenko
- Department of Biochemistry, Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine
| | - Galina Lobintseva
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, 03680 Kyiv, Ukraine
| | - Volodymyr Shablii
- Cell Culture Laboratory, Cryobank, Institute of Cell Therapy, 03680 Kyiv, Ukraine.,Department of Biochemistry, Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 01601 Kyiv, Ukraine
| |
Collapse
|
12
|
Molecular Imaging of Stem Cell Transplantation for Liver Diseases: Monitoring, Clinical Translation, and Theranostics. Stem Cells Int 2016; 2016:4058656. [PMID: 28070195 PMCID: PMC5192340 DOI: 10.1155/2016/4058656] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/01/2016] [Indexed: 02/07/2023] Open
Abstract
Stem cell transplantation has been investigated to rescue experimental liver failure and is promising to offer an alternative therapy to liver transplantation for liver diseases treatment. Several clinical studies in this field have been carried out, but the therapeutic benefit of this treatment is still controversial. A major obstacle to developing stem cell therapies in clinic is being able to visualize the cells in vivo. Imaging modalities allow optimization of delivery, detecting cell survival and functionality by in vivo monitoring these transplanted graft cells. Moreover, theranostic imaging is a brand new field that utilizes nanometer-scale materials to glean diagnostic insight for simultaneous treatment, which is very promising to improve stem cell-based therapy for treatment of liver diseases. The aim of this review was to summarize the various imaging tools that have been explored with advanced molecular imaging probes. We also outline some recent progress of preclinical and clinical studies of liver stem cells transplantation. Finally, we discuss theranostic imaging for stem cells transplantation for liver dysfunction and future opportunities afforded by theranostic imaging.
Collapse
|