1
|
Paola S, Lara G, Michela M, Silvia DC, Serena M, Rosalba P, Maria NA, Eleonora C, Fiorella C, Giulia G, Giovanna T, Giuseppe N, Federica S. When do the pathological signs become evident? Study of human mesenchymal stem cells in MDPL syndrome. Aging (Albany NY) 2024; 16:13505-13525. [PMID: 39611849 DOI: 10.18632/aging.206159] [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: 07/19/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024]
Abstract
Aging syndromes are rare genetic disorders sharing the features of accelerated senescence. Among these, Mandibular hypoplasia, Deafness and Progeroid features with concomitant Lipodystrophy (MDPL; OMIM #615381) is a rare autosomal dominant disease due to a de novo in-frame deletion in POLD1 gene, encoding the catalytic subunit of DNA polymerase delta. Here, we investigated how MSCs may contribute to the phenotypes and progression of premature aging syndromes such as MDPL. In human induced pluripotent stem cells (hiPSCs)-derived MSCs of three MDPL patients we detected several hallmarks of senescence, including (i) abnormal nuclear morphology, (ii) micronuclei presence, (iii) slow cell proliferation and cell cycle progression, (iv) reduced telomere length, and (v) increased levels of mitochondrial reactive oxygen species (ROS). We newly demonstrated that the pathological hallmarks of senescence manifest at an early stage of human development and represent a warning sign for the progression of the disease. Dissecting the mechanisms underlying stem cell dysfunction during aging can thereby contribute to the development of timely pharmacological therapies for ameliorating the pathological phenotype.
Collapse
Affiliation(s)
- Spitalieri Paola
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Guerrieri Lara
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Murdocca Michela
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Di Cesare Silvia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Maccaroni Serena
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Pecorari Rosalba
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | | | - Candi Eleonora
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Colasuonno Fiorella
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
- Department of Science – LIME, Roma Tre University, Rome, Italy
| | - Gori Giulia
- Meyer Children’s Hospital IRCCS, Florence, Italy
| | | | - Novelli Giuseppe
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Sangiuolo Federica
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
2
|
Miyamoto D, Matsuguma K, Nagai K, Miyoshi T, Hara T, Matsushima H, Soyama A, Ochiya T, Miyazaki Y, Eguchi S. Efficacy of chemically induced human hepatic progenitor cells from diseased liver against nonalcoholic steatohepatitis model. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:697-704. [PMID: 39021351 DOI: 10.1002/jhbp.12046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
BACKGROUND Numerous chemical reprogramming techniques have been reported, rendering them applicable to regenerative medicine research. The aim of our study was to evaluate the therapeutic potential of human CLiP derived from clinical specimens transplanted into a nonalcoholic steatohepatitis (NASH) mouse model of liver fibrosis. METHODS We successfully generated chemically induced liver progenitor (CLiP), which exhibited progenitor-like characteristics, through stimulation with low-molecular-weight compounds. We elucidated their cell differentiation ability and therapeutic effects. However, the therapeutic efficacy of human CLiP generated from clinical samples on liver fibrosis, such as liver cirrhosis, remains unproven. RESULTS Following a 4 week period, transplanted human CLiP in the NASH model differentiated into mature hepatocytes and demonstrated suppressive effects on liver injury markers (i.e., aspartate transaminase and alanine transaminase). Although genes related to inflammation and fat deposition did not change in the human CLiP transplantation group, liver fibrosis-related factors (Acta2 and Col1A1) showed suppressive effects on gene expression following transplantation, with approximately a 60% reduction in collagen fibers. Importantly, human CLiP could be efficiently induced from hepatocytes isolated from the cirrhotic liver, underscoring the feasibility of using autologous hepatocytes to produce human CLiP. CONCLUSION Our findings demonstrate the effectiveness of human CLiP transplantation as a viable cellular therapy for liver fibrosis, including NASH liver. These results hold promise for the development of liver antifibrosis therapy utilizing human CLiP within the field of liver regenerative medicine.
Collapse
Affiliation(s)
- Daisuke Miyamoto
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kunihito Matsuguma
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuhiro Nagai
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Clinical Laboratory, NHO Nagasaki Medical Center, Nagasaki, Japan
| | - Takayuki Miyoshi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takanobu Hara
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hajime Matsushima
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akihiko Soyama
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Ochiya
- Department of Molecular Cell Therapy Research, Medical Research Institute, Tokyo Medical University, Tokyo, Japan
| | - Yasushi Miyazaki
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, Japan
- Department of Hematology, Atomic Bomb Disedase Institute, Nagasaki University, Nagasaki, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
3
|
Ikeguchi R, Aoyama T, Noguchi T, Ushimaru M, Amino Y, Nakakura A, Matsuyama N, Yoshida S, Nagai-Tanima M, Matsui K, Arai Y, Torii Y, Miyazaki Y, Akieda S, Matsuda S. Peripheral nerve regeneration following scaffold-free conduit transplant of autologous dermal fibroblasts: a non-randomised safety and feasibility trial. COMMUNICATIONS MEDICINE 2024; 4:12. [PMID: 38278956 PMCID: PMC10817910 DOI: 10.1038/s43856-024-00438-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND The use of Bio 3D nerve conduits is a promising approach for peripheral nerve reconstruction. This study aimed to assess their safety in three patients with peripheral nerve defects in their hands. METHODS We describe a single institution, non-blinded, non-randomised control trial conducted at Kyoto University Hospital. Eligibility criteria included severed peripheral nerve injuries or a defect in the region distal to the wrist joint not caused by a congenital anomaly; a defect with a length of ≤20 mm in a nerve with a diameter ≤2 mm; failed results of sensory functional tests; ability to register in the protocol within 6 months from the day of injury; refusal of artificial nerve or autologous nerve transplantation; age 20-60 years; and willingness to participate and provide informed written consent. Six weeks before transplantation, skin was harvested, dermal fibroblasts were isolated and expanded, and Bio 3D nerve conduits were created using a Bio 3D printer. Bio 3D nerve conduits were transplanted into the patients' nerve defects. The safety of Bio 3D nerve conduits in patients with a peripheral nerve injury in the distal part of the wrist joint were assessed over a 48-week period after transplantation. RESULTS No adverse events related to the use of Bio 3D nerve conduits were observed in any patient, and all three patients completed the trial. CONCLUSIONS Bio 3D nerve conduits were successfully used for clinical nerve reconstruction without adverse events and are a possible treatment option for peripheral nerve injuries.
Collapse
Affiliation(s)
- Ryosuke Ikeguchi
- Department of Rehabilitation Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Tomoki Aoyama
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Noguchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mika Ushimaru
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Yoko Amino
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Akiyoshi Nakakura
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Noriko Matsuyama
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Shiori Yoshida
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Momoko Nagai-Tanima
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiko Matsui
- Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan
| | - Yasuyuki Arai
- Center for Research and Application of Cellular Therapy, Kyoto University Hospital, Kyoto, Japan
| | | | | | | | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
4
|
Eriani K, Desriani D, Suhartono S, Br Sibarani MJ, Ichsan I, Syafrizal D, Asmara H. The differentiation of mesenchymal bone marrow stem cells into nerve cells induced by Chromolaena odorata extracts. F1000Res 2022; 11:252. [PMID: 35811803 PMCID: PMC9214272 DOI: 10.12688/f1000research.108741.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) can differentiate into nerve cells with an induction from chemical compounds in medium culture.
Chromolaena odorata contains active compounds, such as alkaloids and flavonoids, that can initiate the transformation of MSCs into nerve cells. The aim of this study was to determine the potential of methanol extracted
C. odorata leaf to induce the differentiation of bone marrow MSCs into nerve cells. Methods: A serial concentration of
C. odorata leaf extract (0.7–1.0 mg/mL) with two replications was used. The parameters measured were the number of differentiated MSCs into nerve cells (statistically analyzed using ANOVA) and cell confirmation using reverse transcription polymerase chain reaction (RT-PCR). Results: The results showed that the
C. odorata extract had a significant effect on the number MSCs differentiating into nerve cells (
p < 0.05) on the doses of 0.8 mg/ml with 22.6%. Molecular assay with RT-PCR confirmed the presence of the nerve cell gene in all of the samples. Conclusions: In conclusion, this study showed the potential application of
C. odorata leaf extract in stem cell therapy for patients experiencing neurodegeneration by inducing the differentiation of MSCs into nerve cells.
Collapse
Affiliation(s)
- Kartini Eriani
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Desriani Desriani
- Research Center for Biotechnology, National Research and Innovation Agency, Cibinong, Bogor, West Java, 16911, Indonesia
| | - Suhartono Suhartono
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Miftahul Jannah Br Sibarani
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Ichsan Ichsan
- Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Dedy Syafrizal
- Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Hadhymulya Asmara
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
5
|
Mönch D, Koch J, Dahlke MH. Are Mesenchymal Stem Cells Fibroblasts with Benefits? CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00210-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
6
|
Beklemisheva VR, Belokopytova PS, Fishman VS, Menzorov AG. Derivation of Ringed Seal ( Phoca hispida) Induced Multipotent Stem Cells. Cell Reprogram 2021; 23:326-335. [PMID: 34788122 DOI: 10.1089/cell.2021.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Induced pluripotent stem (iPS) cells have been produced just for a few species among order Carnivora: snow leopard, Bengal tiger, serval, jaguar, cat, dog, ferret, and American mink. We applied the iPS cell derivation protocol to the ringed seal (Phoca hispida) fibroblasts. The resulting cell line had the expression of pluripotency marker gene Rex1. Differentiation in embryoid body-like structures allowed us to register expression of AFP, endoderm marker, and Cdx2, trophectoderm marker, but not neuronal (ectoderm) markers. The cells readily differentiated into adipocytes and osteocytes, mesoderm cell types of origin. Transcriptome analysis allowed us to conclude that the cell line does not resemble human pluripotent cells, and, therefore, most probably is not pluripotent. Thus, we produced ringed seal multipotent stem cell line capable of differentiation into adipocytes and osteocytes.
Collapse
Affiliation(s)
- Violetta R Beklemisheva
- Institute of Molecular and Cellular Biology of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Polina S Belokopytova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Veniamin S Fishman
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Aleksei G Menzorov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
7
|
Kwon D, Ahn HJ, Han MJ, Ji M, Ahn J, Seo KW, Kang KS. Human Leukocyte Antigen Class I Pseudo-Homozygous Mesenchymal Stem Cells Derived from Human Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2021; 16:792-808. [PMID: 32712868 DOI: 10.1007/s12015-020-09990-9] [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: 12/17/2022]
Abstract
Mesenchymal stem cells (MSC) are an important type of cell that are highly recognized for their safety and efficacy as a cell therapy agent. In order to obtain MSC, primary tissues (adipose tissue, bone marrow, and umbilical cord blood) must be used; however, these tissues, especially umbilical cord blood, are difficult to obtain due to various reasons, such as the low birth rate trend. In addition, to maximize the safety and efficacy of MSC as allogenic cell therapeutic agents, it is desirable to minimize the possibility of an immune rejection reaction after in vivo transplantation. This study tried to establish a novel method for producing induced pluripotent stem cells (iPSC)-derived MSC in which the human leukocyte antigen (HLA)-class I gene is knocked out. To do so, dermal fibroblast originated iPSC generation using Yamanaka 4-factor, HLA class I gene edited iPSC generation using CRISPR/Cas9, and differentiation from iPSC to MSC using MSC culture medium was utilized. Through this, HLA-A, B, and C pseudo-homozygous iPSC-derived MSC (KO iMSC) were produced by monoallelically knocking out the polymorphic HLA-A, B, and C genes, which are the major causes of immune rejection during allogenic cell transplantation. Produced KO iMSC possesses multipotency and it was safe in vivo to be able to be differentiated to cartilage. In addition, it was not attacked by natural killer cells unlike HLA class I null cells. In conclusion, KO iMSC that do not induce immune rejection during allogenic cell transplantation can be produced. In the future, KO iMSC can be successfully utilized as allogenic cell therapeutic agents for many recipients through HLA screening.
Collapse
Affiliation(s)
- Daekee Kwon
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Hee-Jin Ahn
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Mi-Jung Han
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Minjun Ji
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Jongchan Ahn
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Kwang-Won Seo
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea
| | - Kyung-Sun Kang
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Biomedical Science Building, #81 Seoul National University, Seoul, 08826, South Korea. .,Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
8
|
Chen Q, Yuan C, Jiang S, Heng BC, Zou T, Shen Z, Wang P, Zhang C. Small molecules efficiently reprogram apical papilla stem cells into neuron-like cells. Exp Ther Med 2021; 21:546. [PMID: 33850518 DOI: 10.3892/etm.2021.9978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Stem cell-based therapy may provide a novel approach for neural tissue regeneration. A small molecule cocktail-based culture protocol was previously shown to enhance neurogenic differentiation of stem cells from dental tissues. The present study aimed to investigate the early phase of small molecule-induced neurogenic differentiation of stem cells from the apical papilla (SCAP). SCAP were cultured in neural-induction medium or neural-induction medium with small molecules (NIMS-SCAP) and examined for their cell morphologies. Expression levels of neural progenitor cell-related markers, including Nestin, paired-box gene 6 (Pax6) and Sry-related HMG box 2 (Sox2), were examined using western blotting and immunocytofluorescence. Expression of differentiated neuron-related markers, including neurofilament protein (NFM), neuron-specific nuclear protein (NeuN) and microtubule-associated protein (MAP)-2, were also examined using western blotting, while NFM and MAP2 gene expression and cell proliferation were assessed using reverse transcription-quantitative (RT-q)PCR and Cell Counting Kit (CCK)-8 assays, respectively. SCAP morphology was affected by small molecules after as little as 30 min. Specifically, Nestin, Pax6 and Sox2 expression detected using western blotting was increased by day 3 but then decreased over the course of 7 days with neural induction, while immunocytofluorescence revealed expression of all three markers in NIMS-SCAP. The protein levels of NFM, NeuN and MAP2 on day 7 were significantly upregulated in NIMS-SCAP, as detected using western blotting, while NFM and MAP2 gene expression levels detected using RT-qPCR were significantly increased on days 5 and 7. Proliferation of NIMS-SCAP ceased after 5 days. Electrophysiological analysis showed that only SCAP cultured in NIMS had the functional activity of neuronal cells. Thus, small molecules reprogrammed SCAP into neural progenitor cells within the first 3 days, followed by further differentiation into neuron-like cells.
Collapse
Affiliation(s)
- Qixin Chen
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China.,Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Changyong Yuan
- Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Shan Jiang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Ting Zou
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Zhongshan Shen
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Penglai Wang
- Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Chengfei Zhang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| |
Collapse
|
9
|
Recent Advances in Understandings Towards Pathogenesis and Treatment for Intrauterine Adhesion and Disruptive Insights from Single-Cell Analysis. Reprod Sci 2020; 28:1812-1826. [PMID: 33125685 PMCID: PMC8189970 DOI: 10.1007/s43032-020-00343-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022]
Abstract
Intrauterine adhesion is a major cause of menstrual irregularities, infertility, and recurrent pregnancy losses and the progress towards its amelioration and therapy is slow and unsatisfactory. We aim to summarize and evaluate the current treatment progress and research methods for intrauterine adhesion. We conducted literature review in January 2020 by searching articles at PubMed on prevention and treatment, pathogenesis, the repair of other tissues/organs, cell plasticity, and the stem cell–related therapies for intrauterine adhesion. A total of 110 articles were selected for review. Uterine cell heterogeneity, expression profile, and cell-cell interaction were investigated based on scRNA-seq of uterus provided by Human Cell Landscape (HCL) project. Previous knowledge on intrauterine adhesion (IUA) pathogenesis was mostly derived from correlation studies by differentially expressed genes between endometrial tissue of intrauterine adhesion patients/animal models and normal endometrial tissue. Although the TGF-β1/SMAD pathway was suggested as the key driver for IUA pathogenesis, uterine cell heterogeneity and distinct expression profile among different cell types highlighted the importance of single-cell investigations. Cell-cell interaction in the uterus revealed the central hub of endothelial cells interacting with other cells, with endothelial cells in endothelial to mesenchymal transition and fibroblasts as the strongest interaction partners. The potential of stem cell–related therapies appeared promising, yet suffers from largely animal studies and nonstandard study design. The need to dissect the roles of endometrial cells, endothelial cells, and fibroblasts and their interaction is evident in order to elucidate the molecular and cellular mechanisms in both intrauterine adhesion pathogenesis and treatment.
Collapse
|
10
|
Yurie H, Ikeguchi R, Aoyama T, Ito A, Tanaka M, Noguchi T, Oda H, Takeuchi H, Mitsuzawa S, Ando M, Yoshimoto K, Akieda S, Nakayama K, Matsuda S. Mechanism of Peripheral Nerve Regeneration Using a Bio 3D Conduit Derived from Normal Human Dermal Fibroblasts. J Reconstr Microsurg 2020; 37:357-364. [PMID: 32957155 DOI: 10.1055/s-0040-1716855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND We previously reported the development of a scaffold-free Bio three-dimensional (3D) nerve conduit from normal human dermal fibroblasts (NHDFs). The aim of this study was to investigate the regenerative mechanism of peripheral nerve cells using a Bio 3D conduit in a rat sciatic nerve defect model. METHODS Bio 3D conduits composed of NHDFs were developed, and cell viability was evaluated using a LIVE/DEAD cell viability assay immediately before transplantation and 1-week post-surgery. Tracking analysis using PKH26-labeled NHDFs was performed to assess the distribution of NHDFs within the regenerated nerve and the differentiation of NHDFs into functional Schwann cells (SCs). RESULTS The assessment of the viability of cells within the Bio 3D conduit showed high cell viability both immediately before transplantation and 1-week post-surgery (88.56 ± 1.70 and 87.58 ± 9.11, respectively). A modified Masson's trichrome staining of the Bio 3D conduit revealed the formation of a prominent extracellular matrix (ECM) in between the cells. We observed, via tracking analysis, that the tube-like distribution of the NHDFs remained stable, the majority of the regenerated axons had penetrated this structure and PKH26-labeled cells were also positive for S-100. CONCLUSION Abundant ECM formation resulted in a stable tube-like structure of the Bio 3D conduit with high cell viability. NHDFs in the Bio 3D conduit have the potential to differentiate into SCs-like cells.
Collapse
Affiliation(s)
- Hirofumi Yurie
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Ikeguchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mai Tanaka
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Noguchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroki Oda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hisataka Takeuchi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sadaki Mitsuzawa
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Maki Ando
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koichi Yoshimoto
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Koichi Nakayama
- Department of Regenerative Medicine and Biomedical Engineering Faculty of Medicine, Saga University, Saga, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
11
|
Spehar K, Pan A, Beerman I. Restoring aged stem cell functionality: Current progress and future directions. Stem Cells 2020; 38:1060-1077. [PMID: 32473067 PMCID: PMC7483369 DOI: 10.1002/stem.3234] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/07/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022]
Abstract
Stem cell dysfunction is a hallmark of aging, associated with the decline of physical and cognitive abilities of humans and other mammals [Cell 2013;153:1194]. Therefore, it has become an active area of research within the aging and stem cell fields, and various techniques have been employed to mitigate the decline of stem cell function both in vitro and in vivo. While some techniques developed in model organisms are not directly translatable to humans, others show promise in becoming clinically relevant to delay or even mitigate negative phenotypes associated with aging. This review focuses on diet, treatment, and small molecule interventions that provide evidence of functional improvement in at least one type of aged adult stem cell.
Collapse
Affiliation(s)
- Kevin Spehar
- Epigenetics and Stem Cell Aging Unit, Translational Gerontology Branch, National Institute on Aging, NIH, BRC, Baltimore, Maryland
| | - Andrew Pan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Isabel Beerman
- Epigenetics and Stem Cell Aging Unit, Translational Gerontology Branch, National Institute on Aging, NIH, BRC, Baltimore, Maryland
| |
Collapse
|
12
|
Steens J, Unger K, Klar L, Neureiter A, Wieber K, Hess J, Jakob HG, Klump H, Klein D. Direct conversion of human fibroblasts into therapeutically active vascular wall-typical mesenchymal stem cells. Cell Mol Life Sci 2020; 77:3401-3422. [PMID: 31712992 PMCID: PMC7426315 DOI: 10.1007/s00018-019-03358-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 09/27/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022]
Abstract
Cell-based therapies using adult stem cells are promising options for the treatment of a number of diseases including autoimmune and cardiovascular disorders. Among these, vascular wall-derived mesenchymal stem cells (VW-MSCs) might be particularly well suited for the protection and curative treatment of vascular damage because of their tissue-specific action. Here we report a novel method for the direct conversion of human skin fibroblasts towards MSCs using a VW-MSC-specific gene code (HOXB7, HOXC6 and HOXC8) that directs cell fate conversion bypassing pluripotency. This direct programming approach using either a self-inactivating (SIN) lentiviral vector expressing the VW-MSC-specific HOX-code or a tetracycline-controlled Tet-On system for doxycycline-inducible gene expressions of HOXB7, HOXC6 and HOXC8 successfully mediated the generation of VW-typical MSCs with classical MSC characteristics in vitro and in vivo. The induced VW-MSCs (iVW-MSCs) fulfilled all criteria of MSCs as defined by the International Society for Cellular Therapy (ISCT). In terms of multipotency and clonogenicity, which are important specific properties to discriminate MSCs from fibroblasts, iVW-MSCs behaved like primary ex vivo isolated VW-MSCs and shared similar molecular and DNA methylation signatures. With respect to their therapeutic potential, these cells suppressed lymphocyte proliferation in vitro, and protected mice against vascular damage in a mouse model of radiation-induced pneumopathy in vivo, as well as ex vivo cultured human lung tissue. The feasibility to obtain patient-specific VW-MSCs from fibroblasts in large amounts by a direct conversion into induced VW-MSCs could potentially open avenues towards novel, MSC-based therapies.
Collapse
Affiliation(s)
- Jennifer Steens
- Institute for Cell Biology (Cancer Research), University Hospital Essen, Medical Faculty, University of Duisburg-Essen, Virchowstr. 173, Ger-45122, Essen, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics and Clinical Cooperation Group "Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Lea Klar
- Institute for Cell Biology (Cancer Research), University Hospital Essen, Medical Faculty, University of Duisburg-Essen, Virchowstr. 173, Ger-45122, Essen, Germany
| | - Anika Neureiter
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Karolin Wieber
- Institute for Cell Biology (Cancer Research), University Hospital Essen, Medical Faculty, University of Duisburg-Essen, Virchowstr. 173, Ger-45122, Essen, Germany
| | - Julia Hess
- Research Unit Radiation Cytogenetics and Clinical Cooperation Group "Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Heinz G Jakob
- Department of Thoracic and Cardiovascular Surgery, West-German Heart and Vascular Center Essen, University Duisburg-Essen, Essen, Germany
| | - Hannes Klump
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, Medical Faculty, University of Duisburg-Essen, Virchowstr. 173, Ger-45122, Essen, Germany.
| |
Collapse
|
13
|
Sweat gland regeneration: Current strategies and future opportunities. Biomaterials 2020; 255:120201. [PMID: 32592872 DOI: 10.1016/j.biomaterials.2020.120201] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/21/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022]
Abstract
For patients with extensive skin defects, loss of sweat glands (SwGs) greatly decreases their quality of life. Indeed, difficulties in thermoregulation, ion reabsorption, and maintaining fluid balance might render them susceptible to hyperthermia, heatstroke, or even death. Despite extensive studies on the stem cell biology of the skin in recent years, in-situ regeneration of SwGs with both structural and functional fidelity is still challenging because of the limited regenerative capacity and cell fate control of resident progenitors. To overcome these challenges, one must consider both the intrinsic factors relevant to genetic and epigenetic regulation and cues from the cellular microenvironment. Here, we describe recent progress in molecular biology, developmental pathways, and cellular evolution associated with SwGdevelopment and maturation. This is followed by a summary of the current strategies used for cell-fate modulation, transmembrane drug delivery, and scaffold design associated with SwGregeneration. Finally, we offer perspectives for creating more sophisticated systems to accelerate patients' innate healing capacity and developing engineered skin constructs to treat or replace damaged tissues structurally and functionally.
Collapse
|
14
|
He X, Chi G, Li M, Xu J, Zhang L, Song Y, Wang L, Li Y. Characterisation of extraembryonic endoderm-like cells from mouse embryonic fibroblasts induced using chemicals alone. Stem Cell Res Ther 2020; 11:157. [PMID: 32299508 PMCID: PMC7164364 DOI: 10.1186/s13287-020-01664-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/21/2020] [Accepted: 03/27/2020] [Indexed: 11/10/2022] Open
Abstract
Background The development of somatic reprogramming, especially purely chemical reprogramming, has significantly advanced biological research. And chemical-induced extraembryonic endoderm-like (ciXEN) cells have been confirmed to be an indispensable intermediate stage of chemical reprogramming. They resemble extraembryonic endoderm (XEN) cells in terms of transcriptome, reprogramming potential, and developmental ability in vivo. However, the other characteristics of ciXEN cells and the effects of chemicals and bFGF on the in vitro culture of ciXEN cells have not been systematically reported. Methods Chemicals and bFGF in combination with Matrigel were used to induce the generation of ciXEN cells derived from mouse embryonic fibroblasts (MEFs). RNA sequencing was utilised to examine the transcriptome of ciXEN cells, and PCR/qPCR assays were performed to evaluate the mRNA levels of the genes involved in this study. Hepatic functions were investigated by periodic acid-Schiff staining and indocyanine green assay. Lactate production, ATP detection, and extracellular metabolic flux analysis were used to analyse the energy metabolism of ciXEN cells. Results ciXEN cells expressed XEN-related genes, exhibited high proliferative capacity, had the ability to differentiate into visceral endoderm in vitro, and possessed the plasticity allowing for their differentiation into induced hepatocytes (iHeps). Additionally, the upregulated biological processes of ciXEN cells compared to those in MEFs focused on metabolism, but their energy production was independent of glycolysis. Furthermore, without the cocktail of chemicals and bFGF, which are indispensable for the generation of ciXEN cells, induced XEN (iXEN) cells remained the expression of XEN markers, the high proliferative capacity, and the plasticity to differentiate into iHeps in vitro. Conclusions ciXEN cells had high plasticity, and energy metabolism was reconstructed during chemical reprogramming, but it did not change from aerobic oxidation to glycolysis. And the cocktail of chemicals and bFGF were non-essential for the in vitro culture of ciXEN cells.
Collapse
Affiliation(s)
- Xia He
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Jinying Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Lihong Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Yaolin Song
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, People's Republic of China
| | - Lina Wang
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China.,Department of Paediatrics, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
15
|
Zaffaroni G, Okawa S, Morales-Ruiz M, del Sol A. An integrative method to predict signalling perturbations for cellular transitions. Nucleic Acids Res 2020; 47:e72. [PMID: 30949696 PMCID: PMC6614844 DOI: 10.1093/nar/gkz232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/22/2019] [Accepted: 03/22/2019] [Indexed: 12/19/2022] Open
Abstract
Induction of specific cellular transitions is of clinical importance, as it allows to revert disease cellular phenotype, or induce cellular reprogramming and differentiation for regenerative medicine. Signalling is a convenient way to accomplish such transitions without transfer of genetic material. Here we present the first general computational method that systematically predicts signalling molecules, whose perturbations induce desired cellular transitions. This probabilistic method integrates gene regulatory networks (GRNs) with manually-curated signalling pathways obtained from MetaCore from Clarivate Analytics, to model how signalling cues are received and processed in the GRN. The method was applied to 219 cellular transition examples, including cell type transitions, and overall correctly predicted experimentally validated signalling molecules, consistently outperforming other well-established approaches, such as differential gene expression and pathway enrichment analyses. Further, we validated our method predictions in the case of rat cirrhotic liver, and identified the activation of angiopoietins receptor Tie2 as a potential target for reverting the disease phenotype. Experimental results indicated that this perturbation induced desired changes in the gene expression of key TFs involved in fibrosis and angiogenesis. Importantly, this method only requires gene expression data of the initial and desired cell states, and therefore is suited for the discovery of signalling interventions for disease treatments and cellular therapies.
Collapse
Affiliation(s)
- Gaia Zaffaroni
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
| | - Satoshi Okawa
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
- Integrated BioBank of Luxembourg, Dudelange L-3555, Luxembourg
| | - Manuel Morales-Ruiz
- Biochemistry and Molecular Genetics Department-Hospital Clínic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona 08036, Spain
- Working group for the biochemical assessment of hepatic disease-SEQC, Barcelona 08036, Spain
- Department of Biomedicine-Biochemistry Unit, School of Medicine-University of Barcelona, Barcelona 08036, Spain
| | - Antonio del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
- CIC bioGUNE, Bizkaia Technology Park, Derio 48160, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
- To whom correspondence should be addressed. Tel: +352 46 66 44 6982; Fax: +352 46 66 44 6949;
| |
Collapse
|
16
|
Chen X, Xu H, Hou J, Wang H, Zheng Y, Li H, Cai H, Han X, Dai J. Epithelial cell senescence induces pulmonary fibrosis through Nanog-mediated fibroblast activation. Aging (Albany NY) 2019; 12:242-259. [PMID: 31891567 PMCID: PMC6977687 DOI: 10.18632/aging.102613] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease tightly correlated with aging. The pathological features of IPF include epithelial cell senescence and abundant foci of highly activated pulmonary fibroblasts. However, the underlying mechanism between epithelial cell senescence and pulmonary fibroblast activation remain to be elucidated. In our study, we demonstrated that Nanog, as a pluripotency gene, played an essential role in the activation of pulmonary fibroblasts. In the progression of IPF, senescent epithelial cells could contribute to the activation of pulmonary fibroblasts via increasing the expression of senescence-associated secretory phenotype (SASP). In addition, we found activated pulmonary fibroblasts exhibited aberrant activation of Wnt/β-catenin signalling and elevated expression of Nanog. Further study revealed that the activation of Wnt/β-catenin signalling was responsible for senescent epithelial cell-induced Nanog phenotype in pulmonary fibroblasts. β-catenin was observed to bind to the promoter of Nanog during the activation of pulmonary fibroblasts. Targeted inhibition of epithelial cell senescence or Nanog could effectively suppress the activation of pulmonary fibroblasts and impair the development of pulmonary fibrosis, indicating a potential for the exploration of novel anti-fibrotic strategies.
Collapse
Affiliation(s)
- Xiang Chen
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Hongyang Xu
- Department of Critical Care Medicine, The Affiliated WuXi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Jiwei Hou
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Hui Wang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yi Zheng
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hui Li
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hourong Cai
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jinghong Dai
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
17
|
Kim S, Kim TM. Generation of mesenchymal stem-like cells for producing extracellular vesicles. World J Stem Cells 2019; 11:270-280. [PMID: 31171955 PMCID: PMC6545523 DOI: 10.4252/wjsc.v11.i5.270] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/02/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitor cells with therapeutic potential against autoimmune diseases, inflammation, ischemia, and metabolic disorders. Contrary to the previous conceptions, recent studies have revealed that the tissue repair and immunomodulatory functions of MSCs are largely attributed to their secretome, rather than their potential to differentiate into desired cell types. The composition of MSC secretome encompasses cytokines and growth factors, in addition to the cell-derived structures known as extracellular vesicles (EVs). EVs are membrane-enclosed nanoparticles that are capable of delivering biomolecules, and it is now believed that MSC-derived EVs are the major players that induce biological changes in the target tissues. Based on these EVs’ characteristics, the potential of EVs derived from MSC (MSC-EV) in terms of tissue regeneration and immune modulation has grown during the last decade. However, the use of MSCs for producing sufficient amount of EVs has not been satisfactory due to limitations in the cell growth and large variations among the donor cell types. In this regard, pluripotent stem cells (PSCs)-derived MSC-like cells, which can be robustly induced and expanded in vitro, have emerged as more accessible cell source that can overcome current limitations of using MSCs for EV production. In this review, we have highlighted the methods of generating MSC-like cells from PSCs and their therapeutic outcome in preclinical studies. Finally, we have also discussed future requirements for making this cell-free therapy clinically feasible.
Collapse
Affiliation(s)
- Soo Kim
- Brexogen Research Center, Brexogen Inc., Seoul, Songpa-gu 05718, South Korea
| | - Tae Min Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Gangwon-do, Pyeongchang 25354, South Korea
| |
Collapse
|
18
|
Current Strategies to Generate Human Mesenchymal Stem Cells In Vitro. Stem Cells Int 2018; 2018:6726185. [PMID: 30224922 PMCID: PMC6129345 DOI: 10.1155/2018/6726185] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are heterogeneous multipotent stem cells that are involved in the development of mesenchyme-derived evolving structures and organs during ontogeny. In the adult organism, reservoirs of MSCs can be found in almost all tissues where MSCs contribute to the maintenance of organ integrity. The use of these different MSCs for cell-based therapies has been extensively studied over the past years, which highlights the use of MSCs as a promising option for the treatment of various diseases including autoimmune and cardiovascular disorders. However, the proportion of MSCs contained in primary isolates of adult tissue biopsies is rather low and, thus, vigorous ex vivo expansion is needed especially for therapies that may require extensive and repetitive cell substitution. Therefore, more easily and accessible sources of MSCs are needed. This review summarizes the current knowledge of the different strategies to generate human MSCs in vitro as an alternative method for their applications in regenerative therapy.
Collapse
|
19
|
Soontararak S, Chow L, Johnson V, Coy J, Wheat W, Regan D, Dow S. Mesenchymal Stem Cells (MSC) Derived from Induced Pluripotent Stem Cells (iPSC) Equivalent to Adipose-Derived MSC in Promoting Intestinal Healing and Microbiome Normalization in Mouse Inflammatory Bowel Disease Model. Stem Cells Transl Med 2018; 7:456-467. [PMID: 29635868 PMCID: PMC5980202 DOI: 10.1002/sctm.17-0305] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/19/2018] [Indexed: 02/06/2023] Open
Abstract
Cellular therapy with allogeneic or autologous mesenchymal stem cells (MSC) has emerged as a promising new therapeutic strategy for managing inflammatory bowel disease (IBD). However, MSC therapy ideally requires a convenient and relatively homogenous cell source (typically bone marrow or adipose tissues) and the ability to generate cells with stable phenotype and function. An alternative means of generating allogeneic MSC is to derive them from induced pluripotent stem cells (iPSC), which could in theory provide an indefinite supply of MSC with well-defined phenotype and function. Therefore, we compared the effectiveness of iPSC-derived MSC (iMSC) and adipose-derived MSC (adMSC) in a mouse model of IBD (dextran sodium sulfate-induced colitis), and investigated mechanisms of intestinal protection. We found that iMSC were equivalent to adMSC in terms of significantly improving clinical abnormalities in treated mice and reducing lesion scores and inflammation in the gut. Administration of iMSC also stimulated significant intestinal epithelial cell proliferation, increased in the numbers of Lgr5+ intestinal stem cells, and increased intestinal angiogenesis. In addition, the microbiome alterations present in mice with colitis were partially restored to resemble those of healthy mice following treatment with iMSC or adMSC. Thus, iMSC administration improved overall intestinal health and healing with equivalent potency to treatment with adMSC. This therefore is the first report of the effectiveness of iMSC in the treatment of IBD, along with a description of unique mechanisms of action with respect to intestinal healing and microbiome restoration. Stem Cells Translational Medicine 2018;7:456-467.
Collapse
Affiliation(s)
- Sirikul Soontararak
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Lyndah Chow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Valerie Johnson
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Jonathan Coy
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - William Wheat
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Daniel Regan
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Steven Dow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|
20
|
Chemical compound-based direct reprogramming for future clinical applications. Biosci Rep 2018; 38:BSR20171650. [PMID: 29739872 PMCID: PMC5938430 DOI: 10.1042/bsr20171650] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/29/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022] Open
Abstract
Recent studies have revealed that a combination of chemical compounds enables direct reprogramming from one somatic cell type into another without the use of transgenes by regulating cellular signaling pathways and epigenetic modifications. The generation of induced pluripotent stem (iPS) cells generally requires virus vector-mediated expression of multiple transcription factors, which might disrupt genomic integrity and proper cell functions. The direct reprogramming is a promising alternative to rapidly prepare different cell types by bypassing the pluripotent state. Because the strategy also depends on forced expression of exogenous lineage-specific transcription factors, the direct reprogramming in a chemical compound-based manner is an ideal approach to further reduce the risk for tumorigenesis. So far, a number of reported research efforts have revealed that combinations of chemical compounds and cell-type specific medium transdifferentiate somatic cells into desired cell types including neuronal cells, glial cells, neural stem cells, brown adipocytes, cardiomyocytes, somatic progenitor cells, and pluripotent stem cells. These desired cells rapidly converted from patient-derived autologous fibroblasts can be applied for their own transplantation therapy to avoid immune rejection. However, complete chemical compound-induced conversions remain challenging particularly in adult human-derived fibroblasts compared with mouse embryonic fibroblasts (MEFs). This review summarizes up-to-date progress in each specific cell type and discusses prospects for future clinical application toward cell transplantation therapy.
Collapse
|
21
|
The future of mesenchymal stem cell-based therapeutic approaches for cancer - From cells to ghosts. Cancer Lett 2017; 414:239-249. [PMID: 29175461 DOI: 10.1016/j.canlet.2017.11.025] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/19/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells which can differentiate into a variety of cell types including osteoblasts, adipocytes and chondrocytes. They are normally resident in adipose tissue, bone marrow and the umbilical cord, but can also be found in other tissues and are known to be recruited to sites of wound healing as well as growing tumours. The therapeutic potential of MSCs has been explored in a number of phase I/II and III clinical trials, of which several were targeted against graft-versus-host disease and to support engraftment of haematopoietic stem cells (HSCs), but currently only very few in the oncology field. There are now three clinical trials either ongoing or recruiting patients that use MSCs to treat tumour disease. In these, MSCs target gastrointestinal, lung and ovarian cancer, respectively. The first study uses MSCs loaded with a HSV-TK expression construct under the control of the CCL5 promoter, and has recently reported successful completion of Phase I/II. While no adverse side effects were seen during this study, no outcomes with respect to therapeutic benefits have been published. The other clinical trials targeting lung and ovarian cancer will be using MSCs expressing cytokines as therapeutic payload. Despite these encouraging early steps towards their clinical use, many questions are still unanswered regarding the biology of MSCs in normal and pathophysiological settings. In this review, in addition to summarising the current state of MSC-based therapeutic approaches for cancer, we will describe the remaining questions, obstacles and risks, as well as novel developments such as MSC-derived nanoghosts.
Collapse
|
22
|
Qin H, Zhao A, Fu X. Small molecules for reprogramming and transdifferentiation. Cell Mol Life Sci 2017; 74:3553-3575. [PMID: 28698932 PMCID: PMC11107793 DOI: 10.1007/s00018-017-2586-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/26/2017] [Accepted: 06/28/2017] [Indexed: 01/15/2023]
Abstract
Pluripotency reprogramming and transdifferentiation induced by transcription factors can generate induced pluripotent stem cells, adult stem cells or specialized cells. However, the induction efficiency and the reintroduction of exogenous genes limit their translation into clinical applications. Small molecules that target signaling pathways, epigenetic modifications, or metabolic processes can regulate cell development, cell fate, and function. In the recent decade, small molecules have been widely used in reprogramming and transdifferentiation fields, which can promote the induction efficiency, replace exogenous genes, or even induce cell fate conversion alone. Small molecules are expected as novel approaches to generate new cells from somatic cells in vitro and in vivo. Here, we will discuss the recent progress, new insights, and future challenges about the use of small molecules in cell fate conversion.
Collapse
Affiliation(s)
- Hua Qin
- Tianjin Medical University, Tianjin, 300070, China
| | - Andong Zhao
- Tianjin Medical University, Tianjin, 300070, China
| | - Xiaobing Fu
- Key Laboratory of Wound Repair and Regeneration of PLA, The First Hospital Affiliated to the PLA General Hospital, 51 Fu Cheng Road, Haidian District, Beijing, 100048, China.
| |
Collapse
|
23
|
Elimination of undifferentiated human embryonic stem cells by cardiac glycosides. Sci Rep 2017; 7:5289. [PMID: 28706279 PMCID: PMC5509667 DOI: 10.1038/s41598-017-05616-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022] Open
Abstract
An important safety concern in the use of human pluripotent stem cells (hPSCs) is tumorigenic risk, because these cells can form teratomas after an in vivo injection at ectopic sites. Several thousands of undifferentiated hPSCs are sufficient to induce teratomas in a mouse model. Thus, it is critical to remove all residue-undifferentiated hPSCs that have teratoma potential before the clinical application of hPSC-derived cells. In this study, our data demonstrated the cytotoxic effects of cardiac glycosides, such as digoxin, lanatoside C, bufalin, and proscillaridin A, in human embryonic stem cells (hESCs). This phenomenon was not observed in human bone marrow mesenchymal stem cells (hBMMSCs). Most importantly, digoxin and lanatoside C did not affect the stem cells’ differentiation ability. Consistently, the viability of the hESC-derived MSCs, neurons, and endothelium cells was not affected by the digoxin and lanatoside C treatment. Furthermore, the in vivo experiments demonstrated that digoxin and lanatoside C prevented teratoma formation. To the best of our knowledge, this study is the first to describe the cytotoxicity and tumor prevention effects of cardiac glycosides in hESCs. Digoxin and lanatoside C are also the first FDA-approved drugs that demonstrated cytotoxicity in undifferentiated hESCs.
Collapse
|