1
|
Li C, Wang B, Tu J, Liu C, Wang Y, Chen J, Huang Y, Liu B, Yuan X. ATM inhibition enhance immunotherapy by activating STING signaling and augmenting MHC Class I. Cell Death Dis 2024; 15:519. [PMID: 39033176 PMCID: PMC11271473 DOI: 10.1038/s41419-024-06911-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Accumulating evidence supports the concept that DNA damage response targeted therapies can improve antitumor immune response by increasing the immunogenicity of tumor cells and improving the tumor immune microenvironment. Ataxia telangiectasia mutated (ATM) is a core component of the DNA repair system. Although the ATM gene has a significant mutation rate in many human cancers, including colorectal, prostate, lung, and breast, it remains understudied compared with other DDR-involved molecules such as PARP and ATR. Here, we found that either gene knockout or drug intervention, ATM inhibition activated the cGAS/STING pathway and augmented MHC class I in CRC cells, and these effects could be amplified by radiation. Furthermore, we found that MHC class I upregulation induced by ATM inhibition is dependent on the activation of the NFκB/IRF1/NLRC5 pathway and independent of STING. Animal experiments have shown increasing infiltration and cytotoxic function of T cells and better survival in ATM-deficient tumors. This work indicated that ATM nonsense mutation predicted the clinical benefits of radiotherapy combined with immune checkpoint blockade for patients with CRC. It also provides a molecular mechanism rationale for ATM-targeted agents for patients with CRC.
Collapse
Affiliation(s)
- Chunya Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Boyu Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaofan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Chen Y, Zhou Y, Zhou Z, Fang Y, Ma L, Zhang X, Xiong J, Liu L. Hypoimmunogenic human pluripotent stem cells are valid cell sources for cell therapeutics with normal self-renewal and multilineage differentiation capacity. Stem Cell Res Ther 2023; 14:11. [PMID: 36691086 PMCID: PMC9872349 DOI: 10.1186/s13287-022-03233-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 12/28/2022] [Indexed: 01/25/2023] Open
Abstract
Hypoimmunogenic human pluripotent stem cells (hPSCs) are expected to serve as an unlimited cell source for generating universally compatible "off-the-shelf" cell grafts. However, whether the engineered hypoimmunogenic hPSCs still preserve their advantages of unlimited self-renewal and multilineage differentiation to yield functional tissue cells remains unclear. Here, we systematically studied the self-renewal and differentiation potency of three types of hypoimmunogenic hPSCs, established through the biallelic lesion of B2M gene to remove all surface expression of classical and nonclassical HLA class I molecules (B2Mnull), biallelic homologous recombination of nonclassical HLA-G1 to the B2M loci to knockout B2M while expressing membrane-bound β2m-HLA-G1 fusion proteins (B2MmHLAG), and ectopic expression of soluble and secreted β2m-HLA-G5 fusion proteins in B2MmHLAG hPSCs (B2Mm/sHLAG) in the most widely used WA09 human embryonic stem cells. Our results showed that hypoimmunogenic hPSCs with variable expression patterns of HLA molecules and immune compromising spectrums retained their normal self-renewal capacity and three-germ-layer differentiation potency. More importantly, as exemplified by neurons, cardiomyocytes and hepatocytes, hypoimmunogenic hPSC-derived tissue cells were fully functional as of their morphology, electrophysiological properties, macromolecule transportation and metabolic regulation. Our findings thus indicate that engineered hypoimmunogenic hPSCs hold great promise of serving as an unlimited universal cell source for cell therapeutics.
Collapse
Affiliation(s)
- Yifan Chen
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
| | - Yanjie Zhou
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
| | - Zhongshu Zhou
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
| | - Yujiang Fang
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
| | - Lin Ma
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoqing Zhang
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.
- Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China.
- Tsingtao Advanced Research Institute, Tongji University, Qingdao, China.
| | - Jie Xiong
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.
- Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.
| | - Ling Liu
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.
- Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
3
|
Glioblastoma Embryonic-like Stem Cells Exhibit Immune-Evasive Phenotype. Cancers (Basel) 2022; 14:cancers14092070. [PMID: 35565200 PMCID: PMC9104850 DOI: 10.3390/cancers14092070] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Most glioblastoma (GBM) patients relapse after an initial response to treatment. These aggressive traits are often associated with the presence of glioma stem cells (GSCs) within the tumor bulk, which are thought to participate in GBM therapy resistance. Given GBM cellular heterogeneity, we hypothesized that GSCs might also display cellular hierarchies associated with different degrees of stemness. Based on single-cell RNAseq data from GBM patients, we identified a subpopulation of GSCs, named core-GSCs (c-GSCs), with a similar profile to embryonic stem cells and downregulation of immune-associated pathways. In addition, we developed an in vitro induced c-GSC (ic-GSC) model resembling their tumor counterpart. The characterization of immune-privileged c-GSCs provides a valuable resource to study immune evasion mechanisms in GBM and to identify potential unexplored targets to improve immunotherapy treatments. Abstract Background: Glioma stem cells (GSCs) have self-renewal and tumor-initiating capacities involved in drug resistance and immune evasion mechanisms in glioblastoma (GBM). Methods: Core-GSCs (c-GSCs) were identified by selecting cells co-expressing high levels of embryonic stem cell (ESC) markers from a single-cell RNA-seq patient-derived GBM dataset (n = 28). Induced c-GSCs (ic-GSCs) were generated by reprogramming GBM-derived cells (GBM-DCs) using induced pluripotent stem cell (iPSC) technology. The characterization of ic-GSCs and GBM-DCs was conducted by immunostaining, transcriptomic, and DNA methylation (DNAm) analysis. Results: We identified a GSC population (4.22% ± 0.59) exhibiting concurrent high expression of ESC markers and downregulation of immune-associated pathways, named c-GSCs. In vitro ic-GSCs presented high expression of ESC markers and downregulation of antigen presentation HLA proteins. Transcriptomic analysis revealed a strong agreement of enriched biological pathways between tumor c-GSCs and in vitro ic-GSCs (κ = 0.71). Integration of our epigenomic profiling with 833 functional ENCODE epigenetic maps identifies increased DNA methylation on HLA genes’ regulatory regions associated with polycomb repressive marks in a stem-like phenotype. Conclusions: This study unravels glioblastoma immune-evasive mechanisms involving a c-GSC population. In addition, it provides a cellular model with paired gene expression, and DNA methylation maps to explore potential therapeutic complements for GBM immunotherapy.
Collapse
|
4
|
Dastagir N, Beal Z, Godwin J. Tissue origin of cytotoxic natural killer cells dictates their differential roles in mouse digit tip regeneration and progenitor cell survival. Stem Cell Reports 2022; 17:633-648. [PMID: 35120621 PMCID: PMC9039750 DOI: 10.1016/j.stemcr.2022.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/08/2023] Open
Abstract
Regeneration of amputated digit tips relies on mesenchymal progenitor cells and their differentiation into replacement bone and tissue stroma. Natural killer (NK) cells have well-characterized roles in antigen-independent killing of virally infected, pre-tumorous, or stressed cells; however, the potential for cytotoxic activity against regenerative progenitor cells is unclear. We identified NK cell recruitment to the regenerating digit tip, and NK cytotoxicity was observed against osteoclast and osteoblast progenitors. Adoptive cell transplants of spleen NK (SpNK) or thymus NK (ThNK) donor cells into immunodeficient mice demonstrated ThNK cell-induced apoptosis with a reduction in osteoclasts, osteoblasts, and proliferative cells, resulting in inhibition of regeneration. Adoptive transfer of NK cells deficient in NK cell activation genes identified that promotion of regeneration by SpNK cells requires Ncr1, whereas inhibition by ThNK cells is mediated via Klrk1 and perforin. Successful future therapies aimed at enhancing regeneration will require a deeper understanding of progenitor cell protection from NK cell cytotoxicity.
Collapse
Affiliation(s)
- Nadjib Dastagir
- The Jackson Laboratory, Bar Harbor, ME 04609, USA,Mount Desert Island Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Salisbury Cove, ME 04609, USA,Medical School of Hanover, 30659 Hannover, Germany
| | - Zachery Beal
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - James Godwin
- The Jackson Laboratory, Bar Harbor, ME 04609, USA,Mount Desert Island Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Salisbury Cove, ME 04609, USA,Corresponding author
| |
Collapse
|
5
|
Chen B, Gurung C, Guo J, Kwon C, Guo YL. Pluripotent stem cells are insensitive to the cytotoxicity of TNFα and IFNγ. Reproduction 2021; 160:547-560. [PMID: 32698161 DOI: 10.1530/rep-20-0215] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/17/2020] [Indexed: 01/12/2023]
Abstract
Recent studies have demonstrated that embryonic stem cells (ESCs) have an underdeveloped innate immune system, but the biological implications of this finding are poorly understood. In this study, we compared the responses of mouse ESCs (mESCs) and mESC differentiated fibroblasts (mESC-FBs) to tumor necrosis factor α (TNFα) and interferons (IFNs). Our data revealed that TNFα, IFNα, IFNβ, or IFNγ alone do not cause apparent effects on mESCs and mESC-FBs, but the combination of TNFα and IFNγ (TNFα/IFNγ) showed toxicity to mESC-FBs as indicated by cell cycle inhibition and reduced cell viability, correlating with the expression of inducible nitric oxide synthase (iNOS). However, none of these effects were observed in mESCs that were treated with TNFα/IFNγ. Furthermore, mESC-FBs, but not mESCs, are vulnerable to cytotoxicity resulting from lipopolysaccharide (LPS)-activated macrophages. The insensitivity of mESCs to cytotoxicity in all cases is correlated with their lack of responses to TNFα and IFNγ. Similar to mESCs, human ESCs (hESCs) and iPSCs (hiPSCs) do not respond to TNFα and are not susceptible to the cytotoxicity of TNFα, IFNβ, or IFNγ alone or in combination that significantly affects human foreskin fibroblast (hFBs) and Hela cells. However, unlike mESCs, hESCs and hiPSCs can respond to IFNγ, but this does not cause significant cytotoxicity in hESCs and hiPSCs. Our findings in both mouse and human PSCs together support the hypothesis that attenuated innate immune responses could be a protective mechanism that limits immunologic cytotoxicity resulting from inflammatory and immune responses.
Collapse
Affiliation(s)
- Bohan Chen
- Department of Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Chandan Gurung
- Department of Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Jason Guo
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yan-Lin Guo
- Department of Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| |
Collapse
|
6
|
Strainiene E, Binkis M, Urnikyte S, Stankevicius V, Sasnauskiene A, Kundrotas G, Kazlauskas A, Suziedelis K. Microenvironment dependent gene expression signatures in reprogrammed human colon normal and cancer cell lines. BMC Cancer 2018; 18:222. [PMID: 29482503 PMCID: PMC5827990 DOI: 10.1186/s12885-018-4145-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 02/19/2018] [Indexed: 12/14/2022] Open
Abstract
Background Since the first evidence suggesting existence of stem-like cancer cells, the process of cells reprogramming to the stem cell state remains as an attractive tool for cancer stemness research. Current knowledge in the field of cancer stemness, indicates that the microenvironment is a fundamental regulator of cell behavior. With regard to this, we investigated the changes of genome wide gene expression in reprogrammed human colon normal epithelial CRL-1831 and colon carcinoma DLD1 cell lines grown under more physiologically relevant three-dimensional (3D) cell culture microenvironment compared to 2D monolayer. Methods Whole genome gene expression changes were evaluated in both cell lines cultured under 3D conditions over a 2D monolayer by gene expression microarray analysis. To evaluate the biological significance of gene expression changes, we performed pathway enrichment analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Gene network analysis was used to study relationships between differentially expressed genes (DEGs) in functional categories by the GeneMANIA Cytoscape toolkit. Results In total, we identified 3228 and 2654 differentially expressed genes (DEGs) for colon normal and cancer reprogrammed cell lines, respectively. Furthermore, the expression of 1097 genes was commonly regulated in both cell lines. KEGG enrichment analysis revealed that in total 129 and 101 pathways for iPSC-CRL-1831 and for CSC-DLD1, respectively, were enriched. Next, we grouped these pathways into three functional categories: cancer transformation/metastasis, cell interaction, and stemness. β-catenin (CTNNB1) was confirmed as a hub gene of all three functional categories. Conclusions Our present findings suggest common pathways between reprogrammed human colon normal epithelium (iPSC-CRL-1831) and adenocarcinoma (CSC-DLD1) cells grown under 3D microenvironment. In addition, we demonstrated that pathways important for cancer transformation and tumor metastatic activity are altered both in normal and cancer stem-like cells during the transfer from 2D to 3D culture conditions. Thus, we indicate the potential of cell culture models enriched in normal and cancer stem-like cells for the identification of new therapeutic targets in cancer treatment. Electronic supplementary material The online version of this article (10.1186/s12885-018-4145-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Egle Strainiene
- National Cancer Institute, Santariskiu 1, 08660, Vilnius, LT, Lithuania. .,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania.
| | - Mindaugas Binkis
- National Cancer Institute, Santariskiu 1, 08660, Vilnius, LT, Lithuania.,Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Silvija Urnikyte
- National Cancer Institute, Santariskiu 1, 08660, Vilnius, LT, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Vaidotas Stankevicius
- National Cancer Institute, Santariskiu 1, 08660, Vilnius, LT, Lithuania.,Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ausra Sasnauskiene
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Andrius Kazlauskas
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, MA, 02114, USA
| | - Kestutis Suziedelis
- National Cancer Institute, Santariskiu 1, 08660, Vilnius, LT, Lithuania. .,Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| |
Collapse
|
7
|
Lin Y, Gil CH, Yoder MC. Differentiation, Evaluation, and Application of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Arterioscler Thromb Vasc Biol 2017; 37:2014-2025. [PMID: 29025705 DOI: 10.1161/atvbaha.117.309962] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
The emergence of induced pluripotent stem cell (iPSC) technology paves the way to generate large numbers of patient-specific endothelial cells (ECs) that can be potentially delivered for regenerative medicine in patients with cardiovascular disease. In the last decade, numerous protocols that differentiate EC from iPSC have been developed by many groups. In this review, we will discuss several common strategies that have been optimized for human iPSC-EC differentiation and subsequent studies that have evaluated the potential of human iPSC-EC as a cell therapy or as a tool in disease modeling. In addition, we will emphasize the importance of using in vivo vessel-forming ability and in vitro clonogenic colony-forming potential as a gold standard with which to evaluate the quality of human iPSC-EC derived from various protocols.
Collapse
Affiliation(s)
- Yang Lin
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Chang-Hyun Gil
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Mervin C Yoder
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis.
| |
Collapse
|
8
|
Zhang D, Qiu B, Wang Y, Guan Y, Zhang L, Wu A. Temozolomide increases MHC-I expression via NF-κB signaling in glioma stem cells. Cell Biol Int 2017; 41:680-690. [PMID: 28403532 DOI: 10.1002/cbin.10773] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/08/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Dongyong Zhang
- Department of Neurosurgery; The First Affiliated Hospital of China Medical University; 155 Nanjingbei Street, Heping District Shenyang 110001 China
| | - Bo Qiu
- Department of Neurosurgery; The First Affiliated Hospital of China Medical University; 155 Nanjingbei Street, Heping District Shenyang 110001 China
| | - Yunjie Wang
- Department of Neurosurgery; The First Affiliated Hospital of China Medical University; 155 Nanjingbei Street, Heping District Shenyang 110001 China
| | - Yanlei Guan
- Department of Neurosurgery; The First Affiliated Hospital of China Medical University; 155 Nanjingbei Street, Heping District Shenyang 110001 China
| | - Luyang Zhang
- Department of Neurosurgery; The First Affiliated Hospital of China Medical University; 155 Nanjingbei Street, Heping District Shenyang 110001 China
| | - Anhua Wu
- Department of Neurosurgery; The First Affiliated Hospital of China Medical University; 155 Nanjingbei Street, Heping District Shenyang 110001 China
| |
Collapse
|
9
|
KEILHOFF GERBURG, LUCAS BENJAMIN, UHDE KATJA, FANSA HISHAM. Selected gene profiles of stressed NSC-34 cells and rat spinal cord following peripheral nerve reconstruction and minocycline treatment. Exp Ther Med 2016; 11:1685-1699. [PMID: 27168790 PMCID: PMC4840837 DOI: 10.3892/etm.2016.3130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/17/2015] [Indexed: 12/20/2022] Open
Abstract
The present study was conducted to investigate the effects of minocycline on the expression of selected transcriptional and translational profiles in the rat spinal cord following sciatic nerve (SNR) transection and microsurgical coaptation. The mRNA and protein expression levels of B cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), caspase-3, major histocompatibility complex I (MHC I), tumor necrosis factor-α (TNF-α), activating transcription factor 3 (ATF3), vascular endothelial growth factor (VEGF), matrix metalloproteinase 9 (MMP9), and growth associated protein-43 (GAP-43) were monitored in the rat lumbar spinal cord following microsurgical reconstruction of the sciatic nerves and minocycline treatment. The present study used semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. As a PCR analysis of spinal cord tissue enabled the examination of the expression patterns of all cell types including glia, the motorneuron-like NSC-34 cell line was used to investigate expression level changes in motorneurons. As stressors, oxygen glucose deprivation (OGD) and lipopolysaccharide (LPS) treatment were performed. SNR did not induce significant degeneration of ventral horn motorneurons, whereas microglia activation and synaptic terminal retraction were detectable. All genes were constitutively expressed at the mRNA and protein levels in untreated spinal cord and control cells. SNR significantly increased the mRNA expression levels of all genes, albeit only temporarily. In all genes except MMP9 and GAP-43, the induction was seen ipsilaterally and contralaterally. The effects of minocycline were moderate. The expression levels of MMP9, TNF-α, MHC I, VEGF, and GAP-43 were reduced, whereas those of Bax and Bcl-2 were unaffected. OGD, but not LPS, was toxic for NSC-34 cells. No changes in the expression levels of Bax, caspase-3, MHC I or ATF3 were observed. These results indicated that motorneurons were not preferentially or solely responsible for SNR-mediated upregulation of these genes. MMP9, TNF-α, VEGF and Bcl-2 were stress-activated. These results suggest that a substantial participation of motorneurons in gene expression levels in vivo. Minocycline was also shown to have inhibitory effects. The nuclear factor-κB signalling pathway may be a possible target of minocycline.
Collapse
Affiliation(s)
- GERBURG KEILHOFF
- Institute of Biochemistry and Cell Biology, Otto-Von-Guericke University Magdeburg, Magdeburg D-39120, Germany
| | - BENJAMIN LUCAS
- Institute of Biochemistry and Cell Biology, Otto-Von-Guericke University Magdeburg, Magdeburg D-39120, Germany
- Department of Trauma Surgery, Otto-Von-Guericke University Magdeburg, Magdeburg D-39120, Germany
| | - KATJA UHDE
- Institute of Biochemistry and Cell Biology, Otto-Von-Guericke University Magdeburg, Magdeburg D-39120, Germany
| | - HISHAM FANSA
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld, Bielefeld D-33604, Germany
| |
Collapse
|
10
|
Kruse V, Hamann C, Monecke S, Cyganek L, Elsner L, Hübscher D, Walter L, Streckfuss-Bömeke K, Guan K, Dressel R. Human Induced Pluripotent Stem Cells Are Targets for Allogeneic and Autologous Natural Killer (NK) Cells and Killing Is Partly Mediated by the Activating NK Receptor DNAM-1. PLoS One 2015; 10:e0125544. [PMID: 25950680 PMCID: PMC4423859 DOI: 10.1371/journal.pone.0125544] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/25/2015] [Indexed: 02/07/2023] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) could be used to generate autologous cells for therapeutic purposes, which are expected to be tolerated by the recipient. However, iPSC-derived grafts are at risk of giving rise to teratomas in the host, if residuals of tumorigenic cells are not rejected by the recipient. We have analyzed the susceptibility of hiPSC lines to allogeneic and autologous natural killer (NK) cells. IL-2-activated, in contrast to resting NK cells killed hiPSC lines efficiently (P = 1.69 x 10(-39)). Notably, the specific lysis of the individual hiPSC lines by IL-2-activated NK cells was significantly different (P = 1.72 x 10(-6)) and ranged between 46 % and 64 % in 51Cr-release assays when compared to K562 cells. The hiPSC lines were killed by both allogeneic and autologous NK cells although autologous NK cells were less efficient (P=8.63 x 10(-6)). Killing was partly dependent on the activating NK receptor DNAM-1 (P = 8.22 x 10(-7)). The DNAM-1 ligands CD112 and CD155 as well as the NKG2D ligands MICA and MICB were expressed on the hiPSC lines. Low amounts of human leukocyte antigen (HLA) class I proteins, which serve as ligands for inhibitory and activating NK receptors were also detected. Thus, the susceptibility to NK cell killing appears to constitute a common feature of hiPSCs. Therefore, NK cells might reduce the risk of teratoma formation even after autologous transplantations of pluripotent stem cell-derived grafts that contain traces of pluripotent cells.
Collapse
Affiliation(s)
- Vanessa Kruse
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Carina Hamann
- Department of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Sebastian Monecke
- Department of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Lukas Cyganek
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Leslie Elsner
- Department of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Daniela Hübscher
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Lutz Walter
- Primate Genetics Laboratory, German Primate Center, Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Kaomei Guan
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
- * E-mail: (RD); (KG)
| | - Ralf Dressel
- Department of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
- * E-mail: (RD); (KG)
| |
Collapse
|
11
|
A practical guide to induced pluripotent stem cell research using patient samples. J Transl Med 2015; 95:4-13. [PMID: 25089770 DOI: 10.1038/labinvest.2014.104] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/24/2014] [Accepted: 06/27/2014] [Indexed: 12/13/2022] Open
Abstract
Approximately 3 years ago, we assessed how patient induced pluripotent stem cell (iPSC) research could potentially impact human pathobiology studies in the future. Since then, the field has grown considerably with numerous technical developments, and the idea of modeling diseases 'in a dish' is becoming increasingly popular in biomedical research. Likely, it is even acceptable to include patient iPSCs as one of the standard research tools for disease mechanism studies, just like knockout mice. However, as the field matures, we acknowledge there remain many practical limitations and obstacles for their genuine application to understand diseases, and accept that it has not been as straightforward to model disorders as initially proposed. A major practical challenge has been efficient direction of iPSC differentiation into desired lineages and preparation of the large numbers of specific cell types required for study. Another even larger obstacle is the limited value of in vitro outcomes, which often do not closely represent disease conditions. To overcome the latter issue, many new approaches are underway, including three-dimensional organoid cultures from iPSCs, xenotransplantation of human cells to animal models and in vitro interaction of multiple cell types derived from isogenic iPSCs. Here we summarize the areas where patient iPSC studies have provided truly valuable information beyond existing skepticism, discuss the desired technologies to overcome current limitations and include practical guidance for how to utilize the resources. Undoubtedly, these human patient cells are an asset for experimental pathology studies. The future rests on how wisely we use them.
Collapse
|
12
|
Lu Q, Yu M, Shen C, Chen X, Feng T, Yao Y, Li J, Li H, Tu W. Negligible immunogenicity of induced pluripotent stem cells derived from human skin fibroblasts. PLoS One 2014; 9:e114949. [PMID: 25503995 PMCID: PMC4263724 DOI: 10.1371/journal.pone.0114949] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/16/2014] [Indexed: 12/29/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have potential applications in cell replacement therapy and regenerative medicine. However, limited information is available regarding the immunologic features of iPSCs. In this study, expression of MHC and T cell co-stimulatory molecules in hiPSCs, and the effects on activation, proliferation and cytokine production in allogeneic human peripheral blood mononuclear cells were examined. We found that no-integrate hiPSCs had no MHC-II and T cell co-stimulatory molecules expressions but had moderate level of MHC-I and HLA-G expressions. In contrast to human skin fibroblasts (HSFs) which significantly induced allogeneic T cell activation and proliferation, hiPSCs failed to induce allogeneic CD45+ lymphocyte and CD8+ T cell activation and proliferation but could induce a low level of allogeneic CD4+ T cell proliferation. Unlike HSFs which induced allogeneic lymphocytes to produce high levels of IFN-γ, TNF-α and IL-17, hiPSCs only induced allogeneic lymphocytes to produce IL-2 and IL-10, and promote IL-10-secreting regulatory T cell (Treg) generation. Our study suggests that the integration-free hiPSCs had low or negligible immunogenicity, which may result from their induction of IL-10-secreting Treg.
Collapse
Affiliation(s)
- Qiao Lu
- The Joint Research Center of West China Second University Hospital of Sichuan University and Faculty of Medicine of the University of Hong Kong, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Pediatrics, University Hospital of Hubei University for Nationalities, Enshi, Hubei, 445000, China
| | - Meixing Yu
- The Joint Research Center of West China Second University Hospital of Sichuan University and Faculty of Medicine of the University of Hong Kong, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chongyang Shen
- The Joint Research Center of West China Second University Hospital of Sichuan University and Faculty of Medicine of the University of Hong Kong, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoping Chen
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Ting Feng
- The Joint Research Center of West China Second University Hospital of Sichuan University and Faculty of Medicine of the University of Hong Kong, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yongchao Yao
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jinrong Li
- The Joint Research Center of West China Second University Hospital of Sichuan University and Faculty of Medicine of the University of Hong Kong, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hong Li
- The Joint Research Center of West China Second University Hospital of Sichuan University and Faculty of Medicine of the University of Hong Kong, Sichuan University, Chengdu, Sichuan, 610041, China
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- * E-mail: (HL); (WT)
| | - Wenwei Tu
- The Joint Research Center of West China Second University Hospital of Sichuan University and Faculty of Medicine of the University of Hong Kong, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Pediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- * E-mail: (HL); (WT)
| |
Collapse
|
13
|
Low immunogenicity of neural progenitor cells differentiated from induced pluripotent stem cells derived from less immunogenic somatic cells. PLoS One 2013; 8:e69617. [PMID: 23922758 PMCID: PMC3724937 DOI: 10.1371/journal.pone.0069617] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 06/12/2013] [Indexed: 01/29/2023] Open
Abstract
The groundbreaking discovery of induced pluripotent stem cells (iPS cells) provides a new source for cell therapy. However, whether the iPS derived functional lineages from different cell origins have different immunogenicity remains unknown. It had been known that the cells isolated from extra-embryonic tissues, such as umbilical cord mesenchymal cells (UMCs), are less immunogenic than other adult lineages such as skin fibroblasts (SFs). In this report, we differentiated iPS cells from human UMCs and SFs into neural progenitor cells (NPCs) and analyzed their immunogenicity. Through co-culture with allologous peripheral blood mononuclear cells (PBMCs), we showed that UMCs were indeed less immunogenic than skin cells to simulate proliferation of PBMCs. Surprisingly, we found that the NPCs differentiated from UMC-iPS cells retained low immunogenicity as the parental UMCs based on the PBMC proliferation assay. In cytotoxic expression assay, reactions in most kinds of immune effector cells showed more perforin and granzyme B expression with SF-NPCs stimulation than that with UMC-NPCs stimulation in PBMC co-culture system, in T cell co-culture system as well. Furthermore, through whole genome expression microarray analysis, we showed that over 70 immune genes, including all members of HLA-I, were expressed at lower levels in NPCs derived from UMC-iPS cells than that from SF-iPS cells. Our results demonstrated a phenomenon that the low immunogenicity of the less immunogenic cells could be retained after cell reprogramming and further differentiation, thus provide a new concept to generate functional lineages with lower immunogenicity for regenerative medicine.
Collapse
|
14
|
Study on therapeutic action of bone marrow derived mesenchymal stem cell combined with vitamin E against acute kidney injury in rats. Life Sci 2013; 92:829-37. [PMID: 23499556 DOI: 10.1016/j.lfs.2013.02.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 02/02/2013] [Accepted: 02/23/2013] [Indexed: 02/07/2023]
Abstract
AIMS The study aims to investigate the effect to treat acute kidney injury (AKI) with bone marrow derived mesenchymal stem cells (BMSCs) combined with vitamin E and to develop a new treatment mode for AKI preclinical study. MAIN METHODS BMSCs were separated from rat bone marrow. Gentamicin was used as a damage factor in the culture of renal tubular epithelial cells (RTECs) in vitro. After co-cultured with BMSCs and vitamin E, cell proliferation of each group was detected with CCK-8. In vivo, BMSCs (3.3×10(6)cells/kg) combined with vitamin E (80mg/kg) were administered in AKI rats induced by gentamicin intravenously. The pathological changes, biochemical parameters and apoptosis genes after treatment were investigated furthermore. KEY FINDINGS In co-cultured system, proliferating ability of RTECs was improved by BMSCs or vitamin E, especially for the combined group (P<0.05). The treated rats in combined group presented the lowest serum creatinine and the highest urea nitrogen compared to non-treated rats. The improvement in renal pathological changes was followed by less necrosis, degeneration and expansion of renal tubule. Under transmission electron microscope, unclear cell structure and reduction of endoplasmic reticulum in the cytoplasm of RTECs were ameliorated with the treatment. Most apoptosis genes were up-regulated in model group while down-regulated with the therapy. Further analysis showed that the two treatments may act independently with each other. SIGNIFICANCE Our data demonstrated that both BMSC and vitamin E hold therapeutic action to AKI induced by gentamicin. Especially, the combined treatment is better than BMSC or vitamin E alone.
Collapse
|