1
|
Sun Z, Cai Y, Chen Y, Jin Q, Zhang Z, Zhang L, Li Y, Huang L, Wang J, Yang Y, Lv Q, Han Z, Xie M, Zhu X. Ultrasound-targeted microbubble destruction promotes PDGF-primed bone mesenchymal stem cell transplantation for myocardial protection in acute Myocardial Infarction in rats. J Nanobiotechnology 2023; 21:481. [PMID: 38102643 PMCID: PMC10725038 DOI: 10.1186/s12951-023-02204-7] [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: 06/21/2023] [Accepted: 11/07/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Ultrasound-targeted microbubble destruction (UTMD) has emerged as a promising strategy for the targeted delivery of bone marrow mesenchymal stem cells (MSCs) to the ischemic myocardium. However, the limited migration capacity and poor survival of MSCs remains a major therapeutic barrier. The present study was performed to investigate the synergistic effect of UTMD with platelet-derived growth factor BB (PDGF-BB) on the homing of MSCs for acute myocardial infarction (AMI). METHODS MSCs from male donor rats were treated with PDGF-BB, and a novel microbubble formulation was prepared using a thin-film hydration method. In vivo, MSCs with or without PDGF-BB pretreatment were transplanted by UTMD after inducing AMI in experimental rats. The therapeutic efficacy of PDGF-BB-primed MSCs on myocardial apoptosis, angiogenesis, cardiac function and scar repair was estimated. The effects and molecular mechanisms of PDGF-BB on MSC migration and survival were explored in vitro. RESULTS The results showed that the biological effects of UTMD increased the local levels of stromal-derived factor-1 (SDF-1), which promoted the migration of transplanted MSCs to the ischemic region. Compared with UTMD alone, UTMD combined with PDGF-BB pretreatment significantly increased the cardiac homing of MSCs, which subsequently reduced myocardial apoptosis, promoted neovascularization and tissue repair, and increased cardiac function 30 days after MI. The vitro results demonstrated that PDGF-BB enhanced MSC migration and protected these cells from H2O2-induced apoptosis. Mechanistically, PDGF-BB pretreatment promoted MSC migration and inhibited H2O2-induced MSC apoptosis via activation of the phosphatidylinositol 3-kinase/serine-threonine kinase (PI3K/Akt) pathway. Furthermore, crosstalk between PDGF-BB and stromal-derived factor-1/chemokine receptor 4 (SDF-1/CXCR4) is involved in the PI3K/AKT signaling pathway. CONCLUSION The present study demonstrated that UTMD combined with PDGF-BB treatment could enhance the homing ability of MSCs, thus alleviating AMI in rats. Therefore, UTMD combined with PDGF-BB pretreatment may offer exciting therapeutic opportunities for strengthening MSC therapy in ischemic diseases.
Collapse
Grants
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 2018CFB568 National Natural Science Foundation of Hubei
Collapse
Affiliation(s)
- Zhenxing Sun
- Anhui Medical University, Hefei, 230031, China
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yu Cai
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Ziming Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yuman Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Lei Huang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Zhengyang Han
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China.
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China.
| | - Xiangming Zhu
- Anhui Medical University, Hefei, 230031, China.
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| |
Collapse
|
2
|
Branella GM, Lee JY, Okalova J, Parwani KK, Alexander JS, Arthuzo RF, Fedanov A, Yu B, McCarty D, Brown HC, Chandrakasan S, Petrich BG, Doering CB, Spencer HT. Ligand-based targeting of c-kit using engineered γδ T cells as a strategy for treating acute myeloid leukemia. Front Immunol 2023; 14:1294555. [PMID: 38022523 PMCID: PMC10679681 DOI: 10.3389/fimmu.2023.1294555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
The application of immunotherapies such as chimeric antigen receptor (CAR) T therapy or bi-specific T cell engager (BiTE) therapy to manage myeloid malignancies has proven more challenging than for B-cell malignancies. This is attributed to a shortage of leukemia-specific cell-surface antigens that distinguish healthy from malignant myeloid populations, and the inability to manage myeloid depletion unlike B-cell aplasia. Therefore, the development of targeted therapeutics for myeloid malignancies, such as acute myeloid leukemia (AML), requires new approaches. Herein, we developed a ligand-based CAR and secreted bi-specific T cell engager (sBite) to target c-kit using its cognate ligand, stem cell factor (SCF). c-kit is highly expressed on AML blasts and correlates with resistance to chemotherapy and poor prognosis, making it an ideal candidate for which to develop targeted therapeutics. We utilize γδ T cells as a cytotoxic alternative to αβ T cells and a transient transfection system as both a safety precaution and switch to remove alloreactive modified cells that may hinder successful transplant. Additionally, the use of γδ T cells permits its use as an allogeneic, off-the-shelf therapeutic. To this end, we show mSCF CAR- and hSCF sBite-modified γδ T cells are proficient in killing c-kit+ AML cell lines and sca-1+ murine bone marrow cells in vitro. In vivo, hSCF sBite-modified γδ T cells moderately extend survival of NSG mice engrafted with disseminated AML, but therapeutic efficacy is limited by lack of γδ T-cell homing to murine bone marrow. Together, these data demonstrate preclinical efficacy and support further investigation of SCF-based γδ T-cell therapeutics for the treatment of myeloid malignancies.
Collapse
Affiliation(s)
- Gianna M. Branella
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Jasmine Y. Lee
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Jennifer Okalova
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Molecular Systems Pharmacology Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, United States
| | - Kiran K. Parwani
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Jordan S. Alexander
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Raquel F. Arthuzo
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Andrew Fedanov
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Bing Yu
- Expression Therapeutics, Inc., Tucker, GA, United States
| | - David McCarty
- Expression Therapeutics, Inc., Tucker, GA, United States
| | | | - Shanmuganathan Chandrakasan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | | | - Christopher B. Doering
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Molecular Systems Pharmacology Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, United States
| | - H. Trent Spencer
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Molecular Systems Pharmacology Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, United States
| |
Collapse
|
3
|
Varady ES, Ayala LA, Nguyen PU, Scarfone VM, Karimzadeh A, Zhou C, Chen X, Greilach SA, Walsh CM, Inlay MA. Graft conditioning with fluticasone propionate reduces graft-versus-host disease upon allogeneic hematopoietic cell transplantation in mice. EMBO Mol Med 2023; 15:e17748. [PMID: 37538042 PMCID: PMC10493574 DOI: 10.15252/emmm.202317748] [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: 03/23/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 08/05/2023] Open
Abstract
Hematopoietic cell transplantation (HCT) treats many blood conditions but remains underused due to complications such as graft-versus-host disease (GvHD). In GvHD, donor immune cells attack the patient, requiring powerful immunosuppressive drugs like glucocorticoids (GCs) to prevent death. In this study, we tested the hypothesis that donor cell conditioning with the glucocorticoid fluticasone propionate (FLU) prior to transplantation could increase hematopoietic stem cell (HSC) engraftment and reduce GvHD. Murine HSCs treated with FLU had increased HSC engraftment and reduced severity and incidence of GvHD after transplantation into allogeneic hosts. While most T cells died upon FLU treatment, donor T cells repopulated in the hosts and appeared less inflammatory and alloreactive. Regulatory T cells (Tregs) are immunomodulatory and survived FLU treatment, resulting in an increased ratio of Tregs to conventional T cells. Our results implicate an important role for Tregs in maintaining allogeneic tolerance in FLU-treated grafts and suggest a therapeutic strategy of pre-treating donor cells (and not the patients directly) with GCs to simultaneously enhance engraftment and reduce GvHD upon allogeneic HCT.
Collapse
Affiliation(s)
- Erika S Varady
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
| | - L Angel Ayala
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
| | - Pauline U Nguyen
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
| | - Vanessa M Scarfone
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
| | - Alborz Karimzadeh
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
- Present address:
Joslin Diabetes CenterHarvard Medical SchoolBostonMAUSA
| | - Cuiwen Zhou
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
| | - Xiyu Chen
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
| | - Scott A Greilach
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
| | - Craig M Walsh
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
| | - Matthew A Inlay
- Sue and Bill Gross Stem Cell Research CenterUniversity of California IrvineIrvineCAUSA
- Department of Molecular Biology and BiochemistryUniversity of California IrvineIrvineCAUSA
| |
Collapse
|
4
|
Wieder R. Awakening of Dormant Breast Cancer Cells in the Bone Marrow. Cancers (Basel) 2023; 15:cancers15113021. [PMID: 37296983 DOI: 10.3390/cancers15113021] [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: 02/26/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Up to 40% of patients with breast cancer (BC) have metastatic cells in the bone marrow (BM) at the initial diagnosis of localized disease. Despite definitive systemic adjuvant therapy, these cells survive in the BM microenvironment, enter a dormant state and recur stochastically for more than 20 years. Once they begin to proliferate, recurrent macrometastases are not curable, and patients generally succumb to their disease. Many potential mechanisms for initiating recurrence have been proposed, but no definitive predictive data have been generated. This manuscript reviews the proposed mechanisms that maintain BC cell dormancy in the BM microenvironment and discusses the data supporting specific mechanisms for recurrence. It addresses the well-described mechanisms of secretory senescence, inflammation, aging, adipogenic BM conversion, autophagy, systemic effects of trauma and surgery, sympathetic signaling, transient angiogenic bursts, hypercoagulable states, osteoclast activation, and epigenetic modifications of dormant cells. This review addresses proposed approaches for either eliminating micrometastases or maintaining a dormant state.
Collapse
Affiliation(s)
- Robert Wieder
- Rutgers New Jersey Medical School and the Cancer Institute of New Jersey, 185 South Orange Avenue, MSB F671, Newark, NJ 07103, USA
| |
Collapse
|
5
|
Huang X, Guo H, Wang L, Zhang Z, Zhang W. Biomimetic cell membrane-coated nanocarriers for targeted siRNA delivery in cancer therapy. Drug Discov Today 2023; 28:103514. [PMID: 36736580 DOI: 10.1016/j.drudis.2023.103514] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Small interfering RNA (siRNA) therapeutics for cancer are a focus of increasing research interest. However, the major obstacle to their clinical application is the targeted delivery of siRNA to cancer cells at desirable levels. Cell membrane-coated nanocarriers have the advantage of combining the properties of both cell membranes and nanoparticles (NPs). In this review, we highlight the most common RNAi therapeutics and the extracellular and intracellular barriers to siRNA delivery. Moreover, we discuss clinical applications of different cell membrane-coated nanocarriers for targeted siRNA delivery, including cancer cell membranes (CCMs), platelet membranes, erythrocyte membranes, stem cell membranes, exosome membranes, and hybrid membranes. Taken together, biomimetic cell membrane-coated nanotechnology is a promising strategy for targeted siRNA delivery for cancer treatment.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Haoyu Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lutong Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhicai Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
6
|
Zhao J, Mu X, Hou X, Zhang X, Li P, Jiang J. Synergistic treatment of osteosarcoma with biomimetic nanoparticles transporting doxorubicin and siRNA. Front Oncol 2023; 13:1111855. [PMID: 36756155 PMCID: PMC9900173 DOI: 10.3389/fonc.2023.1111855] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/10/2023] [Indexed: 01/24/2023] Open
Abstract
Introduction Osteosarcoma tumors are the most common malignant bone tumors in children and adolescents. Their treatment usually requires surgical removal of all detectable cancerous tissue and multidrug chemotherapy; however, the prognosis for patients with unresectable or recurrent osteosarcoma is unfavorable. To make chemotherapy safer and more effective for osteosarcoma patients, biomimetic nanoparticles (NPs) camouflaged by mesenchymal stem cell membranes (MSCMs) were synthesized to induce osteosarcoma cell apoptosis by co-delivering the anticancer drug doxorubicin hydrochloride(DOX) and a small interfering RNA (siRNA). Importantly, these NPs have high biocompatibility and tumor-homing ability. This study aimed to improve the efficacy of osteosarcoma therapy by using the synergistic combination of DOX and an siRNA targeting the apoptosis suppressor gene survivin. Methods Biomimetic NPs (DOX/siSUR-PLGA@MSCM NPs) were synthesized by coloading DOX and survivin siRNA (siSUR) into poly (lactide-co-glycolide acid) (PLGA) via a double-emulsion solvent evaporation method. The NPs were camouflaged by MSCMs to deliver both DOX and survivin-targeting siRNA and characterized and evaluated in terms of cellular uptake, in vitro release, in vitro and in vivo antitumor effects, and biosafety. Results DOX/siSUR-PLGA@MSCM NPs had good tumor-homing ability due to the MSCMs modification. The drug-laden biomimetic NPs had good antitumor effects in homozygous MG63 tumor-bearing mice due to the synergistic effect of the drug combination. Conclusion DOX/siSUR-PLGA@MSCM NPs can show improved therapeutic effects in osteosarcoma patients due to the combination of a chemotherapeutic drug and gene therapy based on their good tumor targeting and biosafety.
Collapse
Affiliation(s)
- Jingtong Zhao
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xuejia Hou
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaowen Zhang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Ping Li
- Rheumatology and Immunology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China,*Correspondence: Jinlan Jiang, ; Ping Li,
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China,*Correspondence: Jinlan Jiang, ; Ping Li,
| |
Collapse
|
7
|
Zhang W, Huang X. Stem cell membrane-camouflaged targeted delivery system in tumor. Mater Today Bio 2022; 16:100377. [PMID: 35967738 PMCID: PMC9364095 DOI: 10.1016/j.mtbio.2022.100377] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023] Open
|
8
|
Cell-based drug delivery systems and their in vivo fate. Adv Drug Deliv Rev 2022; 187:114394. [PMID: 35718252 DOI: 10.1016/j.addr.2022.114394] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022]
Abstract
Cell-based drug delivery systems (DDSs) have received attention recently because of their unique biological properties and self-powered functions, such as excellent biocompatibility, low immunogenicity, long circulation time, tissue-homingcharacteristics, and ability to cross biological barriers. A variety of cells, including erythrocytes, stem cells, and lymphocytes, have been explored as functional vectors for the loading and delivery of various therapeutic payloads (e.g., small-molecule and nucleic acid drugs) for subsequent disease treatment. These cell-based DDSs have their own unique in vivo fates, which are attributed to various factors, including their biological properties and functions, the loaded drugs and loading process, physiological and pathological circumstances, and the body's response to these carrier cells, which result in differences in drug delivery efficiency and therapeutic effect. In this review, we summarize the main cell-based DDSs and their biological properties and functions, applications in drug delivery and disease treatment, and in vivo fate and influencing factors. We envision that the unique biological properties, combined with continuing research, will enable development of cell-based DDSs as friendly drug vectors for the safe, effective, and even personalized treatment of diseases.
Collapse
|
9
|
Felker S, Shrestha A, Bailey J, Pillis DM, Siniard D, Malik P. Differential CXCR4 expression on hematopoietic progenitor cells versus stem cells directs homing and engraftment. JCI Insight 2022; 7:151847. [PMID: 35531956 PMCID: PMC9090236 DOI: 10.1172/jci.insight.151847] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Gene therapy involves a substantial loss of hematopoietic stem and progenitor cells (HSPC) during processing and homing. Intra-BM (i.b.m.) transplantation can reduce homing losses, but prior studies have not yielded promising results. We studied the mechanisms involved in homing and engraftment of i.b.m. transplanted and i.v. transplanted genetically modified (GM) human HSPC. We found that i.b.m. HSPC transplantation improved engraftment of hematopoietic progenitor cells (HPC) but not of long-term repopulating hematopoietic stem cells (HSC). Mechanistically, HPC expressed higher functional levels of CXCR4 than HSC, conferring them a retention and homing advantage when transplanted i.b.m. Removing HPC and transplanting an HSC-enriched population i.b.m. significantly increased long-term engraftment over i.v. transplantation. Transient upregulation of CXCR4 on GM HSC-enriched cells, using a noncytotoxic portion of viral protein R (VPR) fused to CXCR4 delivered as a protein in lentiviral particles, resulted in higher homing and long-term engraftment of GM HSC transplanted either i.v. or i.b.m. compared with standard i.v. transplants. Overall, we show a mechanism for why i.b.m. transplants do not significantly improve long-term engraftment over i.v. transplants. I.b.m. transplantation becomes relevant when an HSC-enriched population is delivered. Alternatively, CXCR4 expression on HSC, when transiently increased using a protein delivery method, improves homing and engraftment specifically of GM HSC.
Collapse
Affiliation(s)
- Sydney Felker
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center (CCHMC) and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Experimental Hematology and Cancer Biology and
| | | | - Jeff Bailey
- Division of Experimental Hematology and Cancer Biology and
| | - Devin M Pillis
- Division of Experimental Hematology and Cancer Biology and
| | - Dylan Siniard
- Division of Experimental Hematology and Cancer Biology and
| | - Punam Malik
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center (CCHMC) and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Experimental Hematology and Cancer Biology and
- Division of Hematology, CCHMC, Cincinnati, Ohio, USA
| |
Collapse
|
10
|
Abstract
Metastasis is responsible for a large majority of death from malignant solid tumors. Bone is one of the most frequently affected organs in cancer metastasis, especially in breast and prostate cancer. Development of bone metastasis requires cancer cells to successfully complete a number of challenging steps, including local invasion and intravasation, survival in circulation, extravasation and initial seeding, and finally, formation of metastatic colonies after a period of dormancy or indolent growth. During this process, cancer cells often undergo a series of cellular and molecular changes to gain cellular plasticity that helps them adapt to various environments they encounter along the journey of metastasis. Understanding the mechanisms behind cellular plasticity and adaptation during the formation of bone metastasis is crucial for the development of novel therapies.
Collapse
Affiliation(s)
- Cao Fang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
11
|
Moghadasi MH, Hajifathali A, Azad M, Rahmani M, Soleimani M. Expansion of cord blood stem cells in fibronectin-coated microfluidic bioreactor. Hematol Transfus Cell Ther 2021; 44:504-511. [PMID: 34593367 PMCID: PMC9605910 DOI: 10.1016/j.htct.2021.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/05/2021] [Accepted: 06/16/2021] [Indexed: 11/26/2022] Open
Abstract
Background Hematopoietic stem/progenitor cell transplantation is the main treatment option for hematological malignancies and disorders. One strategy to solve the problem of low stem cell doses used in transplantation is pre-transplant expansion. We hypothesized that using fibronectin-coated microfluidic channels would expand HSPCs and keep self-renewal potential in a three-dimensional environment, compared to the conventional method. We also compared stem cell homing factors expression in microfluidic to conventional cultures. Materials and methods A microfluidic device was created and characterized by scanning electron microscopy. The CD133+ cells were collected from cord blood and purified. They were subsequently cultured in 24-well plates and microfluidic bioreactor systems using the StemSpan serum-free medium. Eventually, we analyzed cell surface expression levels of the CXCR4 molecule and CXCR4 mRNA expression in CD133+ cells cultured in different systems. Results The expansion results showed significant improvement in CD133+ cell expansion in the microfluidic system than the conventional method. The median expression of the CXCR4 in the expanded cell was lower in the conventional system than in the microfluidic system. The CXCR4 gene expression up-regulated in the microfluidic system. Conclusion Utilizing microfluidic systems to expand desired cells effectively is the next step in cell culture. Comparative gene expression profiling provides a glimpse of the effects of culture microenvironments on the genetic program of HSCs grown in different systems.
Collapse
Affiliation(s)
| | - Abbas Hajifathali
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Azad
- Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Masoud Soleimani
- School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
12
|
Fröbel J, Landspersky T, Percin G, Schreck C, Rahmig S, Ori A, Nowak D, Essers M, Waskow C, Oostendorp RAJ. The Hematopoietic Bone Marrow Niche Ecosystem. Front Cell Dev Biol 2021; 9:705410. [PMID: 34368155 PMCID: PMC8339972 DOI: 10.3389/fcell.2021.705410] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
The bone marrow (BM) microenvironment, also called the BM niche, is essential for the maintenance of fully functional blood cell formation (hematopoiesis) throughout life. Under physiologic conditions the niche protects hematopoietic stem cells (HSCs) from sustained or overstimulation. Acute or chronic stress deregulates hematopoiesis and some of these alterations occur indirectly via the niche. Effects on niche cells include skewing of its cellular composition, specific localization and molecular signals that differentially regulate the function of HSCs and their progeny. Importantly, while acute insults display only transient effects, repeated or chronic insults lead to sustained alterations of the niche, resulting in HSC deregulation. We here describe how changes in BM niche composition (ecosystem) and structure (remodeling) modulate activation of HSCs in situ. Current knowledge has revealed that upon chronic stimulation, BM remodeling is more extensive and otherwise quiescent HSCs may be lost due to diminished cellular maintenance processes, such as autophagy, ER stress response, and DNA repair. Features of aging in the BM ecology may be the consequence of intermittent stress responses, ultimately resulting in the degeneration of the supportive stem cell microenvironment. Both chronic stress and aging impair the functionality of HSCs and increase the overall susceptibility to development of diseases, including malignant transformation. To understand functional degeneration, an important prerequisite is to define distinguishing features of unperturbed niche homeostasis in different settings. A unique setting in this respect is xenotransplantation, in which human cells depend on niche factors produced by other species, some of which we will review. These insights should help to assess deviations from the steady state to actively protect and improve recovery of the niche ecosystem in situ to optimally sustain healthy hematopoiesis in experimental and clinical settings.
Collapse
Affiliation(s)
- Julia Fröbel
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Theresa Landspersky
- School of Medicine, Department of Internal Medicine III, Technical University of Munich, Munich, Germany
| | - Gülce Percin
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Christina Schreck
- School of Medicine, Department of Internal Medicine III, Technical University of Munich, Munich, Germany
| | - Susann Rahmig
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Alessandro Ori
- Proteomics of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marieke Essers
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division Inflammatory Stress in Stem Cells, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudia Waskow
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany.,Department of Medicine III, Technical University Dresden, Dresden, Germany
| | - Robert A J Oostendorp
- School of Medicine, Department of Internal Medicine III, Technical University of Munich, Munich, Germany
| |
Collapse
|
13
|
Chappaz S, McArthur K, Kealy L, Law CW, Tailler M, Lane RM, Lieschke A, Ritchie ME, Good-Jacobson KL, Strasser A, Kile BT. Homeostatic apoptosis prevents competition-induced atrophy in follicular B cells. Cell Rep 2021; 36:109430. [PMID: 34289356 DOI: 10.1016/j.celrep.2021.109430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/13/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
While the intrinsic apoptosis pathway is thought to play a central role in shaping the B cell lineage, its precise role in mature B cell homeostasis remains elusive. Using mice in which mature B cells are unable to undergo apoptotic cell death, we show that apoptosis constrains follicular B (FoB) cell lifespan but plays no role in marginal zone B (MZB) cell homeostasis. In these mice, FoB cells accumulate abnormally. This intensifies intercellular competition for BAFF, resulting in a contraction of the MZB cell compartment, and reducing the growth, trafficking, and fitness of FoB cells. Diminished BAFF signaling dampens the non-canonical NF-κB pathway, undermining FoB cell growth despite the concurrent triggering of a protective p53 response. Thus, MZB and FoB cells exhibit a differential requirement for the intrinsic apoptosis pathway. Homeostatic apoptosis constrains the size of the FoB cell compartment, thereby preventing competition-induced FoB cell atrophy.
Collapse
Affiliation(s)
- Stéphane Chappaz
- Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia; ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia.
| | - Kate McArthur
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Liam Kealy
- Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia; Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia
| | - Charity W Law
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Maximilien Tailler
- Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia
| | - Rachael M Lane
- Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia
| | | | - Matthew E Ritchie
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Kim L Good-Jacobson
- Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia; Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia
| | - Andreas Strasser
- Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia
| | - Benjamin T Kile
- Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, 3800 VIC, Australia; ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia; Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, 5005 SA, Australia.
| |
Collapse
|
14
|
Das MK, Lunavat TR, Miletic H, Hossain JA. The Potentials and Pitfalls of Using Adult Stem Cells in Cancer Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1326:139-157. [PMID: 33615422 DOI: 10.1007/5584_2021_619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Stem cells play a pivotal role in the developmental stages of an organism and in adulthood as well. Therefore, it is not surprising that stem cells constitute a focus of extensive research. Indeed, several decades of stem cell research have tremendously increased our knowledge on the mechanistic understandings of stem cell biology. Interestingly, revealing the fundamental principles of stem cell biology has also fostered its application for therapeutic purposes. Many of the attributes that the stem cells possess, some of which are unique, allow multifaceted exploitation of stem cells in the treatment of various diseases. Cancer, the leading cause of mortality worldwide, is one of the disease groups that has been benefited by the potentials of therapeutic applications of the stem cells. While the modi operandi of how stem cells contribute to cancer treatment are many-sided, two major principles can be conceived. One mode involves harnessing the regenerative power of the stem cells to promote the generation of blood-forming cells in cancer patients after cytotoxic regimens. A totally different kind of utility of stem cells has been exercised in another mode where the stem cells can potentially deliver a plethora of anti-cancer therapeutics in a tumor-specific manner. While both these approaches can improve the treatment of cancer patients, there exist several issues that warrant further research. This review summarizes the basic principles of the utility of the stem cells in cancer treatment along with the current trends and pinpoints the major obstacles to focus on in the future for further improvement.
Collapse
Affiliation(s)
- Mrinal K Das
- Department of Molecular Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Taral R Lunavat
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Jubayer A Hossain
- Department of Biomedicine, University of Bergen, Bergen, Norway. .,Department of Pathology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
15
|
Wang M, Xin Y, Cao H, Li W, Hua Y, Webster TJ, Zhang C, Tang W, Liu Z. Recent advances in mesenchymal stem cell membrane-coated nanoparticles for enhanced drug delivery. Biomater Sci 2020; 9:1088-1103. [PMID: 33332490 DOI: 10.1039/d0bm01164a] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Studies of nanomedicine have achieved dramatic progress in recent decades. However, the main challenges that traditional nanomedicine has to overcome include low accumulation at target sites and rapid clearance from the blood circulation. An interesting approach using cell membrane coating technology has emerged as a possible way to overcome these limitations, owing to the enhanced targeted delivery and reduced immunogenicity of cell membrane moieties. Mesenchymal stem cell (MSC) therapy has been investigated for treating various diseases, ranging from inflammatory diseases to tissue damage. Recent studies with engineered modified MSCs or MSC membranes have focused on enhancing cell therapeutic efficacy. Therefore, bioengineering strategies that couple synthetic nanoparticles with MSC membranes have recently received much attention due to their homing ability and tumor tropism. Given the various membrane receptors on their surfaces, MSC membrane-coated nanoparticles are an effective method with selective targeting properties, allowing entry into specific cells. Here, we review recent progress on the use of MSC membrane-coated nanoparticles for biomedical applications, particularly in the two main antitumor and anti-inflammatory fields. The combination of a bioengineered cell membrane and synthesized nanoparticles presents a wide range of possibilities for the further development of targeted drug delivery, showing the potential to enhance the therapeutic efficacy for treating various diseases.
Collapse
Affiliation(s)
- Mian Wang
- Department of Cardiology, Research Center for Translational Medicine, Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Li X, Wei Z, Zhang W, Lv H, Li J, Wu L, Zhang H, Yang B, Zhu M, Jiang J. Anti-Inflammatory Effects of Magnetically Targeted Mesenchymal Stem Cells on Laser-Induced Skin Injuries in Rats. Int J Nanomedicine 2020; 15:5645-5659. [PMID: 32848391 PMCID: PMC7428346 DOI: 10.2147/ijn.s258017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/13/2020] [Indexed: 12/27/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) are a promising resource for tissue regeneration and repair. However, their clinical application is hindered by technical limitations related to MSC enrichment at the target sites. Methods MSCs were labeled with magnetic Fe3O4 nanoparticles (NPs). We analyzed the effects of NP on cell proliferation, stem cell characteristics, and cytokine secretion. Furthermore, we induced NP-labeled MSC migration with an external magnetic field toward laser-induced skin wounds in rats and evaluated the associated anti-inflammatory effects. Results Fe3O4 NP application did not adversely affect MSC characteristics. Moreover, Fe3O4 NP-labeled MSCs presented increased anti-inflammatory cytokine and chemokine production compared with unlabeled MSCs. Furthermore, MSCs accumulated at the injury site and magnetic targeting promoted NP-labeled MSC migration toward burn injury sites in vivo. On day 7 following MSC injection, reduced inflammation and promoted angiogenesis were observed in the magnetically targeted MSC group. In addition, anti-inflammatory factors were upregulated, whereas pro-inflammatory factors were downregulated within the magnetically targeted MSC group compared with those in the PBS group. Conclusion This study demonstrates that magnetically targeted MSCs contribute to cell migration to the site of skin injury, improve anti-inflammatory effects and enhance angiogenesis compared with MSC injection alone. Therefore, magnetically targeted MSC therapy may be an effective treatment approach for epithelial tissue injuries.
Collapse
Affiliation(s)
- Xiuying Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Zhenhong Wei
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Wei Zhang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Huiying Lv
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Jing Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Liya Wu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, People's Republic of China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, People's Republic of China
| | - Mingji Zhu
- Dermatological Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
17
|
Humbert O, Samuelson C, Kiem HP. CRISPR/Cas9 for the treatment of haematological diseases: a journey from bacteria to the bedside. Br J Haematol 2020; 192:33-49. [PMID: 32506752 DOI: 10.1111/bjh.16807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 12/26/2022]
Abstract
Genome editing therapies represent a significant advancement in next-generation, precision medicine for the management of haematological diseases, and CRISPR/Cas9 has to date been the most successful implementation platform. From discovery in bacteria and archaea over three decades ago, through intensive basic research and pre-clinical development phases involving the modification of therapeutically relevant cell types, CRISPR/Cas9 genome editing is now being investigated in ongoing clinic trials. Despite the widespread enthusiasm brought by this new technology, significant challenges remain before genome editing can be routinely recommended and implemented in the clinic. These include risks of genotoxicity resulting from off-target DNA cleavage or chromosomal rearrangement, and suboptimal efficacy of homology-directed repair editing strategies, which thus limit therapeutic options. Practical hurdles such as high costs and inaccessibility to patients outside specialised centres must also be addressed. Future improvements in this rapidly developing field should circumvent current limitations with novel editing platforms and with the simplification of clinical protocols using in vivo delivery of editing reagents.
Collapse
Affiliation(s)
| | | | - Hans-Peter Kiem
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
18
|
Golay H, Jurkovic Mlakar S, Mlakar V, Nava T, Ansari M. The Biological and Clinical Relevance of G Protein-Coupled Receptors to the Outcomes of Hematopoietic Stem Cell Transplantation: A Systematized Review. Int J Mol Sci 2019; 20:E3889. [PMID: 31404983 PMCID: PMC6719093 DOI: 10.3390/ijms20163889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 01/04/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) remains the only curative treatment for several malignant and non-malignant diseases at the cost of serious treatment-related toxicities (TRTs). Recent research on extending the benefits of HSCT to more patients and indications has focused on limiting TRTs and improving immunological effects following proper mobilization and engraftment. Increasing numbers of studies report associations between HSCT outcomes and the expression or the manipulation of G protein-coupled receptors (GPCRs). This large family of cell surface receptors is involved in various human diseases. With ever-better knowledge of their crystal structures and signaling dynamics, GPCRs are already the targets for one third of the current therapeutic arsenal. The present paper assesses the current status of animal and human research on GPCRs in the context of selected HSCT outcomes via a systematized survey and analysis of the literature.
Collapse
Affiliation(s)
- Hadrien Golay
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
| | - Simona Jurkovic Mlakar
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
| | - Vid Mlakar
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
| | - Tiago Nava
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Department of Women-Children-Adolescents, Division of General Pediatrics, Pediatric Onco-Hematology Unit, Geneva University Hospitals (HUG), Avenue de la Roseraie 64, 1205 Geneva, Switzerland
| | - Marc Ansari
- Platform of Pediatric Onco-Hematology research (CANSEARCH Laboratory), Department of Pediatrics, Gynecology, and Obstetrics, University of Geneva, Bâtiment La Tulipe, Avenue de la Roseraie 64, 1205 Geneva, Switzerland.
- Department of Women-Children-Adolescents, Division of General Pediatrics, Pediatric Onco-Hematology Unit, Geneva University Hospitals (HUG), Avenue de la Roseraie 64, 1205 Geneva, Switzerland.
| |
Collapse
|
19
|
Reid JC, Tanasijevic B, Golubeva D, Boyd AL, Porras DP, Collins TJ, Bhatia M. CXCL12/CXCR4 Signaling Enhances Human PSC-Derived Hematopoietic Progenitor Function and Overcomes Early In Vivo Transplantation Failure. Stem Cell Reports 2019; 10:1625-1641. [PMID: 29742393 PMCID: PMC5995456 DOI: 10.1016/j.stemcr.2018.04.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/04/2018] [Accepted: 04/04/2018] [Indexed: 02/03/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) generate hematopoietic progenitor cells (HPCs) but fail to engraft xenograft models used to detect adult/somatic hematopoietic stem cells (HSCs) from donors. Recent progress to derive hPSC-derived HSCs has relied on cell-autonomous forced expression of transcription factors; however, the relationship of bone marrow to transplanted cells remains unknown. Here, we quantified a failure of hPSC-HPCs to survive even 24 hr post transplantation. Across several hPSC-HPC differentiation methodologies, we identified the lack of CXCR4 expression and function. Ectopic CXCR4 conferred CXCL12 ligand-dependent signaling of hPSC-HPCs in biochemical assays and increased migration/chemotaxis, hematopoietic progenitor capacity, and survival and proliferation following in vivo transplantation. This was accompanied by a transcriptional shift of hPSC-HPCs toward somatic/adult sources, but this approach failed to produce long-term HSC xenograft reconstitution. Our results reveal that networks involving CXCR4 should be targeted to generate putative HSCs with in vivo function from hPSCs. Transplant kinetics indicate human PSC-HPCs fail in the first 24 hr in bone marrow hPSC-HPCs aberrantly express chemokine receptors, specifically lacking CXCR4 Ectopic CXCR4 enhances hPSC-HPC function in vitro and transplantation in vivo CXCR4 linked with global transcriptional shift of hPSC-HPCs toward somatic HPCs
Collapse
Affiliation(s)
- Jennifer C Reid
- Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Borko Tanasijevic
- Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Diana Golubeva
- Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Allison L Boyd
- Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Deanna P Porras
- Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Tony J Collins
- Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Mickie Bhatia
- Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
20
|
Li X, Wei Z, Lv H, Wu L, Cui Y, Yao H, Li J, Zhang H, Yang B, Jiang J. Iron oxide nanoparticles promote the migration of mesenchymal stem cells to injury sites. Int J Nanomedicine 2019; 14:573-589. [PMID: 30666115 PMCID: PMC6336032 DOI: 10.2147/ijn.s184920] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Developing new methods to deliver cells to the injured tissue is a critical factor in translating cell therapeutics research into clinical use; therefore, there is a need for improved cell homing capabilities. Materials and methods In this study, we demonstrated the effects of labeling rat bone marrow-derived mesenchymal stem cells (MSCs) with fabricated polydopamine (PDA)-capped Fe3O4 (Fe3O4@PDA) superparticles employing preassembled Fe3O4 nanoparticles as the cores. Results We found that the Fe3O4@PDA composite superparticles exhibited no adverse effects on MSC characteristics. Moreover, iron oxide nanoparticles increased the number of MSCs in the S-phase, their proliferation index and migration ability, and their secretion of vascular endothelial growth factor relative to unlabeled MSCs. Interestingly, nanoparticles not only promoted the expression of C-X-C chemokine receptor 4 but also increased the expression of the migration-related proteins c-Met and C-C motif chemokine receptor 1, which has not been reported previously. Furthermore, the MSC-loaded nanoparticles exhibited improved homing and anti-inflammatory abilities in the absence of external magnetic fields in vivo. Conclusion These results indicated that iron oxide nanoparticles rendered MSCs more favorable for use in injury treatment with no negative effects on MSC properties, suggesting their potential clinical efficacy.
Collapse
Affiliation(s)
- Xiuying Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China,
| | - Zhenhong Wei
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China,
| | - Huiying Lv
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China,
| | - Liya Wu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China,
| | - Yingnan Cui
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China,
| | - Hua Yao
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China,
| | - Jing Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China,
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, People's Republic of China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, People's Republic of China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China,
| |
Collapse
|
21
|
Mesenchymal stem cell-based drug delivery strategy: from cells to biomimetic. J Control Release 2018; 294:102-113. [PMID: 30553849 DOI: 10.1016/j.jconrel.2018.12.019] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022]
Abstract
Owing to the diversity and ease of preparation of nanomaterials, the rational nanocarriers with a rational design have become increasingly popular in medical researches. Although nanoparticle-based drug delivery exhibits great potential, there are some challenges facing like rapid plasma clearance, triggering or aggravation of immune response, etc. Herein, cell-based targeted drug delivery systems have drawn more and more attention owing to low immunogenicity and intrinsic mutation rate, and innate ability to allow targeted delivery. Mesenchymal stem cells (MSCs) have been used in gene and drug delivery. The use of MSCs is a promising approach for the development of gene transfer systems and drug loading strategies because of their intrinsic properties, including homing ability and tumor tropism. By combining the inherent cell properties and merits of synthetic nanoparticles (NPs), cell membrane coated NPs emerge as the time requires. Overall, we provide a comprehensive overview of the utility of MSCs in drug and gene delivery as well as MSC membrane coated nanoparticles for therapy and drug delivery, aiming to figure out the significant room for development and highlight the potential future directions.
Collapse
|
22
|
Liu X, Cao M, Palomares M, Wu X, Li A, Yan W, Fong MY, Chan WC, Wang SE. Metastatic breast cancer cells overexpress and secrete miR-218 to regulate type I collagen deposition by osteoblasts. Breast Cancer Res 2018; 20:127. [PMID: 30348200 PMCID: PMC6198446 DOI: 10.1186/s13058-018-1059-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/04/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Bone is one of the most frequent metastatic sites of advanced breast cancer. Current therapeutic agents aim to inhibit osteoclast-mediated bone resorption but only have palliative effects. During normal bone remodeling, the balance between bone resorption and osteoblast-mediated bone formation is essential for bone homeostasis. One major function of osteoblast during bone formation is to secrete type I procollagen, which will then be processed before being crosslinked and deposited into the bone matrix. METHODS Small RNA sequencing and quantitative real-time PCR were used to detect miRNA levels in patient blood samples and in the cell lysates as well as extracellular vesicles of parental and bone-tropic MDA-MB-231 breast cancer cells. The effects of cancer cell-derived extracellular vesicles isolated by ultracentrifugation and carrying varying levels of miR-218 were examined in osteoblasts by quantitative real-time PCR, Western blot analysis, and P1NP bone formation marker analysis. Cancer cells overexpressing miR-218 were examined by transcriptome profiling through RNA sequencing to identify intrinsic genes and pathways influenced by miR-218. RESULTS We show that circulating miR-218 is associated with breast cancer bone metastasis. Cancer-secreted miR-218 directly downregulates type I collagen in osteoblasts, whereas intracellular miR-218 in breast cancer cells regulates the expression of inhibin β subunits. Increased cancer secretion of inhibin βA results in elevated Timp3 expression in osteoblasts and the subsequent repression of procollagen processing during osteoblast differentiation. CONCLUSIONS Here we identify a twofold function of cancer-derived miR-218, whose levels in the blood are associated with breast cancer metastasis to the bone, in the regulation of type I collagen deposition by osteoblasts. The adaptation of the bone niche mediated by miR-218 might further tilt the balance towards osteolysis, thereby facilitating other mechanisms to promote bone metastasis.
Collapse
Affiliation(s)
- Xuxiang Liu
- City of Hope Irell & Manella Graduate School of Biological Sciences, Duarte, CA, 91010, USA
| | - Minghui Cao
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA
| | | | - Xiwei Wu
- Department of Molecular and Cellular Biology, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Arthur Li
- Division of Biostatistics, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Wei Yan
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA
| | - Miranda Y Fong
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Wing-Chung Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA.
| |
Collapse
|
23
|
Abstract
Bone metastasis, or the development of secondary tumors within the bone of cancer patients, is a debilitating and incurable disease. Despite its morbidity, the biology of bone metastasis represents one of the most complex and intriguing of all oncogenic processes. This complexity derives from the intricately organized bone microenvironment in which the various stages of hematopoiesis, osteogenesis, and osteolysis are jointly regulated but spatially restricted. Disseminated tumor cells (DTCs) from various common malignancies such as breast, prostate, lung, and kidney cancers or myeloma are uniquely primed to subvert these endogenous bone stromal elements to grow into pathological osteolytic or osteoblastic lesions. This colonization process can be separated into three key steps: seeding, dormancy, and outgrowth. Targeting the processes of dormancy and initial outgrowth offers the most therapeutic promise. Here, we discuss the concepts of the bone metastasis niche, from controlling tumor-cell survival to growth into clinically detectable disease.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | - Theresa Guise
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| |
Collapse
|
24
|
Haworth KG, Kiem HP. Next-Generation Conditioning for Bone Marrow Transplantation: Paving the Way for CAR-T Cell-Based Conditioning. Mol Ther 2018; 26:1167-1168. [PMID: 29685383 DOI: 10.1016/j.ymthe.2018.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Kevin G Haworth
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
25
|
Arai Y, Choi U, Corsino CI, Koontz SM, Tajima M, Sweeney CL, Black MA, Feldman SA, Dinauer MC, Malech HL. Myeloid Conditioning with c-kit-Targeted CAR-T Cells Enables Donor Stem Cell Engraftment. Mol Ther 2018; 26:1181-1197. [PMID: 29622475 DOI: 10.1016/j.ymthe.2018.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/28/2018] [Accepted: 03/05/2018] [Indexed: 01/04/2023] Open
Abstract
We report a novel approach to bone marrow (BM) conditioning using c-kit-targeted chimeric antigen receptor T (c-kit CAR-T) cells in mice. Previous reports using anti-c-kit or anti-CD45 antibody linked to a toxin such as saporin have been promising. We developed a distinctly different approach using c-kit CAR-T cells. Initial studies demonstrated in vitro killing of hematopoietic stem cells by c-kit CAR-T cells but poor expansion in vivo and poor migration of CAR-T cells into BM. Pre-treatment of recipient mice with low-dose cyclophosphamide (125 mg/kg) together with CXCR4 transduction in the CAR-T cells enhanced trafficking to and expansion in BM (<1%-13.1%). This resulted in significant depletion of the BM c-kit+ population (9.0%-0.1%). Because congenic Thy1.1 CAR-T cells were used in the Thy1.2-recipient mice, anti-Thy1.1 antibody could be used to deplete CAR-T cells in vivo before donor BM transplant. This achieved 20%-40% multilineage engraftment. We applied this conditioning to achieve an average of 28% correction of chronic granulomatous disease mice by wild-type BM transplant. Our findings provide a proof of concept that c-kit CAR-T cells can achieve effective BM conditioning without chemo-/radiotherapy. Our work also demonstrates that co-expression of a trafficking receptor can enhance targeting of CAR-T cells to a designated tissue.
Collapse
Affiliation(s)
- Yasuyuki Arai
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Uimook Choi
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Cristina I Corsino
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Sherry M Koontz
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Masaki Tajima
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Colin L Sweeney
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Mary A Black
- Surgery Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Steven A Feldman
- Surgery Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Mary C Dinauer
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Harry L Malech
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
26
|
Zuccolo E, Di Buduo C, Lodola F, Orecchioni S, Scarpellino G, Kheder DA, Poletto V, Guerra G, Bertolini F, Balduini A, Rosti V, Moccia F. Stromal Cell-Derived Factor-1α Promotes Endothelial Colony-Forming Cell Migration Through the Ca2+-Dependent Activation of the Extracellular Signal-Regulated Kinase 1/2 and Phosphoinositide 3-Kinase/AKT Pathways. Stem Cells Dev 2018; 27:23-34. [DOI: 10.1089/scd.2017.0114] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Christian Di Buduo
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Laboratory of Biochemistry, Biotechnology, and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Matteo Foundation, Pavia, Italy
| | - Francesco Lodola
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology–Oncology, European Institute of Oncology, Milan, Italy
| | - Giorgia Scarpellino
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Dlzar Ali Kheder
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
- Department of Biology, Faculty of Science, University of Zakho, Zakho, Kurdistan-Region of Iraq
| | - Valentina Poletto
- Laboratory of Biochemistry, Biotechnology, and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Matteo Foundation, Pavia, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences “Vincenzo Tiberio,” University of Molise, Campobasso, Italy
| | - Francesco Bertolini
- Laboratory of Hematology–Oncology, European Institute of Oncology, Milan, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Laboratory of Biochemistry, Biotechnology, and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Matteo Foundation, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Vittorio Rosti
- Laboratory of Biochemistry, Biotechnology, and Advanced Diagnosis, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Matteo Foundation, Pavia, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| |
Collapse
|
27
|
Abstract
Bone metastasis is one of the most common forms of metastasis from a number of different primary carcinomas. MicroRNAs (miRNAs) are short, endogenous RNAs that negatively regulate gene expression to control essential pathways, including those involved in bone organogenesis and homeostasis. As these pathways are often hijacked during bone metastasis, it is not surprising that miRNAs can also influence bone metastasis formation. Areas covered: In this review, we first summarize the major signalling pathways involved in normal bone development and bone metastasis. We will then discuss the overall roles of miRNAs in cancer metastasis and highlight the recent findings on the effects of miRNAs in bone metastasis. To this aim, we have performed a literature search in PubMed by using the search words 'miRNAs' and 'bone metastasis', selecting relevant scientific articles published between 2010 and 2016. Seminal publications before 2010 on the metastatic role of miRNAs have also been considered. Expert commentary: With the lack of current diagnostic biomarkers and effective targeted therapies for bone metastasis, the significant role of miRNAs in the regulation of bone homeostasis and bone metastasis may support the future use of miRNAs as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Douglas G Cheung
- a Department of Cancer Biology and Genetics , The Ohio State University , Columbus , Ohio , USA
| | - Marta Buzzetti
- b Biomedical Research Centre, School of Environment and Life Sciences , University of Salford , Salford , UK
| | - Gianpiero Di Leva
- b Biomedical Research Centre, School of Environment and Life Sciences , University of Salford , Salford , UK
| |
Collapse
|
28
|
Corradetti B, Taraballi F, Martinez JO, Minardi S, Basu N, Bauza G, Evangelopoulos M, Powell S, Corbo C, Tasciotti E. Hyaluronic acid coatings as a simple and efficient approach to improve MSC homing toward the site of inflammation. Sci Rep 2017; 7:7991. [PMID: 28801676 PMCID: PMC5554184 DOI: 10.1038/s41598-017-08687-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 07/13/2017] [Indexed: 12/13/2022] Open
Abstract
A major challenge in regenerative medicine is to improve therapeutic cells' delivery and targeting using an efficient and simple protocol. Mesenchymal stem cells (MSC) are currently employed for the treatment of inflammatory-based diseases, due to their powerful immunosoppressive potential. Here we report a simple and versatile method to transiently overexpress the hyaluronic acid (HA) receptor, CD44, on MSC membranes, to improve their homing potential towards an inflammatory site without affecting their behavior. The effect of HA-coatings on murine MSC was functionally determined both, in vitro and in vivo as a consequence of the transient CD44 overexpression induced by HA. Data obtained from the in vitro migration assay demonstrated a two-fold increase in the migratory potential of HA-treated MSC compared to untreated cells. In an LPS-induced inflamed ear murine model, HA-treated MSC demonstrated a significantly higher inflammatory targeting as observed at 72 hrs as compared to untreated cells. This increased accumulation for HA-treated MSC yielded a substantial reduction in inflammation as demonstrated by the decrease in the expression of pro-inflammatory markers and by the induction of a pro-regenerative environment.
Collapse
Affiliation(s)
- Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60131, Ancona, Italy
| | - Francesca Taraballi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Orthopaedic & Sports Medicine, The Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Jonathan O Martinez
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Silvia Minardi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Nupur Basu
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Guillermo Bauza
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Centre for NanoHealth, Swansea University Medical School, Swansea University Bay, Singleton Park, SA2 8PP, Wales, UK
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Sebastian Powell
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Claudia Corbo
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Centre for NanoHealth, Swansea University Medical School, Swansea University Bay, Singleton Park, SA2 8PP, Wales, UK.
- Department of Orthopaedic & Sports Medicine, The Houston Methodist Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Chen W, Li M, Su G, Zang Y, Yan Z, Cheng H, Pan B, Cao J, Wu Q, Zhao K, Zhu F, Zeng L, Li Z, Xu K. Co-transplantation of Hematopoietic Stem Cells and Cxcr4 Gene-Transduced Mesenchymal Stem Cells Promotes Hematopoiesis. Cell Biochem Biophys 2016; 71:1579-87. [PMID: 25391891 DOI: 10.1007/s12013-014-0381-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) are a promising candidate for cellular therapies. Co-transplantation of MSCs and hematopoietic stem cells (HSCs) promotes successful engraftment and improves hematopoietic recovery. In this study, the effects of co-transplantation of HSCs and mouse bone marrow (BM)-derived MSCs overexpressing CXCR4 (CXCR4-MSC) on CXCR4-MSC homing capacity and the reconstitution potential in lethally irradiated mice were evaluated. Recovery of donor-derived peripheral blood leukocytes and platelets was accelerated when CXCR4-MSCs were co-transplanted with BM cells. The frequency of c-kit(+)Sca(+)Lin(-) HSCs was higher in recipient BM following co-transplantation of CXCR4-MSCs compared with the EGFP-MSC control and the BMT only groups. Surprisingly, the rate of early engraftment of donor-derived BM cells in recipients co-transplanted with CXCR4-MSCs was slightly lower than in the absence of MSCs on day 7. Moreover, co-transplantation of CXCR4-MSCs regulated the balance of T helper cells subsets. Hematopoietic tissue reconstitution was evaluated by histopathological analysis of BM and spleen. Co-transplantation of CXCR4-MSCs was shown to promote the recovery of hematopoietic organs. These findings indicate that co-transplantation of CXCR4-MSCs promotes the early phase of hematopoietic recovery and sustained hematopoiesis.
Collapse
Affiliation(s)
- Wei Chen
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Xuzhou Medical College, Blood Diseases Institute, Xuzhou, 221002, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Miao Li
- Xuzhou Children's Hospital, Sudi Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Guizhen Su
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yu Zang
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Zhiling Yan
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Hai Cheng
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Bin Pan
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Jiang Cao
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Qingyun Wu
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Kai Zhao
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Feng Zhu
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Lingyu Zeng
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Zhenyu Li
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Kailin Xu
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical College, West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Xuzhou Medical College, Blood Diseases Institute, Xuzhou, 221002, People's Republic of China. .,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
30
|
Dothel G, Raschi E, Rimondini R, De Ponti F. Mesenchymal stromal cell-based therapy: Regulatory and translational aspects in gastroenterology. World J Gastroenterol 2016; 22:9057-9068. [PMID: 27895395 PMCID: PMC5107589 DOI: 10.3748/wjg.v22.i41.9057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/09/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023] Open
Abstract
The past decade has witnessed an outstanding scientific production focused towards the possible clinical applications of mesenchymal stromal cells (MSCs) in autoimmune and chronic inflammatory diseases. This raised the need of novel standards to adequately address quality, efficacy and safety issues of this advanced therapy. The development of a streamlined regulation is currently hampered by the complexity of analyzing dynamic biological entities rather than chemicals. Although numerous pieces of evidence show efficacy in reducing intestinal inflammation, some inconsistencies between the mechanisms of action of rodent vs human MSCs suggest caution before assigning translational value to preclinical studies. Preliminary evidence from clinical trials showed efficacy of MSCs in the treatment of fistulizing Crohn’s disease (CD), and preparations of heterologous MSCs for CD treatment are currently tested in ongoing clinical trials. However, safety issues, especially in long-term treatment, still require solid clinical data. In this regard, standardized guidelines for appropriate dosing and methods of infusion could enhance the likelihood to predict more accurately the number of responders and the duration of remission periods. In addition, elucidating MSC mechanisms of action could lead to novel and more reliable formulations such as those derived from the MSCs themselves (e.g., supernatants).
Collapse
|
31
|
Progress and obstacles towards generating hematopoietic stem cells from pluripotent stem cells. Curr Opin Hematol 2016; 22:317-23. [PMID: 26049752 DOI: 10.1097/moh.0000000000000147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Human pluripotent stem cells (PSCs) have the potential to provide an inexhaustible source of hematopoietic stem cells (HSCs) that could be used in disease modeling and in clinical applications such as transplantation. Although the goal of deriving definitive HSCs from PSCs has not been achieved, recent studies indicate that progress is being made. This review will provide information on the current status of deriving HSCs from PSCs, and will highlight existing challenges and obstacles. RECENT FINDINGS Recent strides in HSC generation from PSCs has included derivation of developmental intermediates, identification of transcription factors and small molecules that support hematopoietic derivation, and the development of strategies to recapitulate niche-like conditions. SUMMARY Despite considerable progress in defining the molecular events driving derivation of hematopoietic progenitor cells from PSCs, the generation of robust transplantable HSCs from PSCs remains elusive. We propose that this goal can be facilitated by better understanding of the regulatory pathways governing HSC identity, development of HSC supportive conditions, and examining the marrow homing properties of PSC-derived HSCs.
Collapse
|
32
|
The role of "bone immunological niche" for a new pathogenetic paradigm of osteoporosis. Anal Cell Pathol (Amst) 2015; 2015:434389. [PMID: 26491648 PMCID: PMC4605147 DOI: 10.1155/2015/434389] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/09/2015] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis is characterized by low bone mass and microarchitectural deterioration of bone tissue. The etiology and pathogenetic mechanisms of osteoporosis have not been clearly elucidated. Osteoporosis is linked to bone resorption by the activation of the osteoclastogenic process. The breakdown of homeostasis among pro- and antiosteoclastogenic cells causes unbalanced bone remodeling. The complex interactions among these cells in the bone microenvironment involve several mediators and proinflammatory pathways. Thus, we may consider the bone microenvironment as a complex system in which local and systemic immunity are regulated and we propose to consider it as an "immunological niche." The study of the "bone immunological niche" will permit a better understanding of the complex cell trafficking which regulates bone resorption and disease. The goal of a perfect therapy for osteoporosis would be to potentiate good cells and block the bad ones. In this scenario, additional factors may take part in helping or hindering the proosteoblastogenic factors. Several proosteoblastogenic and antiosteoclastogenic agents have already been identified and some have been developed and commercialized as biological therapies for osteoporosis. Targeting the cellular network of the "bone immunological niche" may represent a successful strategy to better understand and treat osteoporosis and its complications.
Collapse
|
33
|
Wang Z, Wang Y, Wang Z, Gutkind JS, Wang Z, Wang F, Lu J, Niu G, Teng G, Chen X. Engineered mesenchymal stem cells with enhanced tropism and paracrine secretion of cytokines and growth factors to treat traumatic brain injury. Stem Cells 2015; 33:456-67. [PMID: 25346537 DOI: 10.1002/stem.1878] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 07/31/2014] [Accepted: 08/17/2014] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability worldwide. Mesenchymal stem cells (MSCs) are promising for the treatment of various diseases and injuries. Many strategies have been applied to attract MSCs to injury site after systemic infusion. In this study, we evidenced that the CXC chemokine receptor 4 (CXCR4)-SDF1α (stromal cell-derived factor 1α) axis in engineered MSCs serves not only to attract MSC migration to TBI but also to activate Akt kinase signaling pathway in MSCs to promote paracrine secretion of cytokines and growth factors. This leads to enhanced vasculogenesis and neuroprotection at the boundary of TBI for improved blood supply, recovery of axon connectivity, and behavioral ability and results in positive feedback loop to enhance additional MSC tropism to injury. These findings indicate a new aspect of SDF1α in mediating CXCR4 engineered MSCs for brain trauma homing and recovery. This potential mechanism may be applicable to other injuries, where CXCR4-SDF1α interaction is highly associated.
Collapse
Affiliation(s)
- Zhe Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Vertès AA. The potential of cytotherapeutics in hematologic reconstitution and in the treatment and prophylaxis of graft-versus-host disease. Chapter II: emerging transformational cytotherapies. Regen Med 2015; 10:345-73. [DOI: 10.2217/rme.15.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a life-saving treatment for inherited anemias, immunodeficiencies or hematologic malignancies. A major complication of allo-HSCT associated with high transplant-related mortality rates is graft-versus-host disease (GvHD). Current and future clinical benefits in HSCT enabled by advances in hematopoietic stem cells, mesenchymal stem cells, Tregs and natural killer cells technologies are reviewed here and discussed. Among these evolutions, based on the need for mesenchymal stem cells to be recruited by an inflammatory environment, the development and use of novel GvHD biomarkers could be explored further to deliver the right pharmaceutical to the right patient at the right time. The successful commercialization of cytotherapeutics to efficiently manage GvHD will create a virtuous ‘halo’ effect for regenerative medicine.
Collapse
Affiliation(s)
- Alain A Vertès
- Sloan Fellow, London Business School, London, UK
- NxR Biotechnologies GmbH, Basel, Switzerland
| |
Collapse
|
35
|
Li L, Yang M, Wang C, Zhao Q, Liu J, Zhan C, Liu Z, Li X, Wang W, Yang X. Effects of cytokines and chemokines on migration of mesenchymal stem cells following spinal cord injury. Neural Regen Res 2015; 7:1106-12. [PMID: 25722702 PMCID: PMC4340025 DOI: 10.3969/j.issn.1673-5374.2012.14.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 03/20/2012] [Indexed: 01/16/2023] Open
Abstract
We investigated the effects of cytokines and chemokines and their associated signaling pathways on mesenchymal stem cell migration after spinal cord injury, to determine their roles in the curative effects of mesenchymal stem cells. This study reviewed the effects of tumor necrosis factor-α, vascular endothelial growth factor, hepatocyte growth factor, platelet-derived growth factor, basic fibroblast growth factor, insulin like growth factor-1, stromal cell-derived factor and monocyte chemoattractant protein-1, 3 during mesenchymal stem cell migration to damaged sites, and analyzed the signal transduction pathways involved in their effects on mesenchymal stem cell migration. The results confirmed that phosphatidylinositol 3-kinase/serine/threonine protein kinases and nuclear factor-κB play crucial roles in the migration of mesenchymal stem cells induced by cytokines and chemokines.
Collapse
Affiliation(s)
- Longyun Li
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Maoguang Yang
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Chunxin Wang
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Qiheng Zhao
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Jian Liu
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Chuanguo Zhan
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Zhi Liu
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Xuepeng Li
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Weihua Wang
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Xiaoyu Yang
- Department of Orthopedics, China-Japan Friendship Hospital, Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
36
|
Takeo M, Li C, Matsuda M, Nagai H, Hatanaka W, Yamamoto T, Kishimura A, Mori T, Katayama Y. Optimum design of amphiphilic polymers bearing hydrophobic groups for both cell surface ligand presentation and intercellular cross-linking. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2015; 26:353-68. [PMID: 25597323 DOI: 10.1080/09205063.2015.1007414] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Amphiphilic polymers bearing hydrophobic alkyl groups are expected to be applicable for both ligand presentation on the cell surface and intercellular crosslinking. To explore the optimum design for each application, we synthesized eight different acyl-modified dextrans with varying molecular weight, alkyl length, and alkyl modification degree. We found that the behenate-modified polymers retained on the cell surface longer than the palmitate-modified ones. Since the polymers were also modified with biotin, streptavidin can be presented on the cell surface through biotin-streptavidin recognition. The duration of streptavidin on the cell surface is longer in the behenate-modified polymer than the palmitate-modified one. As for the intercellular crosslinking, the palmitate-modified polymers were more efficient than the behenate-modified polymers. The findings in this research will be helpful to design the acyl-modified polymers for the cell surface engineering.
Collapse
Affiliation(s)
- Masafumi Takeo
- a Graduate School of System Life Science , Kyushu University , 744 Motooka, Nishi-ku, Fukuoka 819-0395 , Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lai CY, Yamazaki S, Okabe M, Suzuki S, Maeyama Y, Iimura Y, Onodera M, Kakuta S, Iwakura Y, Nojima M, Otsu M, Nakauchi H. Stage-specific roles for CXCR4 signaling in murine hematopoietic stem/progenitor cells in the process of bone marrow repopulation. Stem Cells 2015; 32:1929-42. [PMID: 24510783 DOI: 10.1002/stem.1670] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/28/2014] [Indexed: 11/07/2022]
Abstract
Hematopoietic cell transplantation has proven beneficial for various intractable diseases, but it remains unclear how hematopoietic stem/progenitor cells (HSPCs) home to the bone marrow (BM) microenvironment, initiate hematopoietic reconstitution, and maintain life-long hematopoiesis. The use of newly elucidated molecular determinants for overall HSPC engraftment should benefit patients. Here, we report that modification of C-X-C chemokine receptor type 4 (Cxcr4) signaling in murine HSPCs does not significantly affect initial homing/lodging events, but leads to alteration in subsequent BM repopulation kinetics, with observations confirmed by both gain- and loss-of-function approaches. By using C-terminal truncated Cxcr4 as a gain-of-function effector, we demonstrated that signal augmentation likely led to favorable in vivo repopulation of primitive cell populations in BM. These improved features were correlated with enhanced seeding efficiencies in stromal cell cocultures and altered ligand-mediated phosphorylation kinetics of extracellular signal-regulated kinases observed in Cxcr4 signal-augmented HSPCs in vitro. Unexpectedly, however, sustained signal enhancement even with wild-type Cxcr4 overexpression resulted in impaired peripheral blood (PB) reconstitution, most likely by preventing release of donor hematopoietic cells from the marrow environment. We thus conclude that timely regulation of Cxcr4/CXCR4 signaling is key in providing donor HSPCs with enhanced repopulation potential following transplantation, whilst preserving the ability to release HSPC progeny into PB for improved transplantation outcomes.
Collapse
Affiliation(s)
- Chen-Yi Lai
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan; Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Yuan A, Lee Y, Choi U, Moeckel G, Karihaloo A. Chemokine receptor Cxcr4 contributes to kidney fibrosis via multiple effectors. Am J Physiol Renal Physiol 2014; 308:F459-72. [PMID: 25537742 DOI: 10.1152/ajprenal.00146.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Kidney fibrosis is the final common pathway for virtually every type of chronic kidney disease and is a consequence of a prolonged healing response that follows tissue inflammation. Chronic kidney inflammation ultimately leads to progressive tissue injury and scarring/fibrosis. Several pathways have been implicated in the progression of kidney fibrosis. In the present study, we demonstrate that G protein-coupled chemokine (C-X-C motif) receptor (CXCR)4 was significantly upregulated after renal injury and that sustained activation of Cxcr4 expression augmented the fibrotic response. We demonstrate that after unilateral ureteral obstruction (UUO), both gene and protein expression of Cxcr4 were highly upregulated in tubular cells of the nephron. The increased Cxcr4 expression in tubules correlated with their increased dedifferentiated state, leading to increased mRNA expression of platelet-derived growth factor (PDGF)-α, transforming growth factor (TGF)-β1, and concurrent loss of bone morphogenetic protein 7 (Bmp7). Ablation of tubular Cxcr4 attenuated UUO-mediated fibrotic responses, which correlated with a significant reduction in PDGF-α and TGF-β1 levels and preservation of Bmp7 expression after UUO. Furthermore, Cxcr4(+) immune cells infiltrated the obstructed kidney and further upregulate their Cxcr4 expression. Genetic ablation of Cxcr4 from macrophages was protective against UUO-induced fibrosis. There was also reduced total kidney TGF-β1, which correlated with reduced Smad activation and α-smooth muscle actin levels. We conclude that chronic high Cxcr4 expression in multiple effector cell types can contribute to the pathogenesis of renal fibrosis by altering their biological profile. This study uncovered a novel cross-talk between Cxcr4-TGF-β1 and Bmp7 pathways and may provide novel targets for interrupting the progression of fibrosis.
Collapse
Affiliation(s)
- Amy Yuan
- Department of Medicine, Section of Nephrology, Yale School of Medicine, New Haven, Connecticut
| | - Yashang Lee
- Department of Medicine, Section of Nephrology, Yale School of Medicine, New Haven, Connecticut
| | - Uimook Choi
- Laboratory of Host Defense, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Gilbert Moeckel
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut; and
| | - Anil Karihaloo
- Department of Medicine, Section of Nephrology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
39
|
Tanenbaum ME, Gilbert LA, Qi LS, Weissman JS, Vale RD. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 2014; 159:635-46. [PMID: 25307933 DOI: 10.1016/j.cell.2014.09.039] [Citation(s) in RCA: 1078] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 09/03/2014] [Accepted: 09/22/2014] [Indexed: 10/24/2022]
Abstract
Signals in many biological processes can be amplified by recruiting multiple copies of regulatory proteins to a site of action. Harnessing this principle, we have developed a protein scaffold, a repeating peptide array termed SunTag, which can recruit multiple copies of an antibody-fusion protein. We show that the SunTag can recruit up to 24 copies of GFP, thereby enabling long-term imaging of single protein molecules in living cells. We also use the SunTag to create a potent synthetic transcription factor by recruiting multiple copies of a transcriptional activation domain to a nuclease-deficient CRISPR/Cas9 protein and demonstrate strong activation of endogenous gene expression and re-engineered cell behavior with this system. Thus, the SunTag provides a versatile platform for multimerizing proteins on a target protein scaffold and is likely to have many applications in imaging and controlling biological outputs.
Collapse
Affiliation(s)
- Marvin E Tanenbaum
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Luke A Gilbert
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Center for RNA Systems Biology, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biomedical Research (QB3), San Francisco, CA 94158, USA
| | - Lei S Qi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Center for RNA Systems Biology, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biomedical Research (QB3), San Francisco, CA 94158, USA
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Center for RNA Systems Biology, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biomedical Research (QB3), San Francisco, CA 94158, USA
| | - Ronald D Vale
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
40
|
Bain O, Detela G, Kim HW, Mason C, Mathur A, Wall IB. Altered hMSC functional characteristics in short-term culture and when placed in low oxygen environments: implications for cell retention at physiologic sites. Regen Med 2014; 9:153-65. [DOI: 10.2217/rme.13.91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: It is very difficult to conserve critical cell characteristics during expansion in culture, particularly those of adult mesenchymal stromal cells (MSCs), whose characteristics can change rapidly even within a short period of expansion. Aim: In this study our aim was to measure cell characteristics that are critical for retention at the injury site after therapeutic delivery. Cells were cultured under conditions typical of current standard best practice. The impact of passage number was assessed and assays were performed in low oxygen (2%) as an in vitro model of physiologic oxygen tension at injury sites. The effect of chemokine preconditioning with SDF1 was also assessed. Materials & methods: Bone marrow mononuclear cells from patients recruited to the REGENERATE Phase II clinical trials, along with MSCs from healthy volunteers subjected to a short period of expansion, were assessed for attachment and migration ability. Using MSCs from healthy donors, the effect of reduced oxygen was also assessed. Results: Short-term expansion resulted in increased cell attachment but decreased rate of migration, whereas attachment and migration of patient-derived bone marrow mononuclear cells was highly heterogeneous. Reduced oxygen impaired MSC attachment but not migration. Finally, SDF1 did not improve any of the responses. Conclusion: The basic functional responses of MSCs required for retention and engraftment alter rapidly even over a relatively short expansion period. This needs careful consideration when expanding cells to achieve clinical quantities for therapy.
Collapse
Affiliation(s)
- Owen Bain
- UCL Department of Biochemical Engineering, Torrington Place, London, WC1E 7JE, UK
| | - Giulia Detela
- UCL Department of Biochemical Engineering, Torrington Place, London, WC1E 7JE, UK
| | - Hae-Won Kim
- Department of Nanobiomedical Science & BK21 Plus NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 330-714, Republic of Korea
- College of Dentistry & Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, Republic of Korea
| | - Chris Mason
- UCL Department of Biochemical Engineering, Torrington Place, London, WC1E 7JE, UK
| | - Anthony Mathur
- Barts Health NIHR Biomedical Research Unit, Department of Cardiology, London Chest Hospital, Bonner Road, London, E2 9JX, UK
| | - Ivan B Wall
- UCL Department of Biochemical Engineering, Torrington Place, London, WC1E 7JE, UK
| |
Collapse
|
41
|
Ankrum JA, Miranda OR, Ng KS, Sarkar D, Xu C, Karp JM. Engineering cells with intracellular agent-loaded microparticles to control cell phenotype. Nat Protoc 2014; 9:233-45. [PMID: 24407352 DOI: 10.1038/nprot.2014.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cell therapies enable unprecedented treatment options to replace tissues, destroy tumors and facilitate regeneration. The greatest challenge facing cell therapy is the inability to control the fate and function of cells after transplantation. We have developed an approach to control cell phenotype in vitro and after transplantation by engineering cells with intracellular depots that continuously release phenotype-altering agents for days to weeks. The platform enables control of cells' secretome, viability, proliferation and differentiation, and the platform can be used to deliver drugs or other factors (e.g., dexamethasone, rhodamine and iron oxide) to the cell's microenvironment. The preparation, efficient internalization and intracellular stabilization of ∼1-μm drug-loaded microparticles are critical for establishing sustained control of cell phenotype. Herein we provide a protocol to generate and characterize micrometer-sized agent-doped poly(lactic-co-glycolic) acid (PLGA) particles by using a single-emulsion evaporation technique (7 h), to uniformly engineer cultured cells (15 h), to confirm particle internalization and to troubleshoot commonly experienced obstacles.
Collapse
Affiliation(s)
- James A Ankrum
- 1] Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA. [2] Harvard-MIT Division of Health Sciences and Technology, Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Oscar R Miranda
- 1] Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA. [2] Harvard-MIT Division of Health Sciences and Technology, Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Kelvin S Ng
- 1] Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA. [2] Harvard-MIT Division of Health Sciences and Technology, Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Debanjan Sarkar
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Chenjie Xu
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Jeffrey M Karp
- 1] Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA. [2] Harvard-MIT Division of Health Sciences and Technology, Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
42
|
Abstract
Cell-based therapy is an emerging field but one that has shown early promise for the treatment of human kidney diseases. The most widely used cell is the mesenchymal stem cell (MSC) given its ability to be harvested from bone marrow, expanded in culture, and used in allogeneic protocols. The beneficial effects of MSCs occur through differentiation-independent pathways that include increased cell survival and proliferation, decreased inflammation, and suppression of immune function. Acute kidney injury and kidney transplantation are the two conditions most frequently treated with MSC infusion. Although initial studies are promising, the long-term efficacy and safety of MSC infusion awaits further study.
Collapse
Affiliation(s)
- Mark E Rosenberg
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
43
|
Yang D, Sun S, Wang Z, Zhu P, Yang Z, Zhang B. Stromal cell-derived factor-1 receptor CXCR4-overexpressing bone marrow mesenchymal stem cells accelerate wound healing by migrating into skin injury areas. Cell Reprogram 2013; 15:206-15. [PMID: 23713431 DOI: 10.1089/cell.2012.0046] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Stromal cell-derived factor-1 (SDF-1) and its membrane receptor C-X-C chemokine receptor type 4 (CXCR4) are involved in the homing and migration of multiple stem cell types, neovascularization, and cell proliferation. This study investigated the hypothesis that bone marrow-derived mesenchymal stem cells (BMSCs) accelerate skin wound healing in the mouse model by overexpression of CXCR4 in BMSCs. We compared SDF-1 expression and skin wound healing times of BALB/c mice, severe combined immunodeficiency (SCID) mice, and immune system-deficient nude mice after (60)Co radiation-induced injury of their bone marrow. The occurrence of transplanted adenovirus-transfected CXCR4-overexpressing male BMSCs in the wound area was compared with the occurrence of untransfected male BALB/c BMSCs in (60)Co-irradiated female mice skin wound healing areas by Y chromosome marker analyses. The wound healing time of BALB/c mice was 14.00±1.41 days, whereas for the nude and SCID mice it was 17.16±1.17 days and 19.83±0.76 days, respectively. Male BMSCs could be detected in the surrounding areas of (60)Co-irradiated female BALB/c mice wounds, and CXCR4-overexpressing BMSCs accelerated the wound healing time. CXCR4-overexpressing BMSCs migrate in an enhanced manner to skin wounds in a SDF-1-expression-dependent manner, thereby reducing the skin wound healing time.
Collapse
Affiliation(s)
- Dazhi Yang
- Department 4, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | | | | | | | | | | |
Collapse
|
44
|
Cutler C, Multani P, Robbins D, Kim HT, Le T, Hoggatt J, Pelus LM, Desponts C, Chen YB, Rezner B, Armand P, Koreth J, Glotzbecker B, Ho VT, Alyea E, Isom M, Kao G, Armant M, Silberstein L, Hu P, Soiffer RJ, Scadden DT, Ritz J, Goessling W, North TE, Mendlein J, Ballen K, Zon LI, Antin JH, Shoemaker DD. Prostaglandin-modulated umbilical cord blood hematopoietic stem cell transplantation. Blood 2013; 122:3074-81. [PMID: 23996087 PMCID: PMC3811179 DOI: 10.1182/blood-2013-05-503177] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/20/2013] [Indexed: 01/16/2023] Open
Abstract
Umbilical cord blood (UCB) is a valuable source of hematopoietic stem cells (HSCs) for use in allogeneic transplantation. Key advantages of UCB are rapid availability and less stringent requirements for HLA matching. However, UCB contains an inherently limited HSC count, which is associated with delayed time to engraftment, high graft failure rates, and early mortality. 16,16-Dimethyl prostaglandin E2 (dmPGE2) was previously identified to be a critical regulator of HSC homeostasis, and we hypothesized that brief ex vivo modulation with dmPGE2 could improve patient outcomes by increasing the "effective dose" of HSCs. Molecular profiling approaches were used to determine the optimal ex vivo modulation conditions (temperature, time, concentration, and media) for use in the clinical setting. A phase 1 trial was performed to evaluate the safety and therapeutic potential of ex vivo modulation of a single UCB unit using dmPGE2 before reduced-intensity, double UCB transplantation. Results from this study demonstrated clear safety with durable, multilineage engraftment of dmPGE2-treated UCB units. We observed encouraging trends in efficacy, with accelerated neutrophil recovery (17.5 vs 21 days, P = .045), coupled with preferential, long-term engraftment of the dmPGE2-treated UCB unit in 10 of 12 treated participants.
Collapse
|
45
|
Esposito M, Kang Y. Targeting tumor-stromal interactions in bone metastasis. Pharmacol Ther 2013; 141:222-33. [PMID: 24140083 DOI: 10.1016/j.pharmthera.2013.10.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 09/05/2013] [Indexed: 12/17/2022]
Abstract
Bone metastasis is a frequent occurrence in late stage solid tumors, including breast cancers, prostate or lung. However, the causes for this proclivity have only recently been elucidated. Significant progress has been made in the past decade toward understanding the molecular underpinnings of bone metastasis, and much of this research reveals a crucial role of the host stroma in each step of the metastatic cascade. Tumor-stromal interactions are crucial in engineering a pre-metastatic niche, accommodating metastatic seeding, and establishing the vicious cycle of bone metastasis. Current treatments in bone metastasis focus on latter steps of the metastatic cascade, with most treatments targeting the process of bone remodeling; however, emerging research identifies many other candidates as promising targets. Host stromal cells including platelets and endothelial cells are important in the early steps of metastatic homing, attachment and extravasation while a variety of immune cells, parenchymal cells and mesenchymal cells of the bone marrow are important in the establishment of overt, immune-suppressed metastatic lesions. Many participants during these steps have been identified and functionally validated. Significant contributors include integrins, (αvβ3, α2β1, α4β1), TGFβ family members, bone resident proteins (BSP, OPG, SPARC, OPN), RANKL, and PTHrP. In this review, we will discuss the contribution of host stromal cells to pre-metastatic niche conditioning, seeding, dormancy, bone-remodeling, immune regulation, and chemotherapeutic shielding in bone metastasis. Research exploring these interactions between bone metastases and stromal cells has yielded many therapeutic targets, and we will discuss both the current and future therapeutic avenues in treating bone metastasis.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, United States
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, United States.
| |
Collapse
|
46
|
Kim DS, Kim YS, Bae WJ, Lee HJ, Chang SW, Kim WS, Kim EC. The role of SDF-1 and CXCR4 on odontoblastic differentiation in human dental pulp cells. Int Endod J 2013; 47:534-41. [DOI: 10.1111/iej.12182] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/12/2013] [Indexed: 01/02/2023]
Affiliation(s)
- D. S. Kim
- Department of Conservative Dentistry; School of Dentistry; Kyung Hee University; Seoul Korea
| | - Y. S. Kim
- Department of Maxillofacial Tissue Regeneration and Research Center for Tooth and Periodontal Regeneration (MRC); School of Dentistry; Kyung Hee University; Seoul Korea
| | - W. J. Bae
- Department of Maxillofacial Tissue Regeneration and Research Center for Tooth and Periodontal Regeneration (MRC); School of Dentistry; Kyung Hee University; Seoul Korea
| | - H. J. Lee
- Department of Maxillofacial Tissue Regeneration and Research Center for Tooth and Periodontal Regeneration (MRC); School of Dentistry; Kyung Hee University; Seoul Korea
| | - S. W. Chang
- Department of Conservative Dentistry; School of Dentistry; Kyung Hee University; Seoul Korea
| | - W. S. Kim
- Department of Periodontology; School of Dentistry; Wonkwang University; Iksan Korea
| | - E. C. Kim
- Department of Maxillofacial Tissue Regeneration and Research Center for Tooth and Periodontal Regeneration (MRC); School of Dentistry; Kyung Hee University; Seoul Korea
| |
Collapse
|
47
|
Sahin AO, Buitenhuis M. Molecular mechanisms underlying adhesion and migration of hematopoietic stem cells. Cell Adh Migr 2012; 6:39-48. [PMID: 22647939 DOI: 10.4161/cam.18975] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem cell transplantation is the most powerful treatment modality for a large number of hematopoietic malignancies, including leukemia. Successful hematopoietic recovery after transplantation depends on homing of hematopoietic stem cells to the bone marrow and subsequent lodging of those cells in specific niches in the bone marrow. Migration of hematopoietic stem cells to the bone marrow is a highly regulated process that requires correct regulation of the expression and activity of various molecules including chemoattractants, selectins and integrins. This review will discuss recent studies that have extended our understanding of the molecular mechanisms underlying adhesion, migration and bone marrow homing of hematopoietic stem cells.
Collapse
Affiliation(s)
- Aysegul Ocal Sahin
- Department of Hematology and Erasmus MC Stem Cell Institute for Regenerative Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
48
|
Xinaris C, Morigi M, Benedetti V, Imberti B, Fabricio AS, Squarcina E, Benigni A, Gagliardini E, Remuzzi G. A novel strategy to enhance mesenchymal stem cell migration capacity and promote tissue repair in an injury specific fashion. Cell Transplant 2012; 22:423-36. [PMID: 22889699 DOI: 10.3727/096368912x653246] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) of bone marrow origin appear to be an attractive candidate for cell-based therapies. However, the major barrier to the effective implementation of MSC-based therapies is the lack of specific homing of exogenously infused cells and overall the inability to drive them to the diseased or damaged tissue. In order to circumvent these limitations, we developed a preconditioning strategy to optimize MSC migration efficiency and potentiate their beneficial effect at the site of injury. Initially, we screened different molecules by using an in vitro injury-migration setting, and subsequently, we evaluated the effectiveness of the different strategies in mice with acute kidney injury (AKI). Our results showed that preconditioning of MSCs with IGF-1 before infusion improved cell migration capacity and restored normal renal function after AKI. The present study demonstrates that promoting migration of MSCs could increase their therapeutic potential and indicates a new therapeutic paradigm for organ repair.
Collapse
Affiliation(s)
- C Xinaris
- "Mario Negri" Institute for Pharmacological Research, Bergamo, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gheisari Y, Azadmanesh K, Ahmadbeigi N, Nassiri SM, Golestaneh AF, Naderi M, Vasei M, Arefian E, Mirab-Samiee S, Shafiee A, Soleimani M, Zeinali S. Genetic modification of mesenchymal stem cells to overexpress CXCR4 and CXCR7 does not improve the homing and therapeutic potentials of these cells in experimental acute kidney injury. Stem Cells Dev 2012; 21:2969-80. [PMID: 22563951 DOI: 10.1089/scd.2011.0588] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The therapeutic potential of bone marrow mesenchymal stem cells (MSCs) in kidney failure has been examined in some studies. However, recent findings indicate that after transplantation, these cells home to kidneys at very low levels. Interaction of stromal derived factor-1 (SDF-1) with its receptor, CXCR4, is of pivotal importance in migration and homing. Recently, CXCR7 has also been recognized as another SDF-1 receptor that interacts with CXCR4 and modulates its functions. In this study, CXCR4 and CXCR7 were separately and simultaneously overexpressed in BALB/c bone marrow MSCs by using a lentiviral vector system and the homing and renoprotective potentials of these cells were evaluated in a mouse model of cisplatin-induced acute kidney injury. Using flow cytometry, immunohistochemistry, and real-time PCR methods for detection of GFP-labeled MSCs, we found that although considerably entrapped in lungs, native MSCs home very rarely to kidneys and bone marrow and this rate cannot be significantly affected by CXCR4 and/or CXCR7 upregulation. Transplantation of neither native nor genetically engineered MSCs ameliorated kidney failure. We concluded that overexpression of CXCR4 and CXCR7 receptors in murine MSCs cannot improve the homing and therapeutic potentials of these cells and it can be due to severe chromosomal abnormalities that these cells bear during ex vivo expansion.
Collapse
Affiliation(s)
- Yousof Gheisari
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Stopp S, Bornhäuser M, Ugarte F, Wobus M, Kuhn M, Brenner S, Thieme S. Expression of the melanoma cell adhesion molecule in human mesenchymal stromal cells regulates proliferation, differentiation, and maintenance of hematopoietic stem and progenitor cells. Haematologica 2012; 98:505-13. [PMID: 22801967 DOI: 10.3324/haematol.2012.065201] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The melanoma cell adhesion molecule defines mesenchymal stromal cells in the human bone marrow that regenerate bone and establish a hematopoietic microenvironment in vivo. The role of the melanoma cell adhesion molecule in primary human mesenchymal stromal cells and the maintenance of hematopoietic stem and progenitor cells during ex vivo culture has not yet been demonstrated. We applied RNA interference or ectopic overexpression of the melanoma cell adhesion molecule in human mesenchymal stromal cells to evaluate the effect of the melanoma cell adhesion molecule on their proliferation and differentiation as well as its influence on co-cultivated hematopoietic stem and progenitor cells. Knockdown and overexpression of the melanoma cell adhesion molecule affected several characteristics of human mesenchymal stromal cells related to osteogenic differentiation, proliferation, and migration. Furthermore, knockdown of the melanoma cell adhesion molecule in human mesenchymal stromal cells stimulated the proliferation of hematopoietic stem and progenitor cells, and strongly reduced the formation of long-term culture-initiating cells. In contrast, melanoma cell adhesion molecule-overexpressing human mesenchymal stromal cells provided a supportive microenvironment for hematopoietic stem and progenitor cells. Expression of the melanoma cell adhesion molecule increased the adhesion of hematopoietic stem and progenitor cells to human mesenchymal stromal cells and their migration beneath the monolayer of human mesenchymal stromal cells. Our results demonstrate that the expression of the melanoma cell adhesion molecule in human mesenchymal stromal cells determines their fate and regulates the maintenance of hematopoietic stem and progenitor cells through direct cell-cell contact.
Collapse
Affiliation(s)
- Sabine Stopp
- Medical Clinic and Policlinic I, Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|