1
|
Taraballi F, Corbo C, Enterria-Rosales J, Martinez JO, Minardi S, Pandolfi L, Wang X, Tasciotti E, Singh KV, Arias CA, Corradetti B. Sugar Functionalized Collagen Material for Local Modulation of Innate Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415364. [PMID: 40411409 DOI: 10.1002/advs.202415364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/19/2025] [Indexed: 05/26/2025]
Abstract
Small alterations during the early stages of the innate immune response to an implant can drive large changes in adaptive immunity. Biomaterials for regenerative purposes can be engineered to modulate this immune response in beneficial ways. This study presents an innovative patch designed and functionalized to target the innate immunity at the implant site. Mannose moieties are incorporated into collagen patches, resulting in a technology called Local Immunotuning Patch (LIP), designed to directly interact with antigen presenting cells through their mannose receptor. In vitro, LIP shows anti-inflammatory effects on bone marrow-derived macrophages and inhibitory properties even on methicillin-resistant bacterial strains. Subcutaneous implantation in mice reveals that LIP modulates multiple pathways related to innate and adaptive immunity, underscoring its role in shaping an immune-engineered environment around the implant. These findings highlight the potential of this strategy to control the foreign body reaction at the implant site, making it applicable for various uses, including wound healing and surgical infection control in reconstructive procedures.
Collapse
Affiliation(s)
- Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Claudia Corbo
- School of Medicine and Surgery, Nanomedicine Center Nanomib, University of Milano-Bicocca, Monza, Italy
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | | | - John Otto Martinez
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Silvia Minardi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Laura Pandolfi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Xing Wang
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Ennio Tasciotti
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Human Longevity Program, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, Rome, Italy
| | - Kavindra V Singh
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, TX, USA
- Division of Infectious Diseases and Department of Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Cesar A Arias
- Center for Infectious Disease, Houston Methodist Research Institute, Houston, TX, USA
- Division of Infectious Diseases and Department of Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Bruna Corradetti
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section Oncology/Hematology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
2
|
Das P, Pal D, Roy S, Chaudhuri S, Kesh SS, Basak P, Nandi SK. Unveiling advanced strategies for therapeutic stem cell interventions in severe burn injuries: a comprehensive review. Int J Surg 2024; 110:6382-6401. [PMID: 38869979 PMCID: PMC11487052 DOI: 10.1097/js9.0000000000001812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
This comprehensive review explores the complex terrain of stem cell therapies as a potential therapeutic frontier in the healing of complicated burn wounds. Serious tissue damage, impaired healing processes, and possible long-term consequences make burn wounds a complex problem. An in-depth review is required since, despite medical progress, existing methods for treating severe burn wounds have significant limitations. Burn wounds are difficult to heal because they cause extensive tissue damage. The challenges of burn injury-induced tissue regeneration and functional recovery are also the subject of this review. Although there is a lot of promise in current stem cell treatments, there are also some limitations with scalability, finding the best way to transport the cells, and finding consistent results across different types of patients. To shed light on how to improve stem cell interventions to heal severe burn wounds, this review covers various stem cell applications in burn wounds and examines these obstacles. To overcome these obstacles, one solution is to enhance methods of stem cell distribution, modify therapies according to the severity of the burn, and conduct more studies on how stem cell therapy affects individual patients. Novel solutions may also be possible through the combination of cutting-edge technologies like nanotechnology and biotechnology. This review seeks to increase stem cell interventions by analyzing present challenges and suggesting strategic improvements. The goal is to provide a more effective and tailored way to repair serious burn wounds.
Collapse
Affiliation(s)
- Pratik Das
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences
- School of Bioscience and Engineering, Jadavpur University
| | - Debajyoti Pal
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences
| | - Sudipta Roy
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences
| | - Shubhamitra Chaudhuri
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - Shyam S. Kesh
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - Piyali Basak
- School of Bioscience and Engineering, Jadavpur University
| | - Samit K. Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences
| |
Collapse
|
3
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [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: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Muhuri AK, Alapan Y, Camargo CP, Thomas SN. Microengineered In Vitro Assays for Screening and Sorting Manufactured Therapeutic T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:199-207. [PMID: 38166247 PMCID: PMC10783858 DOI: 10.4049/jimmunol.2300488] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/10/2023] [Indexed: 01/04/2024]
Abstract
Adoptively transferred T cells constitute a major class of current and emergent cellular immunotherapies for the treatment of disease, including but not limited to cancer. Although key advancements in molecular recognition, genetic engineering, and manufacturing have dramatically enhanced their translational potential, therapeutic potency remains limited by poor homing and infiltration of transferred cells within target host tissues. In vitro microengineered homing assays with precise control over micromechanical and biological cues can address these shortcomings by enabling interrogation, screening, sorting, and optimization of therapeutic T cells based on their homing capacity. In this article, the working principles, application, and integration of microengineered homing assays for the mechanistic study of biophysical and biomolecular cues relevant to homing of therapeutic T cells are reviewed. The potential for these platforms to enable scalable enrichment and screening of next-generation manufactured T cell therapies for cancer is also discussed.
Collapse
Affiliation(s)
- Abir K. Muhuri
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
| | - Yunus Alapan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
| | - Camila P. Camargo
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
| | - Susan N. Thomas
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University
- Winship Cancer Institute, Emory University
| |
Collapse
|
5
|
Lopes-Pacheco M, Rocco PRM. Functional enhancement strategies to potentiate the therapeutic properties of mesenchymal stromal cells for respiratory diseases. Front Pharmacol 2023; 14:1067422. [PMID: 37007034 PMCID: PMC10062457 DOI: 10.3389/fphar.2023.1067422] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Respiratory diseases remain a major health concern worldwide because they subject patients to considerable financial and psychosocial burdens and result in a high rate of morbidity and mortality. Although significant progress has been made in understanding the underlying pathologic mechanisms of severe respiratory diseases, most therapies are supportive, aiming to mitigate symptoms and slow down their progressive course but cannot improve lung function or reverse tissue remodeling. Mesenchymal stromal cells (MSCs) are at the forefront of the regenerative medicine field due to their unique biomedical potential in promoting immunomodulation, anti-inflammatory, anti-apoptotic and antimicrobial activities, and tissue repair in various experimental models. However, despite several years of preclinical research on MSCs, therapeutic outcomes have fallen far short in early-stage clinical trials for respiratory diseases. This limited efficacy has been associated with several factors, such as reduced MSC homing, survival, and infusion in the late course of lung disease. Accordingly, genetic engineering and preconditioning methods have emerged as functional enhancement strategies to potentiate the therapeutic actions of MSCs and thus achieve better clinical outcomes. This narrative review describes various strategies that have been investigated in the experimental setting to functionally potentiate the therapeutic properties of MSCs for respiratory diseases. These include changes in culture conditions, exposure of MSCs to inflammatory environments, pharmacological agents or other substances, and genetic manipulation for enhanced and sustained expression of genes of interest. Future directions and challenges in efficiently translating MSC research into clinical practice are discussed.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- *Correspondence: Miquéias Lopes-Pacheco, ; Patricia R. M. Rocco,
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Miquéias Lopes-Pacheco, ; Patricia R. M. Rocco,
| |
Collapse
|
6
|
Achón Buil B, Tackenberg C, Rust R. Editing a gateway for cell therapy across the blood-brain barrier. Brain 2022; 146:823-841. [PMID: 36397727 PMCID: PMC9976985 DOI: 10.1093/brain/awac393] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Stem cell therapy has been shown to improve stroke outcomes in animal models and is currently advancing towards clinical practice. However, uncertainty remains regarding the optimal route for cell delivery to the injured brain. Local intracerebral injections are effective in precisely delivering cells into the stroke cavity but carry the risk of damaging adjacent healthy tissue. Systemic endovascular injections, meanwhile, are minimally invasive, but most injected cells do not cross CNS barriers and become mechanically trapped in peripheral organs. Although the blood-brain barrier and the blood-CSF barrier tightly limit the entrance of cells and molecules into the brain parenchyma, immune cells can cross these barriers especially under pathological conditions, such as stroke. Deciphering the cell surface signature and the molecular mechanisms underlying this pathophysiological process holds promise for improving the targeted delivery of systemic injected cells to the injured brain. In this review, we describe experimental approaches that have already been developed in which (i) cells are either engineered to express cell surface proteins mimicking infiltrating immune cells; or (ii) cell grafts are preconditioned with hypoxia or incubated with pharmacological agents or cytokines. Modified cell grafts can be complemented with strategies to temporarily increase the permeability of the blood-brain barrier. Although these approaches could significantly enhance homing of stem cells into the injured brain, cell entrapment in off-target organs remains a non-negligible risk. Recent developments in safety-switch systems, which enable the precise elimination of transplanted cells on the administration of a drug, represent a promising strategy for selectively removing stem cells stuck in untargeted organs. In sum, the techniques described in this review hold great potential to substantially improve efficacy and safety of future cell therapies in stroke and may be relevant to other brain diseases.
Collapse
Affiliation(s)
- Beatriz Achón Buil
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ruslan Rust
- Correspondence to: Ruslan Rust Institute for Regenerative Medicine Wagistrasse 12, 8952 Schlieren Zurich, Switzerland E-mail:
| |
Collapse
|
7
|
Khodabukus A, Guyer T, Moore AC, Stevens MM, Guldberg RE, Bursac N. Translating musculoskeletal bioengineering into tissue regeneration therapies. Sci Transl Med 2022; 14:eabn9074. [PMID: 36223445 PMCID: PMC7614064 DOI: 10.1126/scitranslmed.abn9074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Musculoskeletal injuries and disorders are the leading cause of physical disability worldwide and a considerable socioeconomic burden. The lack of effective therapies has driven the development of novel bioengineering approaches that have recently started to gain clinical approvals. In this review, we first discuss the self-repair capacity of the musculoskeletal tissues and describe causes of musculoskeletal dysfunction. We then review the development of novel biomaterial, immunomodulatory, cellular, and gene therapies to treat musculoskeletal disorders. Last, we consider the recent regulatory changes and future areas of technological progress that can accelerate translation of these therapies to clinical practice.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University; Durham, NC, 27708 USA
| | - Tyler Guyer
- Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, 97403 USA
| | - Axel C. Moore
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London; London, SW7 2AZ UK
- Department of Biomedical Engineering, University of Delaware; Newark, DE, 19716 USA
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London; London, SW7 2AZ UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institute; Stockholm, 17177 SE
| | - Robert E. Guldberg
- Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, 97403 USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University; Durham, NC, 27708 USA
| |
Collapse
|
8
|
Chan LY, Dass SA, Tye GJ, Imran SAM, Wan Kamarul Zaman WS, Nordin F. CAR-T Cells/-NK Cells in Cancer Immunotherapy and the Potential of MSC to Enhance Its Efficacy: A Review. Biomedicines 2022; 10:biomedicines10040804. [PMID: 35453554 PMCID: PMC9024487 DOI: 10.3390/biomedicines10040804] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/25/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
The chimeric antigen receptor (CAR) plays a dynamic role in targeting tumour-associated antigens in cancer cells. This novel therapeutic discovery combines fragments of monoclonal antibodies with the signalling and co-stimulatory domains that have been modified to its current fourth generation. CAR has been widely implemented in T-cells and natural killer (NK) cells immunotherapy. The significant advancement in CAR technology is evident based on numerous ongoing clinical trials on CAR-T/-NK cells and successful CAR-related products such as Kymriah (Novartis) and Yescarta (Kite Pharma, Gilead). Another important cell-based therapy is the engineering of mesenchymal stem cells (MSC). Researchers have been exploring MSCs and their innate homing abilities to tumour sites and secretion cytokines that bridge both CAR and MSC technologies as a therapeutic agent. This combination allows for both therapies to overcome each one’s flaw as an immunotherapy intervention. Herein, we have provided a concise review on the background of CAR and its applications in different cancers, as well as MSCs’ unique ability as delivery vectors for cancer therapy and the possibility of enhancing the CAR-immune cells’ activity. Hence, we have highlighted throughout this review the synergistic effects of both interventions.
Collapse
Affiliation(s)
- Ler Yie Chan
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (L.Y.C.); (S.A.M.I.)
- INTEC Education College, Jalan Senangin Satu 17/2A, Seksyen 17, Shah Alam 40200, Malaysia
| | - Sylvia Annabel Dass
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden 11800, Malaysia; (S.A.D.); (G.J.T.)
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden 11800, Malaysia; (S.A.D.); (G.J.T.)
| | - Siti A. M. Imran
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (L.Y.C.); (S.A.M.I.)
| | - Wan Safwani Wan Kamarul Zaman
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia; (L.Y.C.); (S.A.M.I.)
- Correspondence: ; Tel.: +60-3-91457670
| |
Collapse
|
9
|
Schrodt MV, Ankrum JA. Chemomechanically antifibrotic stromal cells. Nat Biomed Eng 2022; 6:6-7. [PMID: 35064245 DOI: 10.1038/s41551-021-00840-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Michael V Schrodt
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, USA
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA. .,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, USA.
| |
Collapse
|
10
|
Yao X, Ma Y, Zhou W, Liao Y, Jiang Z, Lin J, He Q, Wu H, Wei W, Wang X, Björklund M, Ouyang H. In-cytoplasm mitochondrial transplantation for mesenchymal stem cells engineering and tissue regeneration. Bioeng Transl Med 2022; 7:e10250. [PMID: 35111950 PMCID: PMC8780934 DOI: 10.1002/btm2.10250] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/15/2022] Open
Abstract
Stem cell therapies are unsatisfactory due to poor cell survival and engraftment. Stem cell used for therapy must be properly "tuned" for a harsh in vivo environment. Herein, we report that transfer of exogenous mitochondria (mito) to adipose-derived mesenchymal stem cells (ADSCs) can effectively boost their energy levels, enabling efficient cell engraftment. Importantly, the entire process of exogeneous mitochondrial endocytosis is captured by high-content live-cell imaging. Mitochondrial transfer leads to acutely enhanced bioenergetics, with nearly 17% of higher adenosine 5'-triphosphate (ATP) levels in ADSCs treated with high mitochondrial dosage and further results in altered secretome profiles of ADSCs. Mitochondrial transfer also induced the expression of 334 mRNAs in ADSCs, which are mainly linked to signaling pathways associated with DNA replication and cell division. We hypothesize that increase in ATP and cyclin-dependent kinase 1 and 2 expression might be responsible for promoting enhanced proliferation, migration, and differentiation of ADSCs in vitro. More importantly, mito-transferred ADSCs display prolonged cell survival, engraftment and horizontal transfer of exogenous mitochondria to surrounding cells in a full-thickness skin defect rat model with improved skin repair compared with nontreated ADSCs. These results demonstrate that intracellular mitochondrial transplantation is a promising strategy to engineer stem cells for tissue regeneration.
Collapse
Affiliation(s)
- Xudong Yao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- The Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Yuanzhu Ma
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Wenyan Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Youguo Liao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Zongsheng Jiang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Junxin Lin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Qiulin He
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Hongwei Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Wei Wei
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- The Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Xiaozhao Wang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Mikael Björklund
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- Department of Sports MedicineZhejiang University School of MedicineHangzhouChina
- China Orthopedic Regenerative Medicine Group (CORMed)HangzhouChina
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
11
|
Mercer-Smith AR, Findlay IA, Bomba HN, Hingtgen SD. Intravenously Infused Stem Cells for Cancer Treatment. Stem Cell Rev Rep 2021; 17:2025-2041. [PMID: 34138421 DOI: 10.1007/s12015-021-10192-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Despite the recent influx of immunotherapies and small molecule drugs to treat tumors, cancer remains a leading cause of death in the United States, in large part due to the difficulties of treating metastatic cancer. Stem cells, which are inherently tumoritropic, provide a useful drug delivery vehicle to target both primary and metastatic tumors. Intravenous infusions of stem cells carrying or secreting therapeutic payloads show significant promise in the treatment of cancer. Stem cells may be engineered to secrete cytotoxic products, loaded with oncolytic viruses or nanoparticles containing small molecule drugs, or conjugated with immunotherapies. Herein we describe these preclinical and clinical studies, discuss the distribution and migration of stem cells following intravenous infusion, and examine both the limitations of and the methods to improve the migration and therapeutic efficacy of tumoritropic, therapeutic stem cells.
Collapse
Affiliation(s)
- Alison R Mercer-Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Ingrid A Findlay
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Hunter N Bomba
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA. .,Department of Neurosurgery, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA.
| |
Collapse
|
12
|
Dollet PE, Hsu MJ, Ambroise J, Rozzi M, Ravau J, André F, Evraerts J, Najimi M, Sokal E, Lombard C. Evaluation of Strategies Aimed at Improving Liver Progenitor Cell Rolling and Subsequent Adhesion to the Endothelium. Cell Transplant 2021; 29:963689720912707. [PMID: 32425073 PMCID: PMC7444224 DOI: 10.1177/0963689720912707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Adult-derived human liver stem/progenitor cells (ADHLSCs) are a promising
alternative to orthotopic liver transplantation in the treatment of inborn
errors of metabolism. However, as is the case with many mesenchymal stromal
cells, ADHLSCs have shown a low level of engraftment, which could be explained
by the fact that they lack expression of selectin ligand and LFA-1 and only
slightly express VLA- 4, molecules that have been shown to be involved in cell
adhesion to the endothelium. In this paper, we have investigated strategies to
increase their rolling and adhesion during the homing process by (1) adding a
selectin ligand (Sialyl Lewis X) to their surface using
biotinyl-N-hydroxy-succinimide–streptavidin bridges, and
(2) protecting the adhesion proteins from trypsinization-induced damage using a
thermosensitive polymer for cell culture and a nonenzymatic cell dissociation
solution (CDS) for harvest. Despite increasing adhesion of ADHLSCs to E-selectin
during an adhesion test in vitro performed under shear stress,
the addition of Sialyl Lewis X did not increase adhesion to endothelial cells
under the same conditions. Cultivating cells on a thermosensitive polymer and
harvesting them with CDS increased their adhesion to endothelial cells under
noninflammatory conditions, compared to the use of trypsin. However, we were not
able to demonstrate any improvement in cell adhesion to the endothelium
following culture on polymer and harvest with CDS, suggesting that alternative
methods of improving engraftment still need to be evaluated.
Collapse
Affiliation(s)
- Pierre Edouard Dollet
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Mei Ju Hsu
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Jérôme Ambroise
- Centre de Technologies Moléculaires Appliquées (CTMA), Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Milena Rozzi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Joachim Ravau
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Floriane André
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Jonathan Evraerts
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Etienne Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Catherine Lombard
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| |
Collapse
|
13
|
Wechsler ME, Rao VV, Borelli AN, Anseth KS. Engineering the MSC Secretome: A Hydrogel Focused Approach. Adv Healthc Mater 2021; 10:e2001948. [PMID: 33594836 PMCID: PMC8035320 DOI: 10.1002/adhm.202001948] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/21/2021] [Indexed: 02/06/2023]
Abstract
The therapeutic benefits of exogenously delivered mesenchymal stromal/stem cells (MSCs) have been largely attributed to their secretory properties. However, clinical translation of MSC-based therapies is hindered due to loss of MSC regenerative properties during large-scale expansion and low survival/retention post-delivery. These limitations might be overcome by designing hydrogel culture platforms to modulate the MSC microenvironment. Hydrogel systems could be engineered to i) promote MSC proliferation and maintain regenerative properties (i.e., stemness and secretion) during ex vivo expansion, ii) improve MSC survival, retention, and engraftment in vivo, and/or iii) direct the MSC secretory profile using tailored biochemical and biophysical cues. Herein, it is reviewed how hydrogel material properties (i.e., matrix modulus, viscoelasticity, dimensionality, cell adhesion, and porosity) influence MSC secretion, mediated through cell-matrix and cell-cell interactions. In addition, it is highlighted how biochemical cues (i.e., small molecules, peptides, and proteins) can improve and direct the MSC secretory profile. Last, the authors' perspective is provided on future work toward the understanding of how microenvironmental cues influence the MSC secretome, and designing the next generation of biomaterials, with optimized biophysical and biochemical cues, to direct the MSC secretory profile for improved clinical translation outcomes.
Collapse
Affiliation(s)
- Marissa E Wechsler
- Department of Chemical and Biological Engineering, University of Colorado-Boulder, 3415 Colorado Avenue, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado-Boulder, 3415 Colorado Avenue, Boulder, CO, 80303, USA
| | - Varsha V Rao
- Department of Chemical and Biological Engineering, University of Colorado-Boulder, 3415 Colorado Avenue, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado-Boulder, 3415 Colorado Avenue, Boulder, CO, 80303, USA
| | - Alexandra N Borelli
- Department of Chemical and Biological Engineering, University of Colorado-Boulder, 3415 Colorado Avenue, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado-Boulder, 3415 Colorado Avenue, Boulder, CO, 80303, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado-Boulder, 3415 Colorado Avenue, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado-Boulder, 3415 Colorado Avenue, Boulder, CO, 80303, USA
| |
Collapse
|
14
|
Pozo D. Cell-based drug delivery harnesses inflammatory and autoimmune responses in neurodegeneration. J Mol Med (Berl) 2021; 99:673-674. [PMID: 33683375 PMCID: PMC7938677 DOI: 10.1007/s00109-021-02059-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 11/25/2022]
Affiliation(s)
- David Pozo
- Cellular and Molecular Neuroimmunology Laboratory, CABIMER, Andalusian Centre for Molecular Biology and Regenerative Medicine, CSIC-UPO-University of Seville, CABIMER Bldg. Av. Americo Vespucio, 24, Seville, Spain.
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Seville, Spain.
| |
Collapse
|
15
|
Levy O, Rothhammer V, Mascanfroni I, Tong Z, Kuai R, De Biasio M, Wang Q, Majid T, Perrault C, Yeste A, Kenison JE, Safaee H, Musabeyezu J, Heinelt M, Milton Y, Kuang H, Lan H, Siders W, Multon MC, Rothblatt J, Massadeh S, Alaamery M, Alhasan AH, Quintana FJ, Karp JM. A cell-based drug delivery platform for treating central nervous system inflammation. J Mol Med (Berl) 2021; 99:663-671. [PMID: 33398468 DOI: 10.1007/s00109-020-02003-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/22/2020] [Accepted: 10/29/2020] [Indexed: 01/18/2023]
Abstract
Mesenchymal stem cells (MSCs) are promising candidates for the development of cell-based drug delivery systems for autoimmune inflammatory diseases, such as multiple sclerosis (MS). Here, we investigated the effect of Ro-31-8425, an ATP-competitive kinase inhibitor, on the therapeutic properties of MSCs. Upon a simple pretreatment procedure, MSCs spontaneously took up and then gradually released significant amounts of Ro-31-8425. Ro-31-8425 (free or released by MSCs) suppressed the proliferation of CD4+ T cells in vitro following polyclonal and antigen-specific stimulation. Systemic administration of Ro-31-8425-loaded MSCs ameliorated the clinical course of experimental autoimmune encephalomyelitis (EAE), a murine model of MS, displaying a stronger suppressive effect on EAE than control MSCs or free Ro-31-8425. Ro-31-8425-MSC administration resulted in sustained levels of Ro-31-8425 in the serum of EAE mice, modulating immune cell trafficking and the autoimmune response during EAE. Collectively, these results identify MSC-based drug delivery as a potential therapeutic strategy for the treatment of autoimmune diseases. KEY MESSAGES: MSCs can spontaneously take up the ATP-competitive kinase inhibitor Ro-31-8425. Ro-31-8425-loaded MSCs gradually release Ro-31-8425 and exhibit sustained suppression of T cells. Ro-31-8425-loaded MSCs have more sustained serum levels of Ro-31-8425 than free Ro-31-8425. Ro-31-8425-loaded MSCs are more effective than MSCs and free Ro-31-8425 for EAE therapy.
Collapse
Affiliation(s)
- Oren Levy
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Veit Rothhammer
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ivan Mascanfroni
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhixiang Tong
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Rui Kuai
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael De Biasio
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Qingping Wang
- Department of Drug Metabolism and Pharmacokinetics, Sanofi R&D, Waltham, MA, USA
| | - Tahir Majid
- Global Research Program and Portfolio Management, Sanofi-Genzyme, Cambridge, MA, USA
| | - Christelle Perrault
- Sanofi R&D, In Vitro Pharmacology, Integrated Drug Discovery, Centre de Recherche Vitry-Alfortville, Vitry-Sur-Seine, France
| | - Ada Yeste
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica E Kenison
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Helia Safaee
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Juliet Musabeyezu
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Martina Heinelt
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Yuka Milton
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Heidi Kuang
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Haoyue Lan
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - William Siders
- Genzyme R&D, Neuroimmunology Research, Framingham, MA, USA
| | - Marie-Christine Multon
- Sanofi R&D, Translational Sciences, Centre de Recherche Vitry-Alfortville, Vitry-Sur-Seine, France
| | | | - Salam Massadeh
- Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Manal Alaamery
- Developmental Medicine Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Ali H Alhasan
- Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
- National Center of Pharmaceutical Technology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Jeffrey M Karp
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA.
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
16
|
Yu C, Ding S. Therapeutic strategies targeting somatic stem cells: Chemical approaches. Bioorg Med Chem 2020; 28:115824. [PMID: 33126088 DOI: 10.1016/j.bmc.2020.115824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 02/08/2023]
Abstract
Therapeutic modulation of fate and behavior of somatic stem cells can generate safe and functional cells ex vivo for cell-based therapy, or to repair and regenerate damaged tissues in vivo. Chemical approaches involving small molecules have provided promising approaches for modulating cellular fate and function. These strategies offer opportunities that support regenerative medicine. Here, we discuss strategies targeting somatic stem cells through chemical approaches, highlighting their progression as well as future prospects.
Collapse
Affiliation(s)
- Chen Yu
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Sheng Ding
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
17
|
Qi C, Jin Y, Chen Y, Li W, Li Y, Liang K, Li Y, Zhang Y, Du Y. TGase-mediated cell membrane modification and targeted cell delivery to inflammatory endothelium. Biomaterials 2020; 269:120276. [PMID: 32797997 DOI: 10.1016/j.biomaterials.2020.120276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/19/2020] [Accepted: 07/31/2020] [Indexed: 10/23/2022]
Abstract
Targeted cell delivery to lesion sites via minimally invasive approach remains an unmet need in regenerative medicine to endow controlled cell distribution and minimized side-effects. Current cell modification approaches to improve cell delivery tend to have adverse effects on cellular phenotype and functionality. Here, we rationally developed a facile and mild cell modification and targeted delivery strategy leveraging endogenous tissue transglutaminase (TGase) expressed on the surface of MSCs (Mesenchymal Stem Cells) and inflammatory endothelial cells (ECs). Cell modification by functional peptides was accomplished simply via TGase catalyzed cross-linking with naturally-expressed MSCs membrane proteins (e.g. Annexin II), without detectable disturbance of cellular viability and functionality. The modified functional peptides could facilitate adhesion of MSCs to inflammatory ECs (with up-regulated TGase expression compared with normal ECs) in vitro, as demonstrated by a one-fold increase of the MSC-EC adhesion force measured by atomic force microscopy (AFM) and by targeted delivery of modified MSC to inflammatory ECs in a flow chamber assay. When transplanted in vivo, modified MSCs demonstrated a dramatic increase in targeted efficiency to inflammatory endothelium compared with non-modified MSCs in both mice ear inflammation and acute/chronic liver injury models. The cell membrane modification strategy and targeted cell delivery mechanism described here can be readily extended for empowering cell engineering and cell therapy with multifaceted functionalities to combat refractory diseases.
Collapse
Affiliation(s)
- Chunxiao Qi
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China; Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yuhong Jin
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuyang Chen
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjing Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yaqian Li
- Central Laboratories, Department of Scientific Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Kai Liang
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yan Li
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yonghui Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
18
|
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
|
19
|
Melzer C, Jacobs R, Dittmar T, Pich A, von der Ohe J, Yang Y, Hass R. Reversible Growth-Arrest of a Spontaneously-Derived Human MSC-Like Cell Line. Int J Mol Sci 2020; 21:4752. [PMID: 32635395 PMCID: PMC7369918 DOI: 10.3390/ijms21134752] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Life cycle limitation hampers the production of high amounts of primary human mesenchymal stroma-/stem-like cells (MSC) and limits cell source reproducibility for clinical applications. The characterization of permanently growing MSC544 revealed some differentiation capacity and the simultaneous presence of known MSC markers CD73, CD90, and CD105 even after continuous long-term culture for more than one year and 32 passages. The expression of CD13, CD29, CD44, and CD166 were identified as further surface proteins, all of which were also simultaneously detectable in various other types of primary MSC populations derived from the umbilical cord, bone marrow, and placenta suggesting MSC-like properties in the cell line. Proliferating steady state MSC544 exhibited immune-modulatory activity similar to a subpopulation of long-term growth-inhibited MSC544 after 189d of continuous culture in confluency. This confluent connective cell layer with fibroblast-like morphology can spontaneously contract and the generated space is subsequently occupied by new cells with regained proliferative capacity. Accordingly, the confluent and senescence-associated beta-galactosidase-positive MSC544 culture with about 95% G0/G1 growth-arrest resumed re-entry into the proliferative cell cycle within 3d after sub-confluent culture. The MSC544 cells remained viable during confluency and throughout this transition which was accompanied by marked changes in the release of proteins. Thus, expression of proliferation-associated genes was down-modulated in confluent MSC544 and re-expressed following sub-confluent conditions whilst telomerase (hTERT) transcripts remained detectable at similar levels in both, confluent growth-arrested and proliferating MSC544. Together with the capability of connective cell layer formation for potential therapeutic approaches, MSC544 provide a long term reproducible human cell source with constant properties.
Collapse
Affiliation(s)
- Catharina Melzer
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany; (C.M.); (J.v.d.O.); (Y.Y.)
| | - Roland Jacobs
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, 30625 Hannover, Germany;
| | - Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany;
| | - Andreas Pich
- Department of Toxicology, Hannover Medical School, 30625 Hannover, Germany;
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany; (C.M.); (J.v.d.O.); (Y.Y.)
| | - Yuanyuan Yang
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany; (C.M.); (J.v.d.O.); (Y.Y.)
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany; (C.M.); (J.v.d.O.); (Y.Y.)
| |
Collapse
|
20
|
AbuElela AF, Al-Amoodi AS, Ali AJ, Merzaban JS. Fluorescent Multiplex Cell Rolling Assay: Simultaneous Capturing up to Seven Samples in Real-Time Using Spectral Confocal Microscopy. Anal Chem 2020; 92:6200-6206. [PMID: 32264668 DOI: 10.1021/acs.analchem.9b04549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The parallel plate flow chamber assay is widely utilized to study physiological cell-cell adhesive interactions under dynamic flow that mimics the bloodstream. In this technique, the cells are perfused under defined shear stresses over a monolayer of endothelial cells (expressing homing molecules, e.g., selectins) or a surface (expressing recombinant homing molecules). However, with the need to study multiple samples and multiple parameters per sample, using a traditional bright-field microscope-based flow assay allows only one sample at a time to be analyzed, resulting in high interexperiment variability, the need for normalization, waste of materials, and significant consumption of time. We developed a multiplexing approach using a three-color fluorescence staining method, which allowed for up to seven different combination signatures to be run at one time. Using this fluorescent multiplex cell rolling (FMCR) assay, each sample is labeled with a different signature of emission wavelengths and mixed with other samples just minutes before the flow run. Subsequently, real-time images are acquired in a single pass using a line-scanning spectral confocal microscope. To illustrate the glycan-dependent binding of E-selectin, a central molecule in cell migration, to its glycosylated ligands expressed on myeloid-leukemic cells in flow, the FMCR assay was used to analyze E-selectin-ligand interactions following the addition (fucosyltransferase-treatment) or removal (deglycosylation) of key glycans on the flowing cells. The FMCR assay allowed us to analyze the cell-adhesion events from these different treatment conditions simultaneously in a competitive manner and to calculate differences in rolling frequency, velocity, and tethering capability of cells under study.
Collapse
Affiliation(s)
- Ayman F AbuElela
- Cell Migration and Signaling Laboratory, King Abdullah University of Science and Technology, Biological and Environmental Sciences and Engineering Division, Thuwal, Saudi Arabia
| | - Asma S Al-Amoodi
- Cell Migration and Signaling Laboratory, King Abdullah University of Science and Technology, Biological and Environmental Sciences and Engineering Division, Thuwal, Saudi Arabia
| | - Amal J Ali
- Cell Migration and Signaling Laboratory, King Abdullah University of Science and Technology, Biological and Environmental Sciences and Engineering Division, Thuwal, Saudi Arabia
| | - Jasmeen S Merzaban
- Cell Migration and Signaling Laboratory, King Abdullah University of Science and Technology, Biological and Environmental Sciences and Engineering Division, Thuwal, Saudi Arabia
| |
Collapse
|
21
|
Interleukin-1 β Enhances Umbilical Cord Mesenchymal Stem Cell Adhesion Ability on Human Umbilical Vein Endothelial Cells via LFA-1/ICAM-1 Interaction. Stem Cells Int 2019; 2019:7267142. [PMID: 31949440 PMCID: PMC6948307 DOI: 10.1155/2019/7267142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/13/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022] Open
Abstract
The migration of administered mesenchymal stem cells (MSCs) to sites of injury via the bloodstream has been demonstrated. However, the underlying mechanisms of umbilical cord MSC adhesion to endothelial cells during transendothelial migration are still unclear. In this study, our data showed that IL-1β induced LFA-1 expression on MSCs and ICAM-1 expression on HUVECs. We then pretreated MSCs with protein synthesis inhibitor cycloheximide. The results showed that IL-1β induced LFA-1 expression on the surface of MSCs via the protein synthesis pathway. Through the p38 MAPK signaling pathway inhibitor SB 203580, we found that IL-1β induces the expression of LFA-1 through p38 MAPK signaling and enhances ICAM-1 expression in HUVECs. In addition, IL-1β-induced MSC adhesion to HUVECs was found to be inhibited by IL-1RA and the LFA-1 inhibitor lovastatin. These results indicate that IL-1β promotes the cell adhesion of MSCs to HUVECs through LFA-1/ICAM-1 interaction. We address the evidence that the cell adhesion mechanism of IL-1β promotes MSC adhesion to HUVECs. The implications of these findings could enhance the therapeutic potential of MSCs.
Collapse
|
22
|
Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019; 4:22. [PMID: 31815001 PMCID: PMC6889290 DOI: 10.1038/s41536-019-0083-6] [Citation(s) in RCA: 1210] [Impact Index Per Article: 201.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
The terms MSC and MSCs have become the preferred acronym to describe a cell and a cell population of multipotential stem/progenitor cells commonly referred to as mesenchymal stem cells, multipotential stromal cells, mesenchymal stromal cells, and mesenchymal progenitor cells. The MSCs can differentiate to important lineages under defined conditions in vitro and in limited situations after implantation in vivo. MSCs were isolated and described about 30 years ago and now there are over 55,000 publications on MSCs readily available. Here, we have focused on human MSCs whenever possible. The MSCs have broad anti-inflammatory and immune-modulatory properties. At present, these provide the greatest focus of human MSCs in clinical testing; however, the properties of cultured MSCs in vitro suggest they can have broader applications. The medical utility of MSCs continues to be investigated in over 950 clinical trials. There has been much progress in understanding MSCs over the years, and there is a strong foundation for future scientific research and clinical applications, but also some important questions remain to be answered. Developing further methods to understand and unlock MSC potential through intracellular and intercellular signaling, biomedical engineering, delivery methods and patient selection should all provide substantial advancements in the coming years and greater clinical opportunities. The expansive and growing field of MSC research is teaching us basic human cell biology as well as how to use this type of cell for cellular therapy in a variety of clinical settings, and while much promise is evident, careful new work is still needed.
Collapse
|
23
|
Noronha NDC, Mizukami A, Caliári-Oliveira C, Cominal JG, Rocha JLM, Covas DT, Swiech K, Malmegrim KCR. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther 2019; 10:131. [PMID: 31046833 PMCID: PMC6498654 DOI: 10.1186/s13287-019-1224-y] [Citation(s) in RCA: 384] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSC) have been widely explored for cell-based therapy of immune-mediated, inflammatory, and degenerative diseases, due to their immunosuppressive, immunomodulatory, and regenerative potentials. Preclinical studies and clinical trials have demonstrated promising therapeutic results although these have been somewhat limited. Aspects such as low in vivo MSC survival in inhospitable disease microenvironments, requirements for ex vivo cell overexpansion prior to infusions, intrinsic differences between MSC and different sources and donors, variability of culturing protocols, and potency assays to evaluate MSC products have been described as limitations in the field. In recent years, priming approaches to empower MSC have been investigated, thereby generating cellular products with improved potential for different clinical applications. Herein, we review the current priming approaches that aim to increase MSC therapeutic efficacy. Priming with cytokines and growth factors, hypoxia, pharmacological drugs, biomaterials, and different culture conditions, as well as other diverse molecules, are revised from current and future perspectives.
Collapse
Affiliation(s)
- Nádia de Cássia Noronha
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Graduate Program on Bioscience and Biotechnology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Amanda Mizukami
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Juçara Gastaldi Cominal
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Graduate Program on Bioscience and Biotechnology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - José Lucas M Rocha
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Graduate Program on Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kamilla Swiech
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen C R Malmegrim
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil. .,Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/n°, Ribeirão Preto, SP, 14010-903, Brazil.
| |
Collapse
|
24
|
Chen L, Shi K, Andersen TL, Qiu W, Kassem M. KIAA1199 is a secreted molecule that enhances osteoblastic stem cell migration and recruitment. Cell Death Dis 2019; 10:126. [PMID: 30755597 PMCID: PMC6372631 DOI: 10.1038/s41419-018-1202-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022]
Abstract
Factors mediating mobilization of osteoblastic stem and progenitor cells from their bone marrow niche to be recruited to bone formation sites during bone remodeling are poorly known. We have studied secreted factors present in the bone marrow microenvironment and identified KIAA1199 (also known as CEMIP, cell migration inducing hyaluronan binding protein) in human bone biopsies as highly expressed in osteoprogenitor reversal cells (Rv.C) recruited to the eroded surfaces (ES), which are the future bone formation sites. In vitro, KIAA1199 did not affect the proliferation of human osteoblastic stem cells (also known as human bone marrow skeletal or stromal stem cells, hMSCs); but it enhanced cell migration as determined by scratch assay and trans-well migration assay. KIAA1199 deficient hMSCs (KIAA1199down) exhibited significant changes in cell size, cell length, ratio of cell width to length and cell roundness, together with reduction of polymerization actin (F-actin) and changes in phos-CFL1 (cofflin1), phos-LIMK1 (LIM domain kinase 1) and DSTN (destrin), key factors regulating actin cytoskeletal dynamics and cell motility. Moreover, KIAA1199down hMSC exhibited impaired Wnt signaling in TCF-reporter assay and decreased expression of Wnt target genes and these effects were rescued by KIAA1199 treatment. Finally, KIAA1199 regulated the activation of P38 kinase and its associated changes in Wnt-signaling. Thus, KIAA1199 is a mobilizing factor that interacts with P38 and Wnt signaling, and induces changes in actin cytoskeleton, as a mechanism mediating recruitment of hMSC to bone formation sites.
Collapse
Affiliation(s)
- Li Chen
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark.
| | - Kaikai Shi
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology, Vejle Hospital-Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100, Vejle, Denmark
| | - Weimin Qiu
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark.
- The Danish Stem Cell Center (DanStem), University of Copenhagen, 2200, Copenhagen, Denmark.
- Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
25
|
Schweizer MT, Wang H, Bivalacqua TJ, Partin AW, Lim SJ, Chapman C, Abdallah R, Levy O, Bhowmick NA, Karp JM, De Marzo A, Isaacs JT, Brennen WN, Denmeade SR. A Phase I Study to Assess the Safety and Cancer-Homing Ability of Allogeneic Bone Marrow-Derived Mesenchymal Stem Cells in Men with Localized Prostate Cancer. Stem Cells Transl Med 2019; 8:441-449. [PMID: 30735000 PMCID: PMC6477003 DOI: 10.1002/sctm.18-0230] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022] Open
Abstract
Animal models show that systemically administered bone marrow‐derived mesenchymal stem cells (MSCs) home to sites of primary and metastatic prostate cancer (PC)—making them candidates to selectively deliver cytotoxic agents. To further assess this potential as a cell‐based therapeutic vehicle, a phase I study testing homing of systemically infused allogeneic MSCs preprostatectomy was conducted. The primary objective was to assess safety and feasibility and to determine if MSCs accumulate within primary PC tissue. MSCs were quantified using beads, emulsion, amplification, magnetics digital polymerase chain reaction (limit of detection: ≥0.01% MSCs) to measure allogeneic MSC DNA relative to recipient DNA. MSCs were harvested from healthy donors and expanded ex vivo using standard protocols by the Johns Hopkins Cell Therapy Laboratory. PC patients planning to undergo prostatectomy were eligible for MSC infusion. Enrolled subjects received a single intravenous infusion 4–6 days prior to prostatectomy. The first three subjects received 1 x 106 cells per kilogram (maximum 1 x 108 cells), and subsequent four patients received 2 x 106 cells per kilogram (maximum 2 x 108 cells). No dose‐limiting toxicities were observed and all patients underwent prostatectomy without delay. Pathologic assessment of prostate cores revealed ≥70% tumor involvement in cores from four subjects, with benign tissue in the others. MSCs were undetectable in all subjects, and the study was stopped early for futility. MSC infusions appear safe in PC patients. Although intended for eventual use in metastatic PC patients, in this study, MSCs did not home primary tumors in sufficient levels to warrant further development as a cell‐based therapeutic delivery strategy using standard ex vivo expansion protocols. stem cells translational medicine2019;8:441–449
Collapse
Affiliation(s)
- Michael T Schweizer
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Hao Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Trinity J Bivalacqua
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alan W Partin
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Su Jin Lim
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Carolyn Chapman
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rehab Abdallah
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Oren Levy
- Center for Nanomedicine and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Neil A Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jeffrey M Karp
- Center for Nanomedicine and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Angelo De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John T Isaacs
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - W Nathaniel Brennen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samuel R Denmeade
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Manufacturing of primed mesenchymal stromal cells for therapy. Nat Biomed Eng 2019; 3:90-104. [PMID: 30944433 DOI: 10.1038/s41551-018-0325-8] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022]
Abstract
Mesenchymal stromal cells (MSCs) for basic research and clinical applications are manufactured and developed as unique cell products by many different manufacturers and laboratories, often under different conditions. The lack of standardization of MSC identity has limited consensus around which MSC properties are relevant for specific outcomes. In this Review, we examine how the choice of media, cell source, culture environment and storage affects the phenotype and clinical utility of MSC-based products, and discuss the techniques better suited to prime MSCs with specific phenotypes of interest and the need for the continued development of standardized assays that provide quality assurance for clinical-grade MSCs. Bioequivalence between cell products and batches must be investigated rather than assumed, so that the diversity of phenotypes between differing MSC products can be accounted for to identify products with the highest therapeutic potential and to preserve their safety in clinical treatments.
Collapse
|
27
|
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: 53] [Impact Index Per Article: 8.8] [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
|
28
|
Jafari A, Isa A, Chen L, Ditzel N, Zaher W, Harkness L, Johnsen HE, Abdallah BM, Clausen C, Kassem M. TAFA2 Induces Skeletal (Stromal) Stem Cell Migration Through Activation of Rac1-p38 Signaling. Stem Cells 2018; 37:407-416. [PMID: 30485583 PMCID: PMC7379704 DOI: 10.1002/stem.2955] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/17/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022]
Abstract
Understanding the mechanisms regulating recruitment of human skeletal (stromal or mesenchymal) stem cells (hMSC) to sites of tissue injury is a prerequisite for their successful use in cell replacement therapy. Chemokine‐like protein TAFA2 is a recently discovered neurokine involved in neuronal cell migration and neurite outgrowth. Here, we demonstrate a possible role for TAFA2 in regulating recruitment of hMSC to bone fracture sites. TAFA2 increased the in vitro trans‐well migration and motility of hMSC in a dose‐dependent fashion and induced significant morphological changes including formation of lamellipodia as revealed by high‐content‐image analysis at single‐cell level. Mechanistic studies revealed that TAFA2 enhanced hMSC migration through activation of the Rac1‐p38 pathway. In addition, TAFA2 enhanced hMSC proliferation, whereas differentiation of hMSC toward osteoblast and adipocyte lineages was not altered. in vivo studies demonstrated transient upregulation of TAFA2 gene expression during the inflammatory phase of fracture healing in a closed femoral fracture model in mice, and a similar pattern was observed in serum levels of TAFA2 in patients after hip fracture. Finally, interleukin‐1β was found as an upstream regulator of TAFA2 expression. Our findings demonstrate that TAFA2 enhances hMSC migration and recruitment and thus is relevant for regenerative medicine applications. Stem Cells2019;37:407–416
Collapse
Affiliation(s)
- Abbas Jafari
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.,Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Adiba Isa
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Li Chen
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Nicholas Ditzel
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Walid Zaher
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark.,Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Linda Harkness
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Hans E Johnsen
- Department of Haematology, Aalborg University, Aalborg, Denmark
| | - Basem M Abdallah
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark.,Biological Sciences Department, College of Science, King Faisal University, Hofuf, Saudi Arabia
| | | | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.,Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark.,Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
29
|
Krueger TEG, Thorek DLJ, Denmeade SR, Isaacs JT, Brennen WN. Concise Review: Mesenchymal Stem Cell-Based Drug Delivery: The Good, the Bad, the Ugly, and the Promise. Stem Cells Transl Med 2018; 7:651-663. [PMID: 30070053 PMCID: PMC6127224 DOI: 10.1002/sctm.18-0024] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/15/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
The development of mesenchymal stem cells (MSCs) as cell‐based drug delivery vectors for numerous clinical indications, including cancer, has significant promise. However, a considerable challenge for effective translation of these approaches is the limited tumor tropism and broad biodistribution observed using conventional MSCs, which raises concerns for toxicity to nontarget peripheral tissues (i.e., the bad). Consequently, there are a variety of synthetic engineering platforms in active development to improve tumor‐selective targeting via increased homing efficiency and/or specificity of drug activation, some of which are already being evaluated clinically (i.e., the good). Unfortunately, the lack of robust quantification and widespread adoption of standardized methodologies with high sensitivity and resolution has made accurate comparisons across studies difficult, which has significantly impeded progress (i.e., the ugly). Herein, we provide a concise review of active and passive MSC homing mechanisms and biodistribution postinfusion; in addition to in vivo cell tracking methodologies and strategies to enhance tumor targeting with a focus on MSC‐based drug delivery strategies for cancer therapy. Stem Cells Translational Medicine2018;1–13
Collapse
Affiliation(s)
- Timothy E G Krueger
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel L J Thorek
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins, Baltimore, Maryland, USA
| | - Samuel R Denmeade
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins, Baltimore, Maryland, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John T Isaacs
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins, Baltimore, Maryland, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - W Nathaniel Brennen
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Ranganath SH. Bioengineered cellular and cell membrane-derived vehicles for actively targeted drug delivery: So near and yet so far. Adv Drug Deliv Rev 2018; 132:57-80. [PMID: 29935987 DOI: 10.1016/j.addr.2018.06.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/31/2018] [Accepted: 06/18/2018] [Indexed: 12/16/2022]
Abstract
Cellular carriers for drug delivery are attractive alternatives to synthetic nanoparticles owing to their innate homing/targeting abilities. Here, we review molecular interactions involved in the homing of Mesenchymal stem cells (MSCs) and other cell types to understand the process of designing and engineering highly efficient, actively targeting cellular vehicles. In addition, we comprehensively discuss various genetic and non-genetic strategies and propose futuristic approaches of engineering MSC homing using micro/nanotechnology and high throughput small molecule screening. Most of the targeting abilities of a cell come from its plasma membrane, thus, efforts to harness cell membranes as drug delivery vehicles are gaining importance and are highlighted here. We also recognize and report the lack of detailed characterization of cell membranes in terms of safety, structural integrity, targeting functionality, and drug transport. Finally, we provide insights on future development of bioengineered cellular and cell membrane-derived vesicles for successful clinical translation.
Collapse
Affiliation(s)
- Sudhir H Ranganath
- Bio-INvENT Lab, Department of Chemical Engineering, Siddaganga Institute of Technology, B.H. Road, Tumakuru, 572103, Karnataka, India.
| |
Collapse
|
31
|
Coatti GC, Frangini M, Valadares MC, Gomes JP, Lima NO, Cavaçana N, Assoni AF, Pelatti MV, Birbrair A, de Lima ACP, Singer JM, Rocha FMM, Da Silva GL, Mantovani MS, Macedo-Souza LI, Ferrari MFR, Zatz M. Pericytes Extend Survival of ALS SOD1 Mice and Induce the Expression of Antioxidant Enzymes in the Murine Model and in IPSCs Derived Neuronal Cells from an ALS Patient. Stem Cell Rev Rep 2018; 13:686-698. [PMID: 28710685 DOI: 10.1007/s12015-017-9752-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is one of the most common adult-onset motor neuron disease causing a progressive, rapid and irreversible degeneration of motor neurons in the cortex, brain stem and spinal cord. No effective treatment is available and cell therapy clinical trials are currently being tested in ALS affected patients. It is well known that in ALS patients, approximately 50% of pericytes from the spinal cord barrier are lost. In the central nervous system, pericytes act in the formation and maintenance of the blood-brain barrier, a natural defense that slows the progression of symptoms in neurodegenerative diseases. Here we evaluated, for the first time, the therapeutic effect of human pericytes in vivo in SOD1 mice and in vitro in motor neurons and other neuronal cells derived from one ALS patient. Pericytes and mesenchymal stromal cells (MSCs) were derived from the same adipose tissue sample and were administered to SOD1 mice intraperitoneally. The effect of the two treatments was compared. Treatment with pericytes extended significantly animals survival in SOD1 males, but not in females that usually have a milder phenotype with higher survival rates. No significant differences were observed in the survival of mice treated with MSCs. Gene expression analysis in brain and spinal cord of end-stage animals showed that treatment with pericytes can stimulate the host antioxidant system. Additionally, pericytes induced the expression of SOD1 and CAT in motor neurons and other neuronal cells derived from one ALS patient carrying a mutation in FUS. Overall, treatment with pericytes was more effective than treatment with MSCs. Our results encourage further investigations and suggest that pericytes may be a good option for ALS treatment in the future. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Giuliana Castello Coatti
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Miriam Frangini
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Marcos C Valadares
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Juliana Plat Gomes
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Natalia O Lima
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Natale Cavaçana
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Amanda F Assoni
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Mayra V Pelatti
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Alexander Birbrair
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pathology, University Federal of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Julio M Singer
- Department of Statistics, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | | | | | | | - Lucia Inês Macedo-Souza
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Merari F R Ferrari
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil
| | - Mayana Zatz
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of Sao Paulo (USP), Rua do Matāo 106, São Paulo, SP, CEP 05508-030, Brazil.
| |
Collapse
|
32
|
Li X, He X, Yin Y, Wu R, Tian B, Chen F. Administration of signalling molecules dictates stem cell homing for in situ regeneration. J Cell Mol Med 2017; 21:3162-3177. [PMID: 28767189 PMCID: PMC5706509 DOI: 10.1111/jcmm.13286] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 05/29/2017] [Indexed: 12/13/2022] Open
Abstract
Ex vivo-expanded stem cells have long been a cornerstone of biotherapeutics and have attracted increasing attention for treating intractable diseases and improving tissue regeneration. However, using exogenous cellular materials to develop restorative treatments for large numbers of patients has become a major concern for both economic and safety reasons. Advances in cell biological research over the past two decades have expanded the potential for using endogenous stem cells during wound healing processes, and in particular, recent insight into stem cell movement and homing has prompted regenerative research and therapy based on recruiting endogenous cells. Inspired by the natural healing process, artificial administration of specific chemokines as signals systemically or at the injury site, typically using biomaterials as vehicles, is a state-of-the-art strategy that potentiates stem cell homing and recreates an anti-inflammatory and immunomodulatory microenvironment to enhance in situ tissue regeneration. However, pharmacologically coaxing endogenous stem cells to act as therapeutics in the field of biomedicine remains in the early stages; its efficacy is limited by the lack of innovative methodologies for chemokine presentation and release. This review describes how to direct the homing of endogenous stem cells via the administration of specific signals, with a particular emphasis on targeted signalling molecules that regulate this homing process, to enhance in situ tissue regeneration. We also provide an outlook on and critical considerations for future investigations to enhance stem cell recruitment and harness the reparative potential of these recruited cells as a clinically relevant cell therapy.
Collapse
Affiliation(s)
- Xuan Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Xiao‐Tao He
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Yuan Yin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Rui‐Xin Wu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Bei‐Min Tian
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Fa‐Ming Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| |
Collapse
|
33
|
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: 67] [Impact Index Per Article: 8.4] [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
|
34
|
Cortese FAB, Aguiar S, Santostasi G. Induced Cell Turnover: A Novel Therapeutic Modality for In Situ Tissue Regeneration. Hum Gene Ther 2017; 28:703-716. [PMID: 28557533 DOI: 10.1089/hum.2016.167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Induced cell turnover (ICT) is a theoretical intervention in which the targeted ablation of damaged, diseased, and/or nonfunctional cells is coupled with replacement by partially differentiated induced pluripotent stem cells in a gradual and multiphasic manner. Tissue-specific ablation can be achieved using pro-apoptotic small molecule cocktails, peptide mimetics, and/or tissue-tropic adeno-associated virus-delivered suicide genes driven by cell type-specific promoters. Replenishment with new cells can be mediated by systemic administration of cells engineered for homing, robustness, and even enhanced function and disease resistance. Otherwise, the controlled release of cells can be achieved using implanted biodegradable scaffolds, hydrogels, and polymer matrixes. In theory, ICT would enable in situ tissue regeneration without the need for surgical transplantation of organs produced ex vivo, and addresses non-transplantable tissues (such as the vasculature, lymph nodes, and the nervous system). This article outlines several complimentary strategies for overcoming barriers to ICT in an effort to stimulate further research at this promising interface of cell therapy, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Francesco Albert Bosco Cortese
- 1 Biogerontology Research Foundation, Oxford, United Kingdom .,2 Department of Biomedical and Molecular Sciences, Queen's University School of Medicine, Queen's University, Kingston, Canada
| | - Sebastian Aguiar
- 3 Neurobiology Department, Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Giovanni Santostasi
- 4 Department of Neurology, Feinberg School of Medicine, Northwestern University , Chicago, Illinois
| |
Collapse
|
35
|
Nitzsche F, Müller C, Lukomska B, Jolkkonen J, Deten A, Boltze J. Concise Review: MSC Adhesion Cascade-Insights into Homing and Transendothelial Migration. Stem Cells 2017; 35:1446-1460. [DOI: 10.1002/stem.2614] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/13/2017] [Accepted: 02/23/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Franziska Nitzsche
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology, McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh Pennsylvania USA
| | - Claudia Müller
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Barbara Lukomska
- NeuroRepair Department; Mossakowski Medical Research Centre; Warsaw Poland
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern; Kuopio Finland
| | - Alexander Deten
- Translational Centre for Regenerative Medicine, Leipzig University; Leipzig Germany
| | - Johannes Boltze
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Translational Centre for Regenerative Medicine, Leipzig University; Leipzig Germany
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| |
Collapse
|
36
|
Pacioni S, D'Alessandris QG, Giannetti S, Morgante L, Coccè V, Bonomi A, Buccarelli M, Pascucci L, Alessandri G, Pessina A, Ricci-Vitiani L, Falchetti ML, Pallini R. Human mesenchymal stromal cells inhibit tumor growth in orthotopic glioblastoma xenografts. Stem Cell Res Ther 2017; 8:53. [PMID: 28279193 PMCID: PMC5345323 DOI: 10.1186/s13287-017-0516-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 01/27/2017] [Accepted: 02/21/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Mesenchymal stem/stromal cells (MSCs) represent an attractive tool for cell-based cancer therapy mainly because of their ability to migrate to tumors and to release bioactive molecules. However, the impact of MSCs on tumor growth has not been fully established. We previously demonstrated that murine MSCs show a strong tropism towards glioblastoma (GBM) brain xenografts and that these cells are able to uptake and release the chemotherapeutic drug paclitaxel (PTX), maintaining their tropism towards the tumor. Here, we address the therapy-relevant issue of using MSCs from human donors (hMSCs) for local or systemic administration in orthotopic GBM models, including xenografts of patient-derived glioma stem cells (GSCs). METHODS U87MG or GSC1 cells expressing the green fluorescent protein (GFP) were grafted onto the striatum of immunosuppressed rats. Adipose hMSCs (Ad-hMSCs), fluorescently labeled with the mCherry protein, were inoculated adjacent to or into the tumor. In rats bearing U87MG xenografts, systemic injections of Ad-hMSCs or bone marrow (BM)-hMSCs were done via the femoral vein or carotid artery. In each experiment, either PTX-loaded or unloaded hMSCs were used. To characterize the effects of hMSCs on tumor growth, we analyzed survival, tumor volume, tumor cell proliferation, and microvascular density. RESULTS Overall, the AD-hMSCs showed remarkable tropism towards the tumor. Intracerebral injection of Ad-hMSCs significantly improved the survival of rats with U87MG xenografts. This effect was associated with a reduction in tumor growth, tumor cell proliferation, and microvascular density. In GSC1 xenografts, intratumoral injection of Ad-hMSCs depleted the tumor cell population and induced migration of resident microglial cells. Overall, PTX loading did not significantly enhance the antitumor potential of hMSCs. Systemically injected Ad- and BM-hMSCs homed to tumor xenografts. The efficiency of hMSC homing ranged between 0.02 and 0.5% of the injected cells, depending both on the route of cell injection and on the source from which the hMSCs were derived. Importantly, systemically injected PTX-loaded hMSCs that homed to the xenograft induced cytotoxic damage to the surrounding tumor cells. CONCLUSIONS hMSCs have a therapeutic potential in GBM brain xenografts which is also expressed against the GSC population. In this context, PTX loading of hMSCs seems to play a minor role.
Collapse
Affiliation(s)
- Simone Pacioni
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy.,CNR-Institute of Cell Biology and Neurobiology (IBCN), Rome, Italy
| | | | - Stefano Giannetti
- Institute of Anatomy, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Liliana Morgante
- Institute of Anatomy, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Arianna Bonomi
- Department of Cerebrovascular Diseases, Fondazione IRCCS Neurological Institute Carlo Besta, Milan, Italy
| | - Mariachiara Buccarelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Giulio Alessandri
- Department of Cerebrovascular Diseases, Fondazione IRCCS Neurological Institute Carlo Besta, Milan, Italy
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Lucia Ricci-Vitiani
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Roberto Pallini
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
37
|
Liu Z, Lu Y, Xiao Y, Lu Y. Upregulation of miR-21 expression is a valuable predicator of advanced clinicopathological features and poor prognosis in patients with renal cell carcinoma through the p53/p21-cyclin E2-Bax/caspase-3 signaling pathway. Oncol Rep 2017; 37:1437-1444. [DOI: 10.3892/or.2017.5402] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 02/14/2016] [Indexed: 11/06/2022] Open
|
38
|
Yang Z, Concannon J, Ng KS, Seyb K, Mortensen LJ, Ranganath S, Gu F, Levy O, Tong Z, Martyn K, Zhao W, Lin CP, Glicksman MA, Karp JM. Tetrandrine identified in a small molecule screen to activate mesenchymal stem cells for enhanced immunomodulation. Sci Rep 2016; 6:30263. [PMID: 27457881 PMCID: PMC4960598 DOI: 10.1038/srep30263] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 06/24/2016] [Indexed: 12/29/2022] Open
Abstract
Pre-treatment or priming of mesenchymal stem cells (MSC) prior to transplantation can significantly augment the immunosuppressive effect of MSC-based therapies. In this study, we screened a library of 1402 FDA-approved bioactive compounds to prime MSC. We identified tetrandrine as a potential hit that activates the secretion of prostaglandin E2 (PGE2), a potent immunosuppressive agent, by MSC. Tetrandrine increased MSC PGE2 secretion through the NF-κB/COX-2 signaling pathway. When co-cultured with mouse macrophages (RAW264.7), tetrandrine-primed MSC attenuated the level of TNF-α secreted by RAW264.7. Furthermore, systemic transplantation of primed MSC into a mouse ear skin inflammation model significantly reduced the level of TNF-α in the inflamed ear, compared to unprimed cells. Screening of small molecules to pre-condition cells prior to transplantation represents a promising strategy to boost the therapeutic potential of cell therapy.
Collapse
Affiliation(s)
- Zijiang Yang
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, US.,Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Harvard Stem Cell Institute, Cambridge, MA, US.,Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, US.,Advanced Industrial Technology Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - John Concannon
- Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, US
| | - Kelvin S Ng
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, US.,Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Harvard Stem Cell Institute, Cambridge, MA, US
| | - Kathleen Seyb
- Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, US
| | - Luke J Mortensen
- Regenerative Bioscience Center, Department of Animal and Dairy Science, and College of Engineering, University of Georgia, Athens, GA, US
| | - Sudhir Ranganath
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, US.,Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Harvard Stem Cell Institute, Cambridge, MA, US.,Department of Chemical Engineering, Siddaganga Institute of Technology, Tumkur, India
| | - Fangqi Gu
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, US.,Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Harvard Stem Cell Institute, Cambridge, MA, US
| | - Oren Levy
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, US.,Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Harvard Stem Cell Institute, Cambridge, MA, US
| | - Zhixiang Tong
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, US.,Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Harvard Stem Cell Institute, Cambridge, MA, US
| | - Keir Martyn
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, US.,Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Harvard Stem Cell Institute, Cambridge, MA, US
| | - Weian Zhao
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center and Chao Family Comprehensive Cancer Center, Department of Biomedical Engineering, and Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA, US
| | - Charles P Lin
- Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, US
| | - Marcie A Glicksman
- Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, US
| | - Jeffrey M Karp
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, US.,Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, US.,Harvard Stem Cell Institute, Cambridge, MA, US
| |
Collapse
|
39
|
Allen TA, Gracieux D, Talib M, Tokarz DA, Hensley MT, Cores J, Vandergriff A, Tang J, de Andrade JBM, Dinh PU, Yoder JA, Cheng K. Angiopellosis as an Alternative Mechanism of Cell Extravasation. Stem Cells 2016; 35:170-180. [PMID: 27350343 DOI: 10.1002/stem.2451] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/23/2016] [Accepted: 06/07/2016] [Indexed: 12/17/2022]
Abstract
Stem cells possess the ability to home in and travel to damaged tissue when injected intravenously. For the cells to exert their therapeutic effect, they must cross the blood vessel wall and enter the surrounding tissues. The mechanism of extravasation injected stem cells employ for exit has yet to be characterized. Using intravital microscopy and a transgenic zebrafish line Tg(fli1a:egpf) with GFP-expressing vasculature, we documented the detailed extravasation processes in vivo for injected stem cells in comparison to white blood cells (WBCs). While WBCs left the blood vessels by the standard diapedesis process, injected cardiac and mesenchymal stem cells underwent a distinct method of extravasation that was markedly different from diapedesis. Here, the vascular wall undergoes an extensive remodeling to allow the cell to exit the lumen, while the injected cell remains distinctively passive in activity. We termed this process Angio-pello-sis, which represents an alternative mechanism of cell extravasation to the prevailing theory of diapedesis. Stem Cells 2017;35:170-180 Video Highlight: https://youtu.be/i5EI-ZvhBps.
Collapse
Affiliation(s)
- Tyler A Allen
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, North Carolina, USA
| | - David Gracieux
- Department of Biochemistry, North Carolina State University, North Carolina, USA
| | - Maliha Talib
- Department of Biochemistry, North Carolina State University, North Carolina, USA
| | - Debra A Tokarz
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
| | - M Taylor Hensley
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
| | - Jhon Cores
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, North Carolina, USA
| | - Adam Vandergriff
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, North Carolina, USA
| | - Junnan Tang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, North Carolina, USA
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - James B M de Andrade
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
| | - Phuong-Uyen Dinh
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
| | - Jeffrey A Yoder
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, North Carolina, USA
- Molecular Pharmaceutics Division, University of North Carolina at Chapel Hill and North Carolina State University, North Carolina, USA
- Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangshu, People's Republic of China
| |
Collapse
|
40
|
Ezquer FE, Ezquer ME, Vicencio JM, Calligaris SD. Two complementary strategies to improve cell engraftment in mesenchymal stem cell-based therapy: Increasing transplanted cell resistance and increasing tissue receptivity. Cell Adh Migr 2016; 11:110-119. [PMID: 27294313 PMCID: PMC5308221 DOI: 10.1080/19336918.2016.1197480] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Over the past 2 decades, therapies based on mesenchymal stem cells (MSC) have been tested to treat several types of diseases in clinical studies, due to their potential for tissue repair and regeneration. Currently, MSC-based therapy is considered a biologically safe procedure, with the therapeutic results being very promising. However, the benefits of these therapies are not stable in the long term, and the final outcomes manifest with high inter-patient variability. The major cause of these therapeutic limitations results from the poor engraftment of the transplanted cells. Researchers have developed separate strategies to improve MSC engraftment. One strategy aims at increasing the survival of the transplanted MSCs in the recipient tissue, rendering them more resistant to the hostile microenvironment (cell-preconditioning). Another strategy aims at making the damaged tissue more receptive to the transplanted cells, favoring their interactions (tissue-preconditioning). In this review, we summarize several approaches using these strategies, providing an integral and updated view of the recent developments in MSC-based therapies. In addition, we propose that the combined use of these different conditioning strategies could accelerate the process to translate experimental evidences from pre-clinic studies to the daily clinical practice.
Collapse
Affiliation(s)
- Fernando E Ezquer
- a Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo , Santiago , Chile
| | - Marcelo E Ezquer
- a Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo , Santiago , Chile
| | | | - Sebastián D Calligaris
- a Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo , Santiago , Chile
| |
Collapse
|
41
|
Levy O, Brennen WN, Han E, Rosen DM, Musabeyezu J, Safaee H, Ranganath S, Ngai J, Heinelt M, Milton Y, Wang H, Bhagchandani SH, Joshi N, Bhowmick N, Denmeade SR, Isaacs JT, Karp JM. A prodrug-doped cellular Trojan Horse for the potential treatment of prostate cancer. Biomaterials 2016; 91:140-150. [PMID: 27019026 DOI: 10.1016/j.biomaterials.2016.03.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/21/2016] [Accepted: 03/15/2016] [Indexed: 01/10/2023]
Abstract
Despite considerable advances in prostate cancer research, there is a major need for a systemic delivery platform that efficiently targets anti-cancer drugs to sites of disseminated prostate cancer while minimizing host toxicity. In this proof-of-principle study, human mesenchymal stem cells (MSCs) were loaded with poly(lactic-co-glycolic acid) (PLGA) microparticles (MPs) that encapsulate the macromolecule G114, a thapsigargin-based prostate specific antigen (PSA)-activated prodrug. G114-particles (∼950 nm in size) were internalized by MSCs, followed by the release of G114 as an intact prodrug from loaded cells. Moreover, G114 released from G114 MP-loaded MSCs selectively induced death of the PSA-secreting PCa cell line, LNCaP. Finally, G114 MP-loaded MSCs inhibited tumor growth when used in proof-of-concept co-inoculation studies with CWR22 PCa xenografts, suggesting that cell-based delivery of G114 did not compromise the potency of this pro-drug in-vitro or in-vivo. This study demonstrates a potentially promising approach to assemble a cell-based drug delivery platform, which inhibits cancer growth in-vivo without the need of genetic engineering. We envision that upon achieving efficient homing of systemically infused MSCs to cancer sites, this MSC-based platform may be developed into an effective, systemic 'Trojan Horse' therapy for targeted delivery of therapeutic agents to sites of metastatic PCa.
Collapse
Affiliation(s)
- Oren Levy
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - W Nathaniel Brennen
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, United States
| | - Edward Han
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - David Marc Rosen
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, United States
| | - Juliet Musabeyezu
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Helia Safaee
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Sudhir Ranganath
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Jessica Ngai
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Martina Heinelt
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Yuka Milton
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Hao Wang
- Department of Oncology, Division of Biostatistics at the Sidney Kimmel Comprehensive Cancer Center, United States
| | - Sachin H Bhagchandani
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Nitin Joshi
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Neil Bhowmick
- The Samuel Oschin Comprehensive Cancer Institute at the Cedars-Sinai Medical Center, United States
| | - Samuel R Denmeade
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, United States.
| | - John T Isaacs
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, United States.
| | - Jeffrey M Karp
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States.
| |
Collapse
|
42
|
Miyahira AK, Lang JM, Den RB, Garraway IP, Lotan TL, Ross AE, Stoyanova T, Cho SY, Simons JW, Pienta KJ, Soule HR. Multidisciplinary intervention of early, lethal metastatic prostate cancer: Report from the 2015 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2016; 76:125-39. [PMID: 26477609 PMCID: PMC5830186 DOI: 10.1002/pros.23107] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 09/24/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND The 2015 Coffey-Holden Prostate Cancer Academy Meeting, themed: "Multidisciplinary Intervention of Early, Lethal Metastatic Prostate Cancer," was held in La Jolla, California from June 25 to 28, 2015. METHODS The Prostate Cancer Foundation (PCF) sponsors an annual, invitation-only, action-tank-structured meeting on a critical topic concerning lethal prostate cancer. The 2015 meeting was attended by 71 basic, translational, and clinical investigators who discussed the current state of the field, major unmet needs, and ideas for addressing earlier diagnosis and treatment of men with lethal prostate cancer for the purpose of extending lives and making progress toward a cure. RESULTS The questions addressed at the meeting included: cellular and molecular mechanisms of tumorigenesis, evaluating, and targeting the microenvironment in the primary tumor, advancing biomarkers for clinical integration, new molecular imaging technologies, clinical trials, and clinical trial design in localized high-risk and oligometastatic settings, targeting the primary tumor in advanced disease, and instituting multi-modal care of high risk and oligometastatic patients. DISCUSSION This article highlights the current status, greatest unmet needs, and anticipated field changes that were discussed at the meeting toward the goal of optimizing earlier interventions to potentiate cures in high-risk and oligometastatic prostate cancer patients.
Collapse
Affiliation(s)
| | - Joshua M. Lang
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, Wisconsin
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Robert B. Den
- Department of Radiation Oncology, Sidney Kimmel Medical Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Cancer Biology, Sidney Kimmel Medical Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Isla P. Garraway
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles,, California
- Greater Los Angeles VA Healthcare System, Los Angeles, California
| | - Tamara L. Lotan
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ashley E. Ross
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - Tanya Stoyanova
- Department of Microbiology, Immunology, and Molecular Genetics, University of California at Los Angeles, Los Angeles, California
| | - Steve Y. Cho
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, Wisconsin
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | - Kenneth J. Pienta
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, The James Buchanan Brady Urological Institute, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
43
|
Heinelt M, Karp JM, Levy O. Cell therapy--showing cells the way home. Oncotarget 2015; 6:17857-8. [PMID: 26257004 PMCID: PMC4627214 DOI: 10.18632/oncotarget.4883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 07/12/2015] [Indexed: 01/12/2023] Open
Affiliation(s)
- Martina Heinelt
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Cambridge, MA, USA
- Harvard Medical School, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Harvard - MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Jeffrey M Karp
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Cambridge, MA, USA
- Harvard Medical School, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Harvard - MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Oren Levy
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Cambridge, MA, USA
- Harvard Medical School, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Harvard - MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| |
Collapse
|
44
|
Tong Z, Solanki A, Hamilos A, Levy O, Wen K, Yin X, Karp JM. Application of biomaterials to advance induced pluripotent stem cell research and therapy. EMBO J 2015; 34:987-1008. [PMID: 25766254 PMCID: PMC4406648 DOI: 10.15252/embj.201490756] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/25/2015] [Accepted: 02/17/2015] [Indexed: 12/19/2022] Open
Abstract
Derived from any somatic cell type and possessing unlimited self-renewal and differentiation potential, induced pluripotent stem cells (iPSCs) are poised to revolutionize stem cell biology and regenerative medicine research, bringing unprecedented opportunities for treating debilitating human diseases. To overcome the limitations associated with safety, efficiency, and scalability of traditional iPSC derivation, expansion, and differentiation protocols, biomaterials have recently been considered. Beyond addressing these limitations, the integration of biomaterials with existing iPSC culture platforms could offer additional opportunities to better probe the biology and control the behavior of iPSCs or their progeny in vitro and in vivo. Herein, we discuss the impact of biomaterials on the iPSC field, from derivation to tissue regeneration and modeling. Although still exploratory, we envision the emerging combination of biomaterials and iPSCs will be critical in the successful application of iPSCs and their progeny for research and clinical translation.
Collapse
Affiliation(s)
- Zhixiang Tong
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Aniruddh Solanki
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Allison Hamilos
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Oren Levy
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Kendall Wen
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Xiaolei Yin
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Jeffrey M Karp
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| |
Collapse
|