1
|
Park R, Kang MS, Heo G, Shin YC, Han DW, Hong SW. Regulated Behavior in Living Cells with Highly Aligned Configurations on Nanowrinkled Graphene Oxide Substrates: Deep Learning Based on Interplay of Cellular Contact Guidance. ACS NANO 2024; 18:1325-1344. [PMID: 38099607 DOI: 10.1021/acsnano.2c09815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Micro-/nanotopographical cues have emerged as a practical and promising strategy for controlling cell fate and reprogramming, which play a key role as biophysical regulators in diverse cellular processes and behaviors. Extracellular biophysical factors can trigger intracellular physiological signaling via mechanotransduction and promote cellular responses such as cell adhesion, migration, proliferation, gene/protein expression, and differentiation. Here, we engineered a highly ordered nanowrinkled graphene oxide (GO) surface via the mechanical deformation of an ultrathin GO film on an elastomeric substrate to observe specific cellular responses based on surface-mediated topographical cues. The ultrathin GO film on the uniaxially prestrained elastomeric substrate through self-assembly and subsequent compressive force produced GO nanowrinkles with periodic amplitude. To examine the acute cellular behaviors on the GO-based cell interface with nanostructured arrays of wrinkles, we cultured L929 fibroblasts and HT22 hippocampal neuronal cells. As a result, our developed cell-culture substrate obviously provided a directional guidance effect. In addition, based on the observed results, we adapted a deep learning (DL)-based data processing technique to precisely interpret the cell behaviors on the nanowrinkled GO surfaces. According to the learning/transfer learning protocol of the DL network, we detected cell boundaries, elongation, and orientation and quantitatively evaluated cell velocity, traveling distance, displacement, and orientation. The presented experimental results have intriguing implications such that the nanotopographical microenvironment could engineer the living cells' morphological polarization to assemble them into useful tissue chips consisting of multiple cell types.
Collapse
Affiliation(s)
- Rowoon Park
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Gyeonghwa Heo
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Yong Cheol Shin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio 44195, United States
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
- Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
2
|
Oh MS, Lee SG, Lee GH, Kim CY, Song JH, Yu BY, Chung HM. Verification of Therapeutic Effect through Accelerator Mass Spectrometry-Based Single Cell Level Quantification of hESC-Endothelial Cells Distributed into an Ischemic Model. Adv Healthc Mater 2023; 12:e2300476. [PMID: 37068221 DOI: 10.1002/adhm.202300476] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/01/2023] [Indexed: 04/19/2023]
Abstract
As the potential of pluripotent stem cell-derived differentiated cells has been demonstrated in regenerative medicine, differentiated vascular endothelial cells (ECs) are emerging as a therapeutic agent for the cardiovascular system. To verify the therapeutic efficacy of differentiated ECs in an ischemic model, human embryonic stem cells (hESCs) are induced as EC lineage and produce high-purity ECs through fluorescence-activated cell sorting (FACS). When hESC-ECs are transplanted into a hindlimb ischemic model, it is confirmed that blood flow and muscle regeneration are further improved by creating new blood vessels together with autologous ECs than the primary cell as cord blood endothelial progenitor cells (CB-EPCs). In addition, previously reported studies show the detection of transplanted cells engrafted in blood vessels through various tracking methods, but fail to provide accurate quantitative values over time. In this study, it is demonstrated that hESC-ECs are engrafted approximately sevenfold more than CB-EPCs by using an accelerator mass spectrometry (AMS)-based cell tracking technology that can perform quantification at the single cell level. An accurate quantification index is suggested. It has never been reported in in vivo kinetics of hESC-ECs that can act as therapeutic agents.
Collapse
Affiliation(s)
- Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Gwan-Ho Lee
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Jong Han Song
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Byung-Yong Yu
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Mirae Cell Bio Co. Ltd, Seoul, 04795, Republic of Korea
| |
Collapse
|
3
|
Oraee-Yazdani S, Tavanaei R, Rostami F, Hajarizadeh A, Mehrabadi M, Akhlaghpasand M, Tamaddon M, Khannejad S, Yazdani KO, Zali A. Suicide gene therapy using allogeneic adipose tissue-derived mesenchymal stem cell gene delivery vehicles in recurrent glioblastoma multiforme: a first-in-human, dose-escalation, phase I clinical trial. J Transl Med 2023; 21:350. [PMID: 37245011 DOI: 10.1186/s12967-023-04213-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is associated with remarkably poor prognosis, and its treatment is challenging. This investigation aimed to evaluate the safety of suicide gene therapy using allogeneic adipose tissue-derived mesenchymal stem cells (ADSCs) carrying herpes simplex virus-thymidine kinase (HSV-TK) gene for the first time in patients with recurrent GBM. METHODS This study was a first-in-human, open-label, single-arm, phase I clinical trial with a classic 3 + 3 dose escalation design. Patients who did not undergo surgery for their recurrence were included and received this gene therapy protocol. Patients received the intratumoral stereotactic injection of ADSCs according to the assigned dose followed by prodrug administration for 14 days. The first dosing cohort (n = 3) received 2.5 × 105 ADSCs; the second dosing cohort (n = 3) received 5 × 105 ADSCs; the third dosing cohort (n = 6) received 10 × 105 ADSCs. The primary outcome measure was the safety profile of the intervention. RESULTS A total of 12 patients with recurrent GBM were recruited. The median follow-up was 16 (IQR, 14-18.5) months. This gene therapy protocol was safe and well tolerated. During the study period, eleven (91.7%) patients showed tumor progression, and nine (75.0%) died. The median overall survival (OS) was 16.0 months (95% CI 14.3-17.7) and the median progression-free survival (PFS) was 11.0 months (95% CI 8.3-13.7). A total of 8 and 4 patients showed partial response and stable disease, respectively. Moreover, significant changes were observed in volumetric analysis, peripheral blood cell counts, and cytokine profile. CONCLUSIONS The present clinical trial, for the first time, showed that suicide gene therapy using allogeneic ADSCs carrying the HSV-TK gene is safe in patients with recurrent GBM. Future phase II/III clinical trials with multiple arms are warranted to validate our findings and further investigate the efficacy of this protocol compared with standard therapy alone. TRIAL REGISTRATION Iranian Registry of Clinical Trials (IRCT), IRCT20200502047277N2. Registered 8 October 2020, https://www.irct.ir/ .
Collapse
Affiliation(s)
- Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran.
| | - Roozbeh Tavanaei
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Fatemeh Rostami
- Stem Cell Technology Research Center (STRC), Iran university of medical science (IUMS), Tehran, Iran
| | - Atieh Hajarizadeh
- Stem Cell Technology Research Center (STRC), Iran university of medical science (IUMS), Tehran, Iran
| | - Marzieh Mehrabadi
- Stem Cell Technology Research Center (STRC), Iran university of medical science (IUMS), Tehran, Iran
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mohammadhosein Akhlaghpasand
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Mona Tamaddon
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samin Khannejad
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Kaveh Oraii Yazdani
- Department of cardiovascular diseases, Zahedan university of medical science, Zahedan, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| |
Collapse
|
4
|
Sackett SD, Kaplan SJ, Mitchell SA, Brown ME, Burrack AL, Grey S, Huangfu D, Odorico J. Genetic Engineering of Immune Evasive Stem Cell-Derived Islets. Transpl Int 2022; 35:10817. [PMID: 36545154 PMCID: PMC9762357 DOI: 10.3389/ti.2022.10817] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Genome editing has the potential to revolutionize many investigative and therapeutic strategies in biology and medicine. In the field of regenerative medicine, one of the leading applications of genome engineering technology is the generation of immune evasive pluripotent stem cell-derived somatic cells for transplantation. In particular, as more functional and therapeutically relevant human pluripotent stem cell-derived islets (SCDI) are produced in many labs and studied in clinical trials, there is keen interest in studying the immunogenicity of these cells and modulating allogeneic and autoimmune immune responses for therapeutic benefit. Significant experimental work has already suggested that elimination of Human Leukocytes Antigen (HLA) expression and overexpression of immunomodulatory genes can impact survival of a variety of pluripotent stem cell-derived somatic cell types. Limited work published to date focuses on stem cell-derived islets and work in a number of labs is ongoing. Rapid progress is occurring in the genome editing of human pluripotent stem cells and their progeny focused on evading destruction by the immune system in transplantation models, and while much research is still needed, there is no doubt the combined technologies of genome editing and stem cell therapy will profoundly impact transplantation medicine in the future.
Collapse
Affiliation(s)
- Sara D. Sackett
- Division of Transplantation, Department of Surgery, UW Transplant Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States,*Correspondence: Sara D. Sackett,
| | - Samuel J. Kaplan
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States,Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Samantha A. Mitchell
- Division of Transplantation, Department of Surgery, UW Transplant Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Matthew E. Brown
- Division of Transplantation, Department of Surgery, UW Transplant Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Adam L. Burrack
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN,Center for Immunology, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Shane Grey
- Immunology Division, Garvan Institute of Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Danwei Huangfu
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jon Odorico
- Division of Transplantation, Department of Surgery, UW Transplant Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
5
|
Yu H, Commander CW, Stavas JM. Stem Cell-Based Therapies: What Interventional Radiologists Need to Know. Semin Intervent Radiol 2021; 38:523-534. [PMID: 34853498 DOI: 10.1055/s-0041-1736657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
As the basic units of biological organization, stem cells and their progenitors are essential for developing and regenerating organs and tissue systems using their unique self-renewal capability and differentiation potential into multiple cell lineages. Stem cells are consistently present throughout the entire human development, from the zygote to adulthood. Over the past decades, significant efforts have been made in biology, genetics, and biotechnology to develop stem cell-based therapies using embryonic and adult autologous or allogeneic stem cells for diseases without therapies or difficult to treat. Stem cell-based therapies require optimum administration of stem cells into damaged organs to promote structural regeneration and improve function. Maximum clinical efficacy is highly dependent on the successful delivery of stem cells to the target tissue. Direct image-guided locoregional injections into target tissues offer an option to increase therapeutic outcomes. Interventional radiologists have the opportunity to perform a key role in delivering stem cells more efficiently using minimally invasive techniques. This review discusses the types and sources of stem cells and the current clinical applications of stem cell-based therapies. In addition, the regulatory considerations, logistics, and potential roles of interventional Radiology are also discussed with the review of the literature.
Collapse
Affiliation(s)
- Hyeon Yu
- Division of Vascular and Interventional Radiology, Department of Radiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,ProKidney LLC, Winston Salem, North Carolina
| | - Clayton W Commander
- Division of Vascular and Interventional Radiology, Department of Radiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Joseph M Stavas
- Department of Radiology, Creighton University School of Medicine, Omaha, Nebraska
| |
Collapse
|
6
|
Triolo TM, Bellin MD. Lessons from Human Islet Transplantation Inform Stem Cell-Based Approaches in the Treatment of Diabetes. Front Endocrinol (Lausanne) 2021; 12:636824. [PMID: 33776933 PMCID: PMC7992005 DOI: 10.3389/fendo.2021.636824] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is characterized by the body's inability to control blood glucose levels within a physiological range due to loss and/or dysfunction of insulin producing beta cells. Progressive beta cell loss leads to hyperglycemia and if untreated can lead to severe complications and/or death. Treatments at this time are limited to pharmacologic therapies, including exogenous insulin or oral/injectable agents that improve insulin sensitivity or augment endogenous insulin secretion. Cell transplantation can restore physiologic endogenous insulin production and minimize hyper- and hypoglycemic excursions. Islet isolation procedures and management of transplant recipients have advanced over the last several decades; both tight glycemic control and insulin independence are achievable. Research has been conducted in isolating islets, monitoring islet function, and mitigating the immune response. However, this procedure is still only performed in a small minority of patients. One major barrier is the scarcity of human pancreatic islet donors, variation in donor pancreas quality, and variability in islet isolation success. Advances have been made in generation of glucose responsive human stem cell derived beta cells (sBCs) and islets from human pluripotent stem cells using directed differentiation. This is an emerging promising treatment for patients with diabetes because they could potentially serve as an unlimited source of functional, glucose-responsive beta cells. Challenges exist in their generation including long term survival of grafts, safety of transplantation, and protection from the immune response. This review focuses on the progress made in islet allo- and auto transplantation and how these advances may be extrapolated to the sBC context.
Collapse
Affiliation(s)
- Taylor M. Triolo
- The Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Denver, Aurora, CO, United States
- *Correspondence: Taylor M. Triolo,
| | - Melena D. Bellin
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
7
|
Evolution of Stem Cells in Cardio-Regenerative Therapy. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Luck J, Weil BD, Lowdell M, Mosahebi A. Adipose-Derived Stem Cells for Regenerative Wound Healing Applications: Understanding the Clinical and Regulatory Environment. Aesthet Surg J 2020; 40:784-799. [PMID: 31406975 DOI: 10.1093/asj/sjz214] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
There is growing interest in the regenerative potential of adipose-derived stem cells (ADSCs) for wound healing applications. ADSCs have been shown to promote revascularization, activate local stem cell niches, reduce oxidative stress, and modulate immune responses. Combined with the fact that they can be harvested in large numbers with minimal donor site morbidity, ADSC products represent promising regenerative cell therapies. This article provides a detailed description of the defining characteristics and therapeutic potential of ADSCs, with a focus on understanding how ADSCs promote tissue regeneration and repair. It summarizes the current regulatory environment governing the use of ADSC products across Europe and the United States and examines how various adipose-derived products conform to the current UK legislative framework. Advice is given to clinicians and researchers on how novel ADSC therapeutics may be developed in accordance with regulatory guidelines.
Collapse
Affiliation(s)
| | - Benjamin D Weil
- Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital, London, UK
| | | | | |
Collapse
|
9
|
Vishwakarma SK, Jaiswal J, Park K, Lakkireddy C, Raju N, Bardia A, Habeeb MA, Paspala SAB, Khan AA, Dhayal M. TiO
2
Nanoflowers on Conducting Substrates Ameliorate Effective Transdifferentiation of Human Hepatic Progenitor Cells for Long‐Term Hyperglycemia Reversal in Diabetic Mice. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Sandeep Kumar Vishwakarma
- Clinical Research FacilityCSIR‐Centre for Cellular and Molecular Biology Hyderabad Telangana 500007 India
- Central Laboratory for Stem Cell Research and Translational MedicineCentre for Liver Research and Diagnostics, Deccan College of Medical Sciences Kanchanbagh Hyderabad Telangana 500058 India
- Dr. Habeebullah Life Sciences Limited Attapur Hyderabad Telangana 500048 India
| | - Juhi Jaiswal
- Nano‐Cellular Medicine and Biophysics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh 221005 India
| | - Kyung‐Hee Park
- Department of Dental Materials and Hard‐tissue Biointerface Research Center, School of DentistryChonnam National University Gwangju 61186 Republic of Korea
| | - Chandrakala Lakkireddy
- Central Laboratory for Stem Cell Research and Translational MedicineCentre for Liver Research and Diagnostics, Deccan College of Medical Sciences Kanchanbagh Hyderabad Telangana 500058 India
| | - Nagarapu Raju
- Central Laboratory for Stem Cell Research and Translational MedicineCentre for Liver Research and Diagnostics, Deccan College of Medical Sciences Kanchanbagh Hyderabad Telangana 500058 India
- Dr. Habeebullah Life Sciences Limited Attapur Hyderabad Telangana 500048 India
| | - Avinash Bardia
- Central Laboratory for Stem Cell Research and Translational MedicineCentre for Liver Research and Diagnostics, Deccan College of Medical Sciences Kanchanbagh Hyderabad Telangana 500058 India
- Dr. Habeebullah Life Sciences Limited Attapur Hyderabad Telangana 500048 India
| | - Md. Aejaz Habeeb
- Central Laboratory for Stem Cell Research and Translational MedicineCentre for Liver Research and Diagnostics, Deccan College of Medical Sciences Kanchanbagh Hyderabad Telangana 500058 India
- Dr. Habeebullah Life Sciences Limited Attapur Hyderabad Telangana 500048 India
| | - Syed Ameer Basha Paspala
- Central Laboratory for Stem Cell Research and Translational MedicineCentre for Liver Research and Diagnostics, Deccan College of Medical Sciences Kanchanbagh Hyderabad Telangana 500058 India
- Dr. Habeebullah Life Sciences Limited Attapur Hyderabad Telangana 500048 India
| | - Aleem Ahmed Khan
- Central Laboratory for Stem Cell Research and Translational MedicineCentre for Liver Research and Diagnostics, Deccan College of Medical Sciences Kanchanbagh Hyderabad Telangana 500058 India
- Dr. Habeebullah Life Sciences Limited Attapur Hyderabad Telangana 500048 India
| | - Marshal Dhayal
- Clinical Research FacilityCSIR‐Centre for Cellular and Molecular Biology Hyderabad Telangana 500007 India
- Nano‐Cellular Medicine and Biophysics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh 221005 India
| |
Collapse
|
10
|
Aslankoohi N, Mondal D, Rizkalla AS, Mequanint K. Bone Repair and Regenerative Biomaterials: Towards Recapitulating the Microenvironment. Polymers (Basel) 2019; 11:E1437. [PMID: 31480693 PMCID: PMC6780693 DOI: 10.3390/polym11091437] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/24/2019] [Accepted: 08/25/2019] [Indexed: 02/07/2023] Open
Abstract
Biomaterials and tissue engineering scaffolds play a central role to repair bone defects. Although ceramic derivatives have been historically used to repair bone, hybrid materials have emerged as viable alternatives. The rationale for hybrid bone biomaterials is to recapitulate the native bone composition to which these materials are intended to replace. In addition to the mechanical and dimensional stability, bone repair scaffolds are needed to provide suitable microenvironments for cells. Therefore, scaffolds serve more than a mere structural template suggesting a need for better and interactive biomaterials. In this review article, we aim to provide a summary of the current materials used in bone tissue engineering. Due to the ever-increasing scientific publications on this topic, this review cannot be exhaustive; however, we attempted to provide readers with the latest advance without being redundant. Furthermore, every attempt is made to ensure that seminal works and significant research findings are included, with minimal bias. After a concise review of crystalline calcium phosphates and non-crystalline bioactive glasses, the remaining sections of the manuscript are focused on organic-inorganic hybrid materials.
Collapse
Affiliation(s)
- Neda Aslankoohi
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Dibakar Mondal
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Amin S Rizkalla
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
- Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Kibret Mequanint
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
| |
Collapse
|
11
|
Nejadnik H, Tseng J, Daldrup-Link H. Magnetic resonance imaging of stem cell-macrophage interactions with ferumoxytol and ferumoxytol-derived nanoparticles. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1552. [PMID: 30734542 PMCID: PMC6579657 DOI: 10.1002/wnan.1552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/13/2018] [Accepted: 12/19/2018] [Indexed: 01/07/2023]
Abstract
"Off the shelf" allogeneic stem cell transplants and stem cell nano-composites are being used for the treatment of degenerative bone diseases. However, major and minor histocompatibility antigens of therapeutic cell transplants can be recognized as foreign and lead to their rejection by the host immune system. If a host immune response is identified within the first week post-transplant, immune modulating therapies could be applied to prevent graft failure and support engraftment. Ferumoxytol (Feraheme™) is an FDA approved iron oxide nanoparticle preparation for the treatment of anemia in patients. Ferumoxytol can be used "off label" as an magnetic resonance (MR) contrast agent, as these nanoparticles provide measurable signal changes on magnetic resonance imaging (MRI). In this focused review article, we will discuss three methods to localize and identify innate immune responses to stem cell transplants using ferumoxytol-enhanced MRI, which are based on tracking stem cells, tracking macrophages or detecting mediators of cell death: (a) monitor MRI signal changes of ferumoxytol-labeled stem cells in the presence or absence of innate immune responses, (b) monitor influx of ferumoxytol-labeled macrophages into stem cell implants, and (c) monitor apoptosis of stem cell implants with caspase-3 activatable nanoparticles. These techniques can detect transplant failure at an early stage, when immune-modulating interventions can potentially preserve the viability of the cell transplants and thereby improve bone and cartilage repair outcomes. Approaches 1 and 2 are immediately translatable to clinical practice. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Jessica Tseng
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Heike Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| |
Collapse
|
12
|
Sebastian S, Hourd P, Chandra A, Williams DJ, Medcalf N. The management of risk and investment in cell therapy process development: a case study for neurodegenerative disease. Regen Med 2019; 14:465-488. [PMID: 31210581 DOI: 10.2217/rme-2018-0081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cell-based therapies must achieve clinical efficacy and safety with reproducible and cost-effective manufacturing. This study addresses process development issues using the exemplar of a human pluripotent stem cell-based dopaminergic neuron cell therapy product. Early identification and correction of risks to product safety and the manufacturing process reduces the expensive and time-consuming bridging studies later in development. A New Product Introduction map was used to determine the developmental requirements specific to the product. Systematic Risk Analysis is exemplified here. Expected current value-based prioritization guides decisions about the sequence of process studies and whether and if an early abandonment of further research is appropriate. The application of the three tools enabled prioritization of the development studies.
Collapse
Affiliation(s)
- Sujith Sebastian
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Paul Hourd
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Amit Chandra
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - David J Williams
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Nicholas Medcalf
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| |
Collapse
|
13
|
Buduru SD, Gulei D, Zimta AA, Tigu AB, Cenariu D, Berindan-Neagoe I. The Potential of Different Origin Stem Cells in Modulating Oral Bone Regeneration Processes. Cells 2019; 8:cells8010029. [PMID: 30625993 PMCID: PMC6356555 DOI: 10.3390/cells8010029] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 12/22/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering has gained much momentum since the implementation of stem cell isolation and manipulation for regenerative purposes. Despite significant technical improvements, researchers still have to decide which strategy (which type of stem cell) is the most suitable for their specific purpose. Therefore, this short review discusses the advantages and disadvantages of the three main categories of stem cells: embryonic stem cells, mesenchymal stem cells and induced pluripotent stem cells in the context of bone regeneration for dentistry-associated conditions. Importantly, when deciding upon the right strategy, the selection needs to be made in concordance with the morbidity and the life-threatening level of the condition in discussion. Therefore, even when a specific type of stem cell holds several advantages over others, their availability, invasiveness of the collection method and ethical standards become deciding parameters.
Collapse
Affiliation(s)
- Smaranda Dana Buduru
- Prosthetics and Dental Materials, Faculty of Dental Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, 32 Clinicilor Street, 400006 Cluj-Napoca, Romania.
- Stomestet Stomatology Clinic, Calea Manastur 68A Street, 400658 Cluj-Napoca, Romania; .
| | - Diana Gulei
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
| | - Alina-Andreea Zimta
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
| | - Adrian Bogdan Tigu
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
| | - Diana Cenariu
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337 Cluj-Napoca, Romania.
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Republicii 34-36 Street, 400015 Cluj-Napoca, Romania.
| |
Collapse
|
14
|
Suman S, Domingues A, Ratajczak J, Ratajczak MZ. Potential Clinical Applications of Stem Cells in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1201:1-22. [PMID: 31898779 DOI: 10.1007/978-3-030-31206-0_1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The field of regenerative medicine is looking for a pluripotent/multipotent stem cell able to differentiate across germ layers and be safely employed in therapy. Unfortunately, with the exception of hematopoietic stem/progenitor cells (HSPCs) for hematological applications, the current clinical results with stem cells are somewhat disappointing. The potential clinical applications of the more primitive embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have so far been discouraging, as both have exhibited several problems, including genomic instability, a risk of teratoma formation, and the possibility of rejection. Therefore, the only safe stem cells that have so far been employed in regenerative medicine are monopotent stem cells, such as the abovementioned HSPCs or mesenchymal stem cells (MSCs) isolated from postnatal tissues. However, their monopotency, and therefore limited differentiation potential, is a barrier to their broader application in the clinic. Interestingly, results have accumulated indicating that adult tissues contain rare, early-development stem cells known as very small embryonic-like stem cells (VSELs), which can differentiate into cells from more than one germ layer. This chapter addresses different sources of stem cells for potential clinical application and their advantages and problems to be solved.
Collapse
Affiliation(s)
- Suman Suman
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Alison Domingues
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA. .,Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland.
| |
Collapse
|
15
|
Li K, Chan CT, Nejadnik H, Lenkov OD, Wolterman C, Paulmurugan R, Yang H, Gambhir SS, Daldrup-Link HE. Ferumoxytol-based Dual-modality Imaging Probe for Detection of Stem Cell Transplant Rejection. Nanotheranostics 2018; 2:306-319. [PMID: 29977742 PMCID: PMC6030766 DOI: 10.7150/ntno.26389] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/14/2018] [Indexed: 12/24/2022] Open
Abstract
Purpose: Stem cell transplants are an effective approach to repair large bone defects. However, comprehensive techniques to monitor the fate of transplanted stem cells in vivo are lacking. Such strategies would enable corrective interventions at an early stage and greatly benefit the development of more successful tissue regeneration approaches. In this study, we designed and synthesized a dual-modality imaging probe (Feru-AFC) that can simultaneously localize transplanted stem cells and diagnose immune rejection-induced apoptosis at an early stage in vivo. Methods: We used a customized caspase-3 cleavable peptide-dye conjugate to modify the surface of clinically approved ferumoxytol nanoparticles (NPs) to generate the dual-modality imaging probe with fluorescence "light-up" feature. We labeled both mouse mesenchymal stem cells (mMSCs, matched) and pig mesenchymal stem cells (pMSCs, mismatched) with the probe and transplanted the labeled cells with biocompatible scaffold at the calvarial defects in mice. We then employed intravital microscopy (IVM) and magnetic resonance imaging (MRI) to investigate the localization, engraftment, and viability of matched and mismatched stem cells, followed by histological analyses to evaluate the results obtained from in vivo studies. Results: The Feru-AFC NPs showed good cellular uptake efficiency in the presence of lipofectin without cytotoxicity to mMSCs and pMSCs. The fluorescence of Feru-AFC NPs was turned on inside apoptotic cells due to the cleavage of peptide by activated caspase-3 and subsequent release of fluorescence dye molecules. Upon transplantation at the calvarial defects in mice, the intense fluorescence from the cleaved Feru-AFC NPs in apoptotic pMSCs was observed with a concomitant decrease in the overall cell number from days 1 to 6. In contrast, the Feru-AFC NP-treated mMSCs exhibited minimum fluorescence and the cell number also remained similar. Furthermore, in vivo MRI of the Feru-AFC NP-treated mMSC and pMSCs transplants could clearly indicate the localization of matched and mismatched cells, respectively. Conclusions: We successfully developed a dual-modality imaging probe for evaluation of the localization and viability of transplanted stem cells in mouse calvarial defects. Using ferumoxytol NPs as the platform, our Feru-AFC NPs are superparamagnetic and display a fluorescence "light-up" signature upon exposure to activated caspase-3. The results show that the probe is a promising tool for long-term stem cell tracking through MRI and early diagnosis of immune rejection-induced apoptosis through longitudinal fluorescence imaging.
Collapse
Affiliation(s)
- Kai Li
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305.,Institute of Materials Research and Engineering, ASTAR, Singapore, 138634
| | - Carmel T Chan
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Hossein Nejadnik
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Olga D Lenkov
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Cody Wolterman
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Ramasamy Paulmurugan
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford, CA 94305
| | - Sanjiv Sam Gambhir
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Heike E Daldrup-Link
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| |
Collapse
|
16
|
Doğan A. Embryonic Stem Cells in Development and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1079:1-15. [PMID: 29464659 DOI: 10.1007/5584_2018_175] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
After progressive improvement in embryonic stem (ES) cell field, several studies have been conducted to explore the usage of ES cells in regenerative medicine. Unlimited self renewal and pluripoteny properties, combined with encouraging preclinical trials, remark that ES cell technology might be promising for clinical practice. ES cells, which can form three germ layers in vitro, are potential candidates to study development at the cellular and molecular level. Understanding the cell fate decision and differentiation processes during development might enable generating functional progenitor cells for tissue restoration. Progression in gene modifications and tissue engineering technology has facilitated the derivation of desired cells for therapy. Success in differentiation protocols and identification the regulatory pathways simplify the research for clinical applications. Although there are established protocols for cell differentiation in vitro and promising preclinical studies in vivo, many challenges need to be adressed before clinical translation. In this review, ES cells are discussed as a model of development in vitro and as a potential candidate for regenerative medicine. This review also dissusses current challenges for ES cell based therapy.
Collapse
Affiliation(s)
- Ayşegül Doğan
- National Cancer Institute, CDBL, NIH, Frederick, MD, USA.
| |
Collapse
|
17
|
The similarities between smDCs and regDCs in alleviating the immune injury caused by transplantation of hepatocytes differentiated from ESCs. Stem Cell Res Ther 2017; 8:266. [PMID: 29157299 PMCID: PMC5696883 DOI: 10.1186/s13287-017-0712-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 11/15/2022] Open
Abstract
Background This study aimed to investigate the tolerogenic mechanisms induced by semimature dendritic cells (smDCs) and regulatory dendritic cells (regDCs) after transplantation of hepatocytes differentiated from mouse embryonic stem cells (ESCs) and to confirm the low immunogenicity of hepatocytes differentiated from ESCs. Methods Green fluorescent protein-labeled ESCs collected from 129 mice were cultured to differentiate into hepatocytes. smDCs and regDCs were cultured in vitro. The hepatocytes were cultured after being extracted from the livers of 129 mice. After injecting smDCs or regDCs 3 days in advance, these differentiated hepatocytes and normal hepatocytes were transplanted into the livers of BALB/c mice separately. Subsequently, the histopathological features and cytokines in transplant tissues as well as the Foxp3 expression in peripheral blood CD4+ T cells of the recipients were examined. Results The morphological phenotypes of smDCs and regDCs were similar. They both expressed medium levels of MHC-II, CD40, CD80, and CD86, high levels of TGF-β and IL-10, and low levels of IL-2. The survival of differentiated hepatocytes was prolonged and inflammatory infiltration in transplant tissues was reduced in both the smDC and regDC groups. Foxp3 expression in peripheral blood CD4+ T cells of the smDC group increased to 5.38% and that of the regDC group also rose to 3.87%. Moreover, the inflammatory infiltration in the tissues receiving transplanted hepatocytes was more obvious. Conclusions smDCs and regDCs were similar tolerogenic dendritic cells. They both could alleviate the immune injury by inducing CD4+CD25+Foxp3+ regulatory T cells through the medium expression of MHC-II, CD40, CD80, and CD86 and the appropriate secretion of cytokines. Hepatocytes differentiated from ESCs displayed low immunogenicity.
Collapse
|
18
|
The inhibiting effect of neural stem cells on proliferation and invasion of glioma cells. Oncotarget 2017; 8:76949-76960. [PMID: 29100360 PMCID: PMC5652754 DOI: 10.18632/oncotarget.20270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 06/02/2017] [Indexed: 12/11/2022] Open
Abstract
The invasive and infiltrative nature of tumor cells leads to the poor prognosis of glioma. Currently, novel therapeutic means to eliminate the tumor cells without damaging the normal brain tissue are still strongly demanded. Significant attentions had been paid to stem cell-based therapy and neural stem cell (NSC) had been considered as one of the efficient delivery vehicles for targeting therapeutic genes. However, whether the NSCs could directly affect glioma cells remains to be seen. In this study, both rat and human glioma cells (C6 and U251) were co-cultured with normal rat embryonic NSCs directly or in-directly. We found the survival, proliferation, invasion and migration of glioma cells were significantly inhibited, while the differentiation was not affected in the in vitro co-culture system. In nude mice, although no significant difference was observed in the tumor growth, survival status and time of tumor-bearing mice were significantly promoted when U251 cells were subcutaneously injected with NSCs. In coincidence with the suppression of glioma cell growth in vitro, expression of mutant p53 and phosphorylation of AKT, ERK1/2 decreased while the level of caspase-3 increased significantly. Our results suggested that normal NSCs could possess direct anti-glioma properties via inhibiting the glioma cell viability, proliferation, invasion and migration. It could be a very promising candidate for glioma treatment.
Collapse
|
19
|
Abstract
Human embryonic stem cells (hESCs) can undergo unlimited self-renewal and differentiate into all cell types in human body, and therefore hold great potential for cell therapy of currently incurable diseases including neural degenerative diseases, heart failure, and macular degeneration. This potential is further underscored by the promising safety and efficacy data from the ongoing clinical trials of hESC-based therapy of macular degeneration. However, one main challenge for the clinical application of hESC-based therapy is the allogeneic immune rejection of hESC-derived cells by the recipient. The breakthrough of the technology to generate autologous-induced pluripotent stem cells (iPSCs) by nuclear reprogramming of patient’s somatic cells raised the possibility that autologous iPSC-derived cells can be transplanted into the patients without the concern of immune rejection. However, accumulating data indicate that certain iPSC-derived cells can be immunogenic. In addition, the genomic instability associated with iPSCs raises additional safety concern to use iPSC-derived cells in human cell therapy. In this review, we will discuss the mechanism underlying the immunogenicity of the pluripotent stem cells and recent progress in developing immune tolerance strategies of human pluripotent stem cell (hPSC)-derived allografts. The successful development of safe and effective immune tolerance strategy will greatly facilitate the clinical development of hPSC-based cell therapy.
Collapse
Affiliation(s)
- Xin Liu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Wenjuan Li
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuemei Fu
- The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yang Xu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
20
|
Abstract
PURPOSE OF THE REVIEW Type 1 diabetes (T1D) is defined by an autoimmune destruction of insulin producing β-cells located in the endocrine part of the pancreas, the islets of Langerhans. As exogenous insulin administration fails at preventing severe complications associated with this disease, cell replacement therapies are being considered as a means to treat T1D. The purpose of this manuscript is to review the challenges associated with current strategies and discuss the potential of stem cell therapy for the treatment of T1D. RECENT FINDINGS The most prominent therapy offered to T1D patients is exogenous insulin administration which, despite formulations improvement, remains a suboptimal treatment, due to the frequency of injections and the issues associated with precise dosing. As immunotherapy approaches have remained unsuccessful, the only cure for T1D is transplantation of donor-derived pancreas or islets. However, donor scarcity, graft loss, and immune response to the foreign tissue are issues challenging this approach and limiting the number of patients who can benefit from such treatments. In this review, we discuss the causes of T1D and the shortcomings of the current treatments. Furthermore, we summarize the cutting edge research that aims to tackle the current challenges in reaching a quality-controlled product with long-term effects, with a focus on regenerative medicine approaches using human pluripotent stem cells.
Collapse
Affiliation(s)
- Yasaman Aghazadeh
- Toronto General Hospital Research Institute and McEwen Centre for Regenerative Medicine, Toronto, Canada
- University Health Network, Toronto, Ontario, M5G 1L7, Canada
| | - Maria Cristina Nostro
- Toronto General Hospital Research Institute and McEwen Centre for Regenerative Medicine, Toronto, Canada.
- University Health Network, Toronto, Ontario, M5G 1L7, Canada.
- Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
21
|
Daldrup-Link HE, Chan C, Lenkov O, Taghavigarmestani S, Nazekati T, Nejadnik H, Chapelin F, Khurana A, Tong X, Yang F, Pisani L, Longaker M, Gambhir SS. Detection of Stem Cell Transplant Rejection with Ferumoxytol MR Imaging: Correlation of MR Imaging Findings with Those at Intravital Microscopy. Radiology 2017; 284:495-507. [PMID: 28128708 DOI: 10.1148/radiol.2017161139] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Purpose To determine whether endogenous labeling of macrophages with clinically applicable nanoparticles enables noninvasive detection of innate immune responses to stem cell transplants with magnetic resonance (MR) imaging. Materials and Methods Work with human stem cells was approved by the institutional review board and the stem cell research oversight committee, and animal experiments were approved by the administrative panel on laboratory animal care. Nine immunocompetent Sprague-Dawley rats received intravenous injection of ferumoxytol, and 18 Jax C57BL/6-Tg (Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6) 2Bck/J mice received rhodamine-conjugated ferumoxytol. Then, 48 hours later, immune-matched or mismatched stem cells were implanted into osteochondral defects of the knee joints of experimental rats and calvarial defects of Jax mice. All animals underwent serial MR imaging and intravital microscopy (IVM) up to 4 weeks after surgery. Macrophages of Jax C57BL/6-Tg (Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6) 2Bck/J mice express enhanced green fluorescent protein (GFP), which enables in vivo correlation of ferumoxytol enhancement at MR imaging with macrophage quantities at IVM. All quantitative data were compared between experimental groups by using a mixed linear model and t tests. Results Immune-mismatched stem cell implants demonstrated stronger ferumoxytol enhancement than did matched stem cell implants. At 4 weeks, T2 values of mismatched implants were significantly lower than those of matched implants in osteochondral defects of female rats (mean, 10.72 msec for human stem cells and 11.55 msec for male rat stem cells vs 15.45 msec for sex-matched rat stem cells; P = .02 and P = .04, respectively) and calvarial defects of recipient mice (mean, 21.7 msec vs 27.1 msec, respectively; P = .0444). This corresponded to increased recruitment of enhanced GFP- and rhodamine-ferumoxytol-positive macrophages into stem cell transplants, as visualized with IVM and histopathologic examination. Conclusion Endogenous labeling of macrophages with ferumoxytol enables noninvasive detection of innate immune responses to stem cell transplants with MR imaging. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Heike E Daldrup-Link
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Carmel Chan
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Olga Lenkov
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Seyedmeghdad Taghavigarmestani
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Toktam Nazekati
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Hossein Nejadnik
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Fanny Chapelin
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Aman Khurana
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Xinming Tong
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Fan Yang
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Laura Pisani
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Michael Longaker
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Sanjiv Sam Gambhir
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| |
Collapse
|
22
|
D'Angelo W, Gurung C, Acharya D, Chen B, Ortolano N, Gama V, Bai F, Guo YL. The Molecular Basis for the Lack of Inflammatory Responses in Mouse Embryonic Stem Cells and Their Differentiated Cells. THE JOURNAL OF IMMUNOLOGY 2017; 198:2147-2155. [PMID: 28130495 DOI: 10.4049/jimmunol.1601068] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/30/2016] [Indexed: 12/12/2022]
Abstract
We reported previously that mouse embryonic stem cells do not have a functional IFN-based antiviral mechanism. The current study extends our investigation to the inflammatory response in mouse embryonic stem cells and mouse embryonic stem cell-differentiated cells. We demonstrate that LPS, TNF-α, and viral infection, all of which induce robust inflammatory responses in naturally differentiated cells, failed to activate NF-κB, the key transcription factor that mediates inflammatory responses, and were unable to induce the expression of inflammatory genes in mouse embryonic stem cells. Similar results were obtained in human embryonic stem cells. In addition to the inactive state of NF-κB, the deficiency in the inflammatory response in mouse embryonic stem cells is also attributed to the lack of functional receptors for LPS and TNF-α. In vitro differentiation can trigger the development of the inflammatory response mechanism, as indicated by the transition of NF-κB from its inactive to active state. However, a limited response to TNF-α and viral infection, but not to LPS, was observed in mouse embryonic stem cell-differentiated fibroblasts. We conclude that the inflammatory response mechanism is not active in mouse embryonic stem cells, and in vitro differentiation promotes only partial development of this mechanism. Together with our previous studies, the findings described in this article demonstrate that embryonic stem cells are fundamentally different from differentiated somatic cells in their innate immunity, which may have important implications in developmental biology, immunology, and embryonic stem cell-based regenerative medicine.
Collapse
Affiliation(s)
- William D'Angelo
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS 39406; and
| | - Chandan Gurung
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS 39406; and
| | - Dhiraj Acharya
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS 39406; and
| | - Bohan Chen
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS 39406; and
| | - Natalya Ortolano
- Department of Cell and Developmental Biology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| | - Fengwei Bai
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS 39406; and
| | - Yan-Lin Guo
- Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, MS 39406; and
| |
Collapse
|
23
|
van der Torren CR, Zaldumbide A, Duinkerken G, Brand-Schaaf SH, Peakman M, Stangé G, Martinson L, Kroon E, Brandon EP, Pipeleers D, Roep BO. Immunogenicity of human embryonic stem cell-derived beta cells. Diabetologia 2017; 60:126-133. [PMID: 27787618 PMCID: PMC6518073 DOI: 10.1007/s00125-016-4125-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/14/2016] [Indexed: 01/16/2023]
Abstract
AIMS/HYPOTHESIS To overcome the donor shortage in the treatment of advanced type 1 diabetes by islet transplantation, human embryonic stem cells (hESCs) show great potential as an unlimited alternative source of beta cells. hESCs may have immune privileged properties and it is important to determine whether these properties are preserved in hESC-derived cells. METHODS We comprehensively investigated interactions of both innate and adaptive auto- and allo-immunity with hESC-derived pancreatic progenitor cells and hESC-derived endocrine cells, retrieved after in-vivo differentiation in capsules in the subcutis of mice. RESULTS We found that hESC-derived pancreatic endodermal cells expressed relatively low levels of HLA endorsing protection from specific immune responses. HLA was upregulated when exposed to IFNγ, making these endocrine progenitor cells vulnerable to cytotoxic T cells and alloreactive antibodies. In vivo-differentiated endocrine cells were protected from complement, but expressed more HLA and were targets for alloreactive antibody-dependent cellular cytotoxicity and alloreactive cytotoxic T cells. After HLA compatibility was provided by transduction with HLA-A2, preproinsulin-specific T cells killed insulin-producing cells. CONCLUSIONS/INTERPRETATION hESC-derived pancreatic progenitors are hypoimmunogenic, while in vivo-differentiated endocrine cells represent mature targets for adaptive immune responses. Our data support the need for immune intervention in transplantation of hESC-derived pancreatic progenitors. Cell-impermeable macro-encapsulation may suffice.
Collapse
Affiliation(s)
- Cornelis R van der Torren
- Department of Immunohaematology and Blood Transfusion, E3-Q, Leiden University Medical Center, P.O. Box 9600, NL-2300 RC, Leiden, the Netherlands
- JDRF Center for Beta Cell Therapy in Diabetes
| | - Arnaud Zaldumbide
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Gaby Duinkerken
- Department of Immunohaematology and Blood Transfusion, E3-Q, Leiden University Medical Center, P.O. Box 9600, NL-2300 RC, Leiden, the Netherlands
- JDRF Center for Beta Cell Therapy in Diabetes
| | - Simone H Brand-Schaaf
- Department of Immunohaematology and Blood Transfusion, E3-Q, Leiden University Medical Center, P.O. Box 9600, NL-2300 RC, Leiden, the Netherlands
| | - Mark Peakman
- Department of Immunobiology, King's College London School of Medicine, London, UK
| | - Geert Stangé
- JDRF Center for Beta Cell Therapy in Diabetes
- Diabetes Research Center, Brussels Free University-VUB, Brussels, Belgium
| | | | | | | | - Daniel Pipeleers
- JDRF Center for Beta Cell Therapy in Diabetes
- Diabetes Research Center, Brussels Free University-VUB, Brussels, Belgium
| | - Bart O Roep
- Department of Immunohaematology and Blood Transfusion, E3-Q, Leiden University Medical Center, P.O. Box 9600, NL-2300 RC, Leiden, the Netherlands.
- JDRF Center for Beta Cell Therapy in Diabetes, .
- Department of Diabetes Immunology, Diabetes and Metabolism Research Institute at the Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
24
|
Cellular therapy in tuberculosis. Int J Infect Dis 2016; 32:32-8. [PMID: 25809753 DOI: 10.1016/j.ijid.2015.01.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 01/04/2023] Open
Abstract
Cellular therapy now offer promise of potential adjunct therapeutic options for treatment of drug-resistant tuberculosis (TB). We review here the role of Mesenchymal stromal cells, (MSCs), as well as other immune effector cells in the therapy of infectious diseases with a focus on TB. MSCs represent a population of tissue-resident non-hematopoietic adult progenitor cells which home into injured tissues increase the proliferative potential of broncho-alveolar stem cells and restore lung epithelium. MSCs have been shown to be immune-modulatory and anti-inflammatory mediated via cell-cell contacts as well as soluble factors. We discuss the functional profile of MSCs and their potential use for adjunct cellular therapy of multi-drug resistant TB, with the aim of limiting tissue damage, and to convert unproductive inflammatory responses into effective anti-pathogen directed immune responses. Adjunct cellular therapy could potentially offer salvage therapy options for patients with drug-resistant TB, increase clinically relevant anti-M.tuberculosis directed immune responses and possibly shorten the duration of anti-TB therapy.
Collapse
|
25
|
NAMBA HIROKI, KAWAJI HIROSHI, YAMASAKI TOMOHIRO. Use of genetically engineered stem cells for glioma therapy. Oncol Lett 2016; 11:9-15. [PMID: 26870161 PMCID: PMC4726949 DOI: 10.3892/ol.2015.3860] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 09/24/2015] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma, the most common and most malignant type of primary brain tumor, is associated with poor prognosis, even when treated using combined therapies, including surgery followed by concomitant radiotherapy with temozolomide-based chemotherapy. The invasive nature of this type of tumor is a major reason underlying treatment failure. The tumor-tropic ability of neural and mesenchymal stem cells offers an alternative therapeutic approach, where these cells may be used as vehicles for the invasion of tumors. Stem cell-based therapy is particularly attractive due to its tumor selectivity, meaning that the stem cells are able to target tumor cells without harming healthy brain tissue, as well as the extensive tumor tropism of stem cells when delivering anti-tumor substances, even to distant tumor microsatellites. Stem cells have previously been used to deliver cytokine genes, suicide genes and oncolytic viruses. The present review will summarize current trends in experimental studies of stem cell-based gene therapy against gliomas, and discuss the potential concerns for translating these promising strategies into clinical use.
Collapse
Affiliation(s)
- HIROKI NAMBA
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - HIROSHI KAWAJI
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - TOMOHIRO YAMASAKI
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| |
Collapse
|
26
|
The Epigenetic Reprogramming Roadmap in Generation of iPSCs from Somatic Cells. J Genet Genomics 2015; 42:661-70. [PMID: 26743984 DOI: 10.1016/j.jgg.2015.10.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 10/09/2015] [Accepted: 10/15/2015] [Indexed: 12/30/2022]
Abstract
Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) is a comprehensive epigenetic process involving genome-wide modifications of histones and DNA methylation. This process is often incomplete, which subsequently affects iPSC reprogramming, pluripotency, and differentiation capacity. Here, we review the epigenetic changes with a focus on histone modification (methylation and acetylation) and DNA modification (methylation) during iPSC induction. We look at changes in specific epigenetic signatures, aberrations and epigenetic memory during reprogramming and small molecules influencing the epigenetic reprogramming of somatic cells. Finally, we discuss how to improve iPSC generation and pluripotency through epigenetic manipulations.
Collapse
|
27
|
Stem cell based therapies for age-related macular degeneration: The promises and the challenges. Prog Retin Eye Res 2015; 48:1-39. [PMID: 26113213 DOI: 10.1016/j.preteyeres.2015.06.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 12/21/2022]
|
28
|
Schuetz C, Markmann JF. Immunogenicity of β-cells for autologous transplantation in type 1 diabetes. Pharmacol Res 2015; 98:60-8. [DOI: 10.1016/j.phrs.2015.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/05/2015] [Accepted: 03/05/2015] [Indexed: 12/15/2022]
|
29
|
Guo YL, Carmichael GG, Wang R, Hong X, Acharya D, Huang F, Bai F. Attenuated Innate Immunity in Embryonic Stem Cells and Its Implications in Developmental Biology and Regenerative Medicine. Stem Cells 2015; 33:3165-73. [PMID: 26086534 DOI: 10.1002/stem.2079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/16/2015] [Indexed: 12/14/2022]
Abstract
Embryonic stem cells (ESCs) represent a promising cell source for regenerative medicine. Intensive research over the past 2 decades has led to the feasibility of using ESC-differentiated cells (ESC-DCs) in regenerative medicine. However, increasing evidence indicates that ESC-DCs generated by current differentiation methods may not have equivalent cellular functions to their in vivo counterparts. Recent studies have revealed that both human and mouse ESCs as well as some types of ESC-DCs lack or have attenuated innate immune responses to a wide range of infectious agents. These findings raise important concerns for their therapeutic applications since ESC-DCs, when implanted to a wound site of a patient, where they would likely be exposed to pathogens and inflammatory cytokines. Understanding whether an attenuated immune response is beneficial or harmful to the interaction between host and grafted cells becomes an important issue for ESC-based therapy. A substantial amount of recent evidence has demonstrated that the lack of innate antiviral responses is a common feature to ESCs and other types of pluripotent cells. This has led to the hypothesis that mammals may have adapted different antiviral mechanisms at different stages of organismal development. The underdeveloped innate immunity represents a unique and uncharacterized property of ESCs that may have important implications in developmental biology, immunology, and in regenerative medicine.
Collapse
Affiliation(s)
- Yan-Lin Guo
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Gordon G Carmichael
- The Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Ruoxing Wang
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Xiaoxiao Hong
- The Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Dhiraj Acharya
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Faqing Huang
- The Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Fengwei Bai
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| |
Collapse
|
30
|
Abstract
Human adult mesenchymal stem cells are present in fat tissue, which can be obtained using surgical procedures such as liposuction. The multilineage capacity of mesenchymal stem cells makes them very valuable for cell-based medical therapies. In this chapter, we describe how to isolate mesenchymal stem cells from human adult fat tissue, propagate the cells in culture, and cryopreserve the cells for tissue engineering applications. Flow cytometry methods are also described for identification and characterization of adipose-derived stem cells and for cell sorting.
Collapse
Affiliation(s)
- Nastaran Mahmoudifar
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Pauline M Doran
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, P.O. Box 218, Hawthorn, Melbourne, VIC, 3122, Australia.
| |
Collapse
|
31
|
Abstract
Many technologies that underpin tissue engineering as a research field were developed with the aim of producing functional human cartilage in vitro. Much of our practical experience with three-dimensional cultures, tissue bioreactors, scaffold materials, stem cells, and differentiation protocols was gained using cartilage as a model system. Despite these advances, however, generation of engineered cartilage matrix with the composition, structure, and mechanical properties of mature articular cartilage has not yet been achieved. Currently, the major obstacles to synthesis of clinically useful cartilage constructs are our inability to control differentiation to the extent needed, and the failure of engineered and host tissues to integrate after construct implantation. The aim of this chapter is to distil from the large available body of literature the seminal approaches and experimental techniques developed for cartilage tissue engineering and to identify those specific areas requiring further research effort.
Collapse
Affiliation(s)
- Pauline M Doran
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, 218, Hawthorn, Melbourne, VIC, 3122, Australia.
| |
Collapse
|
32
|
Tan Y, Ooi S, Wang L. Immunogenicity and tumorigenicity of pluripotent stem cells and their derivatives: genetic and epigenetic perspectives. Curr Stem Cell Res Ther 2014; 9:63-72. [PMID: 24160683 PMCID: PMC3873036 DOI: 10.2174/1574888x113086660068] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 10/19/2013] [Accepted: 10/22/2013] [Indexed: 12/18/2022]
Abstract
One aim of stem cell-based therapy is to utilize pluripotent stem cells (PSCs) as a supplementary source of cells
to repair or replace tissues or organs that have ceased to function due to severe tissue damage. However, PSC-based therapy
requires extensive research to ascertain if PSC derivatives are functional without the risk of tumorigenicity, and also
do not engender severe immune rejection that threatens graft survival and function. Recently, the suitability of induced
pluripotent stem cells applied for patient-tailored cell therapy has been questioned since the discovery of several genetic
and epigenetic aberrations during the reprogramming process. Hence, it is crucial to understand the effect of these abnormalities
on the immunogenicity and survival of PSC grafts. As induced PSC-based therapy represents a hallmark for the
potential solution to prevent and arrest immune rejection, this review also summarizes several up-to-date key findings in
the field.
Collapse
Affiliation(s)
| | | | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa K1H8M5, Canada.
| |
Collapse
|
33
|
Induced pluripotent stem (iPS) cells: A new source for cell-based therapeutics? J Control Release 2014; 185:37-44. [DOI: 10.1016/j.jconrel.2014.04.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/08/2014] [Accepted: 04/08/2014] [Indexed: 12/18/2022]
|
34
|
Földes G, Mioulane M, Kodagoda T, Lendvai Z, Iqbal A, Ali NN, Schneider MD, Harding SE. Immunosuppressive Agents Modulate Function, Growth, and Survival of Cardiomyocytes and Endothelial Cells Derived from Human Embryonic Stem Cells. Stem Cells Dev 2014; 23:467-76. [DOI: 10.1089/scd.2013.0229] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Gábor Földes
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Maxime Mioulane
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | - Thusharika Kodagoda
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | | | - Adeel Iqbal
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | - Nadire N. Ali
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | - Michael D. Schneider
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| | - Sian E. Harding
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, London, United Kingdom
| |
Collapse
|
35
|
Ljung K, Simonson OE, Felldin U, Wärdell E, Ibarra C, Antonsson L, Kumagai-Braesch M, Hovatta O, Lampela R, Grinnemo KH, Corbascio M. Costimulation blockade induces foxp3(+) regulatory T cells to human embryonic stem cells. Biores Open Access 2013; 2:455-8. [PMID: 24380056 PMCID: PMC3869426 DOI: 10.1089/biores.2013.0039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Transplantation of human embryonic stem cells (hESCs), like other allogeneic cellular transplants, require immunomodulation or immunosuppression in order to be maintained in the recipient. Costimulation blockade applied at the time of transplantation inhibits costimulatory signals in the immunological synapse leading to a state of anergy in the donor reactive T-cell population and a state of immunological tolerance in the host. In models of solid organ transplantation, tolerance is maintained by the infiltration of Foxp3+ regulatory T cells into the graft. In order to study if regulatory T cells could be generated to hESC transplants, costimulation blockade (CTLA4Ig, anti-CD40L, anti-LFA-1) was administered for the first week after transplantation of two different hESC lines implanted under the kidney capsule of wild-type mice. hESC transplants were maintained indefinitely, and when harvested at long-term follow-up, Foxp3+ T-cells were found surrounding the graft, implying the maintenance of tolerance through the induction of regulatory T cells. These results imply that costimulation blockade could be a useful treatment strategy for the induction of tolerance to hESC transplants and may down-modulate immune responses locally around the graft.
Collapse
Affiliation(s)
- Karin Ljung
- Department of Emergency Medicine (Huddinge), Karolinska University Hospital , Stockholm, Sweden
| | - Oscar E Simonson
- Department of Cardiothoracic Surgery & Anesthesiology (Solna), Karolinska University Hospital , Stockholm, Sweden
| | - Ulrika Felldin
- Department of Molecular Medicine & Surgery, Karolinska Institute , Stockholm, Sweden
| | - Ewa Wärdell
- Department of Medicine, Karolinska Institute , Stockholm, Sweden
| | - Cristian Ibarra
- Department of Medical Biochemistry & Biophysics, Karolinska Institute , Stockholm, Sweden
| | - Liselotte Antonsson
- Department of Clinical Science, Intervention, & Technology, Karolinska Institute , Stockholm, Sweden
| | - Makiko Kumagai-Braesch
- Department of Clinical Science, Intervention, & Technology, Karolinska Institute , Stockholm, Sweden
| | - Outi Hovatta
- Department of Clinical Science, Intervention, & Technology, Karolinska Institute , Stockholm, Sweden
| | - Riina Lampela
- Department of Clinical Science, Intervention, & Technology, Karolinska Institute , Stockholm, Sweden
| | - Karl-Henrik Grinnemo
- Department of Cardiothoracic Surgery & Anesthesiology (Solna), Karolinska University Hospital , Stockholm, Sweden
| | - Matthias Corbascio
- Department of Cardiothoracic Surgery & Anesthesiology (Solna), Karolinska University Hospital , Stockholm, Sweden
| |
Collapse
|
36
|
Sinclair K, Yerkovich ST, Chambers DC. Mesenchymal stem cells and the lung. Respirology 2013; 18:397-411. [DOI: 10.1111/resp.12050] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 11/28/2012] [Accepted: 01/01/2013] [Indexed: 12/20/2022]
|
37
|
Li O, English K, Tonlorenzi R, Cossu G, Saverio Tedesco F, Wood KJ. Human iPSC-derived mesoangioblasts, like their tissue-derived counterparts, suppress T cell proliferation through IDO- and PGE-2-dependent pathways. F1000Res 2013; 2:24. [PMID: 24715949 PMCID: PMC3968899 DOI: 10.12688/f1000research.2-24.v1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/12/2013] [Indexed: 11/22/2022] Open
Abstract
Human mesoangioblasts are currently in a phase I/II clinical trial for the treatment of patients with Duchenne muscular dystrophy. However, limitations associated with the finite life span of these cells combined with the significant numbers of mesoangioblasts required to treat all of the skeletal muscles in these patients restricts their therapeutic potential. Induced pluripotent stem cell (iPSC)-derived mesoangioblasts may provide the solution to this problem. Although, the idea of using iPSC-derived cell therapies has been proposed for quite some time, our understanding of how the immune system interacts with these cells is inadequate. Herein, we show that iPSC-derived mesoangioblasts (HIDEMs) from healthy donors and, importantly, limb-girdle muscular dystrophy 2D patients exert immunosuppressive effects on T cell proliferation. Interferon gamma (IFN-γ) and tumour necrosis factor alpha (TNF-α) play crucial roles in the initial activation of HIDEMs and importantly indoleamine 2,3 dioxygenase (IDO) and prostaglandin E2 (PGE-2) were identified as key mechanisms involved in HIDEM suppression of T cell proliferation. Together with recent studies confirming the myogenic function and regenerative potential of these cells, we suggest that HIDEMs could provide an unlimited alternative source for mesoangioblast-based therapies.
Collapse
Affiliation(s)
- Ou Li
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Karen English
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Cellular Immunology Group, Institute of Immunology, National University of Ireland Maynooth, Co. Kildare, Ireland
| | - Rossana Tonlorenzi
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Cossu
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy.,Department of Cell and Developmental Biology and Centre for Stem Cells and Regenerative Medicine, University College London, London, UK
| | - Francesco Saverio Tedesco
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy.,Department of Cell and Developmental Biology and Centre for Stem Cells and Regenerative Medicine, University College London, London, UK
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Al-Adra DP, Anderson CC. Mixed chimerism and split tolerance: mechanisms and clinical correlations. CHIMERISM 2013; 2:89-101. [PMID: 22509425 DOI: 10.4161/chim.2.4.19017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Establishing hematopoietic mixed chimerism can lead to donor-specific tolerance to transplanted organs and may eliminate the need for long-term immunosuppressive therapy, while also preventing chronic rejection. In this review, we discuss central and peripheral mechanisms of chimerism induced tolerance. However, even in the long-lasting presence of a donor organ or donor hematopoietic cells, some allogeneic tissues from the same donor can be rejected; a phenomenon known as split tolerance. With the current goal of creating mixed chimeras using clinically feasible amounts of donor bone marrow and with minimal conditioning, split tolerance may become more prevalent and its mechanisms need to be explored. Some predisposing factors that may increase the likelihood of split tolerance are immunogenicity of the graft, certain donor-recipient combinations, prior sensitization, location and type of graft and minimal conditioning chimerism induction protocols. Additionally, split tolerance may occur due to a differential susceptibility of various types of tissues to rejection. The mechanisms involved in a tissue's differential susceptibility to rejection include the presence of polymorphic tissue-specific antigens and variable sensitivity to indirect pathway effector mechanisms. Finally, we review the clinical attempts at allograft tolerance through the induction of chimerism; studies that are revealing the complex relationship between chimerism and tolerance. This relationship often displays split tolerance, and further research into its mechanisms is warranted.
Collapse
Affiliation(s)
- David P Al-Adra
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AB Canada
| | | |
Collapse
|
39
|
Loewendorf A, Csete M. Concise review: immunologic lessons from solid organ transplantation for stem cell-based therapies. Stem Cells Transl Med 2013; 2:136-42. [PMID: 23349327 DOI: 10.5966/sctm.2012-0125] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Clinical organ transplantation became possible only after powerful immunosuppressive drugs became available to suppress the alloimmune response. After decades of solid organ transplantation, organ rejection is still a major challenge. However, significant insight into allorecognition has emerged from this vast experience and should be used to inform future stem cell-based therapies. For this reason, we review the current understanding of selected topics in transplant immunology that have not been prominent in the stem cell literature, including immune responses to ischemia/reperfusion injuries, natural killer cells, the adaptive immune response, some unresolved issues in T-cell allorecognition, costimulatory molecules, and the anticipated role of regulatory T cells in graft tolerance.
Collapse
Affiliation(s)
- Andrea Loewendorf
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA.
| | | |
Collapse
|
40
|
Hayes M, Curley G, Ansari B, Laffey JG. Clinical review: Stem cell therapies for acute lung injury/acute respiratory distress syndrome - hope or hype? CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:205. [PMID: 22424108 PMCID: PMC3681334 DOI: 10.1186/cc10570] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A growing understanding of the complexity of the pathophysiology of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), coupled with advances in stem cell biology, has led to a renewed interest in the therapeutic potential of stem cells for this devastating disease. Mesenchymal stem cells appear closest to clinical translation, given the evidence that they may favourably modulate the immune response to reduce lung injury, while maintaining host immune-competence and also facilitating lung regeneration and repair. The demonstration that human mesenchymal stem cells exert benefit in the endotoxin-injured human lung is particularly persuasive. Endothelial progenitor cells also demonstrate promise in reducing endothelial damage, which is a key pathophysiological feature of ALI. Embryonic and induced pluripotent stem cells are at an earlier stage in the translational process, but offer the hope of directly replacing injured lung tissue. The lung itself also contains endogenous stem cells, which may ultimately offer the greatest hope for lung diseases, given their physiologic role in replacing and regenerating native lung tissues. However, significant deficits remain in our knowledge regarding the mechanisms of action of stem cells, their efficacy in relevant pre-clinical models, and their safety, particularly in critically ill patients. These gaps need to be addressed before the enormous therapeutic potential of stem cells for ALI/ARDS can be realised.
Collapse
Affiliation(s)
- Mairead Hayes
- Lung Biology Group, Regenerative Medicine Institute, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | | | | | | |
Collapse
|
41
|
English K, Tonlorenzi R, Cossu G, Wood KJ. Mesoangioblasts suppress T cell proliferation through IDO and PGE-2-dependent pathways. Stem Cells Dev 2012; 22:512-23. [PMID: 22913669 DOI: 10.1089/scd.2012.0386] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Human mesoangioblasts are vessel-associated stem cells that are currently in phase I/II clinical trials for the treatment of patients with Duchenne muscular dystrophy. To date, little is known about the effect of mesoangioblasts on human immune cells and vice versa. We hypothesized that mesoangioblasts could modulate the function of immune cells in a similar manner to mesenchymal stromal cells. Human mesoangioblasts did not evoke, but rather potently suppressed human T-cell proliferation and effector function in vitro in a dose- and time-dependent manner. Furthermore, mesoangioblasts exert these inhibitory effects uniformly on human CD4+ and CD8+ T cells in a reversible manner without inducing a state of anergy. Interferon (IFN)-γ and tumor necrosis factor (TNF)-α play crucial roles in the initial activation of mesoangioblasts. Indoleamine 2,3-dioxygenase (IDO) and prostaglandin E-2 (PGE) were identified as key mechanisms of action involved in the mesoangioblast suppression of T-cell proliferation. Together, these data demonstrate a previously unrecognized capacity of mesoangioblasts to modulate immune responses.
Collapse
Affiliation(s)
- Karen English
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | | | | | | |
Collapse
|
42
|
Haag M, Stolk M, Ringe J, Linthout SV, Tschöpe C, Sittinger M, Seifert M. Immune attributes of cardiac-derived adherent proliferating (CAP) cells in cardiac therapy. J Tissue Eng Regen Med 2012; 7:362-70. [DOI: 10.1002/term.531] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 07/06/2011] [Accepted: 09/26/2011] [Indexed: 12/21/2022]
Affiliation(s)
| | - Meaghan Stolk
- Berlin-Brandenburg Centre for Regenerative Therapies; Charité-Universitätsmedizin Berlin; Germany
| | | | | | | | | | | |
Collapse
|
43
|
Binello E, Germano IM. Stem cells as therapeutic vehicles for the treatment of high-grade gliomas. Neuro Oncol 2011; 14:256-65. [PMID: 22166262 DOI: 10.1093/neuonc/nor204] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stem cells have generated great interest in the past decade as potential tools for cell-based treatment of human high-grade gliomas. Thus far, 3 types of stem cells have been tested as vehicles for various therapeutic agents: embryonic, neural, and mesenchymal. The types of therapeutic approaches and/or agents examined in the context of stem cell-based delivery include cytokines, enzyme/prodrug suicide combinations, viral particles, matrix metalloproteinases, and antibodies. Each strategy has specific advantages and disadvantages. Irrespective of the source and/or type of stem cell, there are several areas of concern for their translation to the clinical setting, such as migration in the adult human brain, potential teratogenesis, immune rejection, and regulatory and ethical issues. Nonetheless, a clinical trial is under way using neural stem cell-based delivery of an enzyme/prodrug suicide combination for recurrent high-grade glioma. A proposed future direction could encompass the use of stem cells as vehicles for delivery of agents targeting glioma stem cells, which have been implicated in the resistance of high-grade glioma to treatment. Overall, stem cells are providing an unprecedented opportunity for cell-based approaches in the treatment of high-grade gliomas, which have a persistently dismal prognosis and mandate a continued search for therapeutic options.
Collapse
Affiliation(s)
- Emanuela Binello
- Department of Neurosurgery, Mount Sinai School of Medicine, 1 Gustave L. Levy Place, Box 1136, New York, NY 10029, USA
| | | |
Collapse
|
44
|
Anderson AJ, Haus DL, Hooshmand MJ, Perez H, Sontag CJ, Cummings BJ. Achieving stable human stem cell engraftment and survival in the CNS: is the future of regenerative medicine immunodeficient? Regen Med 2011; 6:367-406. [PMID: 21548741 DOI: 10.2217/rme.11.22] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is potential for a variety of stem cell populations to mediate repair in the diseased or injured CNS; in some cases, this theoretical possibility has already transitioned to clinical safety testing. However, careful consideration of preclinical animal models is essential to provide an appropriate assessment of stem cell safety and efficacy, as well as the basic biological mechanisms of stem cell action. This article examines the lessons learned from early tissue, organ and hematopoietic grafting, the early assumptions of the stem cell and CNS fields with regard to immunoprivilege, and the history of success in stem cell transplantation into the CNS. Finally, we discuss strategies in the selection of animal models to maximize the predictive validity of preclinical safety and efficacy studies.
Collapse
Affiliation(s)
- Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, 845 Health Science Road, UC Irvine, Irvine, CA 92697-1705, USA.
| | | | | | | | | | | |
Collapse
|