1
|
Nyayapathi N, Zheng E, Zhou Q, Doyley M, Xia J. Dual-modal Photoacoustic and Ultrasound Imaging: from preclinical to clinical applications. FRONTIERS IN PHOTONICS 2024; 5:1359784. [PMID: 39185248 PMCID: PMC11343488 DOI: 10.3389/fphot.2024.1359784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Photoacoustic imaging is a novel biomedical imaging modality that has emerged over the recent decades. Due to the conversion of optical energy into the acoustic wave, photoacoustic imaging offers high-resolution imaging in depth beyond the optical diffusion limit. Photoacoustic imaging is frequently used in conjunction with ultrasound as a hybrid modality. The combination enables the acquisition of both optical and acoustic contrasts of tissue, providing functional, structural, molecular, and vascular information within the same field of view. In this review, we first described the principles of various photoacoustic and ultrasound imaging techniques and then classified the dual-modal imaging systems based on their preclinical and clinical imaging applications. The advantages of dual-modal imaging were thoroughly analyzed. Finally, the review ends with a critical discussion of existing developments and a look toward the future.
Collapse
Affiliation(s)
- Nikhila Nyayapathi
- Electrical and Computer Engineering, University of Rochester, Rochester, New York, 14627
| | - Emily Zheng
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York, 14226
| | - Qifa Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90007
| | - Marvin Doyley
- Electrical and Computer Engineering, University of Rochester, Rochester, New York, 14627
| | - Jun Xia
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York, 14226
| |
Collapse
|
2
|
Li SC. Mastering the craft: Creating an insightful and widely-cited literature review. World J Stem Cells 2023; 15:781-786. [PMID: 37700820 PMCID: PMC10494571 DOI: 10.4252/wjsc.v15.i8.781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/25/2023] Open
Abstract
The art of constructing an insightful literature review manuscript has witnessed an exemplar in the work of Oz et al (2023), wherein concept progression harmoniously merges with figures and tables. Reflecting on retrospective data science, it is evident that well-cited articles can wield a transformative influence on the Journal Citation Reports Impact Factor score, as exemplified by Robert Weinberg's landmark on cancer (Hanahan and Weinberg, 2011). Here, we aim to spotlight a commendable contribution by Tuba Oz, Ajeet Kaushik, and Małgorzata Kujawska in this issue while pivoting towards identifying the hallmarks of a subpar literature review-elements that hinder rather than promote advancement. The hurdles and roadblocks encountered within subpar literature reviews are multifold. Anticipation of emerging trends, identification of challenges, and exploration of solutions remain conspicuously absent. Original Contributions fail to surface amidst the vast sea of pre-existing literature, with noticeable gaps amplified by the lack of illustrative figures and tables. The manuscript, at times, assumes a skeletal form, reflecting an attempt to accommodate an excess of references, leading to convoluted sentences laden with citations. In contrast, a potent solution lies in adopting a comprehensive approach. A nuanced and critical evaluation of sources can culminate in a robust discussion, surpassing the mere summarization of conclusions drawn by others. This approach, often dismissed, holds the potential to elevate clarity, coherence, and logical flow, ultimately inviting engaged readership and coveted citations. The critical necessity of integrating visionary insights is underscored and achieved through a rigorous analysis of pivotal concepts and innovative ideas. Examples can be harnessed to elucidate the application of these solutions. We advocate a paradigm shift, urging literature review writers to embrace the readers' perspective. A literature review's purpose extends beyond providing a comprehensive panorama; it should illuminate avenues for concept development within a specific field of interest. By achieving this balance, literature reviews stand to captivate a devoted readership, paving the way for manuscripts that are both widely read and frequently cited. The pathway forward requires a fusion of astute analysis and visionary insights, shaping the future of literature review composition.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Neuro-oncology and Stem Cell Research Laboratory, Children's Hospital of Orange County, Department of Neurology, University of California-Irvine School of Medicine, Orange, CA 92868-3874, United States.
| |
Collapse
|
3
|
Islam J, So KH, Kc E, Moon HC, Kim A, Hyun SH, Kim S, Park YS. Transplantation of human embryonic stem cells alleviates motor dysfunction in AAV2-Htt171-82Q transfected rat model of Huntington's disease. Stem Cell Res Ther 2021; 12:585. [PMID: 34809707 PMCID: PMC8607638 DOI: 10.1186/s13287-021-02653-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human embryonic stem cells (hESCs) transplantation had shown to provide a potential source of cells in neurodegenerative disease studies and lead to behavioral recovery in lentivirus transfected or, toxin-induced Huntington's disease (HD) rodent model. Here, we aimed to observe if transplantation of superparamagnetic iron oxide nanoparticle (SPION)-labeled hESCs could migrate in the neural degenerated area and improve motor dysfunction in an AAV2-Htt171-82Q transfected Huntington rat model. METHODS All animals were randomly allocated into three groups at first: HD group, sham group, and control group. After six weeks, the animals of the HD group and sham group were again divided into two subgroups depending on animals receiving either ipsilateral or contralateral hESCs transplantation. We performed cylinder test and stepping test every two weeks after AAV2-Htt171-82Q injection and hESCs transplantation. Stem cell tracking was performed once per two weeks using T2 and T2*-weighted images at 4.7 Tesla MRI. We also performed immunohistochemistry and immunofluorescence staining to detect the presence of hESCs markers, huntingtin protein aggregations, and iron in the striatum. RESULTS After hESCs transplantation, the Htt virus-injected rats exhibited significant behavioral improvement in behavioral tests. SPION labeled hESCs showed migration with hypointense signal in MRI. The cells were positive with βIII-tubulin, GABA, and DARPP32. CONCLUSION Collectively, our results suggested that hESCs transplantation can be a potential treatment for motor dysfunction of Huntington's disease.
Collapse
Affiliation(s)
- Jaisan Islam
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyoung Ha So
- Institute for Stem Cell & Regenerative Medicine (ISCRM), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Elina Kc
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyeong Cheol Moon
- Department of Neurosurgery, Gammaknife Icon Center, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Aryun Kim
- Department of Neurology, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Sang Hwan Hyun
- Institute for Stem Cell & Regenerative Medicine (ISCRM), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Soochong Kim
- Institute for Stem Cell & Regenerative Medicine (ISCRM), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Young Seok Park
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea.
- Institute for Stem Cell & Regenerative Medicine (ISCRM), College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.
- Department of Neurosurgery, Gammaknife Icon Center, Chungbuk National University Hospital, Cheongju, Republic of Korea.
- Department of Neurosurgery, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, 776, 1 Sunhwanro, Seowon-gu, Cheongju-si, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
4
|
Sun M, Xue X, Li L, Xu D, Li S, Li SC, Su Q. Ectosome biogenesis and release processes observed by using live-cell dynamic imaging in mammalian glial cells. Quant Imaging Med Surg 2021; 11:4604-4616. [PMID: 34737927 DOI: 10.21037/qims-20-1015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/09/2021] [Indexed: 01/08/2023]
Abstract
Background Ectosomes are recognized as shedding from the plasma membranes into the extracellular environment. Recent research has demonstrated that ectosomes are surrounded by phospholipid membranes containing lipid rafts and caveolae. Some ectosomes contain cytokines in the lumen and have high levels of phosphatidylserine exposed to the outer membrane. Intracellular vesicles share both characters with ectosomes. Why the plasma membrane-derived ectosomes have the same characteristics as intracellular vesicles remain largely unknown. Methods Using live-cell dynamic imaging, we recorded the process of ectosome biogenesis and release in primary cultured neural cells. Results Our results show two different ectosome release methods: slow-releasing and fast-releasing. In the slow-releasing, multiple ectosomes emerge almost simultaneously on the cell surface and are released by outward budding from the plasma membrane. In the fast releasing, ectosomes squeeze out of the membrane domain and pinch off from a cell's surface. Using ER-tracker for live-cell imaging, we directly observed the process that intracellular vesicles jump out of the plasma membrane for release. This type of ectosomes has a reverse array of membrane proteins and phospholipids compared to the plasma membrane. So ectosomes should be divided into two groups: plasma membrane-derived and intracellular membrane-derived ectosomes. Conclusions Both slow releasing and fast releasing EVs imply mechanisms of human diseases and for diagnostics and drug delivery.
Collapse
Affiliation(s)
- Mengjiao Sun
- Bioengineering Research Center, School of Medicine, Shenzhen University, Shenzhen, China
| | - Xiufen Xue
- Bioengineering Research Center, School of Medicine, Shenzhen University, Shenzhen, China
| | - Lingyun Li
- Bioengineering Research Center, School of Medicine, Shenzhen University, Shenzhen, China
| | - Dandan Xu
- Nephrology Department, Shenzhen Nanshan People's Hospital, the 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Shihe Li
- Bioengineering Research Center, School of Medicine, Shenzhen University, Shenzhen, China
| | - Shengwen Calvin Li
- Neuro-oncology and Stem Cell Research Laboratory (NSCL), CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC), Orange, CA, USA.,Department of Neurology, University of California-Irvine (UCI) School of Medicine, Orange, CA, USA
| | - Qingning Su
- Bioengineering Research Center, School of Medicine, Shenzhen University, Shenzhen, China
| |
Collapse
|
5
|
Friedrich RP, Cicha I, Alexiou C. Iron Oxide Nanoparticles in Regenerative Medicine and Tissue Engineering. NANOMATERIALS 2021; 11:nano11092337. [PMID: 34578651 PMCID: PMC8466586 DOI: 10.3390/nano11092337] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022]
Abstract
In recent years, many promising nanotechnological approaches to biomedical research have been developed in order to increase implementation of regenerative medicine and tissue engineering in clinical practice. In the meantime, the use of nanomaterials for the regeneration of diseased or injured tissues is considered advantageous in most areas of medicine. In particular, for the treatment of cardiovascular, osteochondral and neurological defects, but also for the recovery of functions of other organs such as kidney, liver, pancreas, bladder, urethra and for wound healing, nanomaterials are increasingly being developed that serve as scaffolds, mimic the extracellular matrix and promote adhesion or differentiation of cells. This review focuses on the latest developments in regenerative medicine, in which iron oxide nanoparticles (IONPs) play a crucial role for tissue engineering and cell therapy. IONPs are not only enabling the use of non-invasive observation methods to monitor the therapy, but can also accelerate and enhance regeneration, either thanks to their inherent magnetic properties or by functionalization with bioactive or therapeutic compounds, such as drugs, enzymes and growth factors. In addition, the presence of magnetic fields can direct IONP-labeled cells specifically to the site of action or induce cell differentiation into a specific cell type through mechanotransduction.
Collapse
|
6
|
Glover JC, Aswendt M, Boulland JL, Lojk J, Stamenković S, Andjus P, Fiori F, Hoehn M, Mitrecic D, Pavlin M, Cavalli S, Frati C, Quaini F. In vivo Cell Tracking Using Non-invasive Imaging of Iron Oxide-Based Particles with Particular Relevance for Stem Cell-Based Treatments of Neurological and Cardiac Disease. Mol Imaging Biol 2021; 22:1469-1488. [PMID: 31802361 DOI: 10.1007/s11307-019-01440-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell-based therapeutics is a rapidly developing field associated with a number of clinical challenges. One such challenge lies in the implementation of methods to track stem cells and stem cell-derived cells in experimental animal models and in the living patient. Here, we provide an overview of cell tracking in the context of cardiac and neurological disease, focusing on the use of iron oxide-based particles (IOPs) visualized in vivo using magnetic resonance imaging (MRI). We discuss the types of IOPs available for such tracking, their advantages and limitations, approaches for labeling cells with IOPs, biological interactions and effects of IOPs at the molecular and cellular levels, and MRI-based and associated approaches for in vivo and histological visualization. We conclude with reviews of the literature on IOP-based cell tracking in cardiac and neurological disease, covering both preclinical and clinical studies.
Collapse
Affiliation(s)
- Joel C Glover
- Laboratory for Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, Norway. .,Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway.
| | - Markus Aswendt
- Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich, Leo-Brandt-Str. 5, 52425, Jülich, Germany
| | - Jean-Luc Boulland
- Laboratory for Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway
| | - Jasna Lojk
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, Ljubljana, Slovenia
| | - Stefan Stamenković
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, PB 52, 10001 Belgrade, Serbia
| | - Pavle Andjus
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, PB 52, 10001 Belgrade, Serbia
| | - Fabrizio Fiori
- Department of Applied Physics, Università Politecnica delle Marche - Di.S.C.O., Via Brecce Bianche, 60131, Ancona, Italy
| | - Mathias Hoehn
- Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich, Leo-Brandt-Str. 5, 52425, Jülich, Germany
| | - Dinko Mitrecic
- Laboratory for Stem Cells, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mojca Pavlin
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, Ljubljana, Slovenia.,Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Stefano Cavalli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | |
Collapse
|
7
|
Zhang XM, Chen DG, Li SC, Zhu B, Li ZJ. Embryonic Origin and Subclonal Evolution of Tumor-Associated Macrophages Imply Preventive Care for Cancer. Cells 2021; 10:903. [PMID: 33919979 PMCID: PMC8071014 DOI: 10.3390/cells10040903] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 01/16/2023] Open
Abstract
Macrophages are widely distributed in tissues and function in homeostasis. During cancer development, tumor-associated macrophages (TAMs) dominatingly support disease progression and resistance to therapy by promoting tumor proliferation, angiogenesis, metastasis, and immunosuppression, thereby making TAMs a target for tumor immunotherapy. Here, we started with evidence that TAMs are highly plastic and heterogeneous in phenotype and function in response to microenvironmental cues. We pointed out that efforts to tear off the heterogeneous "camouflage" in TAMs conduce to target de facto protumoral TAMs efficiently. In particular, several fate-mapping models suggest that most tissue-resident macrophages (TRMs) are generated from embryonic progenitors, and new paradigms uncover the ontogeny of TAMs. First, TAMs from embryonic modeling of TRMs and circulating monocytes have distinct transcriptional profiling and function, suggesting that the ontogeny of TAMs is responsible for the functional heterogeneity of TAMs, in addition to microenvironmental cues. Second, metabolic remodeling helps determine the mechanism of phenotypic and functional characteristics in TAMs, including metabolic bias from macrophages' ontogeny in macrophages' functional plasticity under physiological and pathological conditions. Both models aim at dissecting the ontogeny-related metabolic regulation in the phenotypic and functional heterogeneity in TAMs. We argue that gleaning from the single-cell transcriptomics on subclonal TAMs' origins may help understand the classification of TAMs' population in subclonal evolution and their distinct roles in tumor development. We envision that TAM-subclone-specific metabolic reprogramming may round-up with future cancer therapies.
Collapse
Affiliation(s)
- Xiao-Mei Zhang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing 400037, China;
| | - De-Gao Chen
- Institute of Cancer, The Second Affiliated Hospital, Army Military Medical University, Chongqing 400037, China;
| | - Shengwen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children’s Research Institute, Children’s Hospital of Orange County (CHOC), 1201 West La Veta Ave., Orange, CA 92868, USA
- Department of Neurology, University of California-Irvine School of Medicine, 200 S Manchester Ave., Ste 206, Orange, CA 92868, USA
| | - Bo Zhu
- Institute of Cancer, The Second Affiliated Hospital, Army Military Medical University, Chongqing 400037, China;
| | - Zhong-Jun Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing 400037, China;
| |
Collapse
|
8
|
Song Y, Yan S, Fan W, Zhang M, Liu W, Lu H, Cao M, Hao C, Chen L, Tian F, Zhan Y, Cai L, Xing Y. Identification and Validation of the Immune Subtypes of Lung Adenocarcinoma: Implications for Immunotherapy. Front Cell Dev Biol 2020; 8:550. [PMID: 32719796 PMCID: PMC7348081 DOI: 10.3389/fcell.2020.00550] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is a devastating disease with poor patient survival. Cancer immunotherapy has revolutionized the treatment of LUAD, but only a limited number of patients effectively respond to this treatment. Thus, the work to elucidate the LUAD immune heterogeneity could be crucial in developing new immunotherapeutic strategies with better efficacy. Non-negative matrix factorization-based deconvolution was performed to identify robust clusters of 489 LUAD patients in The Cancer Genome Atlas (TCGA) and verify their reproducibility and stability in an independent LUAD cohort of 439 patients from the Gene Expression Omnibus (GEO). We used the graph learning-based dimensionality reduction to visualize the distribution of individual patients. In this study, four reproducible immune subtypes, Clusters 1-4 (C1-C4) associated with distinct gene module signatures, clinicopathological features, molecular and cellular characteristics were identified and validated. The immune-cold subtype, C3, was associated with the Dead event, the most advanced T stage, N stage, TNM stage and the worst prognosis for LUAD patients. Moreover, C3 exhibited the lowest infiltrating levels of B cells, T cell receptor (TCR) repertoire diversity and the highest level of neoantigen and mutation rate among C1-C4. On the other hand, the immune-hot subtype (C4) exhibited the highest infiltration of six types of infiltrating immune cells as well as the greatest leukocyte fraction, TCR and B cell receptor (BCR) repertoire diversity. C1 and C2 subtypes showed diverse clinicopathological and immunological features. Finally, our investigations discovered a complex immune landscape with a scattered immune subtype profile. This work may help inform immunotherapeutic decision-making and design advanced immunotherapy strategies for the treatment of lung cancer.
Collapse
Affiliation(s)
- Yang Song
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shi Yan
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Weina Fan
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mengyan Zhang
- School of Life Sciences and Technology, Computational Biology Research Center, Harbin Institute of Technology, Harbin, China
| | - Wei Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hailing Lu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mengru Cao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chenguang Hao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lin Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanglin Tian
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuning Zhan
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Li Cai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ying Xing
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
9
|
Vives J, Casademont-Roca A, Martorell L, Nogués N. Beyond chimerism analysis: methods for tracking a new generation of cell-based medicines. Bone Marrow Transplant 2020; 55:1229-1239. [PMID: 32024991 DOI: 10.1038/s41409-020-0822-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 02/06/2023]
Abstract
The analysis of chimerism is crucial to determine the status of patients receiving hematopoietic stem cell transplantation. The variety of relevant techniques available today range from those that analyse nucleic acids (i.e. polymerase chain reaction based, next generation sequencing) and cellular phenotype (i.e. flow cytometry) to sophisticated imaging (particularly multimodal imaging using labelling agents). However, current developments of advanced therapies bring chimerism studies into a new dimension in which methods for detection of donor cells in the patient need to adapt to a wider range of cell- and gene-based medicines, routes of administration, target organs and pathologies. Herein we describe and analyze the toolkit of suitable labelling and detection methodologies with actual examples along with a discussion on challenges ahead and potential solutions. Remarkably, existing methods commonly used in chimerism analysis are suitable for use with new cell- and gene-based medicines. Indeed, new developments may facilitate the evolution and combination of such methodologies to the use of non-invasive and highly informative approaches.
Collapse
Affiliation(s)
- Joaquim Vives
- Servei de Teràpia Cel·lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain.
| | - Aina Casademont-Roca
- Servei de Teràpia Cel·lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain
| | - Lluís Martorell
- Servei de Teràpia Cel·lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain
| | - Núria Nogués
- Departament de Medicina, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain
- Laboratori d'Immunohematologia, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain
| |
Collapse
|
10
|
Zhang J, Su Q, Loudon WG, Lee KL, Luo J, Dethlefs BA, Li SC. Breathing Signature as Vitality Score Index Created by Exercises of Qigong: Implications of Artificial Intelligence Tools Used in Traditional Chinese Medicine. J Funct Morphol Kinesiol 2019; 4:71. [PMID: 31853512 PMCID: PMC6919646 DOI: 10.3390/jfmk4040071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022] Open
Abstract
Rising concerns about the short- and long-term detrimental consequences of administration of conventional pharmacopeia are fueling the search for alternative, complementary, personalized, and comprehensive approaches to human healthcare. Qigong, a form of Traditional Chinese Medicine, represents a viable alternative approach. Here, we started with the practical, philosophical, and psychological background of Ki (in Japanese) or Qi (in Chinese) and their relationship to Qigong theory and clinical application. Noting the drawbacks of the current state of Qigong clinic, herein we propose that to manage the unique aspects of the Eastern 'non-linearity' and 'holistic' approach, it needs to be integrated with the Western "linearity" "one-direction" approach. This is done through developing the concepts of "Qigong breathing signatures," which can define our life breathing patterns associated with diseases using machine learning technology. We predict that this can be achieved by establishing an artificial intelligence (AI)-Medicine training camp of databases, which will integrate Qigong-like breathing patterns with different pathologies unique to individuals. Such an integrated connection will allow the AI-Medicine algorithm to identify breathing patterns and guide medical intervention. This unique view of potentially connecting Eastern Medicine and Western Technology can further add a novel insight to our current understanding of both Western and Eastern medicine, thereby establishing a vitality score index (VSI) that can predict the outcomes of lifestyle behaviors and medical conditions.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Physical Training and Physical Therapy, Shenzhen University, 3688 Nanhai Avenue, Nanshan District, Shenzhen 518060, China
| | - Qingning Su
- Center of Bioengineering, School of Medicine, Shenzhen University, 3688 Nanhai Avenue, Nanshan District, Shenzhen 518060, China
| | - William G. Loudon
- Neuroscience Institute, Children’s Hospital of Orange County, Gamma Knife Center of Southern California, Department of Neurosurgery, University of California-Irvine School of Medicine, Orange, CA 92612, USA
| | - Katherine L. Lee
- School of Social Ecology, University of California-Irvine, 5300 Social and Behavioral Sciences Gateway, Irvine, CA 92697-7050, USA
| | - Jane Luo
- AB Sciex, Inc., Danaher Corporation, 250 South Kraemer Boulevard, Brea, CA 92821-6232, USA
| | - Brent A. Dethlefs
- CHOC Children’s Research Institute, Children’s Hospital of Orange County (CHOC), 1201 W. La Veta Ave., Orange, CA 92868-3874, USA
| | - Shengwen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory (NSCL), CHOC Children’s Research Institute (CCRI), Children’s Hospital of Orange County (CHOC), 1201 W. La Veta Ave., Orange, CA 92868-3874, USA
- Department of Neurology, University of California-Irvine (UCI) School of Medicine, 200 S Manchester Ave Ste 206, Orange, CA 92868, USA
| |
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: 1.4] [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
|
Miao Y, Chen Z, Li SC. Functional endoscopy techniques for tracking stem cell fate. Quant Imaging Med Surg 2019; 9:510-520. [PMID: 31032197 DOI: 10.21037/qims.2019.02.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Tracking and monitoring implanted stem cells are essential to maximize benefits and to minimize the side effects of stem cell therapy for personalized or "precision" medicine. Previously, we proposed a comprehensive biological Global Positioning System (bGPS) to track and monitor stem cells in vivo. Magnetic resonance imaging (MRI), positron emission tomography (PET), bioluminescent imaging, fluorescence imaging, and single-photon emission computerized tomography (SPECT) have been utilized to track labeled or genetically-modified cells in vivo in rats and humans. A large amount of research has been dedicated to the design of reporter genes and molecular probes for imaging and the visualization of the biodistribution of the implanted cells in high resolution. On the other hand, optical-based functional imaging, such as photoacoustic imaging (PAI), optical coherence tomography (OCT), and multiphoton microscopy (MPM), has been implemented into small endoscopes to image cells inside the body. The optical fiber allows miniaturization of the imaging probe while maintaining high resolution due to light-based imaging. Upon summarizing the recent progress in the design and application of functional endoscopy techniques for stem cell monitoring, we offer perspectives for the future development of endoscopic molecular imaging tools for in vivo tracking of spatiotemporal changes in subclonal evolution at the single cell level.
Collapse
Affiliation(s)
- Yusi Miao
- Beckman Laser Institute, University of California Irvine, Irvine, CA, USA
| | - Zhongping Chen
- Beckman Laser Institute, University of California Irvine, Irvine, CA, USA
| | - Shengwen Calvin Li
- Department of Neurology, University of California Irvine School of Medicine, Orange, CA, USA.,Department of Biological Science, California State University, Fullerton, CA, USA.,CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC), University of California Irvine, Orange, CA, USA
| |
Collapse
|
13
|
Onoshima D, Yukawa H, Baba Y. Nanobiodevices for Cancer Diagnostics and Stem Cell Therapeutics. Bioanalysis 2019. [DOI: 10.1007/978-981-13-6229-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
14
|
Santelli J, Lechevallier S, Baaziz H, Vincent M, Martinez C, Mauricot R, Parini A, Verelst M, Cussac D. Multimodal gadolinium oxysulfide nanoparticles: a versatile contrast agent for mesenchymal stem cell labeling. NANOSCALE 2018; 10:16775-16786. [PMID: 30156241 DOI: 10.1039/c8nr03263g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Despite a clear development of innovative therapies based on stem cell manipulation, the availability of new tools to better understand and follow stem cell behavior and improve their biomedical applications is not adequate. Indeed, an ideal tracking device must have good ability to label stem cells as well as complete neutrality relative to their biology. Furthermore, preclinical studies imply in vitro and in vivo approaches that often require several kinds of labeling and/or detection procedures. Consequently, the multimodality concept presented in this work may present a solution to this problem as it has the potential to combine complementary imaging techniques. Spherical europium-doped gadolinium oxysulfide (Gd2O2S:Eu3+) nanoparticles are presented as a candidate as they are detectable by (1) magnetic resonance (MRI), (2) X-ray and (3) photoluminescence imaging. Whole body in vivo distribution, elimination and toxicity evaluation revealed a high tolerance of nanoparticles with a long-lasting MRI signal and slow hepatobiliary and renal clearance. In vitro labeling of a wide variety of cells unveils the nanoparticle potential for efficient and universal cell tracking. Emphasis on mesenchymal stromal cells (MSCs) leads to the definition of optimal conditions for labeling and tracking in the context of cell therapy: concentrations below 50 μg mL-1 and diameters between 170 and 300 nm. Viability, proliferation, migration and differentiation towards mesodermal lineages are preserved under these conditions, and cell labeling appears to be persistent and without any leakage. Ex vivo detection of as few as five thousand Gd2O2S:Eu3+-labeled MSCs by MRI combined with in vitro examination with fluorescence microscopy highlights the feasibility of cell tracking in cell therapy using this new nanoplatform.
Collapse
Affiliation(s)
- Julien Santelli
- CEMES-CNRS, Université de Toulouse, CNRS 29, rue Jeanne Marvig, BP 94347, 31055 Toulouse Cedex 4, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Li SC, Stucky A, Chen X, Kabeer MH, Loudon WG, Plant AS, Torno L, Nangia CS, Cai J, Zhang G, Zhong JF. Single-cell transcriptomes reveal the mechanism for a breast cancer prognostic gene panel. Oncotarget 2018; 9:33290-33301. [PMID: 30279960 PMCID: PMC6161791 DOI: 10.18632/oncotarget.26044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022] Open
Abstract
The clinical benefits of the MammaPrint® signature for breast cancer is well documented; however, how these genes are related to cell cycle perturbation have not been well determined. Our single-cell transcriptome mapping (algorithm) provides details into the fine perturbation of all individual genes during a cell cycle, providing a view of the cell-cycle-phase specific landscape of any given human genes. Specifically, we identified that 38 out of the 70 (54%) MammaPrint® signature genes are perturbated to a specific phase of the cell cycle. The MammaPrint® signature panel derived its clinical prognosis power from measuring the cell cycle activity of specific breast cancer samples. Such cell cycle phase index of the MammaPrint® signature suggested that measurement of the cell cycle index from tumors could be developed into a prognosis tool for various types of cancer beyond breast cancer, potentially improving therapy through targeting a specific phase of the cell cycle of cancer cells.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Neuro-oncology and Stem Cell Research Laboratory, CHOC Children's Research Institute, Children's Hospital of Orange County; Department of Neurology, University of California-Irvine School of Medicine, Orange, CA, USA
| | - Andres Stucky
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Xuelian Chen
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Mustafa H. Kabeer
- Pediatric Surgery, CHOC Children's Hospital, Department of Surgery, University of California-Irvine School of Medicine, Orange, CA, USA
| | - William G. Loudon
- Neuroscience Institute, Children's Hospital of Orange County (CHOC), Gamma Knife Center of Southern California, Department of Neurosurgery, University of California-Irvine School of Medicine, Orange, CA, USA
| | - Ashley S. Plant
- Division of Pediatric Oncology, Children's Hospital of Orange County, Orange, CA, USA
| | - Lilibeth Torno
- Hyundai Cancer Institute at CHOC Children's Hospital, Oncology, Bone and Soft Tissue Tumor Program, After Cancer Treatment Survivorship Program, CHOC Children's Hospital, Orange, CA, USA
| | - Chaitali S. Nangia
- Chan Soon-Shiong Institute for Medicine, Verity Medical Foundation, Laguna Hills, CA, USA
| | - Jin Cai
- Department of Oral and Maxillofacial Surgery, Zhuhai People's Hospital, Zhuhai, China
| | - Gang Zhang
- Department of Oral and Maxillofacial Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jiang F. Zhong
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene, and Division of Biomedical Sciences, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
16
|
Li SC, Kabeer MH. Spatiotemporal switching signals for cancer stem cell activation in pediatric origins of adulthood cancer: Towards a watch-and-wait lifetime strategy for cancer treatment. World J Stem Cells 2018; 10:15-22. [PMID: 29531638 PMCID: PMC5840533 DOI: 10.4252/wjsc.v10.i2.15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 02/06/2023] Open
Abstract
Pediatric origin of cancer stem cell hypothesis holds great promise and potential in adult cancer treatment, however; the road to innovation is full of obstacles as there are plenty of questions left unanswered. First, the key question is to characterize the nature of such stem cells (concept). Second, the quantitative imaging of pediatric stem cells should be implemented (technology). Conceptually, pediatric stem cell origins of adult cancer are based on the notion that plasticity in early life developmental programming evolves local environments to cancer. Technologically, such imaging in children is lacking as all imaging is designed for adult patients. We postulate that the need for quantitative imaging to measure space-time changes of plasticity in early life developmental programming in children may trigger research and development of the imaging technology. Such quantitative imaging of pediatric origin of adulthood cancer will help develop a spatiotemporal monitoring system to determine cancer initiation and progression. Clinical validation of such speculative hypothesis-that cancer originates in a pediatric environment-will help implement a wait-and-watch strategy for cancer treatment.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Neuro-oncology and Stem Cell Research Laboratory, Children’s Hospital of Orange County, Department of Neurology, University of California-Irvine School of Medicine, Orange, CA 92868-3874, United States
| | - Mustafa H Kabeer
- Children's Hospital of Orange County, Department of Surgery, University of California-Irvine School of Medicine, Orange, CA 92868-3874, United States
| |
Collapse
|
17
|
British Society for Gene and Cell Therapy Annual Conference and Joint UK Regenerative Medicine Platform Meeting Royal Welsh College of Music & Drama Cardiff, Wales, United Kingdom Wednesday April 19-Friday April 21, 2017 Conference Abstracts. Hum Gene Ther 2017; 28:A1-A36. [PMID: 28796553 DOI: 10.1089/hum.2017.29044.abstracts] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
18
|
Yukawa H, Baba Y. In Vivo Fluorescence Imaging and the Diagnosis of Stem Cells Using Quantum Dots for Regenerative Medicine. Anal Chem 2017; 89:2671-2681. [PMID: 28194939 DOI: 10.1021/acs.analchem.6b04763] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hiroshi Yukawa
- Department of Applied Chemistry, Graduate School of Engineering, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,ImPACT Research Center for Advanced Nanobiodevices, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Yoshinobu Baba
- Department of Applied Chemistry, Graduate School of Engineering, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,ImPACT Research Center for Advanced Nanobiodevices, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Institute of Innovation for Future Society, Nagoya University , Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.,Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST) , 2217-14, Hayashi-cho, Takamatsu 761-0395, Japan.,College of Pharmacy, Kaohsiung Medical University , Shin-Chuan 1 st Rd., Kaohsiung, 807, Taiwan, R.O.C
| |
Collapse
|
19
|
Stem Cells and Labeling for Spinal Cord Injury. Int J Mol Sci 2016; 18:ijms18010006. [PMID: 28035961 PMCID: PMC5297641 DOI: 10.3390/ijms18010006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that usually results in sudden and long-lasting locomotor and sensory neuron degeneration below the lesion site. During the last two decades, the search for new therapies has been revolutionized with the improved knowledge of stem cell (SC) biology. SCs therapy offers several attractive strategies for spinal cord repair. The transplantation of SCs promotes remyelination, neurite outgrowth and axonal elongation, and activates resident or transplanted progenitor cells across the lesion cavity. However, optimized growth and differentiation protocols along with reliable safety assays should be established prior to the clinical application of SCs. Additionally, the ideal method of SCs labeling for efficient cell tracking after SCI remains a challenging issue that requires further investigation. This review summarizes the current findings on the SCs-based therapeutic strategies, and compares different SCs labeling approaches for SCI.
Collapse
|
20
|
Abstract
Stem cell-based drug delivery for cancer therapy has steadily gained momentum in the past decade as several studies have reported stem cells' inherent tropism towards tumors. Since this science is still in its early stages and there are many factors that could significantly impact tumor tropism of stem cells, some contradictory results have been observed. This review starts by examining a number of proof-of-concept studies that demonstrate the potential application of stem cells in cancer therapy. Studies that illustrate stem cells' tumor tropism and discuss the technical difficulties that could impact the therapeutic outcome are also highlighted. The discussion also emphasizes stem cell imaging/tracking, as it plays a crucial role in performing reliable dose-response studies and evaluating the therapeutic outcome of treatment protocols. In each section, the pros and cons associated with each method are highlighted, limitations are underlined, and potential solutions are discussed. The overall intention is to familiarize the reader with important practical issues related to stem cell cancer tropism and in vivo tracking, underline the shortcomings, and emphasize critical factors that need to be considered for effective translation of this science into the clinic.
Collapse
|
21
|
Zhang T, Jia W, Zhu Y, Yang J. Automatic tracking of neural stem cells in sequential digital images. Biocybern Biomed Eng 2016. [DOI: 10.1016/j.bbe.2015.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
22
|
Kim SM, Jeong CH, Woo JS, Ryu CH, Lee JH, Jeun SS. In vivo near-infrared imaging for the tracking of systemically delivered mesenchymal stem cells: tropism for brain tumors and biodistribution. Int J Nanomedicine 2015; 11:13-23. [PMID: 26719691 PMCID: PMC4690647 DOI: 10.2147/ijn.s97073] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based gene therapy is a promising tool for the treatment of various neurological diseases, including brain tumors. However, the tracking of in vivo stem cell migration, distribution, and survival need to be defined for their clinical application. The systemic routes of stem cell delivery must be determined because direct intracerebral injection as a cure for brain tumors is an invasive method. In this study, we show for the first time that near-infrared (NIR) imaging can reveal the distribution and tumor tropism of intravenously injected MSCs in an intracranial xenograft glioma model. MSCs were labeled with NIR fluorescent nanoparticles, and the effects of the NIR dye on cell proliferation and migratory capacity were evaluated in vitro. We investigated the tumor-targeting properties and tissue distribution of labeled MSCs introduced by intravenous injection and followed by in vivo imaging analysis, histological analysis, and real-time quantitative polymerase chain reaction. We observed no cytotoxicity or change in the overall growth rate and characteristics of labeled MSCs compared with control MSCs. NIR fluorescent imaging showed the organ distribution and targeted tumor tropism of systemically injected human MSCs. A significant number of MSCs accumulated specifically at the tumor site in the mouse brain. These results suggest that NIR-based cell tracking is a potentially useful imaging technique to visualize cell survival, migration, and distribution for the application of MSC-mediated therapies in the treatment of malignant gliomas.
Collapse
Affiliation(s)
- Seong Muk Kim
- Postech-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Chang Hyun Jeong
- Department of Neurosurgery, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Ji Sun Woo
- Department of Neurosurgery, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Chung Heon Ryu
- Postech-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sin-Soo Jeun
- Postech-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea ; Department of Neurosurgery, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
23
|
Onoshima D, Yukawa H, Baba Y. Multifunctional quantum dots-based cancer diagnostics and stem cell therapeutics for regenerative medicine. Adv Drug Deliv Rev 2015; 95:2-14. [PMID: 26344675 DOI: 10.1016/j.addr.2015.08.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/31/2015] [Accepted: 08/31/2015] [Indexed: 12/19/2022]
Abstract
A field of recent diagnostics and therapeutics has been advanced with quantum dots (QDs). QDs have developed into new formats of biomolecular sensing to push the limits of detection in biology and medicine. QDs can be also utilized as bio-probes or labels for biological imaging of living cells and tissues. More recently, QDs has been demonstrated to construct a multifunctional nanoplatform, where the QDs serve not only as an imaging agent, but also a nanoscaffold for diagnostic and therapeutic modalities. This review highlights the promising applications of multi-functionalized QDs as advanced nanosensors for diagnosing cancer and as innovative fluorescence probes for in vitro or in vivo stem cell imaging in regenerative medicine.
Collapse
|
24
|
Direct labeling of 19F-perfluorocarbon onto multilayered cell sheet for MRI-based non-invasive cell tracking. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-014-0092-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
25
|
Suhett GD, de Souza SAL, Carvalho AB, de Pinho Rachid R, da Cunha-E-Silva NL, de Carvalho ACC, da Fonseca LMB, dos Santos Goldenberg RC, Gutfilen B. 99m-Technetium binding site in bone marrow mononuclear cells. Stem Cell Res Ther 2015; 6:115. [PMID: 26041023 PMCID: PMC4473842 DOI: 10.1186/s13287-015-0107-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 05/28/2015] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION The increasing interest in 99m-technetium ((99m)Tc)-labeled stem cells encouraged us to study the (99m)Tc binding sites in stem cell compartments. METHODS Bone marrow mononuclear cells were collected from femurs and tibia of rats. Cells were labeled with (99m)Tc by a direct method, in which reduced molecules react with (99m)Tc with the use of chelating agents, and lysed carefully in an ultrasonic apparatus. The organelles were separated by means of differential centrifugation. At the end of this procedure, supernatants and pellets were counted, and the percentages of radioactivity (in megabecquerels) bound to the different cellular fractions were determined. Percentages were calculated by dividing the radioactivity in each fraction by total radioactivity in the sample. The pellets were separated and characterized by their morphology on electron microscopy. RESULTS The labeling procedure did not affect viability of bone marrow mononuclear cells. Radioactivity distributions in bone marrow mononuclear cell organelles, obtained in five independent experiments, were approximately 38.5 % in the nuclei-rich fraction, 5.3 % in the mitochondria-rich fraction, 2.2 % in microsomes, and 54 % in the cytosol. Our results showed that most of the radioactivity remained in the cytosol; therefore, this is an intracellular labeling procedure that has ribosomes unbound to membrane and soluble molecules as targets. However, approximately 39 % of the radioactivity remained bound to the nuclei-rich fraction. To confirm that cell disruption and organelle separation were efficient, transmission electron microscopy assays of all pellets were performed. CONCLUSIONS Our results showed that most of the radioactivity was present in the cytosol fraction. More studies to elucidate the mechanisms involved in the cellular uptake of (99m)Tc in bone marrow cells are ongoing.
Collapse
Affiliation(s)
- Grazielle Dias Suhett
- Laboratório de Cardiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Bloco G. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-902, Brasil.
| | - Sergio Augusto Lopes de Souza
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco, 255. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-913, Brasil.
| | - Adriana Bastos Carvalho
- Laboratório de Cardiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Bloco G. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-902, Brasil.
| | - Rachel de Pinho Rachid
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Bloco G. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-902, Brasil.
| | - Narcisa Leal da Cunha-E-Silva
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Bloco G. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-902, Brasil.
| | - Antonio Carlos Campos de Carvalho
- Laboratório de Cardiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Bloco G. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-902, Brasil.
| | - Lea Mirian Barbosa da Fonseca
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco, 255. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-913, Brasil.
| | - Regina Coeli dos Santos Goldenberg
- Laboratório de Cardiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Bloco G. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-902, Brasil.
| | - Bianca Gutfilen
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco, 255. Ilha do Fundão, Cidade Universitária, Rio de Janeiro, 21941-913, Brasil.
| |
Collapse
|
26
|
Rueger MA, Schroeter M. In vivo imaging of endogenous neural stem cells in the adult brain. World J Stem Cells 2015; 7:75-83. [PMID: 25621107 PMCID: PMC4300938 DOI: 10.4252/wjsc.v7.i1.75] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/02/2014] [Accepted: 10/29/2014] [Indexed: 02/06/2023] Open
Abstract
The discovery of endogenous neural stem cells (eNSCs) in the adult mammalian brain with their ability to self-renew and differentiate into functional neurons, astrocytes and oligodendrocytes has raised the hope for novel therapies of neurological diseases. Experimentally, those eNSCs can be mobilized in vivo, enhancing regeneration and accelerating functional recovery after, e.g., focal cerebral ischemia, thus constituting a most promising approach in stem cell research. In order to translate those current experimental approaches into a clinical setting in the future, non-invasive imaging methods are required to monitor eNSC activation in a longitudinal and intra-individual manner. As yet, imaging protocols to assess eNSC mobilization non-invasively in the live brain remain scarce, but considerable progress has been made in this field in recent years. This review summarizes and discusses the current imaging modalities suitable to monitor eNSCs in individual experimental animals over time, including optical imaging, magnetic resonance tomography and-spectroscopy, as well as positron emission tomography (PET). Special emphasis is put on the potential of each imaging method for a possible clinical translation, and on the specificity of the signal obtained. PET-imaging with the radiotracer 3’-deoxy-3’-[18F]fluoro-L-thymidine in particular constitutes a modality with excellent potential for clinical translation but low specificity; however, concomitant imaging of neuroinflammation is feasible and increases its specificity. The non-invasive imaging strategies presented here allow for the exploitation of novel treatment strategies based upon the regenerative potential of eNSCs, and will help to facilitate a translation into the clinical setting.
Collapse
|
27
|
Nam SY, Chung E, Suggs LJ, Emelianov SY. Combined ultrasound and photoacoustic imaging to noninvasively assess burn injury and selectively monitor a regenerative tissue-engineered construct. Tissue Eng Part C Methods 2015; 21:557-66. [PMID: 25384558 DOI: 10.1089/ten.tec.2014.0306] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Current biomedical imaging tools have limitations in accurate assessment of the severity of open and deep burn wounds involving excess bleeding and severe tissue damage. Furthermore, sophisticated imaging techniques are needed for advanced therapeutic approaches such as noninvasive monitoring of stem cells seeded and applied in a biomedical 3D scaffold to enhance wound repair. This work introduces a novel application of combined ultrasound (US) and photoacoustic (PA) imaging to assess both burn injury and skin tissue regeneration. Tissue structural damage and bleeding throughout the epidermis and dermis till the subcutaneous skin layer were imaged noninvasively by US/PA imaging. Gold nanoparticle-labeled adipose-derived stem cells (ASCs) within a PEGylated fibrin 3D gel were implanted in a rat model of cutaneous burn injury. ASCs were successfully tracked till 2 weeks and were distinguished from host tissue components (e.g., epidermis, fat, and blood vessels) through spectroscopic PA imaging. The structure and function of blood vessels (vessel density and perfusion) in the wound bed undergoing skin tissue regeneration were monitored both qualitatively and semi-quantitatively by the developed imaging approach. Imaging-based analysis demonstrated ASC localization in the top layer of skin and a higher density of regenerating blood vessels in the treated groups. This was corroborated with histological analysis showing localization of fluorescently labeled ASCs and smooth muscle alpha actin-positive blood vessels. Overall, the US/PA imaging-based strategy coupled with gold nanoparticles has a great potential for stem cell therapies and tissue engineering due to its noninvasiveness, safety, selectivity, and ability to provide long-term monitoring.
Collapse
Affiliation(s)
- Seung Yun Nam
- 1Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas
| | - Eunna Chung
- 2Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Laura J Suggs
- 2Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Stanislav Y Emelianov
- 1Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas.,2Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
28
|
Li SC, Tachiki LML, Kabeer MH, Dethlefs BA, Anthony MJ, Loudon WG. Cancer genomic research at the crossroads: realizing the changing genetic landscape as intratumoral spatial and temporal heterogeneity becomes a confounding factor. Cancer Cell Int 2014; 14:115. [PMID: 25411563 PMCID: PMC4236490 DOI: 10.1186/s12935-014-0115-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 10/24/2014] [Indexed: 02/06/2023] Open
Abstract
The US National Cancer Institute (NCI) and the National Human Genome Research Institute (NHGRI) created the Cancer Genome Atlas (TCGA) Project in 2006. The TCGA's goal was to sequence the genomes of 10,000 tumors to identify common genetic changes among different types of tumors for developing genetic-based treatments. TCGA offered great potential for cancer patients, but in reality has little impact on clinical applications. Recent reports place the past TCGA approach of testing a small tumor mass at a single time-point at a crossroads. This crossroads presents us with the conundrum of whether we should sequence more tumors or obtain multiple biopsies from each individual tumor at different time points. Sequencing more tumors with the past TCGA approach of single time-point sampling can neither capture the heterogeneity between different parts of the same tumor nor catch the heterogeneity that occurs as a function of time, error rates, and random drift. Obtaining multiple biopsies from each individual tumor presents multiple logistical and financial challenges. Here, we review current literature and rethink the utility and application of the TCGA approach. We discuss that the TCGA-led catalogue may provide insights into studying the functional significance of oncogenic genes in reference to non-cancer genetic background. Different methods to enhance identifying cancer targets, such as single cell technology, real time imaging of cancer cells with a biological global positioning system, and cross-referencing big data sets, are offered as ways to address sampling discrepancies in the face of tumor heterogeneity. We predict that TCGA landmarks may prove far more useful for cancer prevention than for cancer diagnosis and treatment when considering the effect of non-cancer genes and the normal genetic background on tumor microenvironment. Cancer prevention can be better realized once we understand how therapy affects the genetic makeup of cancer over time in a clinical setting. This may help create novel therapies for gene mutations that arise during a tumor's evolution from the selection pressure of treatment.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
- />Department of Neurology, University of California Irvine School of Medicine, Irvine, CA 92697-4292 USA
- />Department of Biological Science, California State University, Fullerton, CA 92834 USA
| | - Lisa May Ling Tachiki
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
- />University of California Irvine School of Medicine, Irvine, CA 92697 USA
| | - Mustafa H Kabeer
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
- />Department of Pediatric Surgery, CHOC Children’s Hospital, 1201 West La Veta Ave, Orange, CA 92868 USA
- />Department of Surgery, University of California Irvine School of Medicine, 333 City Blvd. West, Suite 700, Orange, CA 92868 USA
| | - Brent A Dethlefs
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
| | | | - William G Loudon
- />CHOC Children’s Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave, Orange, CA 92868 USA
- />Department of Neurological Surgery, Saint Joseph Hospital, Orange, CA 92868 USA
- />Department of Neurological Surgery, University of California Irvine School of Medicine, Orange, CA 92862 USA
- />Department of Biological Science, California State University, Fullerton, CA 92834 USA
| |
Collapse
|
29
|
Li SC, Kabeer MH, Vu LT, Keschrumrus V, Yin HZ, Dethlefs BA, Zhong JF, Weiss JH, Loudon WG. Training stem cells for treatment of malignant brain tumors. World J Stem Cells 2014; 6:432-440. [PMID: 25258664 PMCID: PMC4172671 DOI: 10.4252/wjsc.v6.i4.432] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 08/09/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
The treatment of malignant brain tumors remains a challenge. Stem cell technology has been applied in the treatment of brain tumors largely because of the ability of some stem cells to infiltrate into regions within the brain where tumor cells migrate as shown in preclinical studies. However, not all of these efforts can translate in the effective treatment that improves the quality of life for patients. Here, we perform a literature review to identify the problems in the field. Given the lack of efficacy of most stem cell-based agents used in the treatment of malignant brain tumors, we found that stem cell distribution (i.e., only a fraction of stem cells applied capable of targeting tumors) are among the limiting factors. We provide guidelines for potential improvements in stem cell distribution. Specifically, we use an engineered tissue graft platform that replicates the in vivo microenvironment, and provide our data to validate that this culture platform is viable for producing stem cells that have better stem cell distribution than with the Petri dish culture system.
Collapse
|
30
|
Ha S, Ahn S, Kim S, Joo Y, Chong YH, Suh YH, Chang KA. In vivo imaging of human adipose-derived stem cells in Alzheimer's disease animal model. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:051206. [PMID: 24297061 DOI: 10.1117/1.jbo.19.5.051206] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/28/2013] [Indexed: 06/02/2023]
Abstract
Stem cell therapy is a promising tool for the treatment of diverse conditions, including neurodegenerative diseases such as Alzheimer's disease (AD). To understand transplanted stem cell biology, in vivo imaging is necessary. Nanomaterial has great potential for in vivo imaging and several noninvasive methods are used, such as magnetic resonance imaging, positron emission tomography, fluorescence imaging (FI) and near-infrared FI. However, each method has limitations for in vivo imaging. To overcome these limitations, multimodal nanoprobes have been developed. In the present study, we intravenously injected human adipose-derived stem cells (hASCs) that were labeled with a multimodal nanoparticle, LEO-LIVE™-Magnoxide 675 or 797 (BITERIALS, Seoul, Korea), into Tg2576 mice, an AD mouse model. After sequential in vivo tracking using Maestro Imaging System, we found fluorescence signals up to 10 days after injection. We also found strong signals in the brains extracted from hASC-transplanted Tg2576 mice up to 12 days after injection. With these results, we suggest that in vivo imaging with this multimodal nanoparticle may provide a useful tool for stem cell tracking and understanding stem cell biology in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sungji Ha
- Gachon University of Medicine and Science, Department of Pharmacology, Incheon, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Sontag CJ, Uchida N, Cummings BJ, Anderson AJ. Injury to the spinal cord niche alters the engraftment dynamics of human neural stem cells. Stem Cell Reports 2014; 2:620-32. [PMID: 24936450 PMCID: PMC4050489 DOI: 10.1016/j.stemcr.2014.03.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 03/12/2014] [Accepted: 03/13/2014] [Indexed: 12/26/2022] Open
Abstract
The microenvironment is a critical mediator of stem cell survival, proliferation, migration, and differentiation. The majority of preclinical studies involving transplantation of neural stem cells (NSCs) into the CNS have focused on injured or degenerating microenvironments, leaving a dearth of information as to how NSCs differentially respond to intact versus damaged CNS. Furthermore, single, terminal histological endpoints predominate, providing limited insight into the spatiotemporal dynamics of NSC engraftment and migration. We investigated the early and long-term engraftment dynamics of human CNS stem cells propagated as neurospheres (hCNS-SCns) following transplantation into uninjured versus subacutely injured spinal cords of immunodeficient NOD-scid mice. We stereologically quantified engraftment, survival, proliferation, migration, and differentiation at 1, 7, 14, 28, and 98 days posttransplantation, and identified injury-dependent alterations. Notably, the injured microenvironment decreased hCNS-SCns survival, delayed and altered the location of proliferation, influenced both total and fate-specific migration, and promoted oligodendrocyte maturation.
Collapse
Affiliation(s)
- Christopher J Sontag
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA 92697, USA ; MIND Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA ; Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | | | - Brian J Cummings
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA 92697, USA ; MIND Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA ; Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA ; Department of Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, CA 92697, USA
| | - Aileen J Anderson
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA 92697, USA ; MIND Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA ; Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA ; Department of Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
32
|
Dedeepiya VD, William JB, Parthiban JKBC, Chidambaram R, Balamurugan M, Kuroda S, Iwasaki M, Preethy S, Abraham SJK. The known-unknowns in spinal cord injury, with emphasis on cell-based therapies - a review with suggestive arenas for research. Expert Opin Biol Ther 2014; 14:617-34. [PMID: 24660978 DOI: 10.1517/14712598.2014.889676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION In spite of extensive research, the progress toward a cure in spinal cord injury (SCI) is still elusive, which holds good for the cell- and stem cell-based therapies. We have critically analyzed seven known gray areas in SCI, indicating the specific arenas for research to improvise the outcome of cell-based therapies in SCI. AREAS COVERED The seven, specific known gray areas in SCI analyzed are: i) the gap between animal models and human victims; ii) uncertainty about the time, route and dosage of cells applied; iii) source of the most efficacious cells for therapy; iv) inability to address the vascular compromise during SCI; v) lack of non-invasive methodologies to track the transplanted cells; vi) need for scaffolds to retain the cells at the site of injury; and vii) physical and chemical stimuli that might be required for synapses formation yielding functional neurons. EXPERT OPINION Further research on scaffolds for retaining the transplanted cells at the lesion, chemical and physical stimuli that may help neurons become functional, a meta-analysis of timing of the cell therapy, mode of application and larger clinical studies are essential to improve the outcome.
Collapse
Affiliation(s)
- Vidyasagar Devaprasad Dedeepiya
- Nichi-In Centre for Regenerative Medicine (NCRM), The Mary-Yoshio Translational Hexagon (MYTH) , PB 1262, Chennai - 600034, Tamil Nadu , India +91 44 24732186 ; ,
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Miura Y, Yoshioka S, Yao H, Takaori-Kondo A, Maekawa T, Ichinohe T. Chimerism of bone marrow mesenchymal stem/stromal cells in allogeneic hematopoietic cell transplantation: is it clinically relevant? CHIMERISM 2013; 4:78-83. [PMID: 23880502 DOI: 10.4161/chim.25609] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Multipotent mesenchymal stem/stromal cells (MSCs) have been extensively used as a transplantable cell source for regenerative medicine and immunomodulatory therapy. Specifically in allogeneic hematopoietic stem cell transplantation (HSCT), co-transplantation or post-transplant infusion of MSCs derived from bone marrow (BM) of non-self donors has been implicated in accelerating hematopoietic recovery, ameliorating graft-vs.-host disease, and promoting tissue regeneration. However, irrespective of the use of MSC co-administration, post-transplant chimerism of BM-derived MSCs after allogeneic HSCT has been reported to remain of host origin, suggesting that the infused donor MSCs are immunologically rejected or not capable of long-term engraftment in the host microenvironment. Also, hematopoietic cell allografts currently used for HSCT do not seem to contain sufficient amount of MSCs or their precursors to reconstitute host BM microenvironment. Since the toxic conditioning employed in allo-HSCT may impair the function of host MSCs to maintain hematopoietic/regenerative stem cell niches and to provide a local immunomodulatory milieu, we propose that new directions for enhancing immunohematopoietic reconstitution and tissue repair after allogeneic HSCT include the development of strategies to support functional replenishment of residual host MSCs or to support more efficient engraftment of infused donor MSCs. Future areas of research should include in vivo tracking of infused MSCs and detection of their microchimeric presence in extra-marrow sites as well as in BM.
Collapse
Affiliation(s)
- Yasuo Miura
- Department of Transfusion Medicine and Cell Therapy; Kyoto University Hospital; Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Freitas ERLD, Santos RLD, Lima ECDO, Guillo LA. Feeder-free culture of human embryonic stem cell line BG01V/hOG using magnetic field-magnetic nanoparticles system. Biomed Pharmacother 2013; 67:17-21. [PMID: 23089476 DOI: 10.1016/j.biopha.2012.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 08/26/2012] [Indexed: 01/04/2023] Open
|
35
|
Li SC, Yin HZ, Loudon WG, Weiss JH. Cultivating stem cells for treating amyotrophic lateral sclerosis. World J Stem Cells 2012; 4:117-9. [PMID: 23516096 PMCID: PMC3600561 DOI: 10.4252/wjsc.v4.i12.117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 09/17/2012] [Accepted: 12/20/2012] [Indexed: 02/06/2023] Open
Abstract
This editorial addresses the current challenges and future directions in the use of stem cells as an approach for treating amyotrophic lateral sclerosis. A wide variety of literature has been reviewed to enlighten the reader on the many facets of stem cell research that are important to consider before using them for a cell based therapy.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Shengwen Calvin Li, William G Loudon, CHOC Children's Hospital, University of California Irvine, Orange, CA 92868, United States
| | | | | | | |
Collapse
|
36
|
de Mel A, Oh JT, Ramesh B, Seifalian AM. Biofunctionalized quantum dots for live monitoring of stem cells: applications in regenerative medicine. Regen Med 2012; 7:335-47. [PMID: 22594327 DOI: 10.2217/rme.12.21] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIM This study aimed to live monitor the degree of endothelial progenitor cell (EPC) integration onto tissue-engineering scaffolds by conjugating relevant antibodies to quantum dots (QDs). MATERIALS & METHODS Biocompatible mercaptosuccinic acid-coated QDs were functionalized with two different antibodies to EPC (CD133 with QDs of 640 nm wavelength [λ] and later-stage mature EPCs; and von Willebrand factor with QDs of λ595 and λ555 nm) using conventional carbomide and N-hydroxysuccinimide chemistry. Biofunctionalization was characterized with Fourier-transform infrared spectroscopy. Cell viability assays and gross morphology observations confirmed cytocompatibility and normal patterns of celluar growth. The antigens corresponding to each state of cell maturation were determined using a single excitation at λ488 nm. RESULTS The optimal concentrations of antibody-QD conjugates were biocompatible, hemocompatible and determined the state of EPC transformation to endothelial cells. CONCLUSION Antibody-functionalized QDs suggest new applications in tissue engineering of polymer-based implants where cell integration can potentially be monitored without requiring the sacrifice of implants.
Collapse
Affiliation(s)
- Achala de Mel
- UCL Centre for Nanotechnology & Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, UK
| | | | | | | |
Collapse
|
37
|
Fenrich KK, Weber P, Hocine M, Zalc M, Rougon G, Debarbieux F. Long-term in vivo imaging of normal and pathological mouse spinal cord with subcellular resolution using implanted glass windows. J Physiol 2012; 590:3665-75. [PMID: 22641787 DOI: 10.1113/jphysiol.2012.230532] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Repeated in vivo two-photon imaging of adult mammalian spinal cords, with subcellular resolution, would be crucial for understanding cellular mechanisms under normal and pathological conditions. Current methods are limited because they require surgery for each imaging session. Here we report a simple glass window methodology avoiding repeated surgical procedures and subsequent inflammation. We applied this strategy to follow axon integrity and the inflammatory response over months by multicolour imaging of adult transgenic mice. We found that glass windows have no significant effect on axon number or structure, cause a transient inflammatory response, and dramatically increase the throughput of in vivo spinal imaging. Moreover, we used this technique to track retraction/degeneration and regeneration of cut axons after a ‘pin-prick' spinal cord injury with high temporal fidelity. We showed that regenerating axons can cross an injury site within 4 days and that their terminals undergo dramatic morphological changes for weeks after injury. Overall the technique can potentially be adapted to evaluate cellular functions and therapeutic strategies in the normal and diseased spinal cord.
Collapse
Affiliation(s)
- Keith K Fenrich
- Aix Marseille University, Developmental Biology Institute of Marseille-Luminy CNRS, 7288 Case 907 Parc Scientifique de Luminy, 13009 Marseille, France.
| | | | | | | | | | | |
Collapse
|
38
|
Nam SY, Ricles LM, Suggs LJ, Emelianov SY. In vivo ultrasound and photoacoustic monitoring of mesenchymal stem cells labeled with gold nanotracers. PLoS One 2012; 7:e37267. [PMID: 22615959 PMCID: PMC3353925 DOI: 10.1371/journal.pone.0037267] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 04/18/2012] [Indexed: 11/25/2022] Open
Abstract
Longitudinal monitoring of cells is required in order to understand the role of delivered stem cells in therapeutic neovascularization. However, there is not an imaging technique that is capable of quantitative, longitudinal assessment of stem cell behaviors with high spatial resolution and sufficient penetration depth. In this study, in vivo and in vitro experiments were performed to demonstrate the efficacy of ultrasound-guided photoacoustic (US/PA) imaging to monitor mesenchymal stem cells (MSCs) labeled with gold nanotracers (Au NTs). The Au NT labeled MSCs, injected intramuscularly in the lower limb of the Lewis rat, were detected and spatially resolved. Furthermore, our quantitative in vitro cell studies indicate that US/PA imaging is capable of high detection sensitivity (1×10⁴ cells/mL) of the Au NT labeled MSCs. Finally, Au NT labeled MSCs captured in the PEGylated fibrin gel system were imaged in vivo, as well as in vitro, over a one week time period, suggesting that longitudinal cell tracking using US/PA imaging is possible. Overall, Au NT labeling of MSCs and US/PA imaging can be an alternative approach in stem cell imaging capable of noninvasive, sensitive, quantitative, longitudinal assessment of stem cell behaviors with high spatial and temporal resolutions at sufficient depths.
Collapse
Affiliation(s)
- Seung Yun Nam
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
- Department of Electrical and Computer Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Laura M. Ricles
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Laura J. Suggs
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Stanislav Y. Emelianov
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
- Department of Electrical and Computer Engineering, University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
39
|
Lee JM, Kim BS, Lee H, Im GI. In vivo tracking of mesechymal stem cells using fluorescent nanoparticles in an osteochondral repair model. Mol Ther 2012; 20:1434-42. [PMID: 22491215 DOI: 10.1038/mt.2012.60] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We devised and tested an in vivo system to monitor the migration of mesenchymal stem cells (MSCs) within the marrow cavity. In vitro studies confirmed that platelet-derived growth factor (PDGF)-AA had the most potent chemotactic effect of the tested factors, and possessed the greatest number of receptors in MSCs. MSCs were labeled with fluorescent nanoparticles and injected into the marrow cavity of nude rats through osteochondral defects created in the distal femur. The defects were sealed with HCF (heparin-conjugated fibrin) or PDGF-AA-loaded HCF. In the HCF-only group, the nanoparticle-labeled MSCs dispersed outside the marrow cavity within 3 days after injection. In the PDGF-AA-loaded HCF group, the labeled cells moved time-dependently for 14 days toward the osteochondral defect. HCF-PDGF in low dose (LD; 8.5 ng/µl) was more effective than HCF-PDGF in high dose (HD; 17 ng/µl) in recruiting the MSCs to the osteochondral defect. After 21 days, the defects treated with PDGF and transforming growth factor (TGF)-β1-loaded HCF showed excellent cartilage repair compared with other groups. Further studies confirmed that this in vivo osteochondral MSCs tracking system (IOMTS) worked for other chemoattractants (chemokine (C-C motif) ligand 2 (CCL2) and PDGF-BB). IOMTS can provide a useful tool to examine the effect of growth factors or chemokines on endogenous cartilage repair.
Collapse
Affiliation(s)
- Jong Min Lee
- Department of Orthopaedics, Dongguk University Ilsan Hospital, Goyang, Korea
| | | | | | | |
Collapse
|
40
|
Boulland JL, Leung DSY, Thuen M, Vik-Mo E, Joel M, Perreault MC, Langmoen IA, Haraldseth O, Glover JC. Evaluation of intracellular labeling with micron-sized particles of iron oxide (MPIOs) as a general tool for in vitro and in vivo tracking of human stem and progenitor cells. Cell Transplant 2012; 21:1743-59. [PMID: 22490338 DOI: 10.3727/096368911x627598] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Magnetic resonance imaging (MRI)-based tracking is increasingly attracting attention as a means of better understanding stem cell dynamics in vivo. Intracellular labeling with micrometer-sized particles of iron oxide (MPIOs) provides a practical MRI-based approach due to superior detectability relative to smaller iron oxide particles. However, insufficient information is available about the general utility across cell types and the effects on cell vitality of MPIO labeling of human stem cells. We labeled six human cell types from different sources: mesenchymal stem cells derived from bone marrow (MSCs), mesenchymal stem cells derived from adipose tissue (ASCs), presumptive adult neural stem cells (ad-NSCs), fetal neural progenitor cells (f-NPCs), a glioma cell line (U87), and glioblastoma tumor stem cells (GSCs), with two different sizes of MPIOs (0.9 and 2.84 µm). Labeling and uptake efficiencies were highly variable among cell types. Several parameters of general cell function were tested in vitro. Only minor differences were found between labeled and unlabeled cells with respect to proliferation rate, mitotic duration, random motility, and capacity for differentiation to specific phenotypes. In vivo behavior was tested in chicken embryos and severe combined immunodeficient (SCID) mice. Postmortem histology showed that labeled cells survived and could integrate into various tissues. MRI-based tracking over several weeks in the SCID mice showed that labeled GSCs and f-NPCs injected into the brain exhibited translocations similar to those seen for unlabeled cells and as expected from migratory behavior described in previous studies. The results support MPIO-based cell tracking as a generally useful tool for studies of human stem cell dynamics in vivo.
Collapse
Affiliation(s)
- Jean-Luc Boulland
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bigham-Sadegh A, Mirshokraei P, Karimi I, Oryan A, Aparviz A, Shafiei-Sarvestani Z. Effects of adipose tissue stem cell concurrent with greater omentum on experimental long-bone healing in dog. Connect Tissue Res 2012; 53:334-42. [PMID: 22268489 DOI: 10.3109/03008207.2012.660585] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Repair of large bone defects resulting from trauma, tumors, and osteitis is a current challenge to surgeons. Adipose-derived adult stem cells (ASCs) are multipotent cells that are able to differentiate into osteoblasts in the presence of certain factors. In this study, the role of greater omentum as a scaffold incorporation of ASCs was evaluated in long-bone defect healing in dog model. Sixteen 3-4-year-old, male adult mongrel dogs, weighing 25.2 ± 3.5 kg, were used in this study. In the control group (n = 4), the defect was left empty. In the omental group (n = 4), the defect was filled with harvested omentum. In the omental-ASCs group (n = 4), the defect was filled with omentum and 1 mL of ASCs was injected into the grafted omentum. In the omental-culture medium group (n = 4), 1 mL of culture medium was injected into the grafted omentum. Finally, the injured radial bones were fixed with plate and screw. Radiographs of each forelimb was taken postoperatively on the first day and at the second, fourth, sixth, and eighth weeks postinjury to evaluate bone formation, union, and remodeling of the defect. The operated radii were removed on the 56th postoperative day and were histopathologically evaluated. In this study, both omental-culture medium and omental-ASCs groups demonstrated superior osteogenic potential in healing the radial bone defect. Compared to those of the omental and control groups, more advanced bone healing criteria were present in the omental-culture medium and omental-ASCs groups at radiological and histopathological levels at 8 weeks postsurgery.
Collapse
Affiliation(s)
- Amin Bigham-Sadegh
- Department of Veterinary Surgery and Radiology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.
| | | | | | | | | | | |
Collapse
|
42
|
Li SC, Lee KL, Luo J, Zhong JF, Loudon WG. Convergence of normal stem cell and cancer stem cell developmental stage: Implication for differential therapies. World J Stem Cells 2011; 3:83-8. [PMID: 22007273 PMCID: PMC3192222 DOI: 10.4252/wjsc.v3.i9.83] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Revised: 09/16/2011] [Accepted: 09/21/2011] [Indexed: 02/06/2023] Open
Abstract
Increased evidence shows that normal stem cells may contribute to cancer development and progression by acting as cancer-initiating cells through their interactions with abnormal environmental elements. We postulate that normal stem cells and cancer stem cells (CSC) possess similar mechanisms of self-renewal and differentiation. CSC can be the key to the elaboration of anti-cancer-based therapy. In this article, we focus on a controversial new theme relating to CSC. Tumorigenesis may have a critical stage characterized as a "therapeutic window", which can be identified by association of molecular, biochemical and biological events. Identifying such a stage can allow the production of more effective therapies (e.g. manipulated stem cells) to treat several cancers. More importantly, confirming the existence of a similar therapeutic window during the conversion of normal stem cells to malignant CSC may lead to targeted therapy specifically against CSC. This conversion information may be derived from investigating the biological behaviour of both normal stem cells and cancerous stem cells. Currently, there is little knowledge about the cellular and molecular mechanisms that govern the initiation and maintenance of CSC. Studies on co-evolution and interdependence of cancer with normal tissues may lead to a useful treatment paradigm of cancer. The crosstalk between normal stem cells and cancer formation may converge developmental stages of different types of stem cells (e.g. normal stem cells, CSC and embryonic stem cells). The differential studies of the convergence may result in novel therapies for treating cancers.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Shengwen Calvin Li, Katherine L Lee, William G Loudon, Neuro-Oncology Research Laboratory, Center for Neuroscience and Stem Cell Research, Children's Hospital of Orange County, University of California Irvine, 455 South Main Street, Orange, CA 92868, United States
| | | | | | | | | |
Collapse
|
43
|
Li SC, Han YP, Dethlefs BA, Loudon WG. Therapeutic window, a critical developmental stage for stem cell therapies. Curr Stem Cell Res Ther 2011. [PMID: 20528752 DOI: 10.2174/157488810793351730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In children, cancers are the deadliest of diseases and second only to accidents as the leading cause of death. The deadliest of the brain cancers are the malignant gliomas. Approximately two-thirds of children can survive less malignant types of brain cancers, however, in ~67% of these survivors recurs under the current regimes of surgery followed by administration of high doses toxic drugs and exposure to high doses of radiation. Even more distressing is that fortunate survivors are generally left with life-long cognitive disabilities. A new medical approach is desperately needed. Stem cells, with their natural ability to seek out brain tumors, could be used to accurately deliver therapy directly to the cancer sparing normal tissues for suppression of tumor growth. Despite exciting initial reports, clinical potency of stem cell therapy in animal brain tumor models has to date proven disappointing. Attempts to extrapolate the animal study results to humans are stymied by the fact that stem cells are heterogeneous, resulting in differences in their efficacy. Indeed, therapeutic success relies on an effective strategy to select for a stem cell sub-population within some particular stage of the development at which they are competitive and capable of targeting brain tumors. To improve this during developmental path, concept of a 'therapeutic window' is proposed. The "therapeutic window" for stem cells or more specifically a "biochemical therapeutic window" can be determined from biochemical assays and a "biological therapeutic window" from biological assays or even a molecular window for genetic description. Taken together, we can use selective processes to generate more effective stem cells to treat cancers as is clearly needed today.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Neuro-Oncology Research Laboratory, Center for Neuroscience and Stem Cell Research, CHOC Children's Hospital, 455 S. Main Street, Orange, CA 92868-3874, USA.
| | | | | | | |
Collapse
|
44
|
Abstract
No other branch of science is quite so laden with expectation. But while corralling stem cells for viable therapies, researchers have found themselves asking an entirely separate question: How do we track them?
Collapse
|
45
|
Bohl SR, Pircher A, Hilbe W. Cancer stem cells: characteristics and their potential role for new therapeutic strategies. ACTA ACUST UNITED AC 2011; 34:269-74. [PMID: 21577035 DOI: 10.1159/000327815] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
With the cancer stem cell (CSC) hypothesis many questions regarding cancer development and drug resistance can be answered more coherently than with the traditional model based on clonal evolution. CSCs are a small subset of cancer cells within the tumour that show stem cell characteristics like self-renewal, the capability to develop into multiple lineages and the potential to proliferate extensively, and are characterised by a typical profile of different markers like CD44 and CD133. In the CSC model, the role of embryonic pathways like Wnt, Hedgehog and Notch is of special interest. This review presents current scientific knowledge on this topic and discusses the potential role of CSC in the resistance against chemotherapy or radiation and presents challenging options for therapeutic interventions.
Collapse
Affiliation(s)
- Stephan R Bohl
- Medical University Innsbruck, Department of Internal Medicine V, Haematology and Oncology, Innsbruck, Austria.
| | | | | |
Collapse
|
46
|
Li KC. Stem cell tracking a perfect application for multimodality imaging. Acad Radiol 2011; 18:1-2. [PMID: 21145024 DOI: 10.1016/j.acra.2010.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 11/08/2010] [Accepted: 11/08/2010] [Indexed: 11/25/2022]
|
47
|
Jiang H, Cheng Z, Tian M, Zhang H. In vivo imaging of embryonic stem cell therapy. Eur J Nucl Med Mol Imaging 2010; 38:774-84. [PMID: 21107558 DOI: 10.1007/s00259-010-1667-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 11/01/2010] [Indexed: 12/11/2022]
Abstract
Embryonic stem cells (ESCs) have the most pluripotent potential of any stem cell. These cells, isolated from the inner cell mass of the blastocyst, are "pluripotent," meaning that they can give rise to all cell types within the developing embryo. As a result, ESCs have been regarded as a leading candidate source for novel regenerative medicine therapies and have been used to derive diverse cell populations, including myocardial and endothelial cells. However, before they can be safely applied clinically, it is important to understand the in vivo behavior of ESCs and their derivatives. In vivo analysis of ESC-derived cells remains critically important to define how these cells may function in novel regenerative medicine therapies. In this review, we describe several available imaging modalities for assessing cell engraftment and discuss their strengths and limitations. We also analyze the applications of these modalities in assessing the utility of ESCs in regenerative medicine therapies.
Collapse
Affiliation(s)
- Han Jiang
- Department of Nuclear Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | | | | | | |
Collapse
|
48
|
Targeting of embryonic stem cells by peptide-conjugated quantum dots. PLoS One 2010; 5:e12075. [PMID: 20711469 PMCID: PMC2919412 DOI: 10.1371/journal.pone.0012075] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 07/18/2010] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Targeting stem cells holds great potential for studying the embryonic stem cell and development of stem cell-based regenerative medicine. Previous studies demonstrated that nanoparticles can serve as a robust platform for gene delivery, non-invasive cell imaging, and manipulation of stem cell differentiation. However specific targeting of embryonic stem cells by peptide-linked nanoparticles has not been reported. METHODOLOGY/PRINCIPAL FINDINGS Here, we developed a method for screening peptides that specifically recognize rhesus macaque embryonic stem cells by phage display and used the peptides to facilitate quantum dot targeting of embryonic stem cells. Through a phage display screen, we found phages that displayed an APWHLSSQYSRT peptide showed high affinity and specificity to undifferentiated primate embryonic stem cells in an enzyme-linked immunoabsorbent assay. These results were subsequently confirmed by immunofluorescence microscopy. Additionally, this binding could be completed by the chemically synthesized APWHLSSQYSRT peptide, indicating that the binding capability was specific and conferred by the peptide sequence. Through the ligation of the peptide to CdSe-ZnS core-shell nanocrystals, we were able to, for the first time, target embryonic stem cells through peptide-conjugated quantum dots. CONCLUSIONS/SIGNIFICANCE These data demonstrate that our established method of screening for embryonic stem cell specific binding peptides by phage display is feasible. Moreover, the peptide-conjugated quantum dots may be applicable for embryonic stem cell study and utilization.
Collapse
|