1
|
Xi J, Zheng W, Chen M, Zou Q, Tang C, Zhou X. Genetically engineered pigs for xenotransplantation: Hopes and challenges. Front Cell Dev Biol 2023; 10:1093534. [PMID: 36712969 PMCID: PMC9878146 DOI: 10.3389/fcell.2022.1093534] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/31/2022] [Indexed: 01/14/2023] Open
Abstract
The shortage of donor resources has greatly limited the application of clinical xenotransplantation. As such, genetically engineered pigs are expected to be an ideal organ source for xenotransplantation. Most current studies mainly focus on genetically modifying organs or tissues from donor pigs to reduce or prevent attack by the human immune system. Another potential organ source is interspecies chimeras. In this paper, we reviewed the progress of the genetically engineered pigs from the view of immunologic barriers and strategies, and discussed the possibility and challenges of the interspecies chimeras.
Collapse
|
2
|
China's Stem Cell Research and Knowledge Levels of Medical Practitioners and Students. Stem Cells Int 2021; 2021:6667743. [PMID: 34113385 PMCID: PMC8154300 DOI: 10.1155/2021/6667743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/05/2021] [Accepted: 04/29/2021] [Indexed: 11/18/2022] Open
Abstract
Over the last few decades, China has greatly expanded its scope of stem cell research, generating various scientific advances and medical applications. However, knowledge of the extent and characteristics of domestic stem cell development, particularly medical workers' opinions, is lacking. This study's purposes were to analyze the growth trends of China's stem cell community and identify the knowledge and attitudes held by Chinese medical workers regarding stem cell research. We found that there are currently 13 high-quality stem cell research centers with more than 400 PhD-level researchers across Mainland China. These centers feature many high-caliber scientists from the stem cell research community. From 1997 through 2019, the National Natural Science Foundation of China allocated roughly $576 million to 8,050 stem cell programs at Chinese universities and research institutions. China's annual publications on stem cells increased from less than 0.6% of the world's total stem cell publications in 1999 to more than 14.1% in 2014. Our survey also revealed that most participants held positive attitudes toward stem cell research, supported further funding, and had high general awareness about stem cells.
Collapse
|
3
|
Lu Y, Zhou Y, Ju R, Chen J. Human-animal chimeras for autologous organ transplantation: technological advances and future perspectives. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:576. [PMID: 31807557 DOI: 10.21037/atm.2019.10.13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Organ transplantation is the most promising curation for end-stage organ disease. However, the donor organ shortage has become a global problem that has limited the development of organ transplantation. Human-animal chimeras provide the ability to produce human organs in other species using autologous stem cells [e.g., induced pluripotent stem cells (iPSCs) or adult stem cells], which would be patient-specific and immune-matched for transplantation. Due to the potential application prospect of interspecies chimeras in basic and translational research, this technology has attracted much interest. This review focuses primarily on technological advances, including options of donor stem cell types and gene editing in donor cells and host animals, in addition to perspectives on human-animal chimeras in clinical and basic research.
Collapse
Affiliation(s)
- Yingfei Lu
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Yu Zhou
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China.,Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Rong Ju
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China.,Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| |
Collapse
|
4
|
Lizier M, Castelli A, Montagna C, Lucchini F, Vezzoni P, Faggioli F. Cell fusion in the liver, revisited. World J Hepatol 2018; 10:213-221. [PMID: 29527257 PMCID: PMC5838440 DOI: 10.4254/wjh.v10.i2.213] [Citation(s) in RCA: 12] [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: 11/21/2017] [Revised: 12/28/2017] [Accepted: 02/06/2018] [Indexed: 02/06/2023] Open
Abstract
There is wide agreement that cell fusion is a physiological process in cells in mammalian bone, muscle and placenta. In other organs, such as the cerebellum, cell fusion is controversial. The liver contains a considerable number of polyploid cells: They are commonly believed to originate by genome endoreplication, although the contribution of cell fusion to polyploidization has not been excluded. Here, we address the topic of cell fusion in the liver from a historical point of view. We discuss experimental evidence clearly supporting the hypothesis that cell fusion occurs in the liver, specifically when bone marrow cells were injected into mice and shown to rescue genetic hepatic degenerative defects. Those experiments-carried out in the latter half of the last century-were initially interpreted to show “transdifferentiation”, but are now believed to demonstrate fusion between donor macrophages and host hepatocytes, raising the possibility that physiologically polyploid cells, such as hepatocytes, could originate, at least partially, through homotypic cell fusion. In support of the homotypic cell fusion hypothesis, we present new data generated using a chimera-based model, a much simpler model than those previously used. Cell fusion as a road to polyploidization in the liver has not been extensively investigated, and its contribution to a variety of conditions, such as viral infections, carcinogenesis and aging, remains unclear.
Collapse
Affiliation(s)
- Michela Lizier
- Istituto di Ricerca Genetica e Biomedica, CNR, Milan 20138, Italy
- Human Genome Laboratory, Humanitas Clinical and Research Center, IRCCS, Milan 20089, Italy
| | - Alessandra Castelli
- Istituto di Ricerca Genetica e Biomedica, CNR, Milan 20138, Italy
- Human Genome Laboratory, Humanitas Clinical and Research Center, IRCCS, Milan 20089, Italy
| | - Cristina Montagna
- Department of Genetics and Pathology Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Franco Lucchini
- Centro Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, Cremona 26100, Italy
| | - Paolo Vezzoni
- Istituto di Ricerca Genetica e Biomedica, CNR, Milan 20138, Italy
- Human Genome Laboratory, Humanitas Clinical and Research Center, IRCCS, Milan 20089, Italy
| | - Francesca Faggioli
- Istituto di Ricerca Genetica e Biomedica, CNR, Milan 20138, Italy
- Human Genome Laboratory, Humanitas Clinical and Research Center, IRCCS, Milan 20089, Italy
| |
Collapse
|
5
|
Tang X, Qin H, Gu X, Fu X. China’s landscape in regenerative medicine. Biomaterials 2017; 124:78-94. [DOI: 10.1016/j.biomaterials.2017.01.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 01/24/2017] [Accepted: 01/28/2017] [Indexed: 12/15/2022]
|
6
|
Nakamura T, Hosoyama T, Kawamura D, Takeuchi Y, Tanaka Y, Samura M, Ueno K, Nishimoto A, Kurazumi H, Suzuki R, Ito H, Sakata K, Mikamo A, Li TS, Hamano K. Influence of aging on the quantity and quality of human cardiac stem cells. Sci Rep 2016; 6:22781. [PMID: 26947751 PMCID: PMC4780032 DOI: 10.1038/srep22781] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/19/2016] [Indexed: 01/01/2023] Open
Abstract
Advanced age affects various tissue-specific stem cells and decreases their regenerative ability. We therefore examined whether aging affected the quantity and quality of cardiac stem cells using cells obtained from 26 patients of various ages (from 2 to 83 years old). We collected fresh right atria and cultured cardiosphere-derived cells (CDCs), which are a type of cardiac stem cell. Then we investigated growth rate, senescence, DNA damage, and the growth factor production of CDCs. All samples yielded a sufficient number of CDCs for experiments and the cellular growth rate was not obviously associated with age. The expression of senescence-associated b-galactosidase and the DNA damage marker, gH2AX, showed a slightly higher trend in CDCs from older patients (≥65 years). The expression of VEGF, HGF, IGF-1, SDF-1, and TGF-b varied among samples, and the expression of these beneficial factors did not decrease with age. An in vitro angiogenesis assay also showed that the angiogenic potency of CDCs was not impaired, even in those from older patients. Our data suggest that the impact of age on the quantity and quality of CDCs is quite limited. These findings have important clinical implications for autologous stem cell transplantation in elderly patients.
Collapse
Affiliation(s)
- Tamami Nakamura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Tohru Hosoyama
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan.,Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Daichi Kawamura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Yuriko Takeuchi
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Yuya Tanaka
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Makoto Samura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Koji Ueno
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan.,Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Arata Nishimoto
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Hiroshi Kurazumi
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Ryo Suzuki
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Surgery, Saiseikai Shimonoseki General Hospital, 8-5-1 Yasuoka, Shimonoseki, Yamaguchi 759-6603, Japan
| | - Kensuke Sakata
- Department of Cardiovascular Surgery, Saiseikai Shimonoseki General Hospital, 8-5-1 Yasuoka, Shimonoseki, Yamaguchi 759-6603, Japan
| | - Akihito Mikamo
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| |
Collapse
|
7
|
Lin H, Woolfson A, Jiang X. New Mouse Models to Investigate the Efficacy of Drug Combinations in Human Chronic Myeloid Leukemia. Methods Mol Biol 2016; 1465:187-205. [PMID: 27581149 DOI: 10.1007/978-1-4939-4011-0_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Chronic myeloid leukemia (CML) comprises a simple and effective paradigm for generating new insights into the cellular origin, pathogenesis, and treatment of many types of human cancer. In particular, mouse models of CML have greatly facilitated the understanding of the underlying molecular mechanisms and pathogenesis of this disease and have led to the identification of new drug targets that in some cases offer the possibility of functional cure. There are currently three established CML mouse models: the BCR-ABL transgenic model, the BCR-ABL retroviral transduction/transplantation model, and the xenotransplant immunodeficient model. Each has its own unique advantages and disadvantages. Depending on the question of interest, some models may be more appropriate than others. In this chapter, we describe a newly developed xenotransplant mouse model to determine the efficacy of novel therapeutic agents, either alone or in combination. The model facilitates the evaluation of the frequency of leukemic stem cells with long-term leukemia-initiating activity, a critical subcellular population that causes disease relapse and progression, through the utilization of primary CD34(+) CML stem/progenitor cells obtained from CML patients at diagnosis and prior to drug treatment. We have also investigated the effectiveness of new combination treatment strategies designed to prevent the development of leukemia in vivo using BCR-ABL (+) blast crisis cells as a model system. These types of in vivo studies are important for the prediction of individual patient responses to drug therapy, and have the potential to facilitate the design of personalized combination therapy strategies.
Collapse
Affiliation(s)
- Hanyang Lin
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, Canada, V5Z 1L3
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Xiaoyan Jiang
- Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, Canada, V5Z 1L3.
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
8
|
Patel AN, Bartlett CE, Ichim TE. Mesenchymal Stem Cells. STEM CELL AND GENE THERAPY FOR CARDIOVASCULAR DISEASE 2016:139-150. [DOI: 10.1016/b978-0-12-801888-0.00011-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
9
|
Banik A, Prabhakar S, Kalra J, Anand A. Effect of human umbilical cord blood derived lineage negative stem cells transplanted in amyloid-β induced cognitive impaired mice. Behav Brain Res 2015; 291:46-59. [PMID: 25989508 DOI: 10.1016/j.bbr.2015.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 05/07/2015] [Accepted: 05/10/2015] [Indexed: 10/23/2022]
Abstract
Alzheimer's disease (AD) is pathologically characterized by extracellular deposition of insoluble amyloid-β (Aβ) plaques and intracellular tangles made up of phosphorylated tau in brain. Several therapeutic approaches are being carried out in animal AD models for testing their safety and efficacy in altering disease pathology and behavioral deficits. Very few studies have examined the effect of human umbilical cord blood (hUCB) derived stem cells in degenerative disease models despite growing number of cord blood banks worldwide. Here we have examined the therapeutic efficacy of hUCB derived lineage negative (Lin -ve) stem cells in alleviating behavioral and neuropathological deficits in a mouse model of cognitive impairment induced by bilateral intrahippocampal injection of Aβ-42. Lin -ve cells were transplanted at two doses (50,000 and 100,000) at the site of injury and examined at 10 and 60 days post transplantation for rescue of memory deficits. These cells were found to ameliorate cognitive impairment in 50,000-60 days and 100,000-10 days groups whereas, 50,000-10 days and 100,000-60 days groups could not exert any significant improvement. Further, mice showing spatial memory improvement were mediated by up-regulation of BDNF, CREB and also by concomitant down regulation of Fas-L in their brain. The transplanted cells were found in the host tissue and survived up to 60 days without expressing markers of neuronal differentiation or reducing Aβ burden in mouse brain. We suggest that these undifferentiated cells could exert neuroprotective effects either through inhibiting apoptosis and/or trophic effects in the brain.
Collapse
Affiliation(s)
- Avijit Banik
- Neuroscience Research Lab, Department of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sudesh Prabhakar
- Neuroscience Research Lab, Department of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jasvinder Kalra
- Department of Obstetrics and Gynecology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
10
|
Li J, Zeng G, Qi Y, Tang X, Zhang J, Wu Z, Liang J, Shi L, Liu H, Zhang P. Xenotransplantation of human adipose-derived stem cells in zebrafish embryos. PLoS One 2015; 10:e0123264. [PMID: 25849455 PMCID: PMC4388839 DOI: 10.1371/journal.pone.0123264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/17/2015] [Indexed: 12/18/2022] Open
Abstract
Zebrafish is a widely used animal model with well-characterized background in developmental biology. The fate of human adipose-derived stem cells (ADSCs) after their xenotransplantation into the developing embryos of zebrafish is unknown. Therefore, human ADSCs were firstly isolated, and then transduced with lentiviral vector system carrying a green fluorescent protein (GFP) reporter gene, and followed by detection of their cell viability and the expression of cell surface antigens. These GFP-expressing human ADSCs were transplanted into the zebrafish embryos at 3.3–4.3 hour post-fertilization (hpf). Green fluorescent signal, the proliferation and differentiation of human ADSCs in recipient embryos were respectively examined using fluorescent microscopy and immunohistochemical staining. The results indicated that human ADSCs did not change their cell viability and the expression levels of cell surface antigens after GFP transduction. Microscopic examination demonstrated that green fluorescent signals of GFP expressed in the transplanted cells were observed in the embryos and larva fish at post-transplantation. The positive staining of Ki-67 revealed the survival and proliferation of human ADSCs in fish larvae after transplantation. The expression of CD105 was observable in the xenotransplanted ADSCs, but CD31 expression was undetectable. Therefore, our results indicate that human ADSCs xenotransplanted in the zebrafish embryos not only can survive and proliferate at across-species circumstance, but also seem to maintain their undifferentiation status in a short term. This xenograft model of zebrafish embryos may provide a promising and useful technical platform for the investigation of biology and physiology of stem cells in vivo.
Collapse
Affiliation(s)
- Jin Li
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Guofang Zeng
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Yawei Qi
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Xudong Tang
- Institute of Biochemistry and Molecular Biology, Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Jingjing Zhang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Zeyong Wu
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Jie Liang
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Lei Shi
- School of Light Industry and Food Science, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Hongwei Liu
- Department of Plastic Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory for Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
- * E-mail: (HL); (PZ)
| | - Peihua Zhang
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
- * E-mail: (HL); (PZ)
| |
Collapse
|
11
|
Long-term deregulated human hematopoiesis in goats transplanted in utero with BCR-ABL-transduced lin(-)CD34(+) cord blood cells. Cell Res 2013; 23:859-62. [PMID: 23628725 DOI: 10.1038/cr.2013.60] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
12
|
Yang CM, Gong XL, Qiu J, Tang HX, Gong ZJ, Huang SZ, Zeng F. Engraftment of genetically modified human amniotic fluid-derived progenitor cells to produce coagulation factor IX after in utero transplantation in mice. Cell Biol Int 2013; 37:420-9. [PMID: 23504762 DOI: 10.1002/cbin.10037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 12/23/2012] [Indexed: 12/29/2022]
Abstract
Human amniotic fluid derived progenitor cells (hAFPCs) may be multipotent and can be considered a potential tool in the field of cell therapy for haemophilia B. Their capacity to express human coagulation factor IX (hFIX) after transduction and their fate after in utero transplantation is unknown. hAFPCs isolated from second trimester pregnancies were assessed for their phenotypic markers, multilineage capacity, and expression of hFIX after transduction. Their engraftment potential was analysed in a mouse model after in utero transplantation at embryonic day 12.5. Immunohistochemistry, fluorescence in situ, ELISA and PCR were used to assess post-transplant chimeras. hAFPCs expressed several pluripotent markers, including NANOG, SOX2, SSEA4 and TRA-1-60, and could differentiate into adipocytes and osteocytes. In vitro, after transduction with hFIX and EGFP cDNAs, constitutive hFIX protein expression and clotting activity were found. Engraftment was achieved in various foetal tissues after in utero transplantation. Safe engraftment without oncogenesis was confirmed, with low donor cell levels, but persistent engraftment, into different organs (liver, heart and lung) through to 12 weeks of age. Transgenic expression of circulating hFIX was detected in recipient mice for up to 12 weeks. hAFPCs can be engrafted long-term in immunocompetent mice after in utero transplantation. Thus, cell transplantation approaches using genetically engineered hAFPCs may prove valuable for the prenatal treatment for haemophilia B.
Collapse
Affiliation(s)
- Chen-Min Yang
- Laboratory of Development Biology, Institute of Medical Science, Shanghai Jiao Tong University School of Medicine, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Shablii VA, Lukash LL, Lobintseva GS. The role of some donor-host cell interactions under a microenvironmental influence during regeneration processes. CYTOL GENET+ 2012. [DOI: 10.3103/s0095452712030097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
14
|
Ratajczak MZ, Zuba-Surma E, Kucia M, Poniewierska A, Suszynska M, Ratajczak J. Pluripotent and multipotent stem cells in adult tissues. Adv Med Sci 2012; 57:1-17. [PMID: 22515973 DOI: 10.2478/v10039-012-0020-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
One of the most intriguing questions in stem cell biology is whether pluripotent stem cells exist in adult tissues. Several groups of investigators employing i) various isolation protocols, ii) detection of surface markers, and iii) experimental in vitro and in vivo models, have reported the presence of cells that possess a pluripotent character in adult tissues. Such cells were assigned various operational abbreviations and names in the literature that added confusion to the field and raised the basic question of whether these are truly distinct or overlapping populations of the same primitive stem cells. Unfortunately, these cells were never characterized side-by-side to address this important issue. Nevertheless, taking into consideration their common features described in the literature, it is very likely that various investigators have described overlapping populations of developmentally early stem cells that are closely related. These different populations of stem cells will be reviewed in this paper.
Collapse
Affiliation(s)
- M Z Ratajczak
- Stem Cell Biology Program, James Graham Brown Cancer Center, University of Louisville, Kentucky, USA.
| | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Estrada J, Li P, Mir B. Multiorgan engraftment of human somatic cells in swine foetuses after intra-blastocyst transplantation. Reprod Domest Anim 2010; 46:630-5. [PMID: 21092066 DOI: 10.1111/j.1439-0531.2010.01718.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Adult human stem cells, mainly from hematopoietic lineage, have been injected into developing pre-immune animal foetuses, and xenogenic engraftment of liver and other organs has been reported. We isolated a rare cell population from adult human liver, fat and skin. Colonies with few cells became visible as early as 2-3 days, and a fully formed colony took 10-14 days to form. These colonies were named as liver-derived cell lines (LDCs), fat-derived cell lines (FDCs) and skin-derived cell lines (SDCs). All these cells express few pluripotency markers like Klf4, c-myc and Sox2. Pig blastocysts were injected with LDCs, FDCs and SDCs and transferred to recipient pigs. We achieved an overall pregnancy rate of 71.4% at day 35. The foetuses were analysed for human cell chimerism in liver, kidney and heart both by RT-PCR and real-time PCR using primers specific to human and pig mitochondrial DNA. The percentage of foetuses showing chimerism was 17.4% (4/23), 12.5% (2/16) and 11.1% (1/9) for LDCs, FDCs and SDCs, respectively. Of these, 42.9% (three out of seven) showed chimerism in liver and 71.4% (five out of seven) showed kidney chimerism. However, we did not detect any chimerism in the heart. The level of chimerism varied and was in the range of one human cell per one hundred thousand to one million pig cells.
Collapse
Affiliation(s)
- J Estrada
- Department of Transplant Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | |
Collapse
|
17
|
Ichim TE, Solano F, Lara F, Rodriguez JP, Cristea O, Minev B, Ramos F, Woods EJ, Murphy MP, Alexandrescu DT, Patel AN, Riordan NH. Combination stem cell therapy for heart failure. Int Arch Med 2010; 3:5. [PMID: 20398245 PMCID: PMC3003238 DOI: 10.1186/1755-7682-3-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 04/14/2010] [Indexed: 02/07/2023] Open
Abstract
Patients with congestive heart failure (CHF) that are not eligible for transplantation have limited therapeutic options. Stem cell therapy such as autologous bone marrow, mobilized peripheral blood, or purified cells thereof has been used clinically since 2001. To date over 1000 patients have received cellular therapy as part of randomized trials, with the general consensus being that a moderate but statistically significant benefit occurs. Therefore, one of the important next steps in the field is optimization. In this paper we discuss three ways to approach this issue: a) increasing stem cell migration to the heart; b) augmenting stem cell activity; and c) combining existing stem cell therapies to recapitulate a "therapeutic niche". We conclude by describing a case report of a heart failure patient treated with a combination stem cell protocol in an attempt to augment beneficial aspects of cord blood CD34 cells and mesenchymal-like stem cells.
Collapse
|
18
|
Chimenti I, Smith RR, Li TS, Gerstenblith G, Messina E, Giacomello A, Marbán E. Relative roles of direct regeneration versus paracrine effects of human cardiosphere-derived cells transplanted into infarcted mice. Circ Res 2010; 106:971-80. [PMID: 20110532 DOI: 10.1161/circresaha.109.210682] [Citation(s) in RCA: 506] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Multiple biological mechanisms contribute to the efficacy of cardiac cell therapy. Most prominent among these are direct heart muscle and blood vessel regeneration from transplanted cells, as opposed to paracrine enhancement of tissue preservation and/or recruitment of endogenous repair. OBJECTIVE Human cardiac progenitor cells, cultured as cardiospheres (CSps) or as CSp-derived cells (CDCs), have been shown to be capable of direct cardiac regeneration in vivo. Here we characterized paracrine effects in CDC transplantation and investigated their relative importance versus direct differentiation of surviving transplanted cells. METHODS AND RESULTS In vitro, many growth factors were found in media conditioned by human adult CSps and CDCs; CDC-conditioned media exerted antiapoptotic effects on neonatal rat ventricular myocytes, and proangiogenic effects on human umbilical vein endothelial cells. In vivo, human CDCs secreted vascular endothelial growth factor, hepatocyte growth factor, and insulin-like growth factor 1 when transplanted into the same SCID mouse model of acute myocardial infarction where they were previously shown to improve function and to produce tissue regeneration. Injection of CDCs in the peri-infarct zone increased the expression of Akt, decreased apoptotic rate and caspase 3 level, and increased capillary density, indicating overall higher tissue resilience. Based on the number of human-specific cells relative to overall increases in capillary density and myocardial viability, direct differentiation quantitatively accounted for 20% to 50% of the observed effects. CONCLUSIONS Together with their spontaneous commitment to cardiac and angiogenic differentiation, transplanted CDCs serve as "role models," recruiting endogenous regeneration and improving tissue resistance to ischemic stress. The contribution of the role model effect rivals or exceeds that of direct regeneration.
Collapse
Affiliation(s)
- Isotta Chimenti
- Cedars-Sinai Heart Institute, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Sun Y, Xiao D, Li HA, Jiang JF, Li Q, Zhang RS, Chen XG. Phenotypic changes of human cells in human-rat liver during partial hepatectomy-induced regeneration. World J Gastroenterol 2009; 15:3611-20. [PMID: 19653338 PMCID: PMC2721234 DOI: 10.3748/wjg.15.3611] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To examine the human hepatic parenchymal and stromal components in rat liver and the phenotypic changes of human cells in liver of human-rat chimera (HRC) generated by in utero transplantation of human cells during partial hepatectomy (PHx)-induced liver regeneration.
METHODS: Human hepatic parenchymal and stromal components and phenotypic changes of human cells during liver regeneration were examined by flow cytometry, in situ hybridization and immunohistochemistry.
RESULTS: ISH analysis demonstrated human Alu-positive cells in hepatic parenchyma and stroma of recipient liver. Functional human hepatocytes generated in this model potentially constituted human hepatic functional units with the presence of donor-derived human endothelial and biliary duct cells in host liver. Alpha fetoprotein (AFP)+, CD34+ and CD45+ cells were observed in the chimeric liver on day 10 after PHx-induced liver regeneration and then disappeared in PHx group, but not in non-PHx group, suggesting that dynamic phenotypic changes of human cells expressing AFP, CD34 and CD45 cells may occur during the chimeric liver regeneration. Additionally, immunostaining for human proliferating cell nuclear antigen (PCNA) showed that the number of PCNA-positive cells in the chimeric liver of PHx group was markedly increased, as compared to that of control group, indicating that donor-derived human cells are actively proliferated during PHx-induced regeneration of HRC liver.
CONCLUSION: HRC liver provides a tool for investigating human liver regeneration in a humanized animal model.
Collapse
|
20
|
Hematopoietic stem cell engraftment by early-stage in utero transplantation in a mouse model. Exp Mol Pathol 2009; 87:173-7. [PMID: 19666020 DOI: 10.1016/j.yexmp.2009.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 07/30/2009] [Indexed: 12/29/2022]
Abstract
A novel intrauterine transplantation (IUT) approach was developed to improve the efficiency of engraftment of hematopoietic stem cells (HSCs). HSCs with a green fluorescent protein (GFP) reporter gene were transplanted in utero on days 12.5, 13.5 and 14.5 post coitum (p.c.). The degree of chimerism of donor cells in recipient newborn mice was examined using fluorescent microscopy, polymerase chain reaction (PCR), fluorescence-activated cell sorting (FACS), and fluorescence in situ hybridization (FISH) analyses. Microscopic examination revealed the presence of green fluorescent signal in the peripheral blood of the chimeric mice. The highest survival rate (47%) as well as the highest chimerism rate (73%) were achieved by our new approach in the newborn mice that were subjected to in utero transplantation (IUT) on day 12.5 p.c. (E12.5) compared to the conventional IUT method. FACS analysis indicated that 1.55+/-1.10% of peripheral blood cells from the newborn mice were GFP-positive donor cells. FISH showed that cells containing the donor-specific GFP sequence were present in the bone marrow (BM) of the chimeric mice. Thus, the efficiency of chimera production with this new method of IUT was significantly improved over the existing IUT techniques and instruments.
Collapse
|
21
|
Association of differential and site-dependent CpG methylation and diverse expression of DNA methyltransferases with the tissue-specific expression of human β-globin gene in transgenic mice. Int J Hematol 2009; 89:414-421. [DOI: 10.1007/s12185-009-0319-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 02/06/2009] [Accepted: 04/07/2009] [Indexed: 01/15/2023]
|
22
|
Brezillon N, Kremsdorf D, Weiss MC. Cell therapy for the diseased liver: from stem cell biology to novel models for hepatotropic human pathogens. Dis Model Mech 2009; 1:113-30. [PMID: 19048074 DOI: 10.1242/dmm.000463] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
It has long been known that hepatocytes possess the potential to replicate through many cell generations because regeneration can be achieved in rodents after serial two-thirds hepatectomy. It has taken considerable time and effort to harness this potential, with liver regeneration models involving hepatocyte transplantation developing over the past 15 years. This review will describe the experiments that have established the models and methodology for liver repopulation, and the use of cells other than adult hepatocytes in liver repopulation, including hepatic cell lines and hematopoietic, cord blood, hepatic and embryonic stem cells. Emphasis will be placed on the characteristics of the models and how they can influence the outcome of the experiments. Finally, an account of the development of murine models that are competent to accept human hepatocytes is provided. In these models, liver deficiencies are induced in immunodeficient mice, where healthy human cells have a selective advantage. These mice with humanized livers provide a powerful new experimental tool for the study of human hepatotropic pathogens.
Collapse
Affiliation(s)
- Nicolas Brezillon
- INSERM, U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris 75015, France
| | | | | |
Collapse
|
23
|
Ratajczak MZ, Kucia M, Ratajczak J, Zuba-Surma EK. A multi-instrumental approach to identify and purify very small embryonic like stem cells (VSELs) from adult tissues. Micron 2009; 40:386-93. [DOI: 10.1016/j.micron.2008.09.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 09/27/2008] [Indexed: 11/17/2022]
|
24
|
Alison MR, Islam S, Lim S. Stem cells in liver regeneration, fibrosis and cancer: the good, the bad and the ugly. J Pathol 2009; 217:282-98. [PMID: 18991329 DOI: 10.1002/path.2453] [Citation(s) in RCA: 168] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The worldwide shortage of donor livers to transplant end stage liver disease patients has prompted the search for alternative cell therapies for intractable liver diseases, such as acute liver failure, cirrhosis and hepatocellular carcinoma (HCC). Under normal circumstances the liver undergoes a low rate of hepatocyte 'wear and tear' renewal, but can mount a brisk regenerative response to the acute loss of two-thirds or more of the parenchymal mass. A body of evidence favours placement of a stem cell niche in the periportal regions, although the identity of such stem cells in rodents and man is far from clear. In animal models of liver disease, adopting strategies to provide a selective advantage for transplanted hepatocytes has proved highly effective in repopulating recipient livers, but the poor success of today's hepatocyte transplants can be attributed to the lack of a clinically applicable procedure to force a similar repopulation of the human liver. The activation of bipotential hepatic progenitor cells (HPCs) is clearly vital for survival in many cases of acute liver failure, and the signals that promote such reactions are being elucidated. Bone marrow cells (BMCs) make, at best, a trivial contribution to hepatocyte replacement after damage, but other BMCs contribute to the hepatic collagen-producing cell population, resulting in fibrotic disease; paradoxically, BMC transplantation may help alleviate established fibrotic disease. HCC may have its origins in either hepatocytes or HPCs, and HCCs, like other solid tumours appear to be sustained by a minority population of cancer stem cells.
Collapse
Affiliation(s)
- M R Alison
- Centre for Diabetes and Metabolic Medicine, St Bartholomew's Hospital and the London School of Medicine and Dentistry, London, UK.
| | | | | |
Collapse
|
25
|
Álvarez-Mercado AI, Sáez-Lara MJ, García-Mediavilla MV, Sánchez-Campos S, Abadía F, Cabello-Donayre M, Gil Á, Gonzalez-Gallego J, Fontana L. Xenotransplantation of Human Umbilical Cord Blood Mononuclear Cells to Rats with D-Galactosamine-Induced Hepatitis. Cell Transplant 2008; 17:845-57. [DOI: 10.3727/096368908786516837] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cord blood is an attractive cell source in regenerative medicine and represents an alternative to bone marrow. The aim of this study was to investigate whether human umbilical cord blood mononuclear (HUCBM) cells might be valuable in hepatic regenerative medicine. HUCBM cells differentiated in vitro into hepatocytes, as suggested by expression of albumin, cytokeratin-18, glutamine synthetase, α-fetoprotein, and cytochrome P450 3A4 at both mRNA and protein levels in a time-dependent fashion. In contrast, the hematopoietic phenotype was gradually lost, as demonstrated by disappearance of CD45 expression. The regenerative potential of HUCBM cells was tested by using a human-to-rat xenotransplant model in which HUCBM cells were intraportally injected into rats with D-galactosamine-induced hepatitis. Liver histology and biochemical markers of hepatic damage were determined. Presence of human cells was detected in blood and liver of both control and D-galactosamine-treated animals. Cell transplantation produced an improvement in both the histological damage and liver function, as demonstrated by plasma values of alkaline phosphatase, γ-glutamyl transferase, lactate dehydrogenase, and total and direct bilirubins. Results obtained suggest that HUCBM cells are capable of hepatic engraftment in this human-to-rat xenotransplant model and that transplantation of HUCBM cells may be a suitable therapy for liver disease.
Collapse
Affiliation(s)
- Ana I. Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain
- Institute of Nutrition and Food Technology, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Avda. del Conocimiento s/n, 18100 Armilla, Granada, Spain
| | - María J. Sáez-Lara
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain
- Institute of Nutrition and Food Technology, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Avda. del Conocimiento s/n, 18100 Armilla, Granada, Spain
| | - María V. García-Mediavilla
- Centro de Investigacion Biomédica en Red de Enferemedades Hepáticas y Digestivas (CIBEREHD), and Institute of Biomedicine, University of Leon, Campus de Vegazana s/n, 24071 Leon, Spain
| | - Sonia Sánchez-Campos
- Centro de Investigacion Biomédica en Red de Enferemedades Hepáticas y Digestivas (CIBEREHD), and Institute of Biomedicine, University of Leon, Campus de Vegazana s/n, 24071 Leon, Spain
| | - Francisco Abadía
- Department of Cell Biology, School of Sciences, University of Granada, Campus de Fuentenueva s/n, 18071 Granada, Spain
| | - María Cabello-Donayre
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain
- Institute of Nutrition and Food Technology, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Avda. del Conocimiento s/n, 18100 Armilla, Granada, Spain
| | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain
- Institute of Nutrition and Food Technology, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Avda. del Conocimiento s/n, 18100 Armilla, Granada, Spain
| | - Javier Gonzalez-Gallego
- Centro de Investigacion Biomédica en Red de Enferemedades Hepáticas y Digestivas (CIBEREHD), and Institute of Biomedicine, University of Leon, Campus de Vegazana s/n, 24071 Leon, Spain
| | - Luis Fontana
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain
- Institute of Nutrition and Food Technology, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Avda. del Conocimiento s/n, 18100 Armilla, Granada, Spain
| |
Collapse
|
26
|
Abstract
The promise of liver stem cells lie in their potential to provide a continual and readily available source of liver cells that can be used for gene therapy, cellular transplant, bioartificial liver-assisted devices, drug toxicology testing and use as an in vitro model to understand the developmental biology of the liver. Both the rodent and human embryonic stem cell, bone marrow hematopoietic stem cell, mesenchymal stem cell, umbilical cord blood cell, fetal liver progenitor cell, adult liver progenitor cell as well as the mature hepatocyte have been reported to be capable of self-renewal, giving rise to daughter hepatocytes both in vivo and in vitro. These cells can repopulate livers in animal models of liver injury and seemingly improve liver function. However, significant challenges still exist before these cells can be used in humans. These include lack of consensus in immunophenotype of liver progenitor cells, uncertainty of the physiological role of reported candidate stem/progenitor cell, practicality in obtaining sufficient quantity of cells for clinical use and concerns over ethics, long-term efficacy and safety. Current molecular techniques of stem cell identification are confounded by cell fusion, horizontal gene transfer, incomplete differentiation and fetal microchimerism. Reports of stem cell transplantation and phase 1 trials of bone marrow transplantation in humans for liver diseases are exciting but require more robust verification. We review the evidence for various candidate stem cells, human clinical trials reported to date and highlight the challenges facing clinicians in their quest to use liver stem cells to save lives.
Collapse
Affiliation(s)
- Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, National University Hospital, Singapore.
| | | |
Collapse
|
27
|
Dinsmore JH, Dib N. Stem cells and cardiac repair: a critical analysis. J Cardiovasc Transl Res 2008; 1:41-54. [PMID: 20559957 DOI: 10.1007/s12265-007-9008-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Accepted: 12/27/2007] [Indexed: 01/11/2023]
Abstract
Utilizing stem cells to repair the damaged heart has seen an intense amount of activity over the last 5 years or so. There are currently multiple clinical studies in progress to test the efficacy of various different cell therapy approaches for the repair of damaged myocardium that were only just beginning to be tested in preclinical animal studies a few years earlier. This rapid transition from preclinical to clinical testing is striking and is not typical of the customary timeframe for the progress of a therapy from bench-to-bedside. Doubtless, there will be many more trials to follow in the upcoming years. With the plethora of trials and cell alternatives, there has come not only great enthusiasm for the potential of the therapy, but also great confusion about what has been achieved. Cell therapy has the potential to do what no drug can: regenerate and replace damaged tissue with healthy tissue. Drugs may be effective at slowing the progression of heart failure, but none can stop or reverse the process. However, tissue repair is not a simple process, although the idea on its surface is quite simple. Understanding cells, the signals that they respond to, and the keys to appropriate survival and tissue formation are orders of magnitude more complicated than understanding the pathways targeted by most drugs. Drugs and their metabolites can be monitored, quantified, and their effects correlated to circulating levels in the body. Not so for most cell therapies. It is quite difficult to measure cell survival except through ex vivo techniques like histological analysis of the target organ. This makes the emphasis on preclinical research all the more important because it is only in the animal studies that research has the opportunity to readily harvest the target tissues and perform the detailed analyses of what has happened with the cells. This need for detailed and usually time-intensive research in animal studies stands in contrast to the rapidity with which therapies have progressed to the clinic. It is now becoming clear through a number of notable examples that progress to the clinic may have occurred too quickly, before adequate testing and independent verification of results could be completed (Check, Nature 446:485-486, 2007; Chien, J Clin Investig 116:1838-1840, 2006; Giles, Nature 442:344-347, 2006). Broad reproducibility and transfer of results from one lab to another has been and always will be essential for the successful application of any cell therapy. So, what is the prognosis for cell therapy to repair heart damage? Will there be an approved cell therapy, or multiple ones, or will it require combinations of more than one cell type to be successful? These are questions often asked. The answers are difficult to know and even more difficult to predict because there are so many variables associated with cell-based therapies. There is much about the biology of cell systems that we still do not understand. Much of the pluripotency or transdifferentiation phenomena (see below) being observed go against accepted and well-tested principles for cell development and fate choice, and has caused a reevaluation of long-accepted theories. Clearly, new pathways for tissue repair and regeneration have been uncovered, but will these new pathways be sufficient to effect significant tissue repair and regeneration? Despite the false starts so far, there is the strong likelihood one or possibly multiple cell therapies will succeed. Clearly, important information has been gained, which should better guide the field to achieving success. When there is the successful verification in patients of a cell therapy, there will be an explosion of technological advances around the approach(es) that succeed. Whatever cells get approved accompanying them will be: more effective delivery methods; growth and storage methods; combination therapies, mixes of cells or cells + gene therapies; combinations with biomaterials and technologies for immune protection, allowing allografting. There are many parallel paths of technology development waiting to be brought together once there is an effective cellular approach. The coming years will no doubt bring some exciting developments.
Collapse
Affiliation(s)
- Jonathan H Dinsmore
- Advanced Cell Technology and Mytogen, Inc., Bldg. 96, 13th St., Charlestown, MA 02129, USA.
| | | |
Collapse
|
28
|
Abstract
Vascular grafts are in large demand for coronary and peripheral bypass surgeries. Although synthetic grafts have been developed, replacement of vessels with purely synthetic polymeric conduits often leads to the failure of such graft, especially in the grafts less than 6 mm in diameter or in the areas of low blood flow, mainly due to the early formation of thrombosis. Moreover, the commonly used materials lack growth potential, and long-term results have revealed several material-related failures, such as stenosis, thromboembolization, calcium deposition and infection. Tissue engineering has become a promising approach for generating a bio-compatible vessel graft with growth potential. Since the first success of constructing blood vessels with collagen and cultured vascular cells by Weinberg and Bell, there has been considerable progress in the area of vessel engineering. To date, tissue- engineered blood vessels (TEBVs) could be successfully constructed in vitro, and be used to repair the vascular defects in animal models. This review describes the major progress in the field, including the seeding cell sources, the biodegradable scaffolds, the construction technologies, as well as the encouraging achievements in clinical applications. The remaining challenges are also discussed.
Collapse
Affiliation(s)
- Wen Jie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, National Tissue Engineering Center of China, Shanghai, China
| | | | | | | |
Collapse
|
29
|
Alison MR, Choong C, Lim S. Application of liver stem cells for cell therapy. Semin Cell Dev Biol 2007; 18:819-26. [PMID: 17997335 DOI: 10.1016/j.semcdb.2007.09.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 09/28/2007] [Indexed: 02/06/2023]
Abstract
The worldwide shortage of donor livers to transplant end stage liver disease patients has prompted the search for alternative cell therapies for intractable liver disease. Embryonic stem cells can be readily differentiated into hepatocytes, and their transplantation into animals has improved liver function in the absence of teratoma formation: their use in bioartificial liver support is an obvious application. In animal models of liver disease, adopting strategies to provide a selective advantage for transplanted foetal or adult hepatocytes have proved highly effective in repopulating recipient livers, but the poor success of today's hepatocyte transplants can be attributed to the lack of a clinically applicable procedure to force a similar repopulation of the human liver. The activation of bipotential hepatic progenitor cells is clearly vital for survival in many cases of acute liver failure, but surprisingly little progress has been made with these cells in terms of transplantation. Finally there is the controversial subject of autologous bone marrow, and while the contribution of these indigenous cells to liver turnover seems at best, trivial, results from a small number of phase 1 studies of transplantation of bone marrow to cirrhotic patients have been moderately encouraging.
Collapse
Affiliation(s)
- Malcolm R Alison
- Centre for Diabetes and Metabolic Medicine, Queen Mary's School of Medicine and Dentistry, ICMS, 4 Newark Street, London E1 2AT, UK.
| | | | | |
Collapse
|
30
|
Xie SY, Ren ZR, Zhang JZ, Guo XB, Wang QX, Wang S, Lin D, Gong XL, Li W, Huang SZ, Zeng F, Zeng YT. Restoration of the balanced alpha/beta-globin gene expression in beta654-thalassemia mice using combined RNAi and antisense RNA approach. Hum Mol Genet 2007; 16:2616-25. [PMID: 17716993 DOI: 10.1093/hmg/ddm218] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The beta-thalassemia is associated with abnormality in beta-globin gene, leading to imbalanced synthesis of alpha-/beta-globin chains. Consequently, the excessive free alpha-globin chains precipitate to the erythrocyte membrane, resulting in hemolytic anemia. We have explored post-transcriptional strategies aiming at alpha-globin reduction and beta-globin enrichment on beta(654) (Hbb(th-4)/Hbb(+)) mouse, carrying a human splicing-deficient beta-globin allele (Hbb(th-4)). Lentiviral vectors of short hairpin RNA (shRNA) targeting alpha-globin and/or antisense RNA facilitating beta-globin correct splicing were microinjected into beta(654) single-cell embryos. Three transgenic strains were generated, as alpha(i)-Hbb(th-4)/Hbb(+)(shRNA), beta(a)-Hbb(th-4)/Hbb(+)(antisense) and alpha(i)beta(a)-Hbb(th-4)/Hbb(+)(both shRNA and antisense). Without notable abnormalities, all the founders and their offsprings showed sustained amelioration of hematologic parameters, ineffective erythropoiesis and extramedullary hematopoiesis. Augmented effects appeared in alpha(i)beta(a)-Hbb(th-4)/Hbb(+), which correlated with a better-balanced alpha-/beta-globin mRNA level. Among the transgenic mice integrated with shRNA and antisense RNA, one homozygous mouse (Hbb(th-4)/Hbb(th-4)) had been viable, and the 3-week survival rate for heterozygotes (Hbb(th-4)/Hbb(+)) was 97%, compared with 45.4% for untreated. Our data have demonstrated the feasibility of techniques for beta-thalassemia therapy by balancing the synthesis of alpha-/beta-globin chains.
Collapse
Affiliation(s)
- Shu-Yang Xie
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Sun Y, Xiao D, Pan XH, Zhang RS, Cui GH, Chen XG. Generation of human/rat xenograft animal model for the study of human donor stem cell behaviors in vivo. World J Gastroenterol 2007; 13:2707-16. [PMID: 17569140 PMCID: PMC4147120 DOI: 10.3748/wjg.v13.i19.2707] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To accurately and realistically elucidate human stem cell behaviors in vivo and the fundamental mechanisms controlling human stem cell fates in vivo, which is urgently required in regenerative medicine and treatments for some human diseases, a surrogate human-rat chimera model was developed.
METHODS: Human-rat chimeras were achieved by in utero transplanting low-density mononuclear cells from human umbilical cord blood into the fetal rats at 9-11 d of gestation, and subsequently, a variety of methods, including flow cytometry, PCR as well as immunohistochemical assay, were used to test the human donor contribution in the recipients.
RESULTS: Of 29 live-born recipients, 19 had the presence of human CD45+ cells in peripheral blood (PB) detected by flow cytometry, while PCR analysis on genomic DNA from 11 different adult tissues showed that 14 selected from flow cytometry-positive 19 animals possessed of donor-derived human cell engraftment in multiple tissues (i.e. liver, spleen, thymus, heart, kidney, blood, lung, muscle, gut and skin) examined at the time of tissue collection, as confirmed by detecting human β2-microglobulin expression using immunohistochemistry. In this xenogeneic system, the engrafted donor-derived human cells persisted in multiple tissues for at least 6 mo after birth. Moreover, transplanted human donor cells underwent site-specific differentiation into CK18-positive human cells in chimeric liver and CD45-positive human cells in chimeric spleen and thymus of recipients.
CONCLUSION: Taken together, these findings suggest that we successfully developed human-rat chimeras, in which xenogeneic human cells exist up to 6 mo later. This humanized small animal model, which offers an in vivo environment more closely resembling to the situations in human, provides an invaluable and effective approach for in vivo investigating human stem cell behaviors, and further in vivo examining fundamental mechanisms controlling human stem cell fates in the future. The potential for new advances in our better understanding the living biological systems in human provided by investigators in humanized animals will remain promising.
Collapse
Affiliation(s)
- Yan Sun
- Center of Experimental Animals, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | | | | | | | | | | |
Collapse
|
32
|
Ratajczak MZ, Machalinski B, Wojakowski W, Ratajczak J, Kucia M. A hypothesis for an embryonic origin of pluripotent Oct-4(+) stem cells in adult bone marrow and other tissues. Leukemia 2007; 21:860-867. [PMID: 17344915 DOI: 10.1038/sj.leu.2404630] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2007] [Revised: 01/29/2007] [Accepted: 01/31/2007] [Indexed: 02/08/2023]
Abstract
Accumulating evidence demonstrates that adult tissues contain a population of stem cells that express early developmental markers such as stage-specific embryonic antigen and transcription factors Oct-4 and Nanog. These are the markers characteristic for embryonic stem cells, epiblast stem cells and primordial germ cells. The presence of these stem cells in adult tissues including bone marrow, epidermis, bronchial epithelium, myocardium, pancreas and testes supports the concept that adult tissues contain some population of pluripotent stem cells that is deposited in embryogenesis during early gastrulation. In this review we will discuss these data and present a hypothesis that these cells could be direct descendants of the germ lineage. The germ lineage in order to pass genes on to the next generations creates soma and thus becomes a 'mother lineage' for all somatic cell lineages present in the adult body.
Collapse
Affiliation(s)
- M Z Ratajczak
- Stem Cell Biology Program at James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| | | | | | | | | |
Collapse
|
33
|
Sun Y, Xiao D, Zhang RS, Cui GH, Wang XH, Chen XG. Formation of human hepatocyte-like cells with different cellular phenotypes by human umbilical cord blood-derived cells in the human-rat chimeras. Biochem Biophys Res Commun 2007; 357:1160-5. [PMID: 17466272 DOI: 10.1016/j.bbrc.2007.04.087] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Accepted: 04/13/2007] [Indexed: 12/21/2022]
Abstract
We took advantage of the proliferative and permissive environment of the developing pre-immune fetus to develop a noninjury human-rat xenograft small animal model, in which the in utero transplantation of low-density mononuclear cells (MNCs) from human umbilical cord blood (hUCB) into fetal rats at 9-11 days of gestation led to the formation of human hepatocyte-like cells (hHLCs) with different cellular phenotypes, as revealed by positive immunostaining for human-specific alpha-fetoprotein (AFP), cytokeratin 19 (CK19), cytokeratin 8 (CK8), cytokeratin 18 (CK18), and albumin (Alb), and with some animals exhibiting levels as high as 10.7% of donor-derived human cells in the recipient liver. More interestingly, donor-derived human cells stained positively for CD34 and CD45 in the liver of 2-month-old rat. Human hepatic differentiation appeared to partially follow the process of hepatic ontogeny, as evidenced by the expression of AFP gene at an early stage and albumin gene at a later stage. Human hepatocytes generated in this model retained functional properties of normal hepatocytes. In this xenogeneic system, the engrafted donor-derived human cells persisted in the recipient liver for at least 6 months after birth. Taken together, these findings suggest that the donor-derived human cells with different cellular phenotypes are found in the recipient liver and hHLCs hold biological activity. This humanized small animal model, which offers an in vivo environment more closely resembling the situations in human, provides an invaluable approach for in vivo investigating human stem cell behaviors, and further in vivo examining fundamental mechanisms controlling human stem cell fates in the future.
Collapse
Affiliation(s)
- Yan Sun
- Center of Experimental Animals, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | | | | | | | | | | |
Collapse
|
34
|
Riordan NH, Chan K, Marleau AM, Ichim TE. Cord blood in regenerative medicine: do we need immune suppression? J Transl Med 2007; 5:8. [PMID: 17261200 PMCID: PMC1796850 DOI: 10.1186/1479-5876-5-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 01/30/2007] [Indexed: 12/31/2022] Open
Abstract
Cord blood is currently used as an alternative to bone marrow as a source of stem cells for hematopoietic reconstitution after ablation. It is also under intense preclinical investigation for a variety of indications ranging from stroke, to limb ischemia, to myocardial regeneration. A major drawback in the current use of cord blood is that substantial morbidity and mortality are associated with pre-transplant ablation of the recipient hematopoietic system. Here we raise the possibility that due to unique immunological properties of both the stem cell and non-stem cell components of cord blood, it may be possible to utilize allogeneic cells for regenerative applications without needing to fully compromise the recipient immune system. Issues raised will include: graft versus host potential, the immunogenicity of the cord blood graft, and the parallels between cord blood transplantation and fetal to maternal trafficking. The previous use of unmatched cord blood in absence of any immune ablation, as well as potential steps for widespread clinical implementation of allogeneic cord blood grafts will also be discussed.
Collapse
Affiliation(s)
| | - Kyle Chan
- Institute for Molecular Medicine, Huntington Beach, California, USA
| | | | | |
Collapse
|
35
|
Abstract
There are a number of promising cell therapy products under development for the treatment of heart failure, whether due to myocardial infarction or cardiomyopathy. Looking forward beyond current products in development, there are a multitude of possibilities that hold significant promise; however, cell-based therapies present challenges that are unique to this platform. Results from transplant studies can often be misleading and need to be interpreted in the context of fundamental biologic properties of cells and development. Provided here is a summary of the current and future developments in the field of cell therapy for cardiac regeneration along with some critical insights to interpret the multitude of studies recently undertaken. Summarized are both clinical and preclinical studies that should serve as a useful entrée into this exciting new field of therapeutic development.
Collapse
Affiliation(s)
- Nabil Dib
- Arizona Heart Institute, Phoenix, AZ 85006, USA.
| | | |
Collapse
|
36
|
Kucia M, Wu W, Ratajczak MZ. Bone marrow-derived very small embryonic-like stem cells: Their developmental origin and biological significance. Dev Dyn 2007; 236:3309-20. [PMID: 17497671 DOI: 10.1002/dvdy.21180] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Data from our and other laboratories provide evidence that bone marrow (BM) contains a population of stem cells that expresses early developmental markers such as (1) stage-specific embryonic antigen (SSEA) and (2) transcription factors Oct-4 and Nanog. These are the markers characteristic for embryonic stem cells, epiblast stem cells, and primordial germ cells (PGC). The presence of these stem cells in adult BM supports the concept that this organ contains some population of pluripotent stem cells that is deposited in embryogenesis during early gastrulation. We hypothesize that these cells could be direct descendants of the germ lineage that, to pass genes on to the next generations, has to create soma and, thus, becomes a "mother lineage" for all somatic cell lineages present in the adult body. Germ potential is established after conception in totipotent zygotes and retained in blastomeres of morula, cells from the inner cell mass of blastocyst, epiblast, and population of PGC. We will present a concept that SSEA(+) Oct-4(+) Nanog(+) cells identified in BM could be descendants of epiblast cells as well as some rare migrating astray PGC.
Collapse
Affiliation(s)
- M Kucia
- Stem Cell Biology Program at James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | |
Collapse
|
37
|
Sáez-Lara MJ, Frecha C, Martín F, Abadía F, Toscano M, Gil A, Fontana L. Transplantation of human CD34+ stem cells from umbilical cord blood to rats with thioacetamide-induced liver cirrhosis. Xenotransplantation 2006; 13:529-35. [PMID: 17059580 DOI: 10.1111/j.1399-3089.2006.00344.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Liver fibrosis results from accumulation of extracellular matrix components and is associated with many chronic hepatic diseases. There is to date no specific therapy for this disease, and patients receive treatment for its associated complications. Specific progenitor cells, known as oval cells, are present in the liver. As oval cells express markers such as CD34, they are thought to arise from a hematopoietic precursor. The aim of this work was to investigate whether transplantation of hematopoietic CD34(+) stem cells could improve hepatic fibrosis by their differentiation into hepatocytes. METHODS CD34(+) stem cells from human umbilical cord blood were purified, transduced with a lentiviral vector containing the green fluorescent protein (GFP) gene and injected via portal vein into rats with liver cirrhosis induced by the 4-month administration of thioacetamide. Rats were killed 15 and 60 days post-transplantation. RESULTS Up to 37% and 22% fluorescent cells were observed in the blood of control and cirrhotic rats, respectively, at 15 days post-transplantation. At 60 days post-transplantation, however, fluorescent cells were completely absent from the blood. Fluorescence was not detected in liver sections at either 15 or 60 days post-transplantation. Polymerase chain-reaction study to detect the GFP gene ruled out silencing of the transgene. CONCLUSIONS These results suggest that the transplanted cells did not engraft in the liver and were eliminated from the rats.
Collapse
Affiliation(s)
- María José Sáez-Lara
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Granada, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Over the past decade, the old idea that the bone marrow contains endothelial cell precursors has become an area of renewed interest. While some still believe that there are no endothelial precursors in the blood, even among those who do, there is no consensus as to what they are or what they do. In this review, we describe the problems in identifying endothelial cells and conclude that expression of endothelial nitric oxide synthase may be the most reliable antigenic indicator of the phenotype. The evidence for two different classes of endothelial precursors is also presented. We suggest that, though there is no single endothelial cell precursor, we may be able to use these phenotypic variations to our advantage in better understanding their biology. We also discuss how a variety of genetic, epigenetic, and methodological differences can account for the seemingly contradictory findings on the physiological relevance of bone marrow-derived precursors in normal vascular maintenance and in response to injury. Data on the impact of tumor type and location on the contribution of bone marrow-derived cells to the tumor vasculature are also presented. These data provide hope that we may ultimately be able to predict those tumors in which bone marrow-derived cells will have a significant contribution and design therapies accordingly. Finally, factors that regulate bone marrow cell recruitment to and function in the endothelium are beginning to be identified, and several of these, including stromal derived factor 1, monocyte chemoattractant factor-1, and vascular endothelial growth factor are discussed.
Collapse
Affiliation(s)
- Gina C Schatteman
- Integrative Physiology FH412, Univ. of Iowa, Iowa City, IA 52242, USA.
| | | | | |
Collapse
|