1
|
LeBleu VS, Kanasaki K, Lovisa S, Alge JL, Kim J, Chen Y, Teng Y, Gerami-Naini B, Sugimoto H, Kato N, Revuelta I, Grau N, Sleeman JP, Taduri G, Kizu A, Rafii S, Hochedlinger K, Quaggin SE, Kalluri R. Genetic reprogramming with stem cells regenerates glomerular epithelial podocytes in Alport syndrome. Life Sci Alliance 2024; 7:e202402664. [PMID: 38561223 PMCID: PMC10985218 DOI: 10.26508/lsa.202402664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
Glomerular filtration relies on the type IV collagen (ColIV) network of the glomerular basement membrane, namely, in the triple helical molecules containing the α3, α4, and α5 chains of ColIV. Loss of function mutations in the genes encoding these chains (Col4a3, Col4a4, and Col4a5) is associated with the loss of renal function observed in Alport syndrome (AS). Precise understanding of the cellular basis for the patho-mechanism remains unknown and a specific therapy for this disease does not currently exist. Here, we generated a novel allele for the conditional deletion of Col4a3 in different glomerular cell types in mice. We found that podocytes specifically generate α3 chains in the developing glomerular basement membrane, and that its absence is sufficient to impair glomerular filtration as seen in AS. Next, we show that horizontal gene transfer, enhanced by TGFβ1 and using allogenic bone marrow-derived mesenchymal stem cells and induced pluripotent stem cells, rescues Col4a3 expression and revive kidney function in Col4a3-deficient AS mice. Our proof-of-concept study supports that horizontal gene transfer such as cell fusion enables cell-based therapy in Alport syndrome.
Collapse
Affiliation(s)
- Valerie S LeBleu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Northwestern University Feinberg School of Medicine and Kellogg School of Management, Chicago, IL, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Keizo Kanasaki
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Sara Lovisa
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph L Alge
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jiha Kim
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Chen
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yingqi Teng
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Behzad Gerami-Naini
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Noritoshi Kato
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Ignacio Revuelta
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Nicole Grau
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jonathan P Sleeman
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Karlsruhe Institute of Technology (IBCS-BIP), Karlsruhe, Germany
| | - Gangadhar Taduri
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Akane Kizu
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Shahin Rafii
- Department of Genetic Medicine and Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY, USA
| | - Konrad Hochedlinger
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Boston, MA, USA
| | - Susan E Quaggin
- Northwestern University Feinberg School of Medicine & Feinberg Cardiovascular and Renal Research Institute, Chicago, IL, USA
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
2
|
Wang W, Zhou L, Li H, Sun T, Wen X, Li W, Esteban MA, Hoffman AR, Hu JF, Cui J. Profiling the role of m6A effectors in the regulation of pluripotent reprogramming. Hum Genomics 2024; 18:33. [PMID: 38566168 PMCID: PMC10986062 DOI: 10.1186/s40246-024-00597-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
The N6-methyladenosine (m6A) RNA modification plays essential roles in multiple biological processes, including stem cell fate determination. To explore the role of the m6A modification in pluripotent reprogramming, we used RNA-seq to map m6A effectors in human iPSCs, fibroblasts, and H9 ESCs, as well as in mouse ESCs and fibroblasts. By integrating the human and mouse RNA-seq data, we found that 19 m6A effectors were significantly upregulated in reprogramming. Notably, IGF2BPs, particularly IGF2BP1, were among the most upregulated genes in pluripotent cells, while YTHDF3 had high levels of expression in fibroblasts. Using quantitative PCR and Western blot, we validated the pluripotency-associated elevation of IGF2BPs. Knockdown of IGF2BP1 induced the downregulation of stemness genes and exit from pluripotency. Proteome analysis of cells collected at both the beginning and terminal states of the reprogramming process revealed that the IGF2BP1 protein was positively correlated with stemness markers SOX2 and OCT4. The eCLIP-seq target analysis showed that IGF2BP1 interacted with the coding sequence (CDS) and 3'UTR regions of the SOX2 transcripts, in agreement with the location of m6A modifications. This study identifies IGF2BP1 as a vital pluripotency-associated m6A effector, providing new insight into the interplay between m6A epigenetic modifications and pluripotent reprogramming.
Collapse
Affiliation(s)
- Wenjun Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
- VA Palo Alto Health Care System, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Lei Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Hui Li
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Tingge Sun
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xue Wen
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Miguel A Esteban
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, PR China
| | - Andrew R Hoffman
- VA Palo Alto Health Care System, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Ji-Fan Hu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
- VA Palo Alto Health Care System, Stanford University School of Medicine, Palo Alto, CA, 94304, USA.
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
3
|
Technical, Biological and Molecular Aspects of Somatic Cell Nuclear Transfer – A Review. ANNALS OF ANIMAL SCIENCE 2022. [DOI: 10.2478/aoas-2021-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Abstract
Since the announcement of the birth of the first cloned mammal in 1997, Dolly the sheep, 24 animal species including laboratory, farm, and wild animals have been cloned. The technique for somatic cloning involves transfer of the donor nucleus of a somatic cell into an enucleated oocyte at the metaphase II (MII) stage for the generation of a new individual, genetically identical to the somatic cell donor. There is increasing interest in animal cloning for different purposes such as rescue of endangered animals, replication of superior farm animals, production of genetically engineered animals, creation of biomedical models, and basic research. However, the efficiency of cloning remains relatively low. High abortion, embryonic, and fetal mortality rates are frequently observed. Moreover, aberrant developmental patterns during or after birth are reported. Researchers attribute these abnormal phenotypes mainly to incomplete nuclear remodeling, resulting in incomplete reprogramming. Nevertheless, multiple factors influence the success of each step of the somatic cloning process. Various strategies have been used to improve the efficiency of nuclear transfer and most of the phenotypically normal born clones can survive, grow, and reproduce. This paper will present some technical, biological, and molecular aspects of somatic cloning, along with remarkable achievements and current improvements.
Collapse
|
4
|
Retinal Lineage Therapeutic Specific Effect of Human Orbital and Abdominal Adipose-Derived Mesenchymal Stem Cells. Stem Cells Int 2021; 2021:7022247. [PMID: 34712333 PMCID: PMC8548122 DOI: 10.1155/2021/7022247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/09/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Retinal degenerative diseases are one of the main causes of complete blindness in aged population. In this study, we compared the therapeutic potential for retinal degeneration of human mesenchymal stem cells derived from abdominal subcutaneous fat (ABASCs) or from orbital fat (OASCs) due to their accessibility and mutual embryonic origin with retinal tissue, respectively. OASCs were found to protect RPE cells from cell death and were demonstrated to increase early RPE precursor markers, while ABASCs showed a raise in retinal precursor marker expression. Subretinal transplantation of OASCs in a mouse model of retinal degeneration led to restoration of the RPE layer while transplantation of ABASCs resulted in a significant restoration of the photoreceptor layer. Taken together, we demonstrated a lineage-specific therapeutic effect for either OASCs or ABASCs in retinal regeneration.
Collapse
|
5
|
Lymphatic Endothelial Cell Progenitors in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1234:87-105. [PMID: 32040857 DOI: 10.1007/978-3-030-37184-5_7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Tumor lymphatics play a key role in cancer progression as they are solely responsible for transporting malignant cells to regional lymph nodes (LNs), a process that precedes and promotes systemic lethal spread. It is broadly accepted that tumor lymphatic sprouting is induced mainly by soluble factors derived from tumor-associated macrophages (TAMs) and malignant cells. However, emerging evidence strongly suggests that a subset of TAMs, myeloid-lymphatic endothelial cell progenitors (M-LECP), also contribute to the expansion of lymphatics through both secretion of paracrine factors and a self-autonomous mode. M-LECP are derived from bone marrow (BM) precursors of the monocyte-macrophage lineage and characterized by unique co-expression of markers identifying lymphatic endothelial cells (LEC), stem cells, M2-type macrophages, and myeloid-derived immunosuppressive cells. This review describes current evidence for the origin of M-LECP in the bone marrow, their recruitment tumors and intratumoral trafficking, similarities to other TAM subsets, and mechanisms promoting tumor lymphatics. We also describe M-LECP integration into preexisting lymphatic vessels and discuss potential mechanisms and significance of this event. We conclude that improved mechanistic understanding of M-LECP functions within the tumor environment may lead to new therapeutic approaches to suppress tumor lymphangiogenesis and metastasis to lymph nodes.
Collapse
|
6
|
Courson JA, Smith I, Do T, Landry PT, Hargrave A, Behzad AR, Hanlon SD, Rumbaut RE, Smith CW, Burns AR. Serial block-face scanning electron microscopy reveals neuronal-epithelial cell fusion in the mouse cornea. PLoS One 2019; 14:e0224434. [PMID: 31721785 PMCID: PMC6853292 DOI: 10.1371/journal.pone.0224434] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/14/2019] [Indexed: 11/28/2022] Open
Abstract
The cornea is the most highly innervated tissue in the body. It is generally accepted that corneal stromal nerves penetrate the epithelial basal lamina giving rise to intra-epithelial nerves. During the course of a study wherein we imaged corneal nerves in mice, we observed a novel neuronal-epithelial cell interaction whereby nerves approaching the epithelium in the cornea fused with basal epithelial cells, such that their plasma membranes were continuous and the neuronal axoplasm freely abutted the epithelial cytoplasm. In this study we sought to determine the frequency, distribution, and morphological profile of neuronal-epithelial cell fusion events within the cornea. Serial electron microscopy images were obtained from the anterior stroma in the paralimbus and central cornea of 8–10 week old C57BL/6J mice. We found evidence of a novel alternative behavior involving a neuronal-epithelial interaction whereby 42.8% of central corneal nerve bundles approaching the epithelium contain axons that fuse with basal epithelial cells. The average surface-to-volume ratio of a penetrating nerve was 3.32, while the average fusing nerve was smaller at 1.39 (p ≤ 0.0001). Despite this, both neuronal-epithelial cell interactions involve similarly sized discontinuities in the basal lamina. In order to verify the plasma membrane continuity between fused neurons and epithelial cells we used the lipophilic membrane tracer DiI. The majority of corneal nerves were labeled with DiI after application to the trigeminal ganglion and, consistent with our ultrastructural observations, fusion sites recognized as DiI-labeled basal epithelial cells were located at points of stromal nerve termination. These studies provide evidence that neuronal-epithelial cell fusion is a cell-cell interaction that occurs primarily in the central cornea, and fusing nerve bundles are morphologically distinct from penetrating nerve bundles. This is, to our knowledge, the first description of neuronal-epithelial cell fusion in the literature adding a new level of complexity to the current understanding of corneal innervation.
Collapse
Affiliation(s)
- Justin A. Courson
- University of Houston, College of Optometry, Houston, TX, United States of America
- * E-mail:
| | - Ian Smith
- University of Houston, College of Optometry, Houston, TX, United States of America
| | - Thao Do
- University of Houston, College of Optometry, Houston, TX, United States of America
| | - Paul T. Landry
- University of Houston, College of Optometry, Houston, TX, United States of America
| | - Aubrey Hargrave
- University of Houston, College of Optometry, Houston, TX, United States of America
| | - Ali R. Behzad
- King Abdullah University of Science and Technology (KAUST), Core Labs, Thuwal, Saudi Arabia
| | - Sam D. Hanlon
- University of Houston, College of Optometry, Houston, TX, United States of America
| | - Rolando E. Rumbaut
- Baylor College of Medicine, Children’s Nutrition Center, Houston, TX, United States of America
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, United States of America
| | - C. Wayne Smith
- Baylor College of Medicine, Children’s Nutrition Center, Houston, TX, United States of America
| | - Alan R. Burns
- University of Houston, College of Optometry, Houston, TX, United States of America
- Baylor College of Medicine, Children’s Nutrition Center, Houston, TX, United States of America
| |
Collapse
|
7
|
Hong YJ, Hong K, Byun S, Choi HW, Do JT. Reprogramming of Extraembryonic Trophoblast Stem Cells into Embryonic Pluripotent State by Fusion with Embryonic Stem Cells. Stem Cells Dev 2018; 27:1350-1359. [PMID: 29993328 DOI: 10.1089/scd.2018.0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pluripotential reprogramming has been examined using various technologies, including nuclear transfer, cell fusion, and direct reprogramming. Many studies have used differentiated cells for reprogramming experiments, and nearly all type of somatic cells can acquire pluripotency. However, within the embryo, other cells types are present in addition to somatic cells. The blastocyst stage embryo consists of two main types of cells, inner cell mass and trophectoderm (TE). TE cells are the first differentiated form of the totipotent zygote and differ from epiblast cells. Thus, we examined whether extraembryonic cells can be reprogrammed using a cell-cell fusion method. Trophoblast stem cells (TSCs), which can be obtained from the TE, are known to acquire pluripotency by transcription factor Oct4 overexpression or somatic cell nuclear transfer. In this study, we demonstrated that TSCs can acquire pluripotent properties by cell fusion with embryonic stem cells (ESCs). TSC-ESC hybrids reactivated Oct4-GFP and displayed self-renewal properties. They expressed the pluripotency markers Oct4 and Nanog, whereas the expression of Cdx2 and Tead4, trophoblast lineage markers, was diminished. Moreover, these cells developed into three germ layers similarly to other pluripotent stem cells. RNA-seq analysis showed that global gene expression patterns of TSC-ESC hybrids are more similar to ESCs than TSCs. Thus, we demonstrated that TSCs successfully complete reprogramming and acquire pluripotency by cell fusion-induced reprogramming.
Collapse
Affiliation(s)
- Yean Ju Hong
- 1 Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology, Konkuk University , Seoul, Republic of Korea
| | - Kwonho Hong
- 1 Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology, Konkuk University , Seoul, Republic of Korea
| | - Seki Byun
- 1 Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology, Konkuk University , Seoul, Republic of Korea
| | - Hyun Woo Choi
- 2 Department of Animal Science, Chonbuk National University , Jeonju-si, Republic of Korea
| | - Jeong Tae Do
- 1 Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology, Konkuk University , Seoul, Republic of Korea
| |
Collapse
|
8
|
Bahadori A, Moreno-Pescador G, Oddershede LB, Bendix PM. Remotely controlled fusion of selected vesicles and living cells: a key issue review. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2018; 81:032602. [PMID: 29369822 DOI: 10.1088/1361-6633/aa9966] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Remote control over fusion of single cells and vesicles has a great potential in biological and chemical research allowing both transfer of genetic material between cells and transfer of molecular content between vesicles. Membrane fusion is a critical process in biology that facilitates molecular transport and mixing of cellular cytoplasms with potential formation of hybrid cells. Cells precisely regulate internal membrane fusions with the aid of specialized fusion complexes that physically provide the energy necessary for mediating fusion. Physical factors like membrane curvature, tension and temperature, affect biological membrane fusion by lowering the associated energy barrier. This has inspired the development of physical approaches to harness the fusion process at a single cell level by using remotely controlled electromagnetic fields to trigger membrane fusion. Here, we critically review various approaches, based on lasers or electric pulses, to control fusion between individual cells or between individual lipid vesicles and discuss their potential and limitations for present and future applications within biochemistry, biology and soft matter.
Collapse
Affiliation(s)
- Azra Bahadori
- Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 Copenhagen, Denmark
| | | | | | | |
Collapse
|
9
|
Abstract
It is extremely rare for a single experiment to be so impactful and timely that it shapes and forecasts the experiments of the next decade. Here, we review how two such experiments-the generation of human induced pluripotent stem cells (iPSCs) and the development of CRISPR/Cas9 technology-have fundamentally reshaped our approach to biomedical research, stem cell biology, and human genetics. We will also highlight the previous knowledge that iPSC and CRISPR/Cas9 technologies were built on as this groundwork demonstrated the need for solutions and the benefits that these technologies provided and set the stage for their success.
Collapse
Affiliation(s)
- Dirk Hockemeyer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Rudolf Jaenisch
- The Whitehead Institute for Biomedical Research and Department of Biology, MIT, Cambridge, MA 02142, USA
| |
Collapse
|
10
|
Wang Q, Stern JH, Temple S. Regenerative Medicine: Solution in Sight. RETINAL DEGENERATIVE DISEASES 2016; 854:543-8. [DOI: 10.1007/978-3-319-17121-0_72] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
11
|
Wei HJ, Nickoloff JA, Chen WH, Liu HY, Lo WC, Chang YT, Yang PC, Wu CW, Williams DF, Gelovani JG, Deng WP. FOXF1 mediates mesenchymal stem cell fusion-induced reprogramming of lung cancer cells. Oncotarget 2015; 5:9514-29. [PMID: 25237908 PMCID: PMC4253450 DOI: 10.18632/oncotarget.2413] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Several reports suggest that malignant cells generate phenotypic diversity through fusion with various types of stromal cells within the tumor microenvironment. Mesenchymal stem cell (MSC) is one of the critical components in the tumor microenvironment and a promising fusogenic candidate, but the underlying functions of MSC fusion with malignant cell have not been fully examined. Here, we demonstrate that MSCs fuse spontaneously with lung cancer cells, and the latter is reprogrammed to slow growth and stem-like state. Transcriptome profiles reveal that lung cancer cells are reprogrammed to a more benign state upon MSC fusion. We further identified FOXF1 as a reprogramming mediator that contributes not only to the reprogramming toward stemness but also to the p21-regulated growth suppression in fusion progeny. Collectively, MSC fusion does not enhance the intrinsic malignancy of lung cancer cells. The anti-malignant effects of MSC fusion-induced reprogramming on lung cancer cells were accomplished by complementation of tumorigenic defects, including restoration of p21 function and normal terminal differentiation pathways as well as up-regulation of FOXF1, a putative tumor suppressor. Such fusion process raises the therapeutic potential that MSC fusion can be utilized to reverse cellular phenotypes in cancer.
Collapse
Affiliation(s)
- Hong-Jian Wei
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan. Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Wei-Hong Chen
- Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Hen-Yu Liu
- Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Cheng Lo
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan. School of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ya-Ting Chang
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Pan-Chyr Yang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Cheng-Wen Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei 112, Taiwan
| | - David F Williams
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC 27157, USA
| | - Juri G Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI 48201, USA Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Win-Ping Deng
- Graduate Institute of Biomedical Materials and Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan. Stem Cell Research Center, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
12
|
Salzman V, Porro V, Bollati-Fogolín M, Aguilar PS. Quantitation of yeast cell-cell fusion using multicolor flow cytometry. Cytometry A 2015; 87:843-54. [DOI: 10.1002/cyto.a.22701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/06/2015] [Accepted: 05/09/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Valentina Salzman
- Laboratorio De Biología Celular De Membranas, Institut Pasteur De Montevideo; Montevideo 11400 Uruguay
| | - Valentina Porro
- Cell Biology Unit, Institut Pasteur De Montevideo; Montevideo 11400 Uruguay
| | | | - Pablo S. Aguilar
- Laboratorio De Biología Celular De Membranas, Institut Pasteur De Montevideo; Montevideo 11400 Uruguay
- Laboratorio de Biología Celular de Membranas, Instituto De Investigaciones Biotecnológicas, Universidad Nacional De San Martín, CONICET; San Martín Buenos Aires Argentina
| |
Collapse
|
13
|
Olmos LC, Nazari H, Rodger DC, Humayun MS. Stem Cell Therapy for the Treatment of Dry Age-Related Macular Degeneration. CURRENT OPHTHALMOLOGY REPORTS 2015. [DOI: 10.1007/s40135-014-0058-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
14
|
Parris GE. Cell-Cell Fusion, Chemotaxis and Metastasis. INTERCELLULAR COMMUNICATION IN CANCER 2015:227-254. [DOI: 10.1007/978-94-017-7380-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
15
|
Bilousova G, Roop DR. Induced pluripotent stem cells in dermatology: potentials, advances, and limitations. Cold Spring Harb Perspect Med 2014; 4:a015164. [PMID: 25368014 DOI: 10.1101/cshperspect.a015164] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of methods for reprogramming adult somatic cells into induced pluripotent stem cells (iPSCs) has raised the possibility of producing truly personalized treatment options for numerous diseases. Similar to embryonic stem cells (ESCs), iPSCs can give rise to any cell type in the body and are amenable to genetic correction by homologous recombination. These ESC properties of iPSCs allow for the development of permanent corrective therapies for many currently incurable disorders, including inherited skin diseases, without using embryonic tissues or oocytes. Here, we review recent progress and limitations of iPSC research with a focus on clinical applications of iPSCs and using iPSCs to model human diseases for drug discovery in the field of dermatology.
Collapse
Affiliation(s)
- Ganna Bilousova
- Department of Dermatology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045 Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Dennis R Roop
- Department of Dermatology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045 Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
16
|
The implications of stem cell applications for diseases of the respiratory system. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2012; 130:39-54. [PMID: 22915199 DOI: 10.1007/10_2012_153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Stem cells possess the unique properties of unlimited self-renewal capability and a broad differentiation spectrum to produce multiple different cell types. This provides many platforms to explore novel multidisciplinary approaches to create and/or restore functional three-dimensional tissues or organs for the treatment of a range of diseases. In this chapter, in the context of respiratory diseases, we review the unique properties of stem cells, and how they have been studied for their therapeutic potential in cell therapy and tissue engineering. In addition, we give a brief overview of the current clinical studies on the use of stem cells for both acute and chronic respiratory diseases.
Collapse
|
17
|
Serov OL, Matveeva NM, Khabarova AA. Reprogramming mediated by cell fusion technology. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 291:155-90. [PMID: 22017976 DOI: 10.1016/b978-0-12-386035-4.00005-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This review is focused on recent advances in fusion-based reprogramming of cells of different pluripotent statuses or lineage origins. Recent findings are discussed from standpoints of both the developmental potency of hybrid cells generated by fusion of pluripotent embryonic stem (ES) cells, embryonal carcinoma (EC) cells, and somatic cells and epigenetic mechanisms and other aspects involved in the reprogramming process. Complete reprogramming occurs at least 5-7 days after fusion and includes at least two steps. (i) initiation at the heterokaryon stage and choice of the direction of reprogramming using an "all-or-none principle" to establish the dominance of one parental genome and (ii) "fixation" of the newly acquired expression profile by epigenetic mechanisms. The first step is realized without cell division, whereas the second requires cell proliferation. Reprogramming in hybrid cells is rapid and complete. Thus, cell fusion is a powerful tool for reprogramming.
Collapse
Affiliation(s)
- Oleg L Serov
- Institute of Cytology and Genetics, Academy of Sciences of Russia, Siberian Branch, Novosibirsk, Russia
| | | | | |
Collapse
|
18
|
Zhou Z, Xu Y, Zhong Q, Zheng J. Phenotypic characteristics of hybrid cells generated by transferring neuronal nuclei into bone marrow stromal cell cytoplasts. Brain Res Bull 2012; 87:303-11. [PMID: 22093690 DOI: 10.1016/j.brainresbull.2011.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 10/09/2011] [Accepted: 11/02/2011] [Indexed: 01/05/2023]
Abstract
Bone marrow stromal cells (BMSCs) are promising donor cells for transplantation therapies for a variety of diseases. However, there still lack efficient ways to induce directional differentiation of BMSCs to promote their practical use in transplantation therapy. In this study, we constructed hybrid cells by transferring neuronal nuclei into BMSC cytoplasts and investigated the proliferative capacity and phenotypic characteristics of the hybrid cells. The neuronal nuclei were labeled with Hoechst 33342 before the transfer process, and the cell membrane antigen CD71 was used as a marker of BMSC cytoplasts. The BMSC cytoplasts and neuronal karyoplasts were separated by Ficoll density gradient ultracentrifugation. The hybrid cells were generated by the polyethylene glycol-mediated fusion of BMSC cytoplasts with neuronal karyoplasts. The hybrid cells exhibited Hoechst 33342 staining in their nuclei and CD71 staining on their cytomembranes, which confirmed the success of cell fusion. The hybrid cells were positive for BrdU immunostaining. Viability analysis of the cultured hybrid cells by the MTT assay demonstrated their proliferative ability. Immunocytochemical staining revealed the expression of the neuron-specific markers NeuN and MAP2 in the third passage hybrid cells, which indicated their neuronal phenotypic characteristics. The results demonstrated that the hybrid cells produced by fusing neuronal karyoplasts with BMSC cytoplasts had proliferative capability and expressed the neuron-specific markers. Further study is required to investigate the phenotype of the hybrid cells both structurally and functionally.
Collapse
Affiliation(s)
- Zhujuan Zhou
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | | | | | | |
Collapse
|
19
|
Avinoam O, Podbilewicz B. Eukaryotic cell-cell fusion families. CURRENT TOPICS IN MEMBRANES 2012; 68:209-34. [PMID: 21771501 DOI: 10.1016/b978-0-12-385891-7.00009-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ori Avinoam
- Department of Biology, Technion, Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
20
|
Cha Y, Moon BH, Lee MO, Ahn HJ, Lee HJ, Lee KA, Fornace AJ, Kim KS, Cha HJ, Park KS. Zap70 functions to maintain stemness of mouse embryonic stem cells by negatively regulating Jak1/Stat3/c-Myc signaling. Stem Cells 2010; 28:1476-86. [PMID: 20641039 PMCID: PMC3164580 DOI: 10.1002/stem.470] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Zeta-chain-associated protein kinase-70 (Zap70), a Syk family tyrosine kinase, has been reported to be present exclusively in normal T-cells, natural killer cells, and B cells, serving as a pivotal regulator of antigen-mediated receptor signaling and development. In this study, we report that Zap70 is expressed in undifferentiated mouse embryonic stem cells (mESCs) and may critically regulate self-renewal and pluripotency in mESCs. We found that Zap70 knocked-down mESCs (Zap70KD) show sustained self-renewal and defective differentiation. In addition, we present evidence that the sustained self-renewal in Zap70KD is associated with enhanced Jak/Stat3 signaling and c-Myc induction. These altered signaling appears to result from upregulated leukemia inhibitory factor receptor and downregulated src homology region 2 domain containing phosphatase 1 (SHP-1) phosphatase activity. On the basis of these results, we propose that in undifferentiated mESCs, Zap70 plays important roles in modulating the balance between self-renewal capacity and pluripotent differentiation ability as a key regulator of the Jak/Stat3/c-Myc signaling pathway.
Collapse
Affiliation(s)
- Young Cha
- Department of Biomedical Science, College of Life Science, CHA University, Pochonsi Gyeonggi-do, 487-010, Korea
| | - Bo-Hyun Moon
- CHA Stem Cell Institute, CHA University, Seoul 135-081, Korea
| | - Mi-Ok Lee
- Department of Biomedical Science, College of Life Science, CHA University, Pochonsi Gyeonggi-do, 487-010, Korea
| | - Hee-Jin Ahn
- Department of Biomedical Science, College of Life Science, CHA University, Pochonsi Gyeonggi-do, 487-010, Korea
| | - Hye-Jin Lee
- CHA Stem Cell Institute, CHA University, Seoul 135-081, Korea
| | - Kyung-Ah Lee
- Department of Biomedical Science, College of Life Science, CHA University, Pochonsi Gyeonggi-do, 487-010, Korea
- CHA Stem Cell Institute, CHA University, Seoul 135-081, Korea
| | - Albert J. Fornace
- Department of Biochemistry and Molecular & Cellular, Georgetown University, Washington DC 20057, USA
| | - Kwang-Soo Kim
- CHA Stem Cell Institute, CHA University, Seoul 135-081, Korea
- Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Hyuk-Jin Cha
- Department of Biomedical Science, College of Life Science, CHA University, Pochonsi Gyeonggi-do, 487-010, Korea
- CHA Stem Cell Institute, CHA University, Seoul 135-081, Korea
| | - Kyung-Soon Park
- Department of Biomedical Science, College of Life Science, CHA University, Pochonsi Gyeonggi-do, 487-010, Korea
- CHA Stem Cell Institute, CHA University, Seoul 135-081, Korea
| |
Collapse
|
21
|
Dodson MV, Hausman GJ, Guan L, Du M, Rasmussen TP, Poulos SP, Mir P, Bergen WG, Fernyhough ME, McFarland DC, Rhoads RP, Soret B, Reecy JM, Velleman SG, Jiang Z. Skeletal muscle stem cells from animals I. Basic cell biology. Int J Biol Sci 2010; 6:465-74. [PMID: 20827399 PMCID: PMC2935669 DOI: 10.7150/ijbs.6.465] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 08/27/2010] [Indexed: 01/14/2023] Open
Abstract
Skeletal muscle stem cells from food-producing animals are of interest to agricultural life scientists seeking to develop a better understanding of the molecular regulation of lean tissue (skeletal muscle protein hypertrophy) and intramuscular fat (marbling) development. Enhanced understanding of muscle stem cell biology and function is essential for developing technologies and strategies to augment the metabolic efficiency and muscle hypertrophy of growing animals potentially leading to greater efficiency and reduced environmental impacts of animal production, while concomitantly improving product uniformity and consumer acceptance and enjoyment of muscle foods.
Collapse
Affiliation(s)
- Michael V Dodson
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Bulla GA, Luong Q, Shrestha S, Reeb S, Hickman S. Genome-wide analysis of hepatic gene silencing in mammalian cell hybrids. Genomics 2010; 96:323-32. [PMID: 20801210 DOI: 10.1016/j.ygeno.2010.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 08/13/2010] [Accepted: 08/17/2010] [Indexed: 12/29/2022]
Abstract
Silencing of tissue-specific gene expression in mammalian somatic cell hybrids is a well-documented epigenetic phenomenon which is both profound (involving a large number of genes) and enigmatic. Our aim was to utilize whole-genome microarray analyses to determine the true extent of gene silencing on a genomic level. By comparing gene expression profiles of hepatoma×fibroblast cell hybrids with those of parental cells, we have identified over 300 liver-enriched genes that are repressed at least 5-fold in the cell hybrids, the majority of which are repressed at least 10-fold. Also, we identify nearly 200 fibroblast-enriched genes that are repressed at least 5-fold. Silenced hepatic genes include several that encode transcription factors and proteins involved in signal transduction pathways. These data suggest that extensive reprogramming occurs in cell hybrids, leading to a nearly global (although not complete) loss of tissue-specific gene expression.
Collapse
Affiliation(s)
- Gary A Bulla
- Department of Biological Sciences, Eastern Illinois University, 600 Lincoln Avenue, Charleston, IL 61920, USA.
| | | | | | | | | |
Collapse
|
23
|
Pristyazhnyuk IE, Matveeva NM, Graphodatskii AS, Serdyukova NA, Serov OL. Chromosome composition of interspecies hybrid embryonic stem cells in mice. ACTA ACUST UNITED AC 2010. [DOI: 10.1134/s1990519x10020021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Lluis F, Cosma MP. Cell-fusion-mediated somatic-cell reprogramming: a mechanism for tissue regeneration. J Cell Physiol 2010; 223:6-13. [PMID: 20049847 DOI: 10.1002/jcp.22003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Spontaneous cell fusion between two cells of different lineages will originate new hybrid cells that have different features from the original parent cells. It has been shown that injury to a tissue can enhance spontaneous cell-cell fusion events. If one of the parent cells of a cell-cell fusion is highly plastic, such as a stem cell, and the other is a somatic cell, their fusion can be followed by reprogramming events that can generate new hybrid pluripotent cells. These, in turn, have the potential to differentiate and regenerate the damaged tissue. However, if this process is deregulated, this would provide a mechanism for cancer development.
Collapse
Affiliation(s)
- Frederic Lluis
- Telethon Institute of Genetics and Medicine and Institute of Genetics and Biophysics, CNR, Naples, Italy
| | | |
Collapse
|
25
|
Lavagnolli TM, Fonseca SA, Serafim RC, Pereira VS, Santos EJ, Abdelmassih S, Kerkis A, Kerkis I. Presumptive germ cells derived from mouse pluripotent somatic cell hybrids. Differentiation 2009; 78:124-30. [DOI: 10.1016/j.diff.2009.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 06/25/2009] [Accepted: 07/01/2009] [Indexed: 10/20/2022]
|
26
|
Yan L, Cai C, Li J, Xu S, Chang Q, Li Y, Wu B. Present status and perspectives of stem cell-based therapies for gastrointestinal diseases. Stem Cell Rev Rep 2009; 5:278-82. [PMID: 19590987 DOI: 10.1007/s12015-009-9070-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Accepted: 04/29/2009] [Indexed: 12/11/2022]
Abstract
In recent years the interest in stem cell-based therapies for gastrointestinal injury has been increasing continuously. From the clinical point of view, transplantation of bone marrow derived stem cells may represent an alternative therapy for gastrointestinal injury, such as radioactive injury, inflammatory bowel disease, and other refractory gastrointestinal tract injury. There were several reports indicated that bone marrow derived stem cells located in the injured gastrointestinal tract and contributed to its regeneration by differentiating into functional epithelia cells or infusing with the gastrointestinal stem cells. Although the concept of cell-based therapy for various diseases of the gastrointestinal is widely accepted, the practical approach in humans remains difficult. Here we discussed the recent published data on clinical and experimental bone marrow stem cell transplantation and the possible role of stem cells in gastrointestinal tissue repair.
Collapse
Affiliation(s)
- Li Yan
- Gastrointestinal Department of Southern Building, PLA general hospital, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Wang W, Li W, Ong LL, Furlani D, Kaminski A, Liebold A, Lützow K, Lendlein A, Wang J, Li RK, Steinhoff G, Ma N. Localized SDF-1alpha gene release mediated by collagen substrate induces CD117 stem cells homing. J Cell Mol Med 2008; 14:392-402. [PMID: 19413887 PMCID: PMC3837586 DOI: 10.1111/j.1582-4934.2008.00624.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Stromal cell-derived factor-1α (SDF-1α) mediated mobilization and homing of stem cells showed promising potential in stem cell based tissue engineering and regenerative medicine. However local and sustained release of SDF-1α is indispensable for stem cell mediated regenerative process due to its short half-life under inflammatory conditions. In this study, a gene activated collagen substrate (GAC) was formed via assembly of plasmid encoding SDF-1α into a collagen substrate to create a microenvironment favoring stem cell homing. Local release of SDF-1α from the transfected cells on GAC and its effect on CD117+ stem cell homing were investigated. Non-viral poly-ethyleneimine (25kDa PEI)/DNA complexes were mixed with rat tail collagen solution to form the GAC. Optimization of GAC was carried out based on collagen effects on the PEI/DNA complexes, viability and luciferase expression of COS7 cells on GAC. CD117+ stem cells homing in response to SDF-1α local expression from transfected cells on GAC were investigated in a flow chamber in vitro and in a mouse hind limb model in vivo. The gene expression, migration of CD117+ stem cells and the induced inflammation were investigated with immunostaining, reverse transcription polymerase chain reaction (RT-PCR) and H&E staining. The optimized parameters for GAC were DNA dosage 10 μg/cm2, molar ratio of PEI nitrogen in primary amine to DNA phosphate (N/P ratio) 4 and mass ratio of collagen to DNA (C/D ratio) 1.0. It kept cell viability above 75% and transfection efficiency around 5.8 × 105 RLU/mg protein. GAC allowed the sustained gene release up to 60 days. GAC mediated SDF-1α gene release induced migration and homing of CD117+ stem cells in vitro and in vivo significantly, and the inflammation of GAC reduced significantly two weeks after transplantation. GAC is a promising stem cell based therapeutic strategy for regenerative medicine.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Cardiac Surgery, University of Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Afanas'yev SA, Sviridov IN, Shakhov VP, Falaleyeva LP, Popov SV, Karpov RS. Colony-forming cells in rat myocardium after destructive exposure and intramyocardial transplantation of bone marrow cells. Bull Exp Biol Med 2008; 145:137-40. [PMID: 19024022 DOI: 10.1007/s10517-008-0027-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The content of colony-forming cells in myocardial cell culture from the perinecrotic zone of rat heart was evaluated on day 40 after cryodestruction. The mean cellularity after cryodestruction was 12-fold lower than in intact animals. Intramyocardial transplantation of bone marrow cells (mononuclears, mesenchymal stem cells, and mesenchymal stem cells treated with 5-azacitidine) into the perinecrotic zone increased the content of colony-forming cells. After transplantation of mesenchymal stem cells and mesenchymal stem cells treated with 5-azacitidine, the number of colonies reached 33 +/- 7 and 11 +/- 3, the mean cellularity being 2975 +/- 80 and 1105 +/- 42 cells/cm2, respectively. Hence, intramyocardial transplantation of mesenchymal stem cells created an appreciable pool of colony-forming cells in the myocardial perinecrotic zone. Treatment with 5-azacitidine reduced survival of mesenchymal stem cells after intramyocardial transplantation.
Collapse
Affiliation(s)
- S A Afanas'yev
- Institute of Cardiology, Tomsk Research Center, Siberian Division of Russian Academy of Medical Sciences, Tomsk
| | | | | | | | | | | |
Collapse
|
29
|
Robinson C, Kolb AF. Analysis of mammary specific gene locus regulation in differentiated cells derived by somatic cell fusion. Exp Cell Res 2008; 315:508-22. [PMID: 19014936 DOI: 10.1016/j.yexcr.2008.10.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 09/19/2008] [Accepted: 10/14/2008] [Indexed: 10/21/2022]
Abstract
The transcriptional regulation of a gene is best analysed in the context of its normal chromatin surroundings. However, most somatic cells, in contrast to embryonic stem cells, are refractory to accurate modification by homologous recombination. We show here that it is possible to introduce precise genomic modifications in ES cells and to analyse the phenotypic consequences in differentiated cells by using a combination of gene targeting, site-specific recombination and somatic cell fusion. To provide a proof of principle, we have analysed the regulation of the casein gene locus in mammary gland cells derived from modified murine ES cells by somatic cell fusion. A beta-galactosidase reporter gene was inserted in place of the beta-casein gene and the modified ES cells, which do not express the reporter gene, were fused with the mouse mammary gland cell line HC11. The resulting cell clones expressed the beta-galactosidase gene to a similar extent and with similar hormone responsiveness as the endogenous gene. However, a reporter gene under the control of a minimal beta-casein promoter (encompassing the two consensus STAT5 binding sites which mediate the hormone response of the casein genes) was unable to replicate expression levels or hormone responsiveness of the endogenous gene when inserted into the same site of the casein locus. As expected, these results implicate sequences other than the STAT5 sites in the regulation of the beta-casein gene.
Collapse
Affiliation(s)
- Claire Robinson
- Molecular Recognition Group, Hannah Research Institute, Ayr KA6 5HL, UK
| | | |
Collapse
|
30
|
Menzorov AG, Matveeva NM, Larkin DM, Zaykin DV, Serov OL. Fate of parental mitochondria in embryonic stem hybrid cells. ACTA ACUST UNITED AC 2008. [DOI: 10.1134/s1990519x08040093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Menzorov AG, Matveeva NM, Larkin DM, Zaykin DV, Serov OL. [Fate of parental mitochondria in embryonic stem hybrid cells]. TSITOLOGIIA 2008; 50:711-8. [PMID: 18822791 PMCID: PMC2775045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
When hybrid cells are created, not only nuclear genomes of parental cells unite but their cytoplasm as well. Mitochondrial DNA (mtDNA) is a convenient marker of cytoplasm allowing one to gain insight into the organization of hybrid cell cytoplasm. We analyzed the parental mtDNAs in hybrid cells resulting from fusion of Mus musculus embryonic stem (ES) cells with splenocytes and fetal fibroblasts of DD/c mice or with splenocytes of M. caroli. Identification of the parental mtDNAs in hybrid cells was based on polymorphism among the parental mtDNAs for certain restrictases. We found that intra- and inter-specific ES cell-splenocyte hybrid cells lost entirely or partially mtDNA derived from the somatic partner, whereas ES cell-fibroblast hybrids retained mtDNAs from both parents in similar ratios with a slight bias. The lost of the "somatic" mitochondria by Es-splenocyte hybrids implies non-random segregation of the parental mitochondria as supported by a computer simulation of genetic drift. In contrast, ES cell-fibroblast hybrids show bilateral random segregation of the parental mitochondria judging from analysis of mtDNA in single cells. Preferential segregation of "somatic" mitochondria does not depend on the differences in sequences of the parental mtDNAs but depends on replicative state of the parental cells.
Collapse
|
32
|
Dai B, Rasmussen TP. Global Epiproteomic Signatures Distinguish Embryonic Stem Cells from Differentiated Cells. Stem Cells 2007; 25:2567-74. [PMID: 17641388 DOI: 10.1634/stemcells.2007-0131] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Complex organisms contain a variety of distinct cell types but only a single genome. Therefore, cellular identity must be specified by the developmentally regulated expression of a subset of genes from an otherwise static genome. In mammals, genomic DNA is modified by cytosine methylation, resulting in a pattern that is distinctive for each cell type (the epigenome). Because nucleosomal histones are subject to a wide variety of post-translational modifications (PTMs), we reasoned that an analogous "epiproteome" might exist that could also be correlated with cellular identity. Here, we show that the quantitative evaluation of nucleosome PTMs yields epiproteomic signatures that are useful for the investigation of stem cell differentiation, chromatin function, cellular identity, and epigenetic responses to pharmacologic agents. We have developed a novel enzyme-linked immunosorbent assay-based method for the quantitative evaluation of the steady-state levels of PTMs and histone variants in preparations of native intact nucleosomes. We show that epiproteomic responses to the histone deacetylase inhibitor trichostatin A trigger changes in histone methylation as well as acetylation, and that the epiproteomic responses differ between mouse embryonic stem cells and mouse embryonic fibroblasts (MEFs). ESCs subjected to retinoic acid-induced differentiation contain reconfigured nucleosomes that include increased content of the histone variant macroH2A and other changes. Furthermore, ESCs can be distinguished from embryonal carcinoma cells and MEFs based purely on their epiproteomic signatures. These results indicate that epiproteomic nucleosomal signatures are useful for the investigation of stem cell identity and differentiation, nuclear reprogramming, epigenetic regulation, chromatin dynamics, and assays for compounds with epigenetic activities. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Bo Dai
- Center for Regenerative Biology, University of Connecticut, Storrs, Connecticut 06269-4243, USA
| | | |
Collapse
|
33
|
Islam MQ, Islam K, Sharp CA. Epigenetic reprogramming of nonreplicating somatic cells for long-term proliferation by temporary cell-cell contact. Stem Cells Dev 2007; 16:253-68. [PMID: 17521237 DOI: 10.1089/scd.2006.0094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Embryonic stem (ES) cells are potential sources of tissue regeneration; however, transplanted ES cells produce tumors in the host tissues. In addition, transplantation between genetically unrelated individuals often results in graft rejection. Although the development of patient specific stem cell lines by somatic cell nuclear transfer (SCNT) represents a means of overcoming the problem of rejection, its human application has ethical dilemmas. Adult stem (AS) cells can also differentiate into specialized cells and may provide an alternative source of cells for human applications. In common with other somatic cells, AS cells have limited capacity for proliferation and cannot be produced in large quantities without genetic manipulation. We demonstrate here that nonreplicating mammalian cells can be reprogrammed for long-term proliferation by temporary cell-cell contact through coculture of AS cells with the GM05267-derived F7 mouse cell line. Subsequent elimination of F7 cells from the co-culture allows proliferation of previously nonreplicating cells, colonies of which can be isolated to produce cell lines. We also demonstrate that the epigenetically reprogrammed AS cells, without the physical transfer of either nuclear or cytoplasmic material from other cells, are capable of long-term proliferation and able to relay signals to other nonreplicating cells to reinitiate proliferation with no addition of recombinant factors. The reported cell amplification procedure is methodologically simple and can be easily reproduced. This procedure allows the production of an unlimited number of cells from a limited number of AS cells, making them an ideal source of cells for applications involving autologous cell transplantation.
Collapse
Affiliation(s)
- M Q Islam
- Laboratory of Cancer Genetics, Laboratory Medicine Center (LMC), University Hospital Linköping, 58216 Linköping, Sweden.
| | | | | |
Collapse
|
34
|
Puzakov MV, Battulin NR, Temirova SA, Matveeva NM, Serdyukova NA, Grafodatsky AS, Serov OL. Analysis of expression of parental alleles Xist and Gla in interspecific embryonic hybrid cells during induced in vitro inactivation of X-chromosomes. Russ J Dev Biol 2007. [DOI: 10.1134/s1062360407030046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
35
|
Islam MQ, Panduri V, Islam K. Generation of somatic cell hybrids for the production of biologically active factors that stimulate proliferation of other cells. Cell Prolif 2007; 40:91-105. [PMID: 17227298 PMCID: PMC6496579 DOI: 10.1111/j.1365-2184.2007.00422.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Some normal somatic cells in culture divide a limited number of times before entering a non-dividing state called replicative senescence and fusion of normal cells with immortal cells claimed to produce hybrid cells of limited proliferation. We reinvestigated the proliferative capacity of hybrid cells between normal cell and immortal cell. MATERIALS AND METHODS Normal pig fibroblast cells and cells of immortal mouse fibroblast cell line F7, a derivative of GM05267, were fused by polyethylene glycol treatment and subsequently the fused cells were cultured in a selective medium containing hypoxanthine-aminopterin-thymidine in order to enrich the hybrid cells. The hybrid cells were then monitored for chromosome content and proliferation. RESULTS Cytogenetic analysis revealed that the hybrid cells contained polyploidy chromosomes derived from normal pig fibroblasts. These hybrid cells exhibit no sign of replicative senescence after more than 190 population doublings in vitro. Instead, these hybrid cells have an accelerated growth and proliferate even in the complete absence of glutamine. In addition, these hybrids produce biologically active factors in the conditioned media, which not only can accelerate their own proliferation but also can reinitiate mitotic activity in the senescent-like normal fibroblast cells. CONCLUSIONS Our results question the validity of cellular senescence as a dominant trait. Additionally, the generation of hybrid cells using the specific mouse cell line can be applied to the generation of hybrids with other normal cell types and can be used to produce tissue-specific growth-factor(s) to extend the lifespan and/or improve the proliferation of various normal cells, including adult stem cells.
Collapse
Affiliation(s)
- M Q Islam
- Laboratory of Cancer Genetics, Laboratory Medicine Center (LMC), University Hospital Linköping, Linköping, Sweden.
| | | | | |
Collapse
|
36
|
Ambrosi DJ, Tanasijevic B, Kaur A, Obergfell C, O'Neill RJ, Krueger W, Rasmussen TP. Genome-wide reprogramming in hybrids of somatic cells and embryonic stem cells. Stem Cells 2007; 25:1104-13. [PMID: 17272499 DOI: 10.1634/stemcells.2006-0532] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Recent experiments demonstrate that somatic nuclei can be reprogrammed to a pluripotent state when fused to ESCs. The resulting hybrids are pluripotent as judged by developmental assays, but detailed analyses of the underlying molecular-genetic control of reprogrammed transcription in such hybrids are required to better understand fusion-mediated reprogramming. We produced hybrids of mouse ESCs and fibroblasts that, although nearly tetraploid, exhibit characteristics of normal ESCs, including apparent immortality in culture, ESC-like colony morphology, and pluripotency. Comprehensive analysis of the mouse embryonic fibroblast/ESC hybrid transcriptome revealed global patterns of gene expression reminiscent of ESCs. However, combined analysis of variance and hierarchical clustering analyses revealed at least seven distinct classes of differentially regulated genes in comparisons of hybrids, ESCs, and somatic cells. The largest class includes somatic genes that are silenced in hybrids and ESCs, but a smaller class includes genes that are expressed at nearly equivalent levels in hybrids and ESCs that contain many genes implicated in pluripotency and chromatin function. Reprogrammed genes are distributed throughout the genome. Reprogramming events include both transcriptional silencing and activation of genes residing on chromosomes of somatic origin. Somatic/ESC hybrid cell lines resemble their pre-fusion ESC partners in terms of behavior in culture and pluripotency. However, they contain unique expression profiles that are similar but not identical to normal ESCs. ESC fusion-mediated reprogramming provides a tractable system for the investigation of mechanisms of reprogramming. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Dominic J Ambrosi
- Center for Regenerative Biology, 1392 Storrs Road, U-4243 University of Connecticut, Storrs, Connecticut 06269-4243, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
French AJ, Wood SH, Trounson AO. Human therapeutic cloning (NTSC). ACTA ACUST UNITED AC 2006; 2:265-76. [PMID: 17848713 DOI: 10.1007/bf02698053] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/15/2022]
Abstract
Human therapeutic cloning or nuclear transfer stem cells (NTSC) to produce patient-specific stem cells, holds considerable promise in the field of regenerative medicine. The recent withdrawal of the only scientific publications claiming the successful generation of NTSC lines afford an opportunity to review the available research in mammalian reproductive somatic cell nuclear transfer (SCNT) with the goal of progressing human NTSC. The process of SCNT is prone to epigenetic abnormalities that contribute to very low success rates. Although there are high mortality rates in some species of cloned animals, most surviving clones have been shown to have normal phenotypic and physiological characteristics and to produce healthy offspring. This technology has been applied to an increasing number of mammals for utility in research, agriculture, conservation, and biomedicine. In contrast, attempts at SCNT to produce human embryonic stem cells (hESCs) have been disappointing. Only one group has published reliable evidence of success in deriving a cloned human blastocyst, using an undifferentiated hESC donor cell, and it failed to develop into a hESC line. When optimal conditions are present, it appears that in vitro development of cloned and parthenogenetic embryos, both of which may be utilized to produce hESCs, may be similar to in vitro fertilized embryos. The derivation of ESC lines from cloned embryos is substantially more efficient than the production of viable offspring. This review summarizes developments in mammalian reproductive cloning, cell-to-cell fusion alternatives, and strategies for oocyte procurement that may provide important clues facilitating progress in human therapeutic cloning leading to the successful application of cell-based therapies utilizing autologous hESC lines.
Collapse
|
38
|
Islam MQ, Ringe J, Reichmann E, Migotti R, Sittinger M, da S Meirelles L, Nardi NB, Magnusson P, Islam K. Functional characterization of cell hybrids generated by induced fusion of primary porcine mesenchymal stem cells with an immortal murine cell line. Cell Tissue Res 2006; 326:123-37. [PMID: 16741712 DOI: 10.1007/s00441-006-0224-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Accepted: 04/11/2006] [Indexed: 12/22/2022]
Abstract
Bone marrow mesenchymal stem cells (MSC) integrate into various organs and contribute to the regeneration of diverse tissues. However, the mechanistic basis of the plasticity of MSC is not fully understood. The change of cell fate has been suggested to occur through cell fusion. We have generated hybrid cell lines by polyethylene-glycol-mediated cell fusion of primary porcine MSC with the immortal murine fibroblast cell line F7, a derivative of the GM05267 cell line. The hybrid cell lines display fibroblastic morphology and proliferate like immortal cells. They contain tetraploid to hexaploid porcine chromosomes accompanied by hypo-diploid murine chromosomes. Interestingly, many hybrid cell lines also express high levels of tissue-nonspecific alkaline phosphatase, which is considered to be a marker of undifferentiated embryonic stem cells. All tested hybrid cell lines retain osteogenic differentiation, a few of them also retain adipogenic potential, but none retain chondrogenic differentiation. Conditioned media from hybrid cells enhance the proliferation of both early-passage and late-passage porcine MSC, indicating that the hybrid cells secrete diffusible growth stimulatory factors. Murine F7 cells thus have the unique property of generating immortal cell hybrids containing unusually high numbers of chromosomes derived from normal cells. These hybrid cells can be employed in various studies to improve our understanding of regenerative biology. This is the first report, to our knowledge, describing the generation of experimentally induced cell hybrids by using normal primary MSC.
Collapse
Affiliation(s)
- M Q Islam
- Laboratory of Cancer Genetics, University Hospital Linkoping, SE-581 85, Linkoping, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Human embryonic stem cells (hESCs) are being rapidly produced from chromosomally euploid, aneuploid, and mutant human embryos that are available from in vitro fertilization clinics treating patients for infertility or preimplantation genetic diagnosis. These hESC lines are an important resource for functional genomics, drug screening, and, perhaps eventually, cell and gene therapy. The methods for deriving hESCs are well established and repeatable and are relatively successful with a ratio of 1:10 to 1:2 new hESC lines produced from 4- to 8-d-old morula and blastocysts and from isolated inner cell mass cell clusters of human blastocysts. The hESCs can be formed and maintained on human somatic cells in humanized serum-free culture conditions and for several passages in cell-free culture systems. The hESCs can be transfected with DNA constructs. Their gene expression profiles are being described and immunological characteristics determined. They may be grown indefinitely in vitro while maintaining their original karyotype and epigenetic status, but this needs to be confirmed from time to time in long-term cultures. hESCs spontaneously differentiate in the absence of the appropriate cell feeder layer, when overgrown in culture and when isolated from the ESC colony. All three major embryonic lineages are produced in differentiating flat attachment cultures and unattached embryoid bodies. Cell progenitors of interest can be identified by markers, expression of reporter genes, and characteristic morphology, and the cells thereafter enriched for progenitor types and further culture to more mature cell types. Directed differentiation systems are well developed for ectodermal pathways that result in neural and glial cells and the mesendodermal pathway for cardiac muscle cells and many other cell types including hematopoietic progenitors and endothelial cells. Directed differentiation into endoderm has been more difficult to achieve, perhaps because of the lack of markers of early progenitors in this lineage. There are reports of enriched cultures of keratinocytes, pigmented retinal epithelium, neural crest cells and motor neurons, hepatic progenitors, and cells that have some markers of gut tissue and pancreatic islet-like cells. The prospects for use of hESC derivatives in regenerative medicine are significant, and there is much optimism for their potential contributions to human regenerative medicine.
Collapse
Affiliation(s)
- Alan Trounson
- Monash Immunology and Stem Cell Laboratories, Monash University, and Australian Stem Cell Centre, Wellington Road, Clayton, Victoria 3800, Australia.
| |
Collapse
|
40
|
Hall VJ, Stojkovic P, Stojkovic M. Using therapeutic cloning to fight human disease: a conundrum or reality? Stem Cells 2006; 24:1628-37. [PMID: 16556706 DOI: 10.1634/stemcells.2005-0592] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The development and transplantation of autologous cells derived from nuclear transfer embryonic stem cell (NT-ESC) lines to treat patients suffering from disease has been termed therapeutic cloning. Human NT is still a developing field, with further research required to improve somatic cell NT and human embryonic stem cell differentiation to deliver safe and effective cell replacement therapies. Furthermore, the implications of transferring mitochondrial heteroplasmic cells, which may harbor aberrant epigenetic gene expression profiles, are of concern. The production of human NT-ESC lines also remains plagued by ethical dilemmas, societal concerns, and controversies. Recently, a number of alternate therapeutic strategies have been proposed to circumvent the moral implications surrounding human nuclear transfer. It will be critical to overcome these biological, legislative, and moral restraints to maximize the potential of this therapeutic strategy and to alleviate human disease.
Collapse
Affiliation(s)
- Vanessa J Hall
- Neuronal Survival Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Centre, Lund University, Sweden.
| | | | | |
Collapse
|