1
|
Miliotou E, de Lázaro I. A Youthful Touch: Reversal of Aging Hallmarks by Cell Reprogramming. Cells Tissues Organs 2024; 213:538-550. [PMID: 38768583 PMCID: PMC11633886 DOI: 10.1159/000539415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 05/16/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND With the elderly population projected to double by 2050, there is an urgent need to address the increasing prevalence of age-related debilitating diseases and ultimately minimize discrepancies between the rising lifespan and stagnant health span. Cellular reprogramming by overexpression of Oct3/4, Klf4, Sox2, and cMyc (OKSM) transcription factors is gaining attention in this context thanks to demonstrated rejuvenating effects in human cell cultures and live mice, many of which can be uncoupled from dedifferentiation and loss of cell identity. SUMMARY Here, we review current evidence of the impact of cell reprogramming on established aging hallmarks and the underlying mechanisms that mediate these effects. We also provide a critical assessment of the challenges in translating these findings and, overall, cell reprogramming technologies into clinically translatable antiaging interventions. KEY MESSAGES Cellular reprogramming has the potential to reverse at least partially some key hallmarks of aging. However, further research is necessary to determine the biological significance and duration of such changes and to ensure the safety of cell reprogramming as a rejuvenation approach. With this review, we hope to stimulate new research directions in the quest to extend health span effectively.
Collapse
Affiliation(s)
- Eleni Miliotou
- Department of Biomedical Engineering, NYU Tandon School of Engineering, New York University, New York, NY, USA
- Cardiovascular Research Center, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Irene de Lázaro
- Department of Biomedical Engineering, NYU Tandon School of Engineering, New York University, New York, NY, USA
- Cardiovascular Research Center, Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
- Harvard John A. Paulson School of Engineering, Harvard University, Cambridge, MA, USA
| |
Collapse
|
2
|
Bekas N, Samiotaki M, Papathanasiou M, Mokos P, Pseftogas A, Xanthopoulos K, Thanos D, Mosialos G, Dafou D. Inactivation of Tumor Suppressor CYLD Inhibits Fibroblast Reprogramming to Pluripotency. Cancers (Basel) 2023; 15:4997. [PMID: 37894364 PMCID: PMC10605754 DOI: 10.3390/cancers15204997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
CYLD is a tumor suppressor gene coding for a deubiquitinating enzyme that has a critical regulatory function in a variety of signaling pathways and biological processes involved in cancer development and progression, many of which are also key modulators of somatic cell reprogramming. Nevertheless, the potential role of CYLD in this process has not been studied. With the dual aim of investigating the involvement of CYLD in reprogramming and developing a better understanding of the intricate regulatory system governing this process, we reprogrammed control (CYLDWT/WT) and CYLD DUB-deficient (CYLDΔ9/Δ9) mouse embryonic fibroblasts (MEFs) into induced pluripotent stem cells (iPSCs) through ectopic overexpression of the Yamanaka factors (Oct3/4, Sox2, Klf4, c-myc). CYLD DUB deficiency led to significantly reduced reprogramming efficiency and slower early reprogramming kinetics. The introduction of WT CYLD to CYLDΔ9/Δ9 MEFs rescued the phenotype. Nevertheless, CYLD DUB-deficient cells were capable of establishing induced pluripotent colonies with full spontaneous differentiation potential of the three germ layers. Whole proteome analysis (Data are available via ProteomeXchange with identifier PXD044220) revealed that the mesenchymal-to-epithelial transition (MET) during the early reprogramming stages was disrupted in CYLDΔ9/Δ9 MEFs. Interestingly, differentially enriched pathways revealed that the primary processes affected by CYLD DUB deficiency were associated with the organization of the extracellular matrix and several metabolic pathways. Our findings not only establish for the first time CYLD's significance as a regulatory component of early reprogramming but also highlight its role as an extracellular matrix regulator, which has profound implications in cancer research.
Collapse
Affiliation(s)
- Nikolaos Bekas
- School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (N.B.); (P.M.); (G.M.)
| | - Martina Samiotaki
- Biomedical Sciences Research Center “Alexander Fleming”, 16672 Vari, Greece;
| | - Maria Papathanasiou
- Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece; (M.P.); (D.T.)
| | - Panagiotis Mokos
- School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (N.B.); (P.M.); (G.M.)
| | - Athanasios Pseftogas
- Division of Experimental Oncology, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy;
| | - Konstantinos Xanthopoulos
- Laboratory of Pharmacology, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Dimitris Thanos
- Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece; (M.P.); (D.T.)
| | - George Mosialos
- School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (N.B.); (P.M.); (G.M.)
| | - Dimitra Dafou
- School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (N.B.); (P.M.); (G.M.)
| |
Collapse
|
3
|
Hu P, Leyton L, Hagood JS, Barker TH. Thy-1-Integrin Interactions in cis and Trans Mediate Distinctive Signaling. Front Cell Dev Biol 2022; 10:928510. [PMID: 35733855 PMCID: PMC9208718 DOI: 10.3389/fcell.2022.928510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
Thy-1 is a cell surface glycosylphosphatidylinositol (GPI)-anchored glycoprotein that bears a broad mosaic of biological roles across various cell types. Thy-1 displays strong physiological and pathological implications in development, cancer, immunity, and tissue fibrosis. Quite uniquely, Thy-1 is capable of mediating integrin-related signaling through direct trans- and cis-interaction with integrins. Both interaction types have shown distinctive roles, even when interacting with the same type of integrin, where binding in trans or in cis often yields divergent signaling events. In this review, we will revisit recent progress and discoveries of Thy-1–integrin interactions in trans and in cis, highlight their pathophysiological consequences and explore other potential binding partners of Thy-1 within the integrin regulation/signaling paradigm.
Collapse
Affiliation(s)
- Ping Hu
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile and Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - James S. Hagood
- Department of Pediatrics, Division of Pulmonology, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
- Program for Rare and Interstitial Lung Disease, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Thomas H. Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States
- *Correspondence: Thomas H. Barker,
| |
Collapse
|
4
|
CCN proteins in the musculoskeletal system: current understanding and challenges in physiology and pathology. J Cell Commun Signal 2021; 15:545-566. [PMID: 34228239 PMCID: PMC8642527 DOI: 10.1007/s12079-021-00631-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
The acronym for the CCN family was recently revised to represent “cellular communication network”. These six, small, cysteine-enriched and evolutionarily conserved proteins are secreted matricellular proteins, that convey and modulate intercellular communication by interacting with structural proteins, signalling factors and cell surface receptors. Their role in the development and physiology of musculoskeletal system, constituted by connective tissues where cells are interspersed in the cellular matrix, has been broadly studied. Previous research has highlighted a crucial balance of CCN proteins in mesenchymal stem cell commitment and a pivotal role for CCN1, CCN2 and their alter ego CCN3 in chondrogenesis and osteogenesis; CCN4 plays a minor role and the role of CCN5 and CCN6 is still unclear. CCN proteins also participate in osteoclastogenesis and myogenesis. In adult life, CCN proteins serve as mechanosensory proteins in the musculoskeletal system providing a steady response to environmental stimuli and participating in fracture healing. Substantial evidence also supports the involvement of CCN proteins in inflammatory pathologies, such as osteoarthritis and rheumatoid arthritis, as well as in cancers affecting the musculoskeletal system and bone metastasis. These matricellular proteins indeed show involvement in inflammation and cancer, thus representing intriguing therapeutic targets. This review discusses the current understanding of CCN proteins in the musculoskeletal system as well as the controversies and challenges associated with their multiple and complex roles, and it aims to link the dispersed knowledge in an effort to stimulate and guide readers to an area that the writers consider to have significant impact and relevant potentialities.
Collapse
|
5
|
Crawford L, Wyatt M, Bryers J, Ratner B. Biocompatibility Evolves: Phenomenology to Toxicology to Regeneration. Adv Healthc Mater 2021; 10:e2002153. [PMID: 33829678 PMCID: PMC8221530 DOI: 10.1002/adhm.202002153] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/26/2021] [Indexed: 12/20/2022]
Abstract
The word "biocompatibility," is inconsistent with the observations of healing for so-called biocompatible biomaterials. The vast majority of the millions of medical implants in humans today, presumably "biocompatible," are walled off by a dense, avascular, crosslinked collagen capsule, hardly suggestive of life or compatibility. In contrast, one is now seeing examples of implant biomaterials that lead to a vascularized reconstruction of localized tissue, a biological reaction different from traditional biocompatible materials that generate a foreign body capsule. Both the encapsulated biomaterials and the reconstructive biomaterials qualify as "biocompatible" by present day measurements of biocompatibility. Yet, this new generation of materials would seem to heal "compatibly" with the living organism, where older biomaterials are isolated from the living organism by the dense capsule. This review/perspective article will explore this biocompatibility etymological conundrum by reviewing the history of the concepts around biocompatibility, today's standard methods for assessing biocompatibility, a contemporary view of the foreign body reaction and finally, a compendium of new biomaterials that heal without the foreign body capsule. A new definition of biocompatibility is offered here to address advances in biomaterials design leading to biomaterials that heal into the body in a facile manner.
Collapse
Affiliation(s)
- Lars Crawford
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Meghan Wyatt
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - James Bryers
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Buddy Ratner
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
6
|
Maali A, Maroufi F, Sadeghi F, Atashi A, Kouchaki R, Moghadami M, Azad M. Induced pluripotent stem cell technology: trends in molecular biology, from genetics to epigenetics. Epigenomics 2021; 13:631-647. [PMID: 33823614 DOI: 10.2217/epi-2020-0409] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology, based on autologous cells' reprogramming to the embryonic state, is a new approach in regenerative medicine. Current advances in iPSC technology have opened up new avenues for multiple applications, from basic research to clinical therapy. Thus, conducting iPSC trials have attracted increasing attention and requires an extensive understanding of the molecular basis of iPSCs. Since iPSC reprogramming is based on the methods inducing the expression of specific genes involved in pluripotency states, it can be concluded that iPSC reprogramming is strongly influenced by epigenetics. In this study, we reviewed the molecular basis of reprogramming, including the reprogramming factors (OCT4, SOX2, KLF4, c-MYC, NANOG, ESRRB, LIN28 as well as their regulatory networks), applied vectors (retroviral vectors, adenoviral vectors, Sendaiviral vectors, episomal plasmids, piggyBac, simple vectors, etc.) and epigenetic modifications (miRNAs, histones and DNA methylation states) to provide a comprehensive guide for reprogramming studies.
Collapse
Affiliation(s)
- Amirhosein Maali
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran.,Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Faezeh Maroufi
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farzin Sadeghi
- Cellular & Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Amir Atashi
- Stem Cells & Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Kouchaki
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mona Moghadami
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
7
|
Abstract
Derivation of induced Pluripotent Stem Cells (iPSCs) by reprogramming somatic cells to a pluripotent state has revolutionized stem cell research. Ensuing this, various groups have used genetic and non-genetic approaches to generate iPSCs from numerous cell types. However, achieving a pluripotent state in most of the reprogramming studies is marred by serious limitations such as low reprogramming efficiency and slow kinetics. These limitations are mainly due to the presence of potent barriers that exist during reprogramming when a mature cell is coaxed to achieve a pluripotent state. Several studies have revealed that intrinsic factors such as non-optimal stoichiometry of reprogramming factors, specific signaling pathways, cellular senescence, pluripotency-inhibiting transcription factors and microRNAs act as a roadblock. In addition, the epigenetic state of somatic cells and specific epigenetic modifications that occur during reprogramming also remarkably impede the generation of iPSCs. In this review, we present a comprehensive overview of the barriers that inhibit reprogramming and the understanding of which will pave the way to develop safe strategies for efficient reprogramming.
Collapse
|
8
|
Zhang ZP, Zhang JT, Huang SC, He XY, Deng LX. Double sperm cloning (DSC) is a promising strategy in mammalian genetic engineering and stem cell research. Stem Cell Res Ther 2020; 11:388. [PMID: 32894201 PMCID: PMC7487873 DOI: 10.1186/s13287-020-01907-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/12/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
Embryonic stem cells (ESCs) derived from somatic cell nuclear transfer (SCNT) and induced pluripotent stem cells (iPSCs) are promising tools for meeting the personalized requirements of regenerative medicine. However, some obstacles need to be overcome before clinical trials can be undertaken. First, donor cells vary, and the reprogramming procedures are diverse, so standardization is a great obstacle regarding SCNT and iPSCs. Second, somatic cells derived from a patient may carry mitochondrial DNA mutations and exhibit telomere instability with aging or disease, and SCNT-ESCs and iPSCs retain the epigenetic memory or epigenetic modification errors. Third, reprogramming efficiency has remained low. Therefore, in addition to improving their success rate, other alternatives for producing ESCs should be explored. Producing androgenetic diploid embryos could be an outstanding strategy; androgenic diploid embryos are produced through double sperm cloning (DSC), in which two capacitated sperms (XY or XX, sorted by flow cytometer) are injected into a denucleated oocyte by intracytoplasmic sperm injection (ICSI) to reconstruct embryo and derive DSC-ESCs. This process could avoid some potential issues, such as mitochondrial interference, telomere shortening, and somatic epigenetic memory, all of which accompany somatic donor cells. Oocytes are naturally activated by sperm, which is unlike the artificial activation that occurs in SCNT. The procedure is simple and practical and can be easily standardized. In addition, DSC-ESCs can overcome ethical concerns and resolve immunological response matching with sperm providers. Certainly, some challenges must be faced regarding imprinted genes, epigenetics, X chromosome inactivation, and dosage compensation. In mice, DSC-ESCs have been produced and have shown excellent differentiation ability. Therefore, the many advantages of DSC make the study of this process worthwhile for regenerative medicine and animal breeding.
Collapse
Affiliation(s)
- Zhi-Ping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Jun-Tao Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Shu-Cheng Huang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Xiu-Yuan He
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Li-Xin Deng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
9
|
Borgohain MP, Haridhasapavalan KK, Dey C, Adhikari P, Thummer RP. An Insight into DNA-free Reprogramming Approaches to Generate Integration-free Induced Pluripotent Stem Cells for Prospective Biomedical Applications. Stem Cell Rev Rep 2020; 15:286-313. [PMID: 30417242 DOI: 10.1007/s12015-018-9861-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
More than a decade ago, a pioneering study reported generation of induced Pluripotent Stem Cells (iPSCs) by ectopic expression of a cocktail of reprogramming factors in fibroblasts. This study has revolutionized stem cell research and has garnered immense interest from the scientific community globally. iPSCs hold tremendous potential for understanding human developmental biology, disease modeling, drug screening and discovery, and personalized cell-based therapeutic applications. The seminal study identified Oct4, Sox2, Klf4 and c-Myc as a potent combination of genes to induce reprogramming. Subsequently, various reprogramming factors were identified by numerous groups. Most of these studies have used integrating viral vectors to overexpress reprogramming factors in somatic cells to derive iPSCs. However, these techniques restrict the clinical applicability of these cells as they may alter the genome due to random viral integration resulting in insertional mutagenesis and tumorigenicity. To circumvent this issue, alternative integration-free reprogramming approaches are continuously developed that eliminate the risk of genomic modifications and improve the prospects of iPSCs from lab to clinic. These methods establish that integration of transgenes into the genome is not essential to induce pluripotency in somatic cells. This review provides a comprehensive overview of the most promising DNA-free reprogramming techniques that have the potential to derive integration-free iPSCs without genomic manipulation, such as sendai virus, recombinant proteins, microRNAs, synthetic messenger RNA and small molecules. The understanding of these approaches shall pave a way for the generation of clinical-grade iPSCs. Subsequently, these iPSCs can be differentiated into desired cell type(s) for various biomedical applications.
Collapse
Affiliation(s)
- Manash P Borgohain
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Krishna Kumar Haridhasapavalan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Chandrima Dey
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Poulomi Adhikari
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
10
|
Chen SN, Chang R, Lin LT, Chern CU, Tsai HW, Wen ZH, Li YH, Li CJ, Tsui KH. MicroRNA in Ovarian Cancer: Biology, Pathogenesis, and Therapeutic Opportunities. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:1510. [PMID: 31035447 PMCID: PMC6539609 DOI: 10.3390/ijerph16091510] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/08/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022]
Abstract
Ovarian cancer comprises one of the three major malignant tumor types in the female reproductive system. The mortality rate of this cancer is the highest among all gynecological tumors, with ovarian cancer metastasis constituting an important cause of death. Therefore, markers for disease prediction and prognosis are highly desirable for early diagnosis as well as for helping optimize and personalize treatment. Recently, microRNAs (miRNAs), which consist of short-sequence RNAs that do not encode a protein, have emerged as new biomarkers in the clinical diagnosis and treatment of ovarian cancer. By pairing with bases specific to the target messenger RNA (mRNA), miRNAs cause degradation of the target mRNA or inhibit its translation, thereby regulating various cellular processes including cell proliferation and adhesion. Increasing numbers of studies have shown that miRNA expression abnormality plays an important role in the development of ovarian cancer. In this review, we discuss the mechanisms of miRNA action, current research regarding their role in the suppression or promotion of ovarian cancer, and their use as markers for diagnosis of prognosis or as therapeutic targets for this disease. Finally, we present future perspectives regarding the clinical management of ovarian cancer and the role for miRNAs therein.
Collapse
Affiliation(s)
- San-Nung Chen
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Renin Chang
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
- Department of Recreation Sports Management, Tajen University, Pingtung 907, Taiwan.
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 840, Taiwan.
| | - Li-Te Lin
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei 112, Taiwan.
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
| | - Chyi-Uei Chern
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Hsiao-Wen Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
- Marine Biomedical Laboratory and Center for Translational Biopharmaceuticals, Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 813, Taiwan.
| | - Yi-Han Li
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung 907, Taiwan.
| |
Collapse
|
11
|
Hagood JS. Thy-1 as an Integrator of Diverse Extracellular Signals. Front Cell Dev Biol 2019; 7:26. [PMID: 30859102 PMCID: PMC6397883 DOI: 10.3389/fcell.2019.00026] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 02/13/2019] [Indexed: 01/11/2023] Open
Abstract
Thy-1 was discovered over 50 years ago, and in that time investigators from a broad variety of fields have described numerous and heterogeneous biological functions of Thy-1 in multiple contexts. As an outwardly facing cell surface molecule, it is well positioned to receive extracellular signals; previously reviewed studies have confirmed an important role in cell-cell and cell-matrix adhesion, cell migration, and regulation of outside-in signaling. More recent studies reviewed here expand the repertoire of Thy-1 effects on signaling pathways, and reveal novel roles in mechanotransduction, cellular differentiation, viral entry, and extracellular vesicle binding and internalization. All of these studies contribute to understanding Thy-1 as a context-dependent integrator of a diverse range of extracellular information, and provide impetus for further studies, some of which are suggested here.
Collapse
Affiliation(s)
- James S. Hagood
- Division of Pulmonology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
12
|
Gomes KMS, Costa IC, Santos JFD, Dourado PMM, Forni MF, Ferreira JCB. Induced pluripotent stem cells reprogramming: Epigenetics and applications in the regenerative medicine. Rev Assoc Med Bras (1992) 2017; 63:180-189. [PMID: 28355380 DOI: 10.1590/1806-9282.63.02.180] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 05/30/2016] [Indexed: 12/20/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are somatic cells reprogrammed into an embryonic-like pluripotent state by the expression of specific transcription factors. iPSC technology is expected to revolutionize regenerative medicine in the near future. Despite the fact that these cells have the capacity to self-renew, they present low efficiency of reprogramming. Recent studies have demonstrated that the previous somatic epigenetic signature is a limiting factor in iPSC performance. Indeed, the process of effective reprogramming involves a complete remodeling of the existing somatic epigenetic memory, followed by the establishment of a "new epigenetic signature" that complies with the new type of cell to be differentiated. Therefore, further investigations of epigenetic modifications associated with iPSC reprogramming are required in an attempt to improve their self-renew capacity and potency, as well as their application in regenerative medicine, with a new strategy to reduce the damage in degenerative diseases. Our review aimed to summarize the most recent findings on epigenetics and iPSC, focusing on DNA methylation, histone modifications and microRNAs, highlighting their potential in translating cell therapy into clinics.
Collapse
Affiliation(s)
- Kátia Maria Sampaio Gomes
- Department of Anatomy, Institute of Biomedical Sciences III, Universidade de São Paulo (ICB III/USP), São Paulo, SP, Brazil
| | - Ismael Cabral Costa
- Department of Anatomy, Institute of Biomedical Sciences III, Universidade de São Paulo (ICB III/USP), São Paulo, SP, Brazil
| | | | | | | | - Julio Cesar Batista Ferreira
- Department of Anatomy, Institute of Biomedical Sciences III, Universidade de São Paulo (ICB III/USP), São Paulo, SP, Brazil
| |
Collapse
|
13
|
Luginbühl J, Sivaraman DM, Shin JW. The essentiality of non-coding RNAs in cell reprogramming. Noncoding RNA Res 2017; 2:74-82. [PMID: 30159423 PMCID: PMC6096403 DOI: 10.1016/j.ncrna.2017.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/03/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023] Open
Abstract
In mammals, short (mi-) and long non-coding (lnc) RNAs are immensely abundant and they are proving to be more functional than ever before. Particularly in cell reprogramming, non-coding RNAs are essential to establish the pluripotent network and are indispensable to reprogram somatic cells to pluripotency. Through systematic screening and mechanistic studies, diverse functional features of both miRNA and lncRNAs have emerged as either scaffolds, inhibitors, or co-activators, necessary to orchestrate the intricacy of gene regulation. Furthermore, the collective characterizations of both miRNA and lncRNA reveal their interdependency (e.g. sequestering the function of the other) to modulate cell reprogramming. This review broadly explores the regulatory processes of cell reprogramming - with key functional examples in neuronal and cardiac differentiations - in the context of both short and long non-coding RNAs.
Collapse
Affiliation(s)
| | | | - Jay W. Shin
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, 230-0045 Japan
| |
Collapse
|
14
|
TGF-β regulates TGFBIp expression in corneal fibroblasts via miR-21, miR-181a, and Smad signaling. Biochem Biophys Res Commun 2016; 472:150-5. [DOI: 10.1016/j.bbrc.2016.02.086] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/21/2016] [Indexed: 12/12/2022]
|
15
|
Abstract
Somatic reprogramming to generate induced pluripotent stem cells, or iPSC, is a powerful tool in developmental biology, disease modeling, and regenerative medicine. microRNAs have been shown to regulate many key pathways in iPSC induction. Here we describe a microRNA mimic enhanced somatic reprogramming process starting from mouse embryonic fibroblast isolation to iPSC induction to colony derivation and characterization.
Collapse
Affiliation(s)
- Jason Dang
- Department of Bioengineering, Department of Pediatrics, University of California San Diego School of Medicine, 9500 Gilman Drive MC 0762, La Jolla, CA, 92093, USA
| | - Tariq M Rana
- Department of Pediatrics, University of California San Diego School of Medicine, 9500 Gilman Drive MC 0762, La Jolla, CA, 92093, USA.
| |
Collapse
|
16
|
Function and significance of MicroRNAs in benign and malignant human stem cells. Semin Cancer Biol 2015; 35:200-11. [DOI: 10.1016/j.semcancer.2015.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/09/2015] [Accepted: 07/13/2015] [Indexed: 12/16/2022]
|
17
|
Effect of siRNA on Wisp-1 gene expression, proliferation, migration and adhesion of mouse hepatocellular carcinoma cells. ASIAN PAC J TROP MED 2015; 8:821-8. [DOI: 10.1016/j.apjtm.2015.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/20/2015] [Accepted: 09/15/2015] [Indexed: 11/20/2022] Open
|
18
|
Clausen MJAM, Melchers LJ, Mastik MF, Slagter-Menkema L, Groen HJM, van der Laan BFAM, van Criekinge W, de Meyer T, Denil S, Wisman GBA, Roodenburg JLN, Schuuring E. Identification and validation of WISP1 as an epigenetic regulator of metastasis in oral squamous cell carcinoma. Genes Chromosomes Cancer 2015; 55:45-59. [PMID: 26391330 DOI: 10.1002/gcc.22310] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 08/19/2015] [Indexed: 12/29/2022] Open
Abstract
Lymph node (LN) metastasis is the most important prognostic factor in oral squamous cell carcinoma (OSCC) patients. However, in approximately one third of OSCC patients nodal metastases remain undetected, and thus are not adequately treated. Therefore, clinical assessment of LN metastasis needs to be improved. The purpose of this study was to identify DNA methylation biomarkers to predict LN metastases in OSCC. Genome wide methylation assessment was performed on six OSCC with (N+) and six without LN metastases (N0). Differentially methylated sequences were selected based on the likelihood of differential methylation and validated using an independent OSCC cohort as well as OSCC from The Cancer Genome Atlas (TCGA). Expression of WISP1 using immunohistochemistry was analyzed on a large OSCC cohort (n = 204). MethylCap-Seq analysis revealed 268 differentially methylated markers. WISP1 was the highest ranking annotated gene that showed hypomethylation in the N+ group. Bisulfite pyrosequencing confirmed significant hypomethylation within the WISP1 promoter region in N+ OSCC (P = 0.03) and showed an association between WISP1 hypomethylation and high WISP1 expression (P = 0.01). Both these results were confirmed using 148 OSCC retrieved from the TCGA database. In a large OSCC cohort, high WISP1 expression was associated with LN metastasis (P = 0.05), disease-specific survival (P = 0.022), and regional disease-free survival (P = 0.027). These data suggest that WISP1 expression is regulated by methylation and WISP1 hypomethylation contributes to LN metastasis in OSCC. WISP1 is a potential biomarker to predict the presence of LN metastases.
Collapse
Affiliation(s)
- Martijn J A M Clausen
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lieuwe J Melchers
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mirjam F Mastik
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lorian Slagter-Menkema
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Otorhinolaryngology/Head & Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Harry J M Groen
- Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bernard F A M van der Laan
- Otorhinolaryngology/Head & Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wim van Criekinge
- Department of Mathematical Modelling Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Tim de Meyer
- Department of Mathematical Modelling Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Simon Denil
- Department of Mathematical Modelling Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - G Bea A Wisman
- Gynecologic Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan L N Roodenburg
- Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
19
|
González F, Huangfu D. Mechanisms underlying the formation of induced pluripotent stem cells. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 5:39-65. [PMID: 26383234 DOI: 10.1002/wdev.206] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/13/2015] [Accepted: 07/21/2015] [Indexed: 12/19/2022]
Abstract
Human pluripotent stem cells (hPSCs) offer unique opportunities for studying human biology, modeling diseases, and therapeutic applications. The simplest approach so far to generate human PSC lines is through reprogramming of somatic cells from an individual by defined factors, referred to simply as reprogramming. Reprogramming circumvents the ethical controversies associated with human embryonic stem cells (hESCs) and nuclear transfer hESCs (nt-hESCs), and the resulting induced pluripotent stem cells (hiPSCs) retain the same basic genetic makeup as the somatic cell used for reprogramming. Since the first report of iPSCs by Takahashi and Yamanaka (Cell 2006, 126:663-676), the molecular mechanisms of reprogramming have been extensively investigated. A better mechanistic understanding of reprogramming is fundamental not only to iPSC biology and improving the quality of iPSCs for therapeutic use, but also to our understanding of the molecular basis of cell identity, pluripotency, and plasticity. Here, we summarize the genetic, epigenetic, and cellular events during reprogramming, and the roles of various factors identified thus far in the reprogramming process. WIREs Dev Biol 2016, 5:39-65. doi: 10.1002/wdev.206 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Federico González
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY, USA
| | - Danwei Huangfu
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY, USA
| |
Collapse
|
20
|
Quesenberry PJ, Aliotta J, Deregibus MC, Camussi G. Role of extracellular RNA-carrying vesicles in cell differentiation and reprogramming. Stem Cell Res Ther 2015; 6:153. [PMID: 26334526 PMCID: PMC4558901 DOI: 10.1186/s13287-015-0150-x] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Growing evidence suggests that transcriptional regulators and secreted RNA molecules encapsulated within membrane vesicles modify the phenotype of target cells. Membrane vesicles, actively released by cells, represent a mechanism of intercellular communication that is conserved evolutionarily and involves the transfer of molecules able to induce epigenetic changes in recipient cells. Extracellular vesicles, which include exosomes and microvesicles, carry proteins, bioactive lipids, and nucleic acids, which are protected from enzyme degradation. These vesicles can transfer signals capable of altering cell function and/or reprogramming targeted cells. In the present review we focus on the extracellular vesicle-induced epigenetic changes in recipient cells that may lead to phenotypic and functional modifications. The relevance of these phenomena in stem cell biology and tissue repair is discussed.
Collapse
Affiliation(s)
- Peter J Quesenberry
- Department of Medicine, Warren Alpert Medical School of Brown University, Box G-A1, Providence, RI, 02912, USA.
| | - Jason Aliotta
- Department of Medicine, Warren Alpert Medical School of Brown University, Box G-A1, Providence, RI, 02912, USA
| | - Maria Chiara Deregibus
- Translational Center for Regenerative Medicine, University of Torino/Fresenius Medical Care, via Nizza 52, 10126, Torino, Italy
- Department of Medical Sciences, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy.
| |
Collapse
|
21
|
Gori M, Trombetta M, Santini D, Rainer A. Tissue engineering and microRNAs: future perspectives in regenerative medicine. Expert Opin Biol Ther 2015. [DOI: 10.1517/14712598.2015.1071349] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
22
|
He Y, Wang J, Wang J, Yung VYW, Hsu E, Li A, Kang Q, Ma J, Han Q, Jin P, Xing R, Lu Y, Sheng J. MicroRNA-135b regulates apoptosis and chemoresistance in colorectal cancer by targeting large tumor suppressor kinase 2. Am J Cancer Res 2015; 5:1382-1395. [PMID: 26101704 PMCID: PMC4473317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/12/2015] [Indexed: 06/04/2023] Open
Abstract
Colorectal cancer remains the third most common cause of death from cancer worldwide. MicroRNA emerges as a good area of research for current cancer therapy. Here, we identified miR-135b to be a contributor to anti-apoptosis and chemoresistance in colorectal cancer. We observed high levels of miR-135b in colorectal cancer cell lines and clinical tissues, compared to colorectal epithelium cell line and noncancerous tissues. Furthermore, enforced expression of miR-135b attenuated doxorubicin-induced apoptosis in colorectal cells. (Doxorubicin alone can trigger significant apoptosis). In elucidating the molecular mechanism by which miR-135b participate in the regulation of apoptosis and chemoresistance in colorectal cancer, we discovered that large tumor suppressor kinase 2 (LATS2) is a direct target of miR-135b. The role of miR-135b was confirmed in colorectal tumor xenograft models. The growth of established tumors was suppressed by an inhibition of miR-135b expression and enhanced apoptosis was further assessed by TUNEL assay. Taken together, our results reveal that miR-135b and LATS2 axis may be a novel therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Yuqi He
- Department of Gastroenterology, Beijing Military General HospitalBeijing 100700, China
| | - Jianxun Wang
- Institute for Translational Medicine, College of Medicine, Qingdao UniversityQingdao 266021, China
| | - Jiheng Wang
- Department of Gastroenterology, Beijing Military General HospitalBeijing 100700, China
| | - Victoria Yee-Wa Yung
- Department of Gastroenterology & Hepatology, University of California Davis, UC Davis Medical CenterSacramento, CA 95817
| | - Emily Hsu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los AngelesLos Angeles, CA 90095
| | - Aiqin Li
- Department of Gastroenterology, Beijing Military General HospitalBeijing 100700, China
| | - Qian Kang
- Department of Gastroenterology, Beijing Military General HospitalBeijing 100700, China
| | - Junbiao Ma
- Department of Coloproctology, The People’s Hospital of Renqiu CityRenqiu 062550, China
| | - Qingfeng Han
- Department of Coloproctology, The People’s Hospital of Renqiu CityRenqiu 062550, China
| | - Peng Jin
- Department of Gastroenterology, Beijing Military General HospitalBeijing 100700, China
| | - Rui Xing
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & InstituteBeijing, 100142, China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & InstituteBeijing, 100142, China
| | - Jianqiu Sheng
- Department of Gastroenterology, Beijing Military General HospitalBeijing 100700, China
| |
Collapse
|