1
|
Shafiei G, Talaei SA, Enderami SE, Mahabady MK, Mahabadi JA. Pluripotent stem cell-derived gametes: A gap for infertility treatment and reproductive medicine in the future. Tissue Cell 2025; 95:102904. [PMID: 40203683 DOI: 10.1016/j.tice.2025.102904] [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: 09/30/2024] [Revised: 03/26/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
Infertility affects 10-15 % of reproductive-age couples worldwide, with male infertility linked to sperm dysfunction and female infertility caused by ovulation disorders and reproductive abnormalities. Stem cell research presents a promising avenue for infertility treatment through germ cell differentiation. However, standardizing differentiation protocols and ensuring the functionality of in vitro-derived gametes remain significant challenges before clinical application becomes feasible.
Collapse
Affiliation(s)
- Golnaz Shafiei
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Sayyed Alireza Talaei
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Ehsan Enderami
- Immunogenetics Research Center, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Amini Mahabadi
- Gametogenesis Research Center, Kashan University of Medical Science, Kashan, Iran.
| |
Collapse
|
2
|
Di A, Zhang X, Song L, Wang S, Liu X, Bai C, Su G, Li G, Yang L. Dppa2 Promotes Early Embryo Development Through Regulating PDH Expression Pattern During Zygotic Genome Activation. Int J Mol Sci 2025; 26:3436. [PMID: 40244397 PMCID: PMC11989748 DOI: 10.3390/ijms26073436] [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: 03/03/2025] [Revised: 03/29/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
During embryonic development, zygotic genome activation (ZGA) is a critical event that determines the rational process and the fate of embryonic cells. The tricarboxylic acid cycle (TCA cycle) provides necessary reactants and energy for biological activities such as genome activation, chromatin opening, and epigenetic modifications during ZGA. Recent studies have shown that during ZGA, core enzymes associated with TCA briefly enter the nucleus and participate in initiating the ZGA process. However, the regulatory relationship between ZGA factors, such as Dux, Dppa2, and Dppa4, and the core enzymes of the TCA cycle remains unknown. In this study, we found that Dppa2 plays a key role in ZGA by directly determining the localization of TCA core enzymes, thereby affecting the early embryonic development. To further investigate the effect of Dppa2 on the localization of pyruvate dehydrogenase (PDH), we followed the establishment of an inducible Dppa2 transgenic mouse model. We found that the "chronoectopic" expression of Dppa2 prior to normal ZGA time could lead to the advanced nuclear localization of PDH. In summary, Dppa2 plays a key role in ZGA, directly determining the location of TCA core enzymes in early embryos. This study provides a theoretical basis for early embryonic development at the metabolic regulation level.
Collapse
Affiliation(s)
- Anqi Di
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Science, Inner Mongolia University, Hohhot 010021, China; (A.D.); (X.Z.); (L.S.); (X.L.); (C.B.); (G.S.)
| | - Xinyi Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Science, Inner Mongolia University, Hohhot 010021, China; (A.D.); (X.Z.); (L.S.); (X.L.); (C.B.); (G.S.)
| | - Lishuang Song
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Science, Inner Mongolia University, Hohhot 010021, China; (A.D.); (X.Z.); (L.S.); (X.L.); (C.B.); (G.S.)
| | - Song Wang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Xuefei Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Science, Inner Mongolia University, Hohhot 010021, China; (A.D.); (X.Z.); (L.S.); (X.L.); (C.B.); (G.S.)
| | - Chunling Bai
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Science, Inner Mongolia University, Hohhot 010021, China; (A.D.); (X.Z.); (L.S.); (X.L.); (C.B.); (G.S.)
| | - Guanghua Su
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Science, Inner Mongolia University, Hohhot 010021, China; (A.D.); (X.Z.); (L.S.); (X.L.); (C.B.); (G.S.)
| | - Guangpeng Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Science, Inner Mongolia University, Hohhot 010021, China; (A.D.); (X.Z.); (L.S.); (X.L.); (C.B.); (G.S.)
| | - Lei Yang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Science, Inner Mongolia University, Hohhot 010021, China; (A.D.); (X.Z.); (L.S.); (X.L.); (C.B.); (G.S.)
| |
Collapse
|
3
|
Scheifer ST, Michels AC, Modolo F, Carla Dos Santos E, Scariot R, Aguiar MCF, Ignácio SA, de Noronha L, Martins LT, Frigeri HR, de Souza CM, Jham BC, Marins MH, Johann ACBR. Analysis of genetic polymorphism and expression of SOX2 in oral leukoplakia: a case-control study. Oral Surg Oral Med Oral Pathol Oral Radiol 2025; 139:430-438. [PMID: 39648056 DOI: 10.1016/j.oooo.2024.11.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/10/2024]
Abstract
PURPOSE To investigate the association of SOX2 polymorphisms with oral leukoplakia with dysplasia (OLD) and compare it with the immunohistochemical expression of SOX-2. METHODS The samples comprised 64 patients with oral leukoplakia and 20 with normal oral mucosa who were subjected to SOX2 polymorphism rs77677339 genotyping by real-time polymerase chain reaction and immunohistochemistry for SOX-2 (basal epithelium expression, suprabasal and total; nuclear area and intensity). Statistical tests included the Chi-square and Fisher's exact tests. RESULTS No significant difference was observed in genotype distribution between the OLD and control groups. The GG genotype (96.9%) was observed in the OLD group and 100% in the control group. The GA genotype was not observed in the control group. Statistical comparisons between the immunohistochemistry and genetic results were not statistically significant. No association was identified between rs77677339 and immunohistochemistry in OLD; however, the presence of allele A in heterozygotes with OLD suggests that this allele may serve as a risk marker. CONCLUSION The variant rs77677339 is localized in a region that contains different micro-RNA-binding sites, which can lead to changes in gene expression, contributing to OLD development through unclear molecular mechanisms. This study presents the preliminary results for this single nucleotide polymorphism in the literature.
Collapse
Affiliation(s)
- Suelen Teixeira Scheifer
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | - Arieli Carini Michels
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | - Filipe Modolo
- Universidade Federal de Santa Catarina, Eng. Agronômico Andrei Cristian Ferreira, s/n - Trindade, Florianópolis, Santa Catarina, Brazil, 88040-900
| | - Emanuela Carla Dos Santos
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | - Rafaela Scariot
- Universidade Federal do Paraná, Avenida Pref. Lothário Meissner, 632, Jardim Botânico, Curitiba, Paraná, Brazil, 80210-170
| | - Maria Cassia Ferreira Aguiar
- Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Pampulha, Belo Horizonte, Minas Gerais, Brazil, 31270-901
| | - Sergio Aparecido Ignácio
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | - Lucia de Noronha
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | - Laysa Toschi Martins
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | - Henrique Ravanhol Frigeri
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | - Cleber Machado de Souza
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | - Bruno Correia Jham
- College of Dental Medicine - Illinois, Midwestern University, 555 31st Street, Cardinal Hall, Room 594, Downers Grove, Illinois, USA, 60515
| | - Mariana Hornung Marins
- Pontifícia Universidade Católica do Paraná, Imaculada Conceição, 1155, Prado Velho, Curitiba, Paraná, Brazil, 80215-901
| | | |
Collapse
|
4
|
Liu S, Yu H, Zhao Z. The molecular pathogenesis of SOX2 in prostate cancer. Discov Oncol 2025; 16:215. [PMID: 39976818 PMCID: PMC11842661 DOI: 10.1007/s12672-025-01972-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
SOX2 is one of the members of the SOX transcription factor family, which is believed to be an important transcription factor that plays a role in embryonic development, maintenance of stem cells, cancer progression, and resistance to cancer treatment. There is increasing evidence suggesting that SOX2 is crucial for the initiation, progression, invasion, metastasis, and treatment resistance of prostate cancer, therefore understanding the mechanism of SOX2 in prostate cancer can provide better targets for the treatment of prostate cancer. This article reviews the structural domains, normal physiological functions, and role in prostate cancer progression of SOX2, providing potential targets for prostate cancer treatment.
Collapse
Affiliation(s)
- Shixue Liu
- Jining Medical University, Jining, 272067, Shandong, China
| | - Honglian Yu
- Department of Biochemistry, Jining Medical University, 133 Hehua Road, Jining, 272067, Shandong, China.
| | - Zhankui Zhao
- Department of Urology, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong, China.
- Department of Urology, Seventh People'S Hospital of Shanghai University of TCM, Shanghai, 200137, Shanghai, China.
| |
Collapse
|
5
|
Liang Y, Yao X, Han J, Wang J, Zhang X, Zhao D, Jiang C, Geng L, Lv S, Liu Z, Mu Y. Establishment of a CRISPR-Based Lentiviral Activation Library for Transcription Factor Screening in Porcine Cells. Animals (Basel) 2024; 15:19. [PMID: 39794961 PMCID: PMC11718943 DOI: 10.3390/ani15010019] [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: 10/30/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/13/2025] Open
Abstract
Transcription factors play important roles in the growth and development of various tissues in pigs, such as muscle, fat, and bone. A transcription-factor-scale activation library based on the clustered, regularly interspaced, short palindromic repeat (CRISPR)/CRISPR-associated endonuclease Cas9 (Cas9) system could facilitate the discovery and functional characterization of the transcription genes involved in a specific gene network. Here, we have designed and constructed a CRISPR activation (CRISPRa) sgRNA library, containing 5056 sgRNAs targeting the promoter region of 1264 transcription factors in pigs. The sgRNA library, including sgRNA with MS2 loops, is a single-vector system and is packaged with lentivirus for cell screening. Porcine PK15 cells expressing the porcine OCT4 promoter driving EGFP, dCas9 fused with VP64, and MS2-binding protein-p65-HSF1 were constructed, and then, the sgRNA activation library was used to screen the transcription factors regulating OCT4 expression. After the lentiviral transduction and deep sequencing of the CRISPR sgRNAs library, the highest ranking candidate genes were identified, including 31 transcription factors activating OCT4 gene expression and 5 transcription factors inhibiting OCT4 gene expression. The function and gene regulation of the candidate genes were further confirmed by the CRISPR activation system in PK15 cells. The CRISPR activation library targeting pig transcription factors provides a promising platform for the systematic discovery and study of genes that determine cell fate.
Collapse
Affiliation(s)
- Yingjuan Liang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Xiaoxia Yao
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Jingxin Han
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Jinpeng Wang
- Key Laboratory of Public Nutrition and Health, National Health Commission of the Peoples’ Republic of China, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 155 Changbai Road, Changping District, Beijing 102206, China;
| | - Xiao Zhang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Donglin Zhao
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Chaoqian Jiang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
| | - Lishuang Geng
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Shihao Lv
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Zhonghua Liu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| | - Yanshuang Mu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (X.Y.); (J.H.); (X.Z.); (C.J.); (L.G.); (S.L.)
- College of Life Science, Northeast Agricultural University, Harbin 150030, China;
| |
Collapse
|
6
|
Kazeminia S, Eirin A. Role of mitochondria in endogenous renal repair. Clin Sci (Lond) 2024; 138:963-973. [PMID: 39076039 PMCID: PMC11410300 DOI: 10.1042/cs20231331] [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: 04/24/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024]
Abstract
Renal tubules have potential to regenerate and repair after mild-to-moderate injury. Proliferation of tubular epithelial cells represents the initial step of this reparative process. Although for many years, it was believed that proliferating cells originated from a pre-existing intra-tubular stem cell population, there is now consensus that surviving tubular epithelial cells acquire progenitor properties to regenerate the damaged kidney. Scattered tubular-like cells (STCs) are dedifferentiated adult renal tubular epithelial cells that arise upon injury and contribute to renal self-healing and recovery by replacing lost neighboring tubular epithelial cells. These cells are characterized by the co-expression of the stem cell surface markers CD133 and CD24, as well as mesenchymal and kidney injury markers. Previous studies have shown that exogenous delivery of STCs ameliorates renal injury and dysfunction in murine models of acute kidney injury, underscoring the regenerative potential of this endogenous repair system. Although STCs contain fewer mitochondria than their surrounding terminally differentiated tubular epithelial cells, these organelles modulate several important cellular functions, and their integrity and function are critical to preserve the reparative capacity of STCs. Recent data suggest that the microenviroment induced by cardiovascular risk factors, such as obesity, hypertension, and renal ischemia may compromise STC mitochondrial integrity and function, limiting the capacity of these cells to repair injured renal tubules. This review summarizes current knowledge of the contribution of STCs to kidney repair and discusses recent insight into the key role of mitochondria in modulating STC function and their vulnerability in the setting of cardiovascular disease.
Collapse
Affiliation(s)
- Sara Kazeminia
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, U.S.A
| | - Alfonso Eirin
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, U.S.A
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, U.S.A
| |
Collapse
|
7
|
Wang Z, Gong W, Yao Z, Jin K, Niu Y, Li B, Zuo Q. Mechanisms of Embryonic Stem Cell Pluripotency Maintenance and Their Application in Livestock and Poultry Breeding. Animals (Basel) 2024; 14:1742. [PMID: 38929361 PMCID: PMC11201147 DOI: 10.3390/ani14121742] [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: 05/09/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Embryonic stem cells (ESCs) are remarkably undifferentiated cells that originate from the inner cell mass of the blastocyst. They possess the ability to self-renew and differentiate into multiple cell types, making them invaluable in diverse applications such as disease modeling and the creation of transgenic animals. In recent years, as agricultural practices have evolved from traditional to biological breeding, it has become clear that pluripotent stem cells (PSCs), either ESCs or induced pluripotent stem cells (iPSCs), are optimal for continually screening suitable cellular materials. However, the technologies for long-term in vitro culture or establishment of cell lines for PSCs in livestock are still immature, and research progress is uneven, which poses challenges for the application of PSCs in various fields. The establishment of a robust in vitro system for these cells is critically dependent on understanding their pluripotency maintenance mechanisms. It is believed that the combined effects of pluripotent transcription factors, pivotal signaling pathways, and epigenetic regulation contribute to maintaining their pluripotent state, forming a comprehensive regulatory network. This article will delve into the primary mechanisms underlying the maintenance of pluripotency in PSCs and elaborate on the applications of PSCs in the field of livestock.
Collapse
Affiliation(s)
- Ziyu Wang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Wei Gong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zeling Yao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Kai Jin
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yingjie Niu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Bichun Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
8
|
Kong D, Zha L, Yao Y, Zhang Z, Gao J, Zhang R, Chen L, Wang Z. Effects of HMGA2 on the biological characteristics and stemness acquisition of gastric cancer cells. Arab J Gastroenterol 2024; 25:135-142. [PMID: 38378354 DOI: 10.1016/j.ajg.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 09/01/2023] [Accepted: 01/02/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND AND STUDY AIMS The high mobility group A2 (HMGA2), a nonhistone nuclear binding protein, modulates transcription by altering the chromatin architecture of the target gene DNA in its specific AT-hooks region. HMGA2 overexpression has been observed in embryonic tissue and many malignant neoplasms. This study sought to verify whether HMGA2 plays a role in the biological functions of gastric cancer cells, such as cell proliferation, invasiveness, migration, and stem cell acquisition, and to provide some ideas for further research on the metastatic mechanism of gastric cancer. PATIENTS AND METHODS HMGA2's effects on the proliferation, invasiveness, and migration capabilities of gastric cancer cells were individually detected by BrdU, Transwell, and wound healing assays. Western blotting and immunofluorescence were used to evaluate whether HMGA2 could promote the acquisition of gastric cancer cells. Biostatistical analyses were performed using SPSS 17.0 for Windows. RESULTS HMGA2 expression levels in gastric cancer cell lines were significantly higher than those in human immortalized gastric epithelial cell lines (p < 0.01). Gastric cancer cell proliferation was inhibited when HMGA2 was overexpressed (p < 0.05). The invasiveness and migration capabilities of gastric cancer cells with HMGA2 overexpression were enhanced more than those of the corresponding control groups (p < 0.05). HMGA2 overexpression promotes the stemness acquisition of stem cells from gastric cancer cells. CONCLUSIONS This study verified that the HMGA2 structural transcription factor promotes invasiveness, migration, and acquisition of gastric cancer cells. Furthermore, our findings provide significant insight for further research on the metastatic mechanism of gastric cancer.
Collapse
Affiliation(s)
- Dequan Kong
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China.
| | - Lang Zha
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yaben Yao
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Zhenyu Zhang
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Jun Gao
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Rui Zhang
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Lei Chen
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
9
|
Gadwal A, Purohit P, Khokhar M, Vishnoi JR, Pareek P, Choudhary R, Elhence P, Banerjee M, Sharma P. GALNT6, GALNT14, and Gal-3 in association with GDF-15 promotes drug resistance and stemness of breast cancer via β-catenin axis. Growth Factors 2024; 42:84-100. [PMID: 38889447 DOI: 10.1080/08977194.2024.2368907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
N-acetylgalactosaminyltransferases (GALNTs) are a polypeptide responsible for aberrant glycosylation in breast cancer (BC), but the mechanism is unclear. In this study, expression levels of GALNT6, GALNT14, and Gal-3 were assessed in BC, and their association with GDF-15, β-catenin, stemness (SOX2 and OCT4), and drug resistance marker (ABCC5) was evaluated. Gene expression of GALNT6, GALNT14, Gal-3, GDF-15, OCT4, SOX2, ABCC5, and β-catenin in tumor and adjacent non-tumor tissues (n = 30) was determined. The same was compared with GEO-microarray datasets. A significant increase in the expression of candidate genes was observed in BC tumor compared to adjacent non-tumor tissue; and in pre-therapeutic patients compared to post-therapeutic. GALNT6, GALNT14, Gal-3, and GDF-15 showed positive association with β-catenin, SOX2, OCT4, and ABCC5 and were significantly associated with poor Overall Survival. Our findings were also validated via in silico analysis. Our study suggests that GALNT6, GALNT14, and Gal-3 in association with GDF-15 promote stemness and intrinsic drug resistance in BC, possibly by β-catenin signaling pathway.
Collapse
Affiliation(s)
- Ashita Gadwal
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| | - Jeewan Ram Vishnoi
- Department of Oncosurgery, All India Institute of Medical Sciences, Jodhpur, India
| | - Puneet Pareek
- Department of Radiation Oncology, All India Institute of Medical Sciences, Jodhpur, India
| | - Ramkaran Choudhary
- Department of General Surgery, All India Institute of Medical Sciences, Jodhpur, India
| | - Poonam Elhence
- Department of Pathology, All India Institute of Medical Sciences, Jodhpur, India
| | - Mithu Banerjee
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, India
| |
Collapse
|
10
|
Giri A, Kar S. Interlinked bi-stable switches govern the cell fate commitment of embryonic stem cells. FEBS Lett 2024; 598:915-934. [PMID: 38408774 DOI: 10.1002/1873-3468.14832] [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: 10/05/2023] [Revised: 12/23/2023] [Accepted: 02/03/2024] [Indexed: 02/28/2024]
Abstract
The development of embryonic stem (ES) cells to extraembryonic trophectoderm and primitive endoderm lineages manifests distinct steady-state expression patterns of two key transcription factors-Oct4 and Nanog. How dynamically such kind of steady-state expressions are maintained remains elusive. Herein, we demonstrate that steady-state dynamics involving two bistable switches which are interlinked via a stepwise (Oct4) and a mushroom-like (Nanog) manner orchestrate the fate specification of ES cells. Our hypothesis qualitatively reconciles various experimental observations and elucidates how different feedback and feedforward motifs orchestrate the extraembryonic development and stemness maintenance of ES cells. Importantly, the model predicts strategies to optimize the dynamics of self-renewal and differentiation of embryonic stem cells that may have therapeutic relevance in the future.
Collapse
Affiliation(s)
- Amitava Giri
- Department of Chemistry, IIT Bombay, Powai, India
| | - Sandip Kar
- Department of Chemistry, IIT Bombay, Powai, India
| |
Collapse
|
11
|
Dehghanian F, Bovio PP, Gather F, Probst S, Naghsh-Nilchi A, Vogel T. ZFP982 confers mouse embryonic stem cell characteristics by regulating expression of Nanog, Zfp42, and Dppa3. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119686. [PMID: 38342310 DOI: 10.1016/j.bbamcr.2024.119686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Understanding the genetic underpinnings of protein networks conferring stemness is of broad interest for basic and translational research. METHODS We used multi-omics analyses to identify and characterize stemness genes, and focused on the zinc finger protein 982 (Zfp982) that regulates stemness through the expression of Nanog, Zfp42, and Dppa3 in mouse embryonic stem cells (mESC). RESULTS Zfp982 was expressed in stem cells, and bound to chromatin through a GCAGAGKC motif, for example near the stemness genes Nanog, Zfp42, and Dppa3. Nanog and Zfp42 were direct targets of ZFP982 that decreased in expression upon knockdown and increased upon overexpression of Zfp982. We show that ZFP982 expression strongly correlated with stem cell characteristics, both on the transcriptional and morphological levels. Zfp982 expression decreased with progressive differentiation into ecto-, endo- and mesodermal cell lineages, and knockdown of Zfp982 correlated with morphological and transcriptional features of differentiated cells. Zfp982 showed transcriptional overlap with members of the Hippo signaling pathway, one of which was Yap1, the major co-activator of Hippo signaling. Despite the observation that ZFP982 and YAP1 interacted and localized predominantly to the cytoplasm upon differentiation, the localization of YAP1 was not influenced by ZFP982 localization. CONCLUSIONS Together, our study identified ZFP982 as a transcriptional regulator of early stemness genes, and since ZFP982 is under the control of the Hippo pathway, underscored the importance of the context-dependent Hippo signals for stem cell characteristics.
Collapse
Affiliation(s)
- Fariba Dehghanian
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, Isfahan 81746-73441, Iran; Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany.
| | - Patrick Piero Bovio
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Fabian Gather
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Simone Probst
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Amirhosein Naghsh-Nilchi
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, Isfahan 81746-73441, Iran
| | - Tanja Vogel
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Centre for Basics in Neuromodulation (Neuromodul Basics), Freiburg, Germany.
| |
Collapse
|
12
|
Chen YH, van Zon S, Adams A, Schmidt-Arras D, Laurence ADJ, Uhlig HH. The Human GP130 Cytokine Receptor and Its Expression-an Atlas and Functional Taxonomy of Genetic Variants. J Clin Immunol 2023; 44:30. [PMID: 38133879 PMCID: PMC10746620 DOI: 10.1007/s10875-023-01603-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 10/30/2023] [Indexed: 12/23/2023]
Abstract
Genetic variants in IL6ST encoding the shared cytokine receptor for the IL-6 cytokine family GP130 have been associated with a diverse number of clinical phenotypes and disorders. We provide a molecular classification for 59 reported rare IL6ST pathogenic or likely pathogenic variants and additional polymorphisms. Based on loss- or gain-of-function, cytokine selectivity, mono- and biallelic associations, and variable cellular mosaicism, we grade six classes of IL6ST variants and explore the potential for additional variants. We classify variants according to the American College of Medical Genetics and Genomics criteria. Loss-of-function variants with (i) biallelic complete loss of GP130 function that presents with extended Stüve-Wiedemann Syndrome; (ii) autosomal recessive hyper-IgE syndrome (HIES) caused by biallelic; and (iii) autosomal dominant HIES caused by monoallelic IL6ST variants both causing selective IL-6 and IL-11 cytokine loss-of-function defects; (iv) a biallelic cytokine-specific variant that exclusively impairs IL-11 signaling, associated with craniosynostosis and tooth abnormalities; (v) somatic monoallelic mosaic constitutively active gain-of-function variants in hepatocytes that present with inflammatory hepatocellular adenoma; and (vi) mosaic constitutively active gain-of-function variants in hematopoietic and non-hematopoietic cells that are associated with an immune dysregulation syndrome. In addition to Mendelian IL6ST coding variants, there are common non-coding cis-acting variants that modify gene expression, which are associated with an increased risk of complex immune-mediated disorders and trans-acting variants that affect GP130 protein function. Our taxonomy highlights IL6ST as a gene with particularly strong functional and phenotypic diversity due to the combinatorial biology of the IL-6 cytokine family and predicts additional genotype-phenotype associations.
Collapse
Affiliation(s)
- Yin-Huai Chen
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Sarah van Zon
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Alex Adams
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Dirk Schmidt-Arras
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | | | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
- Biomedical Research Centre, University of Oxford, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Choi J, Lee H. NFIB-MLL1 complex is required for the stemness and Dlx5-dependent osteogenic differentiation of C3H10T1/2 mesenchymal stem cells. J Biol Chem 2023; 299:105193. [PMID: 37633334 PMCID: PMC10519831 DOI: 10.1016/j.jbc.2023.105193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023] Open
Abstract
Despite significant progress in our understanding of the molecular mechanism of mesenchymal stem cell (MSC) differentiation, less is known about the factors maintaining the stemness and plasticity of MSCs. Here, we show that the NFIB-MLL1 complex plays key roles in osteogenic differentiation and stemness of C3H10T1/2 MSCs. We find that depletion of either NFIB or MLL1 results in a severely hampered osteogenic potential and failed activation of key osteogenic transcription factors, such as Dlx5, Runx2, and Osx, following osteogenic stimuli. In addition, the NFIB-MLL1 complex binds directly to the promoter of Dlx5, and exogenous expression of Myc-Dlx5, but not the activation of either the BMP- or the Wnt-signaling pathway, is sufficient to restore the osteogenic potential of cells depleted of NFIB or MLL1. Moreover, chromatin immunoprecipitation (ChIP) and ChIP-sequencing analysis showed that the NFIB-MLL1 complex mediates the deposition of trimethylated histone H3K4 at both Dlx5 and Cebpa, key regulator genes that function at the early stages of osteogenic and adipogenic differentiation, respectively, in uncommitted C3H10T1/2 MSCs. Surprisingly, the depletion of either NFIB or MLL1 leads to decreased trimethylated histone H3K4 and results in elevated trimethylated histone H3K9 at those developmental genes. Furthermore, gene expression profiling and ChIP-sequencing analysis revealed lineage-specific changes in chromatin landscape and gene expression in response to osteogenic stimuli. Taken together, these data provide evidence for the hitherto unknown role of the NFIB-MLL1 complex in the maintenance and lineage-specific differentiation of C3H10T1/2 MSCs and support the epigenetic regulatory mechanism underlying the stemness and plasticity of MSCs.
Collapse
Affiliation(s)
- Janghyun Choi
- Department of Biological Sciences, College of Natural Science, Inha University, Incheon, South Korea.
| | - Hansol Lee
- Department of Biological Sciences, College of Natural Science, Inha University, Incheon, South Korea.
| |
Collapse
|
14
|
Cierpikowski P, Leszczyszyn A, Bar J. The Role of Hedgehog Signaling Pathway in Head and Neck Squamous Cell Carcinoma. Cells 2023; 12:2083. [PMID: 37626893 PMCID: PMC10453169 DOI: 10.3390/cells12162083] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth leading malignancy worldwide, with a poor prognosis and limited treatment options. Molecularly targeted therapies for HNSCC are still lacking. However, recent reports provide novel insights about many molecular alterations in HNSCC that may be useful in future therapies. Therefore, it is necessary to identify new biomarkers that may provide a better prediction of the disease and promising targets for personalized therapy. The poor response of HNSCC to therapy is attributed to a small population of tumor cells called cancer stem cells (CSCs). Growing evidence indicates that the Hedgehog (HH) signaling pathway plays a crucial role in the development and maintenance of head and neck tissues. The HH pathway is normally involved in embryogenesis, stem cell renewal, and tissue regeneration. However, abnormal activation of the HH pathway is also associated with carcinogenesis and CSC regulation. Overactivation of the HH pathway was observed in several tumors, including basal cell carcinoma, that are successfully treated with HH inhibitors. However, clinical studies about HH pathways in HNSCC are still rare. In this review, we summarize the current knowledge and recent advances regarding the HH pathway in HNSCC and discuss its possible implications for prognosis and future therapy.
Collapse
Affiliation(s)
- Piotr Cierpikowski
- Department of Maxillofacial Surgery, The Ludwik Rydygier Specialist Hospital, Osiedle Zlotej Jesieni 1, 31-826 Krakow, Poland
| | - Anna Leszczyszyn
- Dental Surgery Outpatient Clinic, 4th Military Clinical Hospital, Weigla 5, 53-114 Wroclaw, Poland;
| | - Julia Bar
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland
| |
Collapse
|
15
|
Sagnak Yilmaz Z, Sarioglu S. Molecular Pathology of Micropapillary Carcinomas: Is Characteristic Morphology Related to Molecular Mechanisms? Appl Immunohistochem Mol Morphol 2023; 31:267-277. [PMID: 37036419 DOI: 10.1097/pai.0000000000001123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/13/2023] [Indexed: 04/11/2023]
Abstract
Micropapillary carcinoma is an entity defined histologically in many organs. It is associated with lymph node metastasis and poor prognosis. The main mechanism for its histopathologic appearance is reverse polarization. Although the studies on this subject are limited, carcinomas with micropapillary morphology observed in different organs are examined by immunohistochemical and molecular methods. Differences are shown in these tumors compared with conventional carcinomas regarding the rate of somatic mutations, mRNA and miRNA expressions, and protein expression levels. TP53 , PIK3CA , TERT , KRAS , EGFR , MYC , FGFR1 , BRAF , AKT1 , HER2/ERBB2 , CCND1 , and APC mutations, which genes frequently detected in solid tumors, have also been detected in invasive micropapillary carcinoma (IMPC) in various organs. 6q chromosome loss, DNAH9 , FOXO3 , SEC. 63 , and FMN2 gene mutations associated with cell polarity or cell structure and skeleton have also been detected in IMPCs. Among the proteins that affect cell polarity, RAC1, placoglobin, as well as CLDNs, LIN7A, ZEB1, CLDN1, DLG1, CDH1 (E-cadherin), OCLN, AFDN/AF6, ZEB1, SNAI2, ITGA1 (integrin alpha 1), ITGB1 (integrin beta 1), RHOA, Jagged-1 (JAG1) mRNAs differentially express between IMPC and conventional carcinomas. Prediction of prognosis and targeted therapy may benefit from the understanding of molecular mechanisms of micropapillary morphology. This review describes the molecular pathologic mechanisms underlying the micropapillary changes of cancers in various organs in a cell polarity-related dimension.
Collapse
Affiliation(s)
- Zeynep Sagnak Yilmaz
- Department of Molecular Pathology, Dokuz Eylül University Graduate School of Health Sciences
- Pathology Department, Karadeniz Technical University Faculty of Medicine, Trabzon, Turkey
| | - Sulen Sarioglu
- Department of Molecular Pathology, Dokuz Eylül University Graduate School of Health Sciences
- Pathology Department, Dokuz Eylül University Faculty of Medicine, Izmir
| |
Collapse
|
16
|
Dwivedi S, Choudhary P, Gupta A, Singh S. Therapeutical growth in oligodendroglial fate induction via transdifferentiation of stem cells for neuroregenerative therapy. Biochimie 2023; 211:35-56. [PMID: 36842627 DOI: 10.1016/j.biochi.2023.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/20/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
The merits of stem cell therapy and research are undisputed due to their widespread usage in the treatment of neurodegenerative diseases and demyelinating disorders. Cell replacement therapy especially revolves around stem cells and their induction into different cell lineages both adult and progenitor - belonging to each germ layer, prior to transplantation or disease modeling studies. The nervous system is abundant in glial cells and among these are oligodendrocytes capable of myelinating new-born neurons and remyelination of axons with lost or damaged myelin sheath. But demyelinating diseases generate tremendous deficit between myelin loss and recovery. To compensate for this loss, analyze the defects in remyelination mechanisms as well as to trigger full recovery in such patients mesenchymal stem cells (MSCs) have been induced to transdifferentiate into oligodendrocytes. But such experiments are riddled with problems like prolonged, tenuous and complicated protocols that stretch longer than the time taken for the spread of demyelination-associated after-effects. This review delves into such protocols and the combinations of different molecules and factors that have been recruited to derive bona fide oligodendrocytes from in vitro differentiation of embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and MSCs with special focus on MSC-derived oligodendrocytes.
Collapse
Affiliation(s)
- Shrey Dwivedi
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Princy Choudhary
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Ayushi Gupta
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Sangeeta Singh
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India.
| |
Collapse
|
17
|
Ozden Akkaya O, Dikmen T, Nawaz S, Kibria AG, Altunbaş K, Yağci A, Erdoğan M, Yaprakci MV. Comparison of proliferation and osteogenic differentiation potential of bovine adipose tissue and bone marrow derived stem cells. Biotech Histochem 2023; 98:267-279. [PMID: 36815431 DOI: 10.1080/10520295.2023.2177347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Bone marrow derived stem cells (BMSC) are the most utilized cell type in the field of bone regeneration. Although BMSC are both safe and efficacious, the search for alternative sources for stem cells continues. We investigated bovine BMSC and adipose tissue derived mesenchymal stem cells (ATSC) using immunofluorescence and PCR. We further compared the osteogenic differentiation potentials of both sources of stem cells. We assessed alkaline phosphatase (ALP) enzyme levels and calcium deposition in differentiating cells at days 7, 14 and 21 to compare the osteogenic differentiation capability of both cell types. We found that ATSC expressed significantly higher ALP levels compared to BMSC throughout osteogenic differentiation. Calcium deposition was greater in ATSC than BMSC at days 7 and 14. By the end of day 21, BMSC produced greater calcium deposition. We found that ATSC undergo osteogenic differentiation more rapidly than BMSC, but BMSC provide greater mineralization over longer periods.
Collapse
Affiliation(s)
- Ozlem Ozden Akkaya
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Tayfun Dikmen
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Shah Nawaz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye.,Department of Anatomy, Faculty of Veterinary Science, University of Agriculture, Faisalabad, Pakistan
| | - Asm Golam Kibria
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye.,Department of Anatomy and Histology, Chattogram University of Veterinary and Animal Sciences, Chattogram, Bangladesh
| | - Korhan Altunbaş
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Artay Yağci
- Department of Histology and Embryology, Milas Veterinary Faculty, Mugla Sıtkı Kocman University, Mugla, Türkiye
| | - Metin Erdoğan
- Department of Veterinary Biology and Genetics, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Mustafa Volkan Yaprakci
- Department of Surgery, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| |
Collapse
|
18
|
Lei K, Zhang W, Chen J, McKinney SA, Ross EJ, Lee HC, Sánchez Alvarado A. Pluripotency retention and exogenous mRNA introduction in planarian stem cells in culture. iScience 2023; 26:106001. [PMID: 36866042 PMCID: PMC9971864 DOI: 10.1016/j.isci.2023.106001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/31/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Planarians possess naturally occurring pluripotent adult somatic stem cells (neoblasts) required for homeostasis and whole-body regeneration. However, no reliable neoblast culture methods are currently available, hindering mechanistic studies of pluripotency and the development of transgenic tools. We report robust methods for neoblast culture and delivery of exogenous mRNAs. We identify optimal culture media for the short-term maintenance of neoblasts in vitro and show via transplantation that cultured stem cells retain pluripotency for two days. We developed a procedure that significantly improves neoblast yield and purity by modifying standard flow cytometry methods. These methods enable the introduction and expression of exogenous mRNAs in neoblasts, overcoming a key hurdle impeding the application of transgenics in planarians. The advances in cell culture reported here create new opportunities for mechanistic studies of planarian adult stem cell pluripotency, and provide a systematic framework to develop cell culture techniques in other emerging research organisms.
Collapse
Affiliation(s)
- Kai Lei
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Wenya Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Jiajia Chen
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Sean A. McKinney
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Eric J. Ross
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Howard Hughes Medical Institute, Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Heng-Chi Lee
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Alejandro Sánchez Alvarado
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Howard Hughes Medical Institute, Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| |
Collapse
|
19
|
Herzog AE, Somayaji R, Nör JE. Bmi-1: A master regulator of head and neck cancer stemness. FRONTIERS IN ORAL HEALTH 2023; 4:1080255. [PMID: 36726797 PMCID: PMC9884974 DOI: 10.3389/froh.2023.1080255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
Head and neck cancers are composed of a diverse group of malignancies, many of which exhibit an unacceptably low patient survival, high morbidity and poor treatment outcomes. The cancer stem cell (CSC) hypothesis provides an explanation for the substantial patient morbidity associated with treatment resistance and the high frequency of tumor recurrence/metastasis. Stem cells are a unique population of cells capable of recapitulating a heterogenous organ from a single cell, due to their capacity to self-renew and differentiate into progenitor cells. CSCs share these attributes, in addition to playing a pivotal role in cancer initiation and progression by means of their high tumorigenic potential. CSCs constitute only a small fraction of tumor cells but play a major role in tumor initiation and therapeutic evasion. The shift towards stem-like phenotype fuels many malignant features of a cancer cell and mediates resistance to conventional chemotherapy. Bmi-1 is a master regulator of stem cell self-renewal as part of the polycomb repressive complex 1 (PRC1) and has emerged as a prominent player in cancer stem cell biology. Bmi-1 expression is upregulated in CSCs, which is augmented by tumor-promoting factors and various conventional chemotherapies. Bmi-1+ CSCs mediate chemoresistance and metastasis. On the other hand, inhibiting Bmi-1 rescinds CSC function and re-sensitizes cancer cells to chemotherapy. Therefore, elucidating the functional role of Bmi-1 in CSC-mediated cancer progression may unveil an attractive target for mechanism-based, developmental therapeutics. In this review, we discuss the parallels in the role of Bmi-1 in stem cell biology of health and disease and explore how this can be leveraged to advance clinical treatment strategies for head and neck cancer.
Collapse
Affiliation(s)
- Alexandra E. Herzog
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Ritu Somayaji
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Jacques E. Nör
- Department of Cariology, Restorative Sciences, Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States,Department of Otolaryngology – Head and Neck Surgery, University of Michigan Medical School; Ann Arbor, MI, United States,Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, United States,Universityof Michigan Rogel Cancer Center, Ann Arbor, MI, United States
| |
Collapse
|
20
|
Zhang J, Lv X, Wei B, Gong X, Chen L. CHD4 mediates SOX2 transcription through TRPS1 in luminal breast cancer. Cell Signal 2022; 100:110464. [PMID: 36075559 DOI: 10.1016/j.cellsig.2022.110464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/26/2022] [Accepted: 08/31/2022] [Indexed: 11/26/2022]
Abstract
Chromodomain helicase DNA binding protein 4 (CHD4), as a core component of the nucleosome remodeling and deactetylase (NuRD) complex, participated in the inititation and development of myriad cancers. However, little is known about the linkage between CHD4 and breast cancer stemness. Here, we found that CHD4 repress the expression of SOX2, a key regulator of cancer stem cells (CSCs), to suppress cancer stemness in breast cancer. Mechanistically, CHD4 binds to the promoter of SOX2 depend on TRPS1. And CHD4 transcriptional activation of SOX2 was abolished by TRPS1. These findings identify CHD4 as a regulator of SOX2 linked to breast cancer stemness and provide detailed mechanistic of CHD4 in CSC functions.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiang Lv
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Bo Wei
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xue Gong
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China.
| | - Liming Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
21
|
DiNatale A, Castelli MS, Nash B, Meucci O, Fatatis A. Regulation of Tumor and Metastasis Initiation by Chemokine Receptors. J Cancer 2022; 13:3160-3176. [PMID: 36118530 PMCID: PMC9475358 DOI: 10.7150/jca.72331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/10/2022] [Indexed: 12/13/2022] Open
Abstract
Tumor-initiating cells (TICs) are a rare sub-population of cells within the bulk of a tumor that are major contributors to tumor initiation, metastasis, and chemoresistance. TICs have a stem-cell-like phenotype that is dictated by the expression of master regulator transcription factors, including OCT4, NANOG, and SOX2. These transcription factors are expressed via activation of multiple signaling pathways that drive cancer initiation and progression. Importantly, these same signaling pathways can be activated by select chemokine receptors. Chemokine receptors are increasingly being revealed as major drivers of the TIC phenotype, as their signaling can lead to activation of stemness-controlling transcription factors. Additionally, the cell surface expression of chemokine receptors provides a unique therapeutic target to disrupt signaling pathways that control the expression of master regulator transcription factors and the TIC phenotype. This review summarizes the master regulator transcription factors known to dictate the TIC phenotype, along with the complex signaling pathways that can mediate their expression and the chemokine receptors that are most upstream of this phenotype.
Collapse
Affiliation(s)
- Anthony DiNatale
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.,Present Address: Janssen Oncology, Spring House, PA, USA
| | - Maria Sofia Castelli
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.,Present address: Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.,Program in Immune Cell Regulation & Targeting, Sidney Kimmel Cancer Center of Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alessandro Fatatis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.,Program in Translational and Cellular Oncology, Sidney Kimmel Cancer Center of Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
22
|
Rivas SR, Valdez MJM, Govindarajan V, Seetharam D, Doucet-O’Hare TT, Heiss JD, Shah AH. The Role of HERV-K in Cancer Stemness. Viruses 2022; 14:v14092019. [PMID: 36146825 PMCID: PMC9504571 DOI: 10.3390/v14092019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 12/15/2022] Open
Abstract
Human endogenous retrovirus-K (HERV-K) is the most recently integrated retrovirus in the human genome, with implications for multiple disorders, including cancer. Although typically transcriptionally silenced in normal adult cells, dysregulation of HERV-K (HML-2) elements has been observed in cancer, including breast, germ cell tumors, pancreatic, melanoma, and brain cancer. While multiple methods of carcinogenesis have been proposed, here we discuss the role of HERV-K (HML-2) in the promotion and maintenance of the stem-cell in cancer. Aberrant expression of HERV-K has been shown to promote expression of stem cell markers and promote dedifferentiation. In this review, we discuss HERV-K (HML-2) as a potential therapeutic target based on evidence that some tumors depend on the expression of its proteins for survival.
Collapse
Affiliation(s)
- Sarah R. Rivas
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892, USA
- Correspondence: (S.R.R.); (A.H.S.)
| | - Mynor J. Mendez Valdez
- Section of Virology and Immunotherapy, Department of Neurosurgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Vaidya Govindarajan
- Section of Virology and Immunotherapy, Department of Neurosurgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Deepa Seetharam
- Section of Virology and Immunotherapy, Department of Neurosurgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Tara T. Doucet-O’Hare
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - John D. Heiss
- Surgical Neurology Branch, National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892, USA
| | - Ashish H. Shah
- Section of Virology and Immunotherapy, Department of Neurosurgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Correspondence: (S.R.R.); (A.H.S.)
| |
Collapse
|
23
|
Oct4 facilitates chondrogenic differentiation of mesenchymal stem cells by mediating CIP2A expression. Cell Tissue Res 2022; 389:11-21. [PMID: 35435493 DOI: 10.1007/s00441-022-03619-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Bone development and cartilage formation require strict modulation of gene expression for mesenchymal stem cells (MSCs) to progress through their differentiation stages. Octamer-binding transcription factor 4 (Oct4) expression is generally restricted to developing embryonic pluripotent cells, but its role in chondrogenic differentiation (CD) of MSCs remains unclear. We therefore investigated the role of Oct4 in CD using a microarray, quantitative real-time polymerase chain reaction, and western blotting. The expression of Oct4 was elevated when the CD of cultured MSCs was induced. Silencing Oct4 damaged MSC growth and proliferation and decreased CD, indicated by decreased cartilage matrix formation and the expression of Col2a1, Col10a1, Acan, and Sox9. We found a positive correlation between the expression of CIP2A, a natural inhibitor of protein phosphatase 2A (PP2A) and that of Oct4. Cellular inhibitor of PP2A (CIP2A) expression gradually increased after CD. Overexpression of CIP2A in MSCs with Oct4 depletion promoted cartilage matrix deposition as well as Col2a1, Col10a1, Acan, and Sox9 expression. The chondrogenic induction triggered c-Myc, Akt, ERK, and MEK phosphorylation and upregulated c-Myc and mTOR expression, which was downregulated upon Oct4 knockdown and restored by CIP2A overexpression. These findings indicated that Oct4 functions as an essential chondrogenesis regulator, partly via the CIP2A/PP2A pathway.
Collapse
|
24
|
Abbasi A, Imaichi S, Ling V, Shukla A. Mesenchymal Stem Cell Behavior on Soft Hydrogels with Aligned Surface Topographies. ACS APPLIED BIO MATERIALS 2022; 5:1890-1900. [PMID: 35199983 DOI: 10.1021/acsabm.1c01260] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human mesenchymal stem cells (HMSCs) are important for cell-based therapies. However, the success of HMSC therapy requires large-scale in vitro expansion of these multipotent cells. The traditional expansion of HMSCs on tissue-culture-treated stiff polystyrene induces significant changes in their shape, multipotency, and secretome, leading to early senescence and subdued paracrine activity. To enhance their therapeutic potential, here, we have developed two-dimensional soft hydrogels with imprinted microscale aligned grooves for use as HMSC culture substrates. We showed that, depending on the dimensions of the topographical features, these substrates led to lower cellular spreading and cytoskeletal tension, maintaining multipotency and osteogenic and adipogenic differentiate potential, while lowering cellular senescence. We also observed a greater capacity of HMSCs to produce anti-inflammatory cytokines after short-term priming on these hydrogel substrates. Overall, these soft hydrogels with unique surface topography have shown great promise as in vitro culture substrates to maximize the therapeutic potential of HMSCs.
Collapse
Affiliation(s)
- Akram Abbasi
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Sachiko Imaichi
- Takeda Pharmaceuticals, Cambridge, Massachusetts 02139, United States
| | - Vincent Ling
- Takeda Pharmaceuticals, Cambridge, Massachusetts 02139, United States
| | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
25
|
Borojević A, Jauković A, Kukolj T, Mojsilović S, Obradović H, Trivanović D, Živanović M, Zečević Ž, Simić M, Gobeljić B, Vujić D, Bugarski D. Vitamin D3 Stimulates Proliferation Capacity, Expression of Pluripotency Markers, and Osteogenesis of Human Bone Marrow Mesenchymal Stromal/Stem Cells, Partly through SIRT1 Signaling. Biomolecules 2022; 12:biom12020323. [PMID: 35204824 PMCID: PMC8868595 DOI: 10.3390/biom12020323] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/02/2022] Open
Abstract
The biology of vitamin D3 is well defined, as are the effects of its active metabolites on various cells, including mesenchymal stromal/stem cells (MSCs). However, the biological potential of its precursor, cholecalciferol (VD3), has not been sufficiently investigated, although its significance in regenerative medicine—mainly in combination with various biomaterial matrices—has been recognized. Given that VD3 preconditioning might also contribute to the improvement of cellular regenerative potential, the aim of this study was to investigate its effects on bone marrow (BM) MSC functions and the signaling pathways involved. For that purpose, the influence of VD3 on BM-MSCs obtained from young human donors was determined via MTT test, flow cytometric analysis, immunocytochemistry, and qRT-PCR. Our results revealed that VD3, following a 5-day treatment, stimulated proliferation, expression of pluripotency markers (NANOG, SOX2, and Oct4), and osteogenic differentiation potential in BM-MSCs, while it reduced their senescence. Moreover, increased sirtuin 1 (SIRT1) expression was detected upon treatment with VD3, which mediated VD3-promoted osteogenesis and, partially, the stemness features through NANOG and SOX2 upregulation. In contrast, the effects of VD3 on proliferation, Oct4 expression, and senescence were SIRT1-independent. Altogether, these data indicate that VD3 has strong potential to modulate BM-MSCs’ features, partially through SIRT1 signaling, although the precise mechanisms merit further investigation.
Collapse
Affiliation(s)
- Ana Borojević
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (Ž.Z.); (M.S.); (B.G.); (D.V.)
- Correspondence: ; Tel.: +381-11-3108-175
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (T.K.); (S.M.); (H.O.); (D.T.); (M.Ž.); (D.B.)
| | - Tamara Kukolj
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (T.K.); (S.M.); (H.O.); (D.T.); (M.Ž.); (D.B.)
| | - Slavko Mojsilović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (T.K.); (S.M.); (H.O.); (D.T.); (M.Ž.); (D.B.)
| | - Hristina Obradović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (T.K.); (S.M.); (H.O.); (D.T.); (M.Ž.); (D.B.)
| | - Drenka Trivanović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (T.K.); (S.M.); (H.O.); (D.T.); (M.Ž.); (D.B.)
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics, Röntgenring 11, 97070 Würzburg, Germany
- Bernhard-Heine-Center for Locomotion Research, University Würzburg, Sanderring 2, 97070 Würzburg, Germany
| | - Milena Živanović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (T.K.); (S.M.); (H.O.); (D.T.); (M.Ž.); (D.B.)
| | - Željko Zečević
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (Ž.Z.); (M.S.); (B.G.); (D.V.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marija Simić
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (Ž.Z.); (M.S.); (B.G.); (D.V.)
| | - Borko Gobeljić
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (Ž.Z.); (M.S.); (B.G.); (D.V.)
| | - Dragana Vujić
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (Ž.Z.); (M.S.); (B.G.); (D.V.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Diana Bugarski
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (T.K.); (S.M.); (H.O.); (D.T.); (M.Ž.); (D.B.)
| |
Collapse
|
26
|
Pock T, Schulte K, Schlatt S, Boiani M, Nordhoff V. GM-CSF perturbs cell identity in mouse pre-implantation embryos. PLoS One 2022; 17:e0263793. [PMID: 35143564 PMCID: PMC8830693 DOI: 10.1371/journal.pone.0263793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/26/2022] [Indexed: 12/03/2022] Open
Abstract
Growth factors became attractive candidates for medium supplementation to further improve the quality of embryo culture and to mimic in vivo nutrition. Granulocyte macrophage colony-stimulating factor (GM-CSF) is a cytokine influencing the maternal-fetal interface and supporting placental development in mouse and human. It is expressed in epithelial cells of the endometrium under the regulation of estrogens. The factor is already in clinical use and a large clinical trial showed that, if supplemented to an embryo culture medium, it leads to increased survival of embryos, especially in women with previous miscarriages. Animal and cell culture studies on isolated trophectoderm cells support an effect mainly on cellular expansion. Aim of this study was to investigate, if the supplementation of GM-CSF either in a human ART medium or in a mouse optimized medium, leads to a change in cell number and cell lineages in the early pre-implantation mouse embryo. Our data shows that mouse GM-CSF increased total cell numbers with increasing concentrations. This increase of cell number has not been found in embryos cultured in ART media with or without human GM-CSF (hGM-CSF) or in a mouse medium supplemented with different concentrations of hGM-CSF. The changes were caused by a marked difference in TE and primitive endoderm cell numbers but not due to a change in epiblast cell numbers. Additionally, results show an ectopic expression of NANOG among trophectoderm cells in both, human ART media (with and without GM-CSF) and at increasing concentrations in the mouse and the human GM-CSF supplemented media. In conclusion, we could show that GM-CSF has an effect on cell identity in mice, which might probably also occur in the human. Therefore, we would like to rare awareness that the use of supplements without proper research could bare risks for the embryo itself and probably also in the post-implantation phase.
Collapse
Affiliation(s)
- Tim Pock
- Centre of Reproductive Medicine and Andrology (CeRA), University of Münster, Münster, Germany
| | - Katharina Schulte
- Central Animal Facility of the Faculty of Medicine, University of Münster, Münster, Germany
| | - Stefan Schlatt
- Centre of Reproductive Medicine and Andrology (CeRA), University of Münster, Münster, Germany
| | - Michele Boiani
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Verena Nordhoff
- Centre of Reproductive Medicine and Andrology (CeRA), University of Münster, Münster, Germany
- * E-mail:
| |
Collapse
|
27
|
Shahoumi LA. Oral Cancer Stem Cells: Therapeutic Implications and Challenges. FRONTIERS IN ORAL HEALTH 2022; 2:685236. [PMID: 35048028 PMCID: PMC8757826 DOI: 10.3389/froh.2021.685236] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is currently one of the 10 most common malignancies worldwide, characterized by a biologically highly diverse group of tumors with non-specific biomarkers and poor prognosis. The incidence rate of HNSCC varies widely throughout the world, with an evident prevalence in developing countries such as those in Southeast Asia and Southern Africa. Tumor relapse and metastasis following traditional treatment remain major clinical problems in oral cancer management. Current evidence suggests that therapeutic resistance and metastasis of cancer are mainly driven by a unique subpopulation of tumor cells, termed cancer stem cells (CSCs), or cancer-initiating cells (CICs), which are characterized by their capacity for self-renewal, maintenance of stemness and increased tumorigenicity. Thus, more understanding of the molecular mechanisms of CSCs and their behavior may help in developing effective therapeutic interventions that inhibit tumor growth and progression. This review provides an overview of the main signaling cascades in CSCs that drive tumor repropagation and metastasis in oral cancer, with a focus on squamous cell carcinoma. Other oral non-SCC tumors, including melanoma and malignant salivary gland tumors, will also be considered. In addition, this review discusses some of the CSC-targeted therapeutic strategies that have been employed to combat disease progression, and the challenges of targeting CSCs, with the aim of improving the clinical outcomes for patients with oral malignancies. Targeting of CSCs in head and neck cancer (HNC) represents a promising approach to improve disease outcome. Some CSC-targeted therapies have already been proven to be successful in pre-clinical studies and they are now being tested in clinical trials, mainly in combination with conventional treatment regimens. However, some studies revealed that CSCs may not be the only players that control disease relapse and progression of HNC. Further, clinical research studying a combination of therapies targeted against head and neck CSCs may provide significant advances.
Collapse
Affiliation(s)
- Linah A Shahoumi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
28
|
MicroRNA 630 Represses NANOG Expression through Transcriptional and Post-Transcriptional Regulation in Human Embryonal Carcinoma Cells. Int J Mol Sci 2021; 23:ijms23010046. [PMID: 35008480 PMCID: PMC8744645 DOI: 10.3390/ijms23010046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022] Open
Abstract
The pluripotent transcription factor NANOG is essential for maintaining embryonic stem cells and driving tumorigenesis. We previously showed that PKC activity is involved in the regulation of NANOG expression. To explore the possible involvement of microRNAs in regulating the expression of key pluripotency factors, we performed a genome-wide analysis of microRNA expression in the embryonal carcinoma cell line NT2/D1 in the presence of the PKC activator, PMA. We found that MIR630 was significantly upregulated in PMA-treated cells. Experimentally, we showed that transfection of MIR630 mimic into embryonal carcinoma cell lines directly targeted the 3′UTR of OCT4, SOX2, and NANOG and markedly suppressed their expression. RNAhybrid and RNA22 algorithms were used to predict miRNA target sites in the NANOG 3′UTR, four possible target sites of MIR630 were identified. To examine the functional interaction between MIR630 and NANOG mRNA, the predicted MIR630 target sites in the NANOG 3′UTR were deleted and the activity of the reporters were compared. After targeted mutation of the predicted MIR630 target sites, the MIR630 mimic inhibited NANOG significantly less than the wild-type reporters. It is worth noting that mutation of a single putative binding site in the 3′UTR of NANOG did not completely abolish MIR630-mediated suppression, suggesting that MIR630 in the NANOG 3′UTR may have multiple binding sites and act together to maximally repress NANOG expression. Interestingly, MIR630 mimics significantly downregulated NANOG gene transcription. Exogenous expression of OCT4, SOX2, and NANOG lacking the 3′UTR almost completely rescued the reduced transcriptional activity of MIR630. MIR630 mediated the expression of differentiation markers in NT2/D1 cells, suggesting that MIR630 leads to the differentiation of NT2/D1 cell. Our findings show that MIR630 represses NANOG through transcriptional and post-transcriptional regulation, suggesting a direct link between core pluripotency factors and MIR630.
Collapse
|
29
|
Ding Z, Dai C, Shan W, Liu R, Lu W, Gao W, Zhang H, Huang W, Guan J, Yin Z. TNF-α up-regulates Nanog by activating NF-κB pathway to induce primary rat spinal cord astrocytes dedifferentiation. Life Sci 2021; 287:120126. [PMID: 34758295 DOI: 10.1016/j.lfs.2021.120126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/10/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
AIMS Astrocytes re-acquire stem cell potential upon inflammation, thereby becoming a promising source of cells for regenerative medicine. Nanog is an essential transcription factor to maintain the characteristics of stem cells. We aimed to investigate the role of Nanog in astrocyte dedifferentiation. MAIN METHODS TNF-α was used to induce the dedifferentiation of primary rat spinal cord astrocytes. The expression of immature markers CD44 and Musashi-1 was detected by qRT-PCR and immunofluorescence. The Nanog gene is knocked down by small interference RNA. Nanog expression was measured by qRT-PCR and western blotting. BAY 11-7082 was used to suppress NF-κB signals in astrocytes. NF-κB signaling was evaluated by Western blotting. KEY FINDINGS Our results showed that TNF-α promoted the re-expression of CD44 and Musashi-1 in astrocytes. Dedifferentiated astrocytes could be induced to differentiate into oligodendrocyte lineage cells indicating that the astrocytes had pluripotency. In addition, TNF-α treatment activated NF-κB signaling pathway and up-regulated Nanog. Knockdown of Nanog reversed the increase of CD44 and Musashi-1 induced by TNF-α without affecting the activation of NF-κB signaling. Importantly, blocking NF-κB signaling by BAY 11-7082 inhibited the expression of immature markers suggesting that TNF-α induces dedifferentiation of astrocytes through the NF-κB signaling pathway. BAY 11-7082 could also inhibit the expression of Nanog, which indicated that Nanog was regulated by NF-κB signaling pathway. SIGNIFICANCE These findings indicate that activation of the NF-κB signaling pathway through TNF-α leads to astrocytes dedifferentiation via Nanog. These results expand our understanding of the mechanism of astrocytes dedifferentiation.
Collapse
Affiliation(s)
- Zhenfei Ding
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, 287#Chang Huai Road, Bengbu 230071, Anhui, China; Department of Orthopaedics, The Second People's Hospital of Hefei, Intersection of Guangde Road and Leshui Road, Hefei 230011, Anhui, China
| | - Ce Dai
- Department of Orthopaedics, The Second People's Hospital of Hefei, Intersection of Guangde Road and Leshui Road, Hefei 230011, Anhui, China
| | - Wenshan Shan
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Rui Liu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Wei Lu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Weilu Gao
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Hui Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Jianzhong Guan
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, 287#Chang Huai Road, Bengbu 230071, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, 2600#Dong Hai Avenue, Bengbu 233030, Anhui, China.
| | - Zongsheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China.
| |
Collapse
|
30
|
Randhawa V, Kumar M. An integrated network analysis approach to identify potential key genes, transcription factors, and microRNAs regulating human hematopoietic stem cell aging. Mol Omics 2021; 17:967-984. [PMID: 34605522 DOI: 10.1039/d1mo00199j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hematopoietic stem cells (HSCs) undergo functional deterioration with increasing age that causes loss of their self-renewal and regenerative potential. Despite various efforts, significant success in identifying molecular regulators of HSC aging has not been achieved, one prime reason being the non-availability of appropriate human HSC samples. To demonstrate the scope of integrating and re-analyzing the HSC transcriptomics data available, we used existing tools and databases to structure a sequential data analysis pipeline to predict potential candidate genes, transcription factors, and microRNAs simultaneously. This sequential approach comprises (i) collecting matched young and aged mice HSC sample datasets, (ii) identifying differentially expressed genes, (iii) identifying human homologs of differentially expressed genes, (iv) inferring gene co-expression network modules, and (v) inferring the microRNA-transcription factor-gene regulatory network. Systems-level analyses of HSC interaction networks provided various insights based on which several candidates were predicted. For example, 16 HSC aging-related candidate genes were predicted (e.g., CD38, BRCA1, AGTR1, GSTM1, etc.) from GCN analysis. Following this, the shortest path distance-based analyses of the regulatory network predicted several novel candidate miRNAs and TFs. Among these, miR-124-3p was a common regulator in candidate gene modules, while TFs MYC and SP1 were identified to regulate various candidate genes. Based on the regulatory interactions among candidate genes, TFs, and miRNAs, a potential regulation model of biological processes in each of the candidate modules was predicted, which provided systems-level insights into the molecular complexity of each module to regulate HSC aging.
Collapse
Affiliation(s)
- Vinay Randhawa
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific & Industrial Research, Chandigarh-160036, India.
| | - Manoj Kumar
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific & Industrial Research, Chandigarh-160036, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
31
|
Sahoo DP, Van Winkle LJ, Díaz de la Garza RI, Dubrovsky JG. Interkingdom Comparison of Threonine Metabolism for Stem Cell Maintenance in Plants and Animals. Front Cell Dev Biol 2021; 9:672545. [PMID: 34557481 PMCID: PMC8454773 DOI: 10.3389/fcell.2021.672545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/11/2021] [Indexed: 01/12/2023] Open
Abstract
In multicellular organisms, tissue generation, maintenance, and homeostasis depend on stem cells. Cellular metabolic status is an essential component of different differentiated states, from stem to fully differentiated cells. Threonine (Thr) metabolism has emerged as a critical factor required to maintain pluripotent/multipotent stem cells in both plants and animals. Thus, both kingdoms conserved or converged upon this fundamental feature of stem cell function. Here, we examine similarities and differences in Thr metabolism-dependent mechanisms supporting stem cell maintenance in these two kingdoms. We then consider common features of Thr metabolism in stem cell maintenance and predict and speculate that some knowledge about Thr metabolism and its role in stem cell function in one kingdom may apply to the other. Finally, we outline future research directions to explore these hypotheses.
Collapse
Affiliation(s)
- Debee Prasad Sahoo
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Lon J. Van Winkle
- Department of Biochemistry, Midwestern University, Downers Grove, IL, United States
- Department of Medical Humanities, Rocky Vista University, Parker, CO, United States
| | | | - Joseph G. Dubrovsky
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
32
|
Fütterer A, Talavera-Gutiérrez A, Pons T, de Celis J, Gutiérrez J, Domínguez Plaza V, Martínez-A C. Impaired stem cell differentiation and somatic cell reprogramming in DIDO3 mutants with altered RNA processing and increased R-loop levels. Cell Death Dis 2021; 12:637. [PMID: 34155199 PMCID: PMC8217545 DOI: 10.1038/s41419-021-03906-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 02/08/2023]
Abstract
Embryonic stem cell (ESC) differentiation and somatic cell reprogramming are biological processes governed by antagonistic expression or repression of a largely common set of genes. Accurate regulation of gene expression is thus essential for both processes, and alterations in RNA processing are predicted to negatively affect both. We show that truncation of the DIDO gene alters RNA splicing and transcription termination in ESC and mouse embryo fibroblasts (MEF), which affects genes involved in both differentiation and reprogramming. We combined transcriptomic, protein interaction, and cellular studies to identify the underlying molecular mechanism. We found that DIDO3 interacts with the helicase DHX9, which is involved in R-loop processing and transcription termination, and that DIDO3-exon16 deletion increases nuclear R-loop content and causes DNA replication stress. Overall, these defects result in failure of ESC to differentiate and of MEF to be reprogrammed. MEF immortalization restored impaired reprogramming capacity. We conclude that DIDO3 has essential functions in ESC differentiation and somatic cell reprogramming by supporting accurate RNA metabolism, with its exon16-encoded domain playing the main role.
Collapse
Affiliation(s)
- Agnes Fütterer
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), 28049, Madrid, Spain
| | - Amaia Talavera-Gutiérrez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), 28049, Madrid, Spain
| | - Tirso Pons
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), 28049, Madrid, Spain
| | - Jesús de Celis
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), 28049, Madrid, Spain
| | - Julio Gutiérrez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), 28049, Madrid, Spain
| | - Verónica Domínguez Plaza
- Transgenesis Unit, CNB & Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Carlos Martínez-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), 28049, Madrid, Spain.
| |
Collapse
|
33
|
Niu X, Deng K, Liu L, Yang K, Hu X. A statistical framework for predicting critical regions of p53-dependent enhancers. Brief Bioinform 2021; 22:bbaa053. [PMID: 32392580 PMCID: PMC8138796 DOI: 10.1093/bib/bbaa053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
P53 is the 'guardian of the genome' and is responsible for regulating cell cycle and apoptosis. The genomic p53 binding regions, where activating transcriptional factors and cofactors like p300 simultaneously bind, are called 'p53-dependent enhancers', which play an important role in tumorigenesis. Current experimental assays generally provide a broad peak of each enhancer element, leaving our knowledge about critical enhancer regions (CERs) limited. Under the inspiration of enhancer dissection by CRISPR-Cas9 screen library on genome-wide p53 binding sites, here we introduce a statistical framework called 'Computational CRISPR Strategy' (CCS), to predict whether a given DNA fragment will be a p53-dependent CER by employing 7-mer as feature extractions along with random forest as the regressor. When training on a p53 CRISPR enhancer dataset, CCS not only accurately fitted the top-ranked enriched single guide RNAs (sgRNAs) but also successfully reproduced two known CERs that were validated by experiments. When applying it to an independent testing dataset on a tilling of a 2K-b genomic region of CRISPR-deCDKN1A-Lib, the trained model shows great generalizability by identifying a CER containing five top-ranked sgRNAs. A feature importance analysis further indicates that top-ranked 7-mers are mapped onto informative TF motifs including POU5F1 and SOX5, which are differentially enriched in p53-dependent CERs and are potential factors to make a general p53 binding site to form a p53-dependent CER, providing the interpretability of the trained model. Our results demonstrate that CCS is an alternative way of the CRISPR experiment to screen the genome for mapping p53-dependent CERs.
Collapse
Affiliation(s)
| | | | | | | | - Xuehai Hu
- Corresponding author: Xuehai Hu, College of Informatics, Hubei Key Laboratory of Agricultural Bioinformatics, Huazhong Agricultural University, Wuhan, Hubei, 430070, P.R. China. Tel.: +86-18171282783; Fax: +86-27-87288509; E-mail:
| |
Collapse
|
34
|
Worku MG. Pluripotent and Multipotent Stem Cells and Current Therapeutic Applications: Review. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2021; 14:3-7. [PMID: 33880040 PMCID: PMC8052119 DOI: 10.2147/sccaa.s304887] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/29/2021] [Indexed: 12/17/2022]
Abstract
There is numerous evidence for the presence of stem cells, which is important for the treatment of a wide variety of disease conditions. Stem cells have a great therapeutic effect on different degenerative diseases through the development of specialized cells. Embryonic stem (ES) cells are derived from preimplantation embryos, which have a natural karyotype. This cell has the capacity of proliferation indefinitely and undifferentiated. Stem cells are very crucial for the treatment of different chronic and degenerative diseases. For instance, stem cell clinical trials have been done for ischemic heart disease. Also, the olfactory cells for spinal cord lesions and human fetal pancreatic cells for diabetes mellitus are the other clinical importance of stem cell therapy. Extracellular matrix (ECM) and other environmental factors influence the fate and activity of stem cells.
Collapse
Affiliation(s)
- Misganaw Gebrie Worku
- Department of Human Anatomy, University of Gondar, College of Medicine and Health Science, School of Medicine, Gondar, Ethiopia
| |
Collapse
|
35
|
Sevilla A, Papatsenko D, Mazloom AR, Xu H, Vasileva A, Unwin RD, LeRoy G, Chen EY, Garrett-Bakelman FE, Lee DF, Trinite B, Webb RL, Wang Z, Su J, Gingold J, Melnick A, Garcia BA, Whetton AD, MacArthur BD, Ma'ayan A, Lemischka IR. An Esrrb and Nanog Cell Fate Regulatory Module Controlled by Feed Forward Loop Interactions. Front Cell Dev Biol 2021; 9:630067. [PMID: 33816475 PMCID: PMC8017264 DOI: 10.3389/fcell.2021.630067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/17/2021] [Indexed: 01/02/2023] Open
Abstract
Cell fate decisions during development are governed by multi-factorial regulatory mechanisms including chromatin remodeling, DNA methylation, binding of transcription factors to specific loci, RNA transcription and protein synthesis. However, the mechanisms by which such regulatory “dimensions” coordinate cell fate decisions are currently poorly understood. Here we quantified the multi-dimensional molecular changes that occur in mouse embryonic stem cells (mESCs) upon depletion of Estrogen related receptor beta (Esrrb), a key pluripotency regulator. Comparative analyses of expression changes subsequent to depletion of Esrrb or Nanog, indicated that a system of interlocked feed-forward loops involving both factors, plays a central part in regulating the timing of mESC fate decisions. Taken together, our meta-analyses support a hierarchical model in which pluripotency is maintained by an Oct4-Sox2 regulatory module, while the timing of differentiation is regulated by a Nanog-Esrrb module.
Collapse
Affiliation(s)
- Ana Sevilla
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Departament de Biología Cellular, Fisiología i Immunología, Facultat de Biología, Universitat de Barcelona, Barcelona, Spain
| | - Dimitri Papatsenko
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Amin R Mazloom
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Huilei Xu
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ana Vasileva
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Richard D Unwin
- Stem Cell and Leukaemia Proteomics Laboratory, School of Cancer and Enabling Sciences, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom.,Academic Health Science Centre, Wolfson Molecular Imaging Centre, Manchester, United Kingdom.,Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust, Institute of Human Development, Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Gary LeRoy
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Edward Y Chen
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Francine E Garrett-Bakelman
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Dung-Fang Lee
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Benjamin Trinite
- Institut de Recerca de La Sida, IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute, Hospital Universitari Germans Trias I Pujol, Catalonia, Spain
| | - Ryan L Webb
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zichen Wang
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jie Su
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Julian Gingold
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ari Melnick
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Benjamin A Garcia
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Anthony D Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, School of Cancer and Enabling Sciences, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom.,Academic Health Science Centre, Wolfson Molecular Imaging Centre, Manchester, United Kingdom
| | - Ben D MacArthur
- The Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, United Kingdom
| | - Avi Ma'ayan
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ihor R Lemischka
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
36
|
Abstract
In the past several decades, the establishment of in vitro models of pluripotency has ushered in a golden era for developmental and stem cell biology. Research in this arena has led to profound insights into the regulatory features that shape early embryonic development. Nevertheless, an integrative theory of the epigenetic principles that govern the pluripotent nucleus remains elusive. Here, we summarize the epigenetic characteristics that define the pluripotent state. We cover what is currently known about the epigenome of pluripotent stem cells and reflect on the use of embryonic stem cells as an experimental system. In addition, we highlight insights from super-resolution microscopy, which have advanced our understanding of the form and function of chromatin, particularly its role in establishing the characteristically "open chromatin" of pluripotent nuclei. Further, we discuss the rapid improvements in 3C-based methods, which have given us a means to investigate the 3D spatial organization of the pluripotent genome. This has aided the adaptation of prior notions of a "pluripotent molecular circuitry" into a more holistic model, where hotspots of co-interacting domains correspond with the accumulation of pluripotency-associated factors. Finally, we relate these earlier hypotheses to an emerging model of phase separation, which posits that a biophysical mechanism may presuppose the formation of a pluripotent-state-defining transcriptional program.
Collapse
Affiliation(s)
| | - Eran Meshorer
- Department of Genetics, the Institute of Life Sciences
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Jerusalem, Israel 9190400
| |
Collapse
|
37
|
Markers of Stem Cells. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
38
|
Sohn EJ, Moon HJ, Lim JK, Kim DS, Kim JH. Regulation of the protein stability and transcriptional activity of OCT4 in stem cells. Adv Biol Regul 2020; 79:100777. [PMID: 33451972 DOI: 10.1016/j.jbior.2020.100777] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 12/31/2022]
Abstract
OCT4 (also known as Oct3 and Oct3/4), which is encoded by Pou5f1, is expressed in early embryonic cells and plays an important role in early development, pluripotency maintenance, and self-renewal of embryonic stem cells. It also regulates the reprogramming of somatic cells into induced pluripotent stem cells. Several OCT4-binding proteins, including SOX2 and NANOG, reportedly regulate gene transcription in stem cells. An increasing number of evidence suggests that not only gene transcription but also post-translational modifications of OCT4 play a pivotal role in regulating the expression and activity of OCT4. For instance, ubiquitination and sumoylation have been reported to regulate OCT4 protein stability. In addition, the phosphorylation of Ser347 in OCT4 also stabilizes the OCT4 protein level. Recently, we identified KAP1 as an OCT4-binding protein and reported the KAP1-mediated regulation of OCT4 protein stability. KAP1 overexpression led to an increased proliferation of mouse embryonic stem cells and promoted the reprogramming of somatic cells resulting in induced pluripotent stem cells. In this review, we discuss how the protein stability and function of OCT4 are regulated by protein-protein interaction in stem cells.
Collapse
Affiliation(s)
- Eun Jung Sohn
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, 50612, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hye Ji Moon
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, 50612, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Jae Kyong Lim
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, 50612, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Da Sol Kim
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, 50612, Republic of Korea; Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Jae Ho Kim
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
39
|
Yaglova N, Obernikhin S, Nazimova S, Yaglov V. Developmental exposure to endocrine disrupter dichlorodiphenyltrichloroethane alters transcriptional regulation of postnatal morphogenesis of adrenal zona fasciculata. Saudi J Biol Sci 2020; 27:3655-3659. [PMID: 33304177 PMCID: PMC7714961 DOI: 10.1016/j.sjbs.2020.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022] Open
Abstract
The present study is aimed to validate expression of transcriptional factors mediating postnatal development of adrenal zona fasciculata in rats exposed to low doses of endocrine disrupter dichlorodiphenyltrichloroethane prenatally and postnatally. Histological and immunohistochemical examination of the adrenals was performed. Impaired blood circulation, dystrophy and cell death were found in zona fasciculata of pubertal rats after developmental exposure to low doses of dichlorodiphenyltrichloroethane. Reparation of zona fasciculata was associated with increased number of Sonic hedgehog- and Oct4-expressing adrenal cortical cells but not in areas of regeneration. These data suggest that cell death may promote upregulation of factors inducing and maintaining pluripotent state in fasciculata cells for restoration of tissue homeostasis. Termination of growth of the adrenals after puberty was associated with upregulation of antiproliferative factor Hhex and decrease of cell proliferation. Dichlorodiphenyltrichloroethane exposure disrupted transcriptional control of cell proliferation by downregulation of Hhex expression in fasciculata cells. Decrease of proliferation in the exposed rats was mediated by inhibition of Sonic hedgehog and Oct4 expression and suppression of canonical Wnt signaling. The present study elucidated an alternative mechanism of proliferation control activated by endocrine disrupter dichlorodiphenyltrichloroethane through transition of fasciculata cells from pluripotent state and higher proliferative potential to differentiation. Activation of the alternative mechanism of growth control may probably affect maintenance of tissue homeostasis of zona fasciculata in postnatal development.
Collapse
Affiliation(s)
- Nataliya Yaglova
- Laboratory of Endocrine System Development, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Sergey Obernikhin
- Laboratory of Endocrine System Development, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Svetlana Nazimova
- Laboratory of Endocrine System Development, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Valentin Yaglov
- Laboratory of Endocrine System Development, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| |
Collapse
|
40
|
Ahn YT, Kim MS, Kim YS, An WG. Astaxanthin Reduces Stemness Markers in BT20 and T47D Breast Cancer Stem Cells by Inhibiting Expression of Pontin and Mutant p53. Mar Drugs 2020; 18:md18110577. [PMID: 33233699 PMCID: PMC7699712 DOI: 10.3390/md18110577] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/28/2022] Open
Abstract
Astaxanthin (AST) is a product made from marine organisms that has been used as an anti-cancer supplement. It reduces pontin expression and induces apoptosis in SKBR3, a breast cancer cell line. Using Western blotting and qRT-PCR analyses, this study revealed that in the T47D and BT20 breast cancer cell lines, AST inhibits expression of pontin and mutp53, as well as the Oct4 and Nanog cancer stem cell (CSC) stemness genes. In addition, we explored the mechanism by which AST eradicates breast cancer cells using pontin siRNAs. Pontin knockdown by pontin siRNA reduced proliferation, Oct4 and Nanog expression, colony and spheroid formation, and migration and invasion abilities in breast cancer cells. In addition, reductions in Oct4, Nanog, and mutp53 expression following rottlerin treatment confirmed the role of pontin in these cells. Therefore, pontin may play a central role in the regulation of CSC properties and in cell proliferation following AST treatment. Taken together, these findings demonstrate that AST can repress CSC stemness genes in breast cancer cells, which implies that AST therapy could be used to improve the efficacy of other anti-cancer therapies against breast cancer cells.
Collapse
Affiliation(s)
- Yong Tae Ahn
- Research Institute for Longevity and Well-Being, Pusan National University, Busan 46241, Korea;
| | - Min Sung Kim
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea;
| | - Youn Sook Kim
- Gene & Cell Therapy Research Center for Vessel-Associated Diseases, Pusan National University, Yangsan 50612, Korea;
| | - Won Gun An
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea;
- Correspondence: ; Tel.: +82-51-510-8455
| |
Collapse
|
41
|
Kudryavtsev GY, Kudryavtseva LV, Mikhaleva LM, Kudryavtseva Y, Solovyeva NA, Osipov VA, Babichenko II. Immunohistochemical expression of Nanog protein in prostate cancer cells of distinct grade groups. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2020. [DOI: 10.24075/brsmu.2020.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prostate cancer is the most common type of cancer among men, which is mainly due to extensive use of screening tests and high total number of prostate biopsies. Verification of tumors with poorer prognosis is the primary goal of prostate cancer management. The study was aimed to determine the clinical and morphological associations and the prognostic value of the Nanog protein expression in prostate cancer of distinct Grade Groups. We used the prostate tissue specimens obtained during surgery, and the biopsy specimens, the total of 89 cases. Histological and immunohistochemical assessment was performed using antibodies to Ki-67 and Nanog. Correlations between the expression of markers and the Grade Groups were revealed using the Spearman's rank correlation coefficient, and the correlation with clinical and morphological characteristics was determined using the chi-squared test (χ2). There was a positive correlation between the expression of Ki-67 and Nanog, and the Grade Group numerical order (rs = 0.619, p < 0.001 and rs = 0.786, p < 0.001 respectively). We managed to find the relationship between the high Nanog expression and the extraprostatic extension (p = 0.041). High expression of Nanog protein in the prostate cancer cells was associated with a higher-grade adenocarcinoma and indicated a poor prognosis.
Collapse
Affiliation(s)
- GYu Kudryavtsev
- Peoples' Friendship University of Russia, Moscow, Russia; Hospital for War Veterans № 2, Moscow, Russia
| | | | - LM Mikhaleva
- Research Institute of Human Morphology, Moscow, Russia
| | | | - NA Solovyeva
- Peoples' Friendship University of Russia, Moscow, Russia
| | - VA Osipov
- Hospital for War Veterans # 2, Moscow, Russia
| | - II Babichenko
- Peoples' Friendship University of Russia, Moscow, Russia
| |
Collapse
|
42
|
Zargari S, Negahban Khameneh S, Rad A, Forghanifard MM. MEIS1 promotes expression of stem cell markers in esophageal squamous cell carcinoma. BMC Cancer 2020; 20:789. [PMID: 32819319 PMCID: PMC7441725 DOI: 10.1186/s12885-020-07307-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND MEIS1 (Myeloid ecotropic viral integration site 1) as a homeobox (HOX) transcription factor plays regulatory roles in a variety of cellular processes including development, differentiation, survival, apoptosis and hematopoiesis, as well as stem cell regulation. Few studies have established pluripotency and self-renewal regulatory roles for MEIS1 in human esophageal squamous cell carcinoma (ESCC), and our aim in this study was to evaluate the functional correlation between MEIS1 and the stemness markers in ESCC patients and cell line KYSE-30. METHODS Expression pattern of MEIS1 and SALL4 gene expression was analyzed in different pathological features of ESCC patients. shRNA in retroviral vector was used for constantly silencing of MEIS1 mRNA in ESCC line (KYSE-30). Knockdown of MEIS1 gene and the expression pattern of selected stemness markers including SALL4, OCT4, BMI-1, HIWI, NANOG, PLK1, and KLF4 were evaluated using real-time PCR. RESULTS Significant correlations were observed between MEIS1 and stemness marker SALL4 in different early pathological features of ESCC including non-invaded tumors, and the tumors with primary stages of progression. Retroviral knockdown of MEIS1 in KYSE-30 cells caused a noteworthy underexpression of both MEIS1 and major involved markers in stemness state of the cells including SALL4, OCT4, BMI-1, HIWI and KLF4. CONCLUSIONS The results highlight the important potential role of MEIS1 in modulating stemness properties of ESCCs and cells KYSE-30. These findings may confirm the linkage between MEIS1 and self-renewal capacity in ESCC and support probable oncogenic role for MEIS1 in the disease.
Collapse
Affiliation(s)
- Selma Zargari
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shabnam Negahban Khameneh
- Department of Biology, Damghan branch, Islamic Azad University, P.O.Box: 3671639998, Cheshmeh-Ali Boulevard, Sa'dei Square, Damghan, Islamic Republic of Iran
| | - Abolfazl Rad
- Cellular and Molecular Research center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Mohammad Mahdi Forghanifard
- Department of Biology, Damghan branch, Islamic Azad University, P.O.Box: 3671639998, Cheshmeh-Ali Boulevard, Sa'dei Square, Damghan, Islamic Republic of Iran.
| |
Collapse
|
43
|
Alba G, Martínez R, Postigo-Corrales F, López S, Santa-María C, Jiménez J, Cahuana GM, Soria B, Bedoya FJ, Tejedo JR. AICAR Stimulates the Pluripotency Transcriptional Complex in Embryonic Stem Cells Mediated by PI3K, GSK3β, and β-Catenin. ACS OMEGA 2020; 5:20270-20282. [PMID: 32832780 PMCID: PMC7439381 DOI: 10.1021/acsomega.0c02137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/23/2020] [Indexed: 05/03/2023]
Abstract
Pluripotent stem cells maintain the property of self-renewal and differentiate into all cell types under clear environments. Though the gene regulatory mechanism for pluripotency has been investigated in recent years, it is still not completely understood. Here, we show several signaling pathways involved in the maintenance of pluripotency. To investigate whether AMPK is involved in maintaining the pluripotency in mouse embryonic stem cells (mESCs) and elucidating the possible molecular mechanisms, implicated D3 and R1/E mESC lines were used in this study. Cells were cultured in the absence or presence of LIF and treated with 1 mM and 0.5 mM 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR), 2 mM metformin, compound C, and the PI3K inhibitor LY294002 for 24, 72, and 120 h. The levels of Nanog, Oct3/4, and REX1 and Brachyury, Notch2, and Gata4 mRNAs and Nanog or OCT3/4 protein levels were analyzed. Alkaline phosphatase and the cellular cycle were determined. The pGSK3β, GSK3β, p-β-catenin, and β-catenin protein levels were also investigated. We found that AMPK activators such as AICAR and metformin increase mRNA expression of pluripotency markers and decrease mRNA expression of differentiation markers in R1/E and D3 ES cells. AICAR increases phosphatase activity and arrests the cellular cycle in the G1 phase in these cells. We describe that AICAR effects were mediated by AMPK activation using a chemical inhibitor or by silencing this gene. AICAR effects were also mediated by PI3K, GSK3β, and β-catenin in R1/E ES cells. According to our findings, we provide a mechanism by which AICAR increases and maintains a pluripotency state through enhanced Nanog expression, involving AMPK/PI3K and p-GSK3β Ser21/9 pathways backing up the AICAR function as a potential target for this drug controlling pluripotency. The highlights of this study are that AICAR (5-aminoimidazole-4-carboxamied-1-b-riboside), an AMP protein kinase (AMPK) activator, blocks the ESC differentiation and AMPK is a key enzyme for pluripotency and shows valuable data to clarify the molecular pluripotency mechanism.
Collapse
Affiliation(s)
- Gonzalo Alba
- Department
of Medical Biochemistry and Molecular Biology, Universidad de Sevilla, Seville 41009, Spain
- . Telephone: +34-955421044. Fax: +34-954907048
| | - Raquel Martínez
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
| | - Fátima Postigo-Corrales
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
| | - Soledad López
- Department
of Medical Biochemistry and Molecular Biology, Universidad de Sevilla, Seville 41009, Spain
| | - Consuelo Santa-María
- Department
of Biochemistry and Molecular Biology, Universidad
de Sevilla, Seville 41009, Spain
| | - Juan Jiménez
- Department
of Medical Biochemistry and Molecular Biology, Universidad de Sevilla, Seville 41009, Spain
| | - Gladys M. Cahuana
- Department
of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville 41013, Spain
- Biomedical
Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM,
Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Bernat Soria
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
- Biomedical
Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM,
Instituto de Salud Carlos III, Madrid 28029, Spain
- Cell
Therapy
Network, Madrid (RED-TERCEL), Instituto
de Salud Carlos III, Madrid 28029, Spain
- Universidad
Miguel Hernández, Alicante 03550, Spain
| | - Francisco J. Bedoya
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
- Department
of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville 41013, Spain
- Biomedical
Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM,
Instituto de Salud Carlos III, Madrid 28029, Spain
- Cell
Therapy
Network, Madrid (RED-TERCEL), Instituto
de Salud Carlos III, Madrid 28029, Spain
| | - Juan R. Tejedo
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
- Department
of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville 41013, Spain
- Biomedical
Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM,
Instituto de Salud Carlos III, Madrid 28029, Spain
- Cell
Therapy
Network, Madrid (RED-TERCEL), Instituto
de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
44
|
Balzano F, Garroni G, Cruciani S, Bellu E, Dei Giudici S, Oggiano A, Capobianco G, Dessole S, Ventura C, Maioli M. Behavioral Changes in Stem-Cell Potency by HepG2-Exhausted Medium. Cells 2020; 9:1890. [PMID: 32806709 PMCID: PMC7547384 DOI: 10.3390/cells9081890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/05/2020] [Accepted: 08/09/2020] [Indexed: 12/16/2022] Open
Abstract
Wharton jelly mesenchymal stem cells (WJ-MSCs) are able to differentiate into different cell lineages upon stimulation. This ability is closely related to the perfect balance between the pluripotency-related genes, which control stem-cell proliferation, and genes able to orchestrate the appearance of a specific phenotype. Here we studied the expression of stemness-related genes, epigenetic regulators (DNMT1, SIRT1), miRNAs (miR-145, miR-148, and miR-185) related to stemness, exosomes, the cell-cycle regulators p21 (WAF1/CIP1) and p53, and the senescence-associated genes (p16, p19, and hTERT). Cells were cultured in the presence or absence of the human hepatocarcinoma cell line HepG2-exhausted medium, to evaluate changes in stemness, differentiation capability, and senescence sensibility. Our results showed the overexpression of SIRT1 and reduced levels of p21 mRNA. Moreover, we observed a downregulation of DNMT1, and a simultaneous overexpression of Oct-4 and c-Myc. These findings suggest that WJ-MSCs are more likely to retain a stem phenotype and sometimes to switch to a highly undifferentiable proliferative-like behavior if treated with medium exhausted by human HepG2 cell lines.
Collapse
Affiliation(s)
- Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.B.); (G.G.); (S.C.); (E.B.)
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.B.); (G.G.); (S.C.); (E.B.)
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.B.); (G.G.); (S.C.); (E.B.)
| | - Emanuela Bellu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.B.); (G.G.); (S.C.); (E.B.)
| | - Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100 Sassari, Italy; (S.D.G.); (A.O.)
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100 Sassari, Italy; (S.D.G.); (A.O.)
| | - Giampiero Capobianco
- Department of Medical, Surgical and experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, 07100 Sassari, Italy; (G.C.); (S.D.)
| | - Salvatore Dessole
- Department of Medical, Surgical and experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, 07100 Sassari, Italy; (G.C.); (S.D.)
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)-Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy;
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (F.B.); (G.G.); (S.C.); (E.B.)
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, 09042 Cagliari, Italy
- Center for developmental biology and reprogramming-CEDEBIOR, Department of Biomedical Sciences, University of Sassari Viale San Pietro 43/B, 07100 Sassari, Italy
| |
Collapse
|
45
|
Nieto-Nicolau N, de la Torre RM, Fariñas O, Savio A, Vilarrodona A, Casaroli-Marano RP. Extrinsic modulation of integrin α6 and progenitor cell behavior in mesenchymal stem cells. Stem Cell Res 2020; 47:101899. [PMID: 32659733 DOI: 10.1016/j.scr.2020.101899] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/11/2020] [Accepted: 06/24/2020] [Indexed: 01/07/2023] Open
Abstract
Mesenchymal stem cells (MSC) are heterogeneous cells of complex nature that show different potentials while different culture conditions can modify their functionalities through interactions with the microenviroment. Here, we found that bone marrow (BM) MSC from different donor sources and passages that expressed higher levels of α6 integrin subunit (ITGA6), showed higher clonogenicity, migration and differentiation potential. ITGA6 showed important roles improving these potentials and regulating proliferation through protein kinase B (AKT) pathway and cell cycle inhibitor proteins p53 and p21. Moreover, ITGA6 downregulation impaired migration. Cell confluence regulated ITGA6, increasing its expression in low density cultures and decreasing in high density cultures. Besides, ITGA6- cells expressed ITGA6 when seeded at low densities. We found higher ITGA6 expression on fibronectin substrates at lower confluency. Fibronectin increased proliferation, clonogenicity, activation of AKT, decreased cell cycle inhibitor proteins and augmented growth factors expression. Spheres-derived MSC showed higher ITGA6 expression and enhanced potentials for migration, clonogenicity and proliferation. In conclusion, though there is an intrinsic regulation of ITGA6 expression, associated to the progenitor potential of BM-MSC, this expression is regulated by culture conditions and is translated in changes in cell behavior and proliferation. This knowledge could be used to enhance the potential of BM-MSC for clinical application.
Collapse
Affiliation(s)
- Nuria Nieto-Nicolau
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; CellTec-UB, University of Barcelona, Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | | | - Oscar Fariñas
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Andrés Savio
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Anna Vilarrodona
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Ricardo P Casaroli-Marano
- Barcelona Tissue Bank (BTB) & Donor Center, Banc de Sang i Teixits (BST), Barcelona, Spain; CellTec-UB, University of Barcelona, Barcelona, Spain; Department of Surgery, School of Medicine & Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain; Institute of Biomedical Research Sant Pau (IIB-Sant Pau), Barcelona, Spain.
| |
Collapse
|
46
|
Duan Y, Li X, Zhang S, Wang S, Wang T, Chen H, Yang Y, Jia S, Chen G, Tian W. Therapeutic potential of HERS spheroids in tooth regeneration. Theranostics 2020; 10:7409-7421. [PMID: 32642002 PMCID: PMC7330840 DOI: 10.7150/thno.44782] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/25/2020] [Indexed: 02/05/2023] Open
Abstract
Hertwig's epithelial root sheath (HERS) plays indispensable roles in tooth root development, including controlling the shape and number of roots, dentin formation, and helping generate the cementum. Based on these characteristics, HERS cell is a potential seed cell type for tooth-related tissue regeneration. However, the application is severely limited by a lack of appropriate culture methods and small cell numbers. Methods: Here, we constructed a 3D culture method to expand functional HERS cells into spheroids, and investigated characteristics and application of dental tissue regeneration of these spheroids. HERS spheroids and HERS cells (2D monolayer culture) were compared in terms of biological characteristics (such as proliferation, self-renewal capacity, and stemness) in vitro and functions (including differentiation potential and inductive ability of dentin formation) both in vitro and in vivo. Further, transcriptome analysis was utilized to reveal the molecular mechanisms of their obvious differences. Results: HERS spheroids showed obvious superiority in biological characteristics and functions compared to 2D monolayers of HERS cells in vitro. In vivo, HERS spheroids generated more mineralized tissue; when combined with dental papilla cells (DPCs), HERS spheroids contributed to dentin-like tissue formation. Moreover, the generation and expansion of HERS spheroids rely to some degree on the HIF-1 pathway. Conclusion: HERS spheroid generation is beneficial for functional HERS cell expansion and can provide a useful cell source for further tooth regeneration and mechanistic research. Notably, HIF-1 pathway plays a critical role in HERS spheroid formation and function.
Collapse
Affiliation(s)
- Yufeng Duan
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuebing Li
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sicheng Zhang
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shikai Wang
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tao Wang
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong Chen
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Yang
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sixun Jia
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guoqing Chen
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Weidong Tian
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
Jing R, Guo X, Yang Y, Chen W, Kang J, Zhu S. Long noncoding RNA Q associates with Sox2 and is involved in the maintenance of pluripotency in mouse embryonic stem cells. Stem Cells 2020; 38:834-848. [PMID: 32277787 DOI: 10.1002/stem.3180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/11/2020] [Accepted: 03/17/2020] [Indexed: 11/11/2022]
Abstract
Large intergenic noncoding RNAs (lincRNAs) in ESCs may play an important role in the maintenance of pluripotency. The identification of stem cell-specific lincRNAs and their interacting partners will deepen our understanding of the maintenance of stem cell pluripotency. We identified a lincRNA, LincQ, which is specifically expressed in ESCs and is regulated by core pluripotent transcription factors. It was rapidly downregulated during the differentiation process. Knockdown of LincQ in ESCs led to differentiation, downregulation of pluripotency-related genes, and upregulation of differentiation-related genes. We found that exon 1 of LincQ can specifically bind to Sox2. The Soxp region in Sox2, rather than the high mobility group domain, is responsible for LincQ binding. Importantly, the interaction between LincQ and Sox2 is required for the maintenance of pluripotency in ESCs and the transcription of pluripotency genes. Esrrb and Tfcp2l1 are key downstream targets of LincQ and Sox2, since overexpression of Esrrb and Tfcp2l1 can restore the loss of ESC pluripotency that is induced by LincQ depletion. In summary, we found that LincQ specifically interacts with Sox2 and contributes to the maintenance of pluripotency, highlighting the critical role of lincRNA in the pluripotency regulatory network.
Collapse
Affiliation(s)
- Ruiqi Jing
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China.,Institute of Biophysics, Chinese Academy of Sciences, Beijing, People's Republic of China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xudong Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China.,Institute for Advanced Study, Tongji University, Shanghai, People's Republic of China
| | - Yiwei Yang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Wen Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Jiuhong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, People's Republic of China
| | - Songcheng Zhu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
48
|
Dash S, Aydin Y, Widmer KE, Nayak L. Hepatocellular Carcinoma Mechanisms Associated with Chronic HCV Infection and the Impact of Direct-Acting Antiviral Treatment. J Hepatocell Carcinoma 2020; 7:45-76. [PMID: 32346535 PMCID: PMC7167284 DOI: 10.2147/jhc.s221187] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatitis C virus (HCV) infection is the major risk factor for liver cirrhosis and hepatocellular carcinoma (HCC). The mechanisms of HCC initiation, growth, and metastasis appear to be highly complex due to the decade-long interactions between the virus, immune system, and overlapping bystander effects of host metabolic liver disease. The lack of a readily accessible animal model system for HCV is a significant obstacle to understand the mechanisms of viral carcinogenesis. Traditionally, the primary prevention strategy of HCC has been to eliminate infection by antiviral therapy. The success of virus elimination by antiviral treatment is determined by the SVR when the HCV is no longer detectable in serum. Interferon-alpha (IFN-α) and its analogs, pegylated IFN-α (PEG-IFN-α) alone with ribavirin (RBV), have been the primary antiviral treatment of HCV for many years with a low cure rate. The cloning and sequencing of HCV have allowed the development of cell culture models, which accelerated antiviral drug discovery. It resulted in the selection of highly effective direct-acting antiviral (DAA)-based combination therapy that now offers incredible success in curing HCV infection in more than 95% of all patients, including those with cirrhosis. However, several emerging recent publications claim that patients who have liver cirrhosis at the time of DAAs treatment face the risk of HCC occurrence and recurrence after viral cure. This remains a substantial challenge while addressing the long-term benefit of antiviral medicine. The host-related mechanisms that drive the risk of HCC in the absence of the virus are unknown. This review describes the multifaceted mechanisms that create a tumorigenic environment during chronic HCV infection. In addition to the potential oncogenic programming that drives HCC after viral clearance by DAAs, the current status of a biomarker development for early prediction of cirrhosis regression and HCC detection post viral treatment is discussed. Since DAAs treatment does not provide full protection against reinfection or viral transmission to other individuals, the recent studies for a vaccine development are also reviewed.
Collapse
Affiliation(s)
- Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA70119, USA
- Department of Medicine, Division of Gastroenterology, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - Yucel Aydin
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - Kyle E Widmer
- Southeast Louisiana Veterans Health Care System, New Orleans, LA70119, USA
| | - Leela Nayak
- Southeast Louisiana Veterans Health Care System, New Orleans, LA70119, USA
| |
Collapse
|
49
|
Rak Kwon D, Jung S, Jang J, Park GY, Suk Moon Y, Lee SC. A 3-Dimensional Bioprinted Scaffold With Human Umbilical Cord Blood-Mesenchymal Stem Cells Improves Regeneration of Chronic Full-Thickness Rotator Cuff Tear in a Rabbit Model. Am J Sports Med 2020; 48:947-958. [PMID: 32167836 DOI: 10.1177/0363546520904022] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Chronic full-thickness rotator cuff tears (FTRCTs) represent a major clinical concern because they show highly compromised healing capacity. PURPOSE To evaluate the efficacy of using a 3-dimensional (3D) bioprinted scaffold with human umbilical cord blood (hUCB)-mesenchymal stem cells (MSCs) for regeneration of chronic FTRCTs in a rabbit model. STUDY DESIGN Controlled laboratory study. METHODS A total of 32 rabbits were randomly assigned to 4 treatment groups (n = 8 per group) at 6 weeks after a 5-mm FTRCT was created on the supraspinatus tendon. Group 1 (G1-SAL) was transplanted with normal saline. Group 2 (G2-MSC) was transplanted with hUCB-MSCs (0.2 mL, 1 × 106) into FTRCTs. Group 3 (G3-3D) was transplanted with a 3D bioprinted construct without MSCs, and group 4 (G4-3D+MSC) was transplanted with a 3D bioprinted construct containing hUCB-MSCs (0.2 mL, 1 × 106 cells) into FTRCTs. All 32 rabbits were euthanized at 4 weeks after treatment. Examination of gross morphologic changes and histologic results was performed on all rabbits after sacrifice. Motion analysis was also performed before and after treatment. RESULTS In G4-3D+MSC, newly regenerated collagen type 1 fibers, walking distance, fast walking time, and mean walking speed were greater than those in G2-MSC based on histochemical and motion analyses. In addition, when compared with G3-3D, G4-3D+MSC showed more prominent regenerated tendon fibers and better parameters of motion analysis. However, there was no significant difference in gross tear size among G2-MSC, G3-3D, and G4-3D+MSC, although these groups showed significant decreases in tear size as compared with the control group (G1-SAL). CONCLUSION Findings of this study show that a tissue engineering strategy based on a 3D bioprinted scaffold filled with hUCB-MSCs can improve the microenvironment for regenerative processes of FTRCT without any surgical repair. CLINICAL RELEVANCE In the case of rotator cuff tear, the cell loss of the external MSCs can be increased by exposure to synovial fluid. Therefore, a 3D bioprinted scaffold in combination with MSCs without surgical repair may be effective in increasing cell retention in FTRCT.
Collapse
Affiliation(s)
- Dong Rak Kwon
- Department of Rehabilitation Medicine, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Seungman Jung
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea.,Department of Creative IT Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea.,Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Gi-Young Park
- Department of Rehabilitation Medicine, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Yong Suk Moon
- Department of Anatomy, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Sang Chul Lee
- Department and Research Institute of Rehabilitation Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
50
|
Cherubini A, Barilani M, Rossi RL, Jalal M, Rusconi F, Buono G, Ragni E, Cantarella G, Simpson H, Péault B, Lazzari L. FOXP1 circular RNA sustains mesenchymal stem cell identity via microRNA inhibition. Nucleic Acids Res 2019; 47:5325-5340. [PMID: 30937446 PMCID: PMC6547427 DOI: 10.1093/nar/gkz199] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/11/2019] [Accepted: 03/18/2019] [Indexed: 12/31/2022] Open
Abstract
Stem cell identity and plasticity are controlled by master regulatory genes and complex circuits also involving non-coding RNAs. Circular RNAs (circRNAs) are a class of RNAs generated from protein-coding genes by backsplicing, resulting in stable RNA structures devoid of free 5’ and 3’ ends. Little is known of the mechanisms of action of circRNAs, let alone in stem cell biology. In this study, for the first time, we determined that a circRNA controls mesenchymal stem cell (MSC) identity and differentiation. High-throughput MSC expression profiling from different tissues revealed a large number of expressed circRNAs. Among those, circFOXP1 was enriched in MSCs compared to differentiated mesodermal derivatives. Silencing of circFOXP1 dramatically impaired MSC differentiation in culture and in vivo. Furthermore, we demonstrated a direct interaction between circFOXP1 and miR-17–3p/miR-127–5p, which results in the modulation of non-canonical Wnt and EGFR pathways. Finally, we addressed the interplay between canonical and non-canonical Wnt pathways. Reprogramming to pluripotency of MSCs reduced circFOXP1 and non-canonical Wnt, whereas canonical Wnt was boosted. The opposing effect was observed during generation of MSCs from human pluripotent stem cells. Our results provide unprecedented evidence for a regulatory role for circFOXP1 as a gatekeeper of pivotal stem cell molecular networks.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Mario Barilani
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, 20122, Italy
| | - Riccardo L Rossi
- Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, 20122 Italy
| | - Murtadhah M K Jalal
- Department of Orthopaedic Surgery, The Royal Infirmary of Edinburgh, Edinburgh, EH16 4SA, UK
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Francesco Rusconi
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Giuseppe Buono
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Enrico Ragni
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Giovanna Cantarella
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, 20122, Italy
- Department of Otolaryngology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Hamish A R W Simpson
- Department of Orthopaedic Surgery, The Royal Infirmary of Edinburgh, Edinburgh, EH16 4SA, UK
| | - Bruno Péault
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, EH16 4UU, UK
- Orthopaedic Hospital Research Centre, David Geffen School of Medicine, University of California at Los Angeles, California, 90095, USA
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
- To whom correspondence should be addressed. Tel: +39 0255034053;
| |
Collapse
|