1
|
Higuchi Y, Teo JL, Yi D, Kahn M. Safely Targeting Cancer, the Wound That Never Heals, Utilizing CBP/Beta-Catenin Antagonists. Cancers (Basel) 2025; 17:1503. [PMID: 40361430 PMCID: PMC12071182 DOI: 10.3390/cancers17091503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Stem cells, both normal somatic (SSC) and cancer stem cells (CSC) exist in minimally two states, i.e., quiescent and activated. Regulation of these two states, including their reliance on different metabolic processes, i.e., FAO and glycolysis in quiescent versus activated stem cells respectively, involves the analysis of a complex array of factors (nutrient and oxygen levels, adhesion molecules, cytokines, etc.) to initiate the epigenetic changes to either depart or enter quiescence. Quiescence is a critical feature of SSC that is required to maintain the genomic integrity of the stem cell pool, particularly in long lived complex organisms. Quiescence in CSC, whether they are derived from mutations arising in SSC, aberrant microenvironmental regulation, or via dedifferentiation of more committed progenitors, is a critical component of therapy resistance and disease latency and relapse. At the beginning of vertebrate evolution, approximately 450 million years ago, a gene duplication generated the two members of the Kat3 family, CREBBP (CBP) and EP300 (p300). Despite their very high degree of homology, these two Kat3 coactivators play critical and non-redundant roles at enhancers and super-enhancers via acetylation of H3K27, thereby controlling stem cell quiescence versus activation and the cells metabolic requirements. In this review/perspective, we discuss the unique regulatory roles of CBP and p300 and how specifically targeting the CBP/β-catenin interaction utilizing small molecule antagonists, can correct lineage infidelity and safely eliminate quiescent CSC.
Collapse
Affiliation(s)
- Yusuke Higuchi
- Beckman Research Institute, City of Hope, Duarte, CA 91010, USA;
| | - Jia-Ling Teo
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (J.-L.T.); (D.Y.)
| | - Daniel Yi
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (J.-L.T.); (D.Y.)
| | - Michael Kahn
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA; (J.-L.T.); (D.Y.)
| |
Collapse
|
2
|
Shen X, Yan H, Hu M, Zhou H, Zhang Q, Gao R, Liu Q, Sun Q. A detailed transcriptome study uncovers the epigenetic characteristics associated with Aromatase inhibitor-induced masculinization in Takifugu rubripes larvae gonads. BMC Genomics 2025; 26:380. [PMID: 40240894 PMCID: PMC12001447 DOI: 10.1186/s12864-025-11375-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/17/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Takifugu rubripes is an economically valuable fish species in Asia. The implementation of all-male culture for T. rubripes is highly anticipated in aquaculture. Aromatase inhibitor (AI, letrozole) treatment was found to be an efficient method to induced masculinization in T. rubripes, as reported in our previous study. Here, to further explore the underlying regulation mechanism of AI-induced masculinization, a whole-transcriptome analysis comparing was conducted between AI-induced masculinized XX (AI-XX) gonads and control (Con) gonads in T. rubripes. RESULTS In Con-XX/Con-XY comparison, 1,172 differential expression (DE) mRNAs, 129 DEmiRNAs, 210 DElncRNAs, and 4 DEcircRNAs were identified. In the Con-XX/AI-XX comparison, 1,329 DEmRNAs, 174 DEmiRNAs, 6 DEcircRNAs and 280 DElncRNAs were found. Con-XX/Con-XY and Con-XX/AI-XX comparisons shared 690 DEmRNAs, 50 DEmiRNAs, 3 DEcircRNAs, and 105 DElncRNAs. The analyses of protein-protein interaction (PPI) and competitive endogenous RNA (ceRNA) network identified interactions among these shared DERNAs. Kcnh2b, trim27, cnnm2b, reln, cckbra, pkd1l2, steap4, gsg1l, hamp, and foxg1c were predicted as the top ten of hub genes. miRNAs included miRNA-27 family and miRNA-489 family showed targeting relationship with hub genes. GO and KEGG functional enrichment analysis showed that the targeted genes were mainly enriched in GO:0065008 regulation of biological quality and TGF-beta signaling pathway. qPCR validation confirmed the differential expression of selected mRNAs, and ncRNAs. CONCLUSIONS This research comprehensively reveals the potential regulatory effects of ncRNAs on cellular motility, fate regulation, and hormonal regulation during gonadal masculinization in T. rubripes. It may provide significant insights into the regulation mechanisms underlying sex reversal in fish.
Collapse
Affiliation(s)
- Xufang Shen
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, Liaoning, 116023, China
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University), Ministry of Education, Dalian, 116023, China
| | - Hongwei Yan
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, Liaoning, 116023, China.
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University), Ministry of Education, Dalian, 116023, China.
| | - Mingtao Hu
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, Liaoning, 116023, China
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University), Ministry of Education, Dalian, 116023, China
| | - Huiting Zhou
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, Liaoning, 116023, China
| | - Qi Zhang
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, Liaoning, 116023, China
| | - Rui Gao
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University), Ministry of Education, Dalian, 116023, China
- College of Marine Science and Environment Engineering, Dalian Ocean University, Dalian, Liaoning, 116023, China
| | - Qi Liu
- Key Laboratory of Environment Controlled Aquaculture (Dalian Ocean University), Ministry of Education, Dalian, 116023, China
- College of Marine Science and Environment Engineering, Dalian Ocean University, Dalian, Liaoning, 116023, China
| | - Qunwen Sun
- Dalian Tian Zheng Co., Ltd, Dalian, Liaoning, China
| |
Collapse
|
3
|
Hao W, Luo Y, Tian J, Lu Y, Cui Y, Zhang Y, Jin X, Ye H, Lu M, Song J, Zhou W, Zhang W, He Z. Scale-Up of Human Amniotic Epithelial Cells Through Regulation of Epithelial-Mesenchymal Plasticity Under Defined Conditions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408581. [PMID: 39804851 PMCID: PMC11923953 DOI: 10.1002/advs.202408581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/12/2024] [Indexed: 01/16/2025]
Abstract
Human amniotic epithelial cells (hAECs) have shown excellent efficacy in clinical research and have prospective applications in the treatment of many diseases. However, the properties of the hAECs and their proliferative mechanisms remain unclear. Here, single-cell RNA sequencing (scRNA-seq) is performed on hAECs obtained from amniotic tissues at different gestational ages and passages during in vitro culture. The results showed that the proliferation of hAECs is associated with epithelial-mesenchymal plasticity (EMP) during amniogenesis. Freshly isolated, full-term hAECs are identified as mature epithelial cells. Once cultured in vitro, they are observed to rapidly undergo epithelial-mesenchymal transition (EMT) and enter a partial epithelial-mesenchymal transition (pEMT) state to regain their EMP properties and proliferation capacities. With the continuous development of EMT, hAECs eventually enter a senescent state. The addition of SB431542 and microcarrier screening enabled the effective 3D expansion of hAECs by 50 fold while maintaining the EMP status in hAECs for further proliferation. This study not only elucidated the central proliferation mechanism of hAECs during development and expansion but also optimized the in vitro culture system so that it is sufficient to generate hAECs for 50 patients from a single donor amniotic membrane.
Collapse
Affiliation(s)
- Wangping Hao
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Yi Luo
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
| | - Jia Tian
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- College of Chemical EngineeringUniversity of the Chinese Academy of SciencesBeijing101408P. R. China
| | - Yuefeng Lu
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Yangyang Cui
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Ying Zhang
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Xiao Jin
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
| | - Hongjuan Ye
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
| | - Mengqi Lu
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
- Postgraduate Training Base of Shanghai East HospitalJinzhou Medical UniversityJinzhouLiaoning121001P. R. China
| | - Jinjia Song
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
| | - Weiqing Zhou
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- Key Laboratory of Biopharmaceutical Preparation and DeliveryInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- College of Chemical EngineeringUniversity of the Chinese Academy of SciencesBeijing101408P. R. China
| | - Wencheng Zhang
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
| | - Zhiying He
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200123P. R. China
- Shanghai iCELL Biotechnology Co., LtdShanghai200335P. R. China
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghai200335P. R. China
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghai200120P. R. China
- Postgraduate Training Base of Shanghai East HospitalJinzhou Medical UniversityJinzhouLiaoning121001P. R. China
| |
Collapse
|
4
|
Gupta S, Zhang E, Sinha S, Martin LM, Varghese TS, Forck NG, Sinha PR, Ericsson AC, Hesemann NP, Mohan RR. Analysis of Smad3 in the modulation of stromal extracellular matrix proteins in corneal scarring after alkali injury. Mol Vis 2024; 30:448-464. [PMID: 39959170 PMCID: PMC11829792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/28/2024] [Indexed: 02/18/2025] Open
Abstract
Purpose During ocular trauma, excessive proliferation and transdifferentiation of corneal stromal fibroblasts cause haze/fibrosis in the cornea. Transforming growth factor β (TGFβ) plays a key role in corneal fibrosis through the Smad signaling pathway. The aberrant activity of TGFβ signaling during ocular trauma (viz. mechanical, infectious, chemical, or surgically altered TGFβ/Smad signaling) leads to regulating the predominant expression of myogenic proteins and the extracellular matrix (ECM). We sought to investigate the functional role of Smad3 in corneal wound repair and stromal ECM assembly using Smad3+/+ wild-type and Smad3-/- deficient mice. Methods Corneal injury was introduced with the topical application of an alkali-soaked 2-mm filter disc on the central cornea in the Smad3+/+ (C57BL/6J) and Smad3-/- (129-Smad3tm1Par/J) mouse strains. Slit-lamp and stereo microscopy were used for clinical assessment and corneal haze grading in live animals. Hematoxylin and eosin and Masson's trichrome staining were used to study comparative morphology and collagen level alterations between the groups. Real-time qRT-PCR, western blot, and immunohistochemistry were used to measure changes in profibrotic genes at the mRNA and protein levels. Results Slit-lamp clinical exams and stereo microscopy detected notably less opaque cornea in the eyes of Smad3-/- compared with Smad3+/+ mice at 3 weeks (p<0.01) in live animals. Corneal tissue sections of Smad3-/- mice showed significantly fewer α-smooth muscle actin-positive cells compared with those of the Smad3+/+ animals (p<0.05). The corneas of the Smad3-/- mice showed significantly lower mRNA levels of pro-fibrotic genes, α-smooth muscle actin, fibronectin, and collagen I (p<0.05, p<0.01, and p<0.001). In addition, the matrix metalloproteinase and tissue inhibitors of metalloproteinase levels were significantly increased (p<0.001) in the corneal tissue during alkali injury in both Smad3+/+ wild-type and Smad3-/- deficient mice. Conclusions The significant changes in profibrotic genes and stromal ECM proteins revealed a direct role of Smad3 in stromal ECM proteins and TGFβ/Smad-driven wound healing. Smad3 appears to be an attractive molecular target for limiting abnormal stroma wound healing to treat corneal fibrosis in vivo.
Collapse
Affiliation(s)
- Suneel Gupta
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Eric Zhang
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO
| | - Sampann Sinha
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Lynn M. Martin
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Thomas S. Varghese
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO
| | - Nathan G. Forck
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Prashant R. Sinha
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Aaron C. Ericsson
- Departments of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO
| | - Nathan P. Hesemann
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO
| |
Collapse
|
5
|
Karami N, Taei A, Eftekhari-Yazdi P, Hassani F. Signaling pathway regulators in preimplantation embryos. J Mol Histol 2024; 56:57. [PMID: 39729177 DOI: 10.1007/s10735-024-10338-7] [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: 08/31/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Embryonic development during the preimplantation stages is highly sensitive and critically dependent on the reception of signaling cues. The precise coordination of diverse pathways and signaling factors is essential for successful embryonic progression. Even minor disruptions in these factors can result in physiological dysfunction, fetal malformations, or embryonic arrest. This issue is particularly evident in assisted reproductive technologies, such as in vitro fertilization, where embryonic arrest is frequently observed. A detailed understanding of these pathways enhances insight into the fundamental mechanisms underlying cellular processes and their contributions to embryonic development. The significance of elucidating signaling pathways and their regulatory factors in preimplantation development cannot be overstated. The application of this knowledge in laboratory settings has the potential to support strategies for modeling developmental stages and diseases, drug screening, therapeutic discovery, and reducing embryonic arrest. Furthermore, using various factors, small molecules, and pharmacological agents can enable the development or optimization of culture media for enhanced embryonic viability. While numerous pathways influence preimplantation development, this study examines several critical signaling pathways in this contex.
Collapse
Affiliation(s)
- Narges Karami
- MSc., Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O.Box 16635-148, Tehran, Iran
| | - Fatemeh Hassani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O.Box 16635-148, Tehran, Iran.
| |
Collapse
|
6
|
Kaitsuka T. The Unique Roles of Ion Channels in Pluripotent Stem Cells in Response to Biological Stimuli. BIOLOGY 2024; 13:1043. [PMID: 39765710 PMCID: PMC11673299 DOI: 10.3390/biology13121043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Ion channels are essential for mineral ion homeostasis in mammalian cells, and these are activated or inhibited by environmental stimuli such as heat, cold, mechanical, acidic, or basic stresses. These expressions and functions are quite diverse between cell types. The function and importance of ion channels are well-studied in neurons and cardiac cells, while those functions in pluripotent stem cells (PSCs) were not fully understood. Some sodium, potassium, chloride, calcium, transient receptor potential channels and mechanosensitive Piezo channels are found to be expressed and implicated in pluripotency and self-renewal capacity in PSCs. This review summarizes present and previous reports about ion channels and their response to environmental stimuli in PSCs. Furthermore, we compare the expressions and roles between PSCs and their differentiated embryoid bodies. We then discuss those contributions to pluripotency and differentiation.
Collapse
Affiliation(s)
- Taku Kaitsuka
- School of Pharmacy at Fukuoka, International University of Health and Welfare, Enokizu 137-1, Okawa 831-8501, Fukuoka, Japan
| |
Collapse
|
7
|
Bi R, Pan LN, Dai H, Sun C, Li C, Lin HJ, Xie LP, Ma HX, Li L, Xie H, Guo K, Hou CH, Yao YG, Chen LN, Zheng P. Epigenetic characterization of adult rhesus monkey spermatogonial stem cells identifies key regulators of stem cell homeostasis. Nucleic Acids Res 2024; 52:13644-13664. [PMID: 39535033 DOI: 10.1093/nar/gkae1013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Spermatogonial stem cells (SSCs) play crucial roles in the preservation of male fertility. However, successful ex vivo expansion of authentic human SSCs remains elusive due to the inadequate understanding of SSC homeostasis regulation. Using rhesus monkeys (Macaca mulatta) as a representative model, we characterized SSCs and progenitor subsets through single-cell RNA sequencing using a cell-specific network approach. We also profiled chromatin status and major histone modifications (H3K4me1, H3K4me3, H3K27ac, H3K27me3 and H3K9me3), and subsequently mapped promoters and active enhancers in TSPAN33+ putative SSCs. Comparing the epigenetic changes between fresh TSPAN33+ cells and cultured TSPAN33+ cells (resembling progenitors), we identified the regulatory elements with higher activity in SSCs, and the potential transcription factors and signaling pathways implicated in SSC regulation. Specifically, TGF-β signaling is activated in monkey putative SSCs. We provided evidence supporting its role in promoting self-renewal of monkey SSCs in culture. Overall, this study outlines the epigenetic landscapes of monkey SSCs and provides clues for optimization of the culture condition for primate SSCs expansion.
Collapse
Affiliation(s)
- Rui Bi
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
| | - Lin-Nuo Pan
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, No. 320 Yue Yang Road, Shanghai 200031, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Hao Dai
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, No. 320 Yue Yang Road, Shanghai 200031, China
| | - Chunli Sun
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| | - Cong Li
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| | - Hui-Juan Lin
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
| | - Lan-Ping Xie
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Huai-Xiao Ma
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
| | - Lin Li
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Heng Xie
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Kun Guo
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| | - Chun-Hui Hou
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Yong-Gang Yao
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| | - Luo-Nan Chen
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, No. 320 Yue Yang Road, Shanghai 200031, China
- Key Laboratory of Systems Biology, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, No. 1 Xiangshan Branch Lane, Xihu District, Hangzhou 310024, China
| | - Ping Zheng
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Baohua Road, Kunming 650107, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Kunming, Yunnan 650204, China
| |
Collapse
|
8
|
Bolkent S. Cellular and molecular mechanisms of asymmetric stem cell division in tissue homeostasis. Genes Cells 2024; 29:1099-1110. [PMID: 39379096 PMCID: PMC11609605 DOI: 10.1111/gtc.13172] [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: 02/15/2024] [Revised: 09/09/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024]
Abstract
The asymmetric cell division determines cell diversity and distinct sibling cell fates by mechanisms linked to mitosis. Many adult stem cells divide asymmetrically to balance self-renewal and differentiation. The process of asymmetric cell division involves an axis of polarity and, second, the localization of cell fate determinants at the cell poles. Asymmetric division of stem cells is achieved by intrinsic and extrinsic fate determinants such as signaling molecules, epigenetics factors, molecules regulating gene expression, and polarized organelles. At least some stem cells perform asymmetric and symmetric cell divisions during development. Asymmetric division ensures that the number of stem cells remains constant throughout life. The asymmetric division of stem cells plays an important role in biological events such as embryogenesis, tissue regeneration and carcinogenesis. This review summarizes recent advances in the regulation of asymmetric stem cell division in model organisms.
Collapse
Affiliation(s)
- Sema Bolkent
- Cerrahpaşa Faculty of Medicine, Department of Medical BiologyIstanbul University‐CerrahpaşaCerrahpaşaIstanbulTurkey
| |
Collapse
|
9
|
Liu S, Ren J, Hu Y, Zhou F, Zhang L. TGFβ family signaling in human stem cell self-renewal and differentiation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:26. [PMID: 39604763 PMCID: PMC11602941 DOI: 10.1186/s13619-024-00207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Human stem cells are undifferentiated cells with the capacity for self-renewal and differentiation into distinct cell lineages, playing important role in the development and maintenance of diverse tissues and organs. The microenvironment of stem cell provides crucial factors and components that exert significant influence over the determination of cell fate. Among these factors, cytokines from the transforming growth factor β (TGFβ) superfamily, including TGFβ, bone morphogenic protein (BMP), Activin and Nodal, have been identified as important regulators governing stem cell maintenance and differentiation. In this review, we present a comprehensive overview of the pivotal roles played by TGFβ superfamily signaling in governing human embryonic stem cells, somatic stem cells, induced pluripotent stem cells, and cancer stem cells. Furthermore, we summarize the latest research and advancements of TGFβ family in various cancer stem cells and stem cell-based therapy, discussing their potential clinical applications in cancer therapy and regeneration medicine.
Collapse
Affiliation(s)
- Sijia Liu
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiang Ren
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanmei Hu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Long Zhang
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
10
|
Li YY, Qian FC, Zhang GR, Li XC, Zhou LW, Yu ZM, Liu W, Wang QY, Li CQ. FunlncModel: integrating multi-omic features from upstream and downstream regulatory networks into a machine learning framework to identify functional lncRNAs. Brief Bioinform 2024; 26:bbae623. [PMID: 39602828 PMCID: PMC11601888 DOI: 10.1093/bib/bbae623] [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: 08/26/2024] [Revised: 10/26/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Accumulating evidence indicates that long noncoding RNAs (lncRNAs) play important roles in molecular and cellular biology. Although many algorithms have been developed to reveal their associations with complex diseases by using downstream targets, the upstream (epi)genetic regulatory information has not been sufficiently leveraged to predict the function of lncRNAs in various biological processes. Therefore, we present FunlncModel, a machine learning-based interpretable computational framework, which aims to screen out functional lncRNAs by integrating a large number of (epi)genetic features and functional genomic features from their upstream/downstream multi-omic regulatory networks. We adopted the random forest method to mine nearly 60 features in three categories from >2000 datasets across 11 data types, including transcription factors (TFs), histone modifications, typical enhancers, super-enhancers, methylation sites, and mRNAs. FunlncModel outperformed alternative methods for classification performance in human embryonic stem cell (hESC) (0.95 Area Under Curve (AUROC) and 0.97 Area Under the Precision-Recall Curve (AUPRC)). It could not only infer the most known lncRNAs that influence the states of stem cells, but also discover novel high-confidence functional lncRNAs. We extensively validated FunlncModel's efficacy by up to 27 cancer-related functional prediction tasks, which involved multiple cancer cell growth processes and cancer hallmarks. Meanwhile, we have also found that (epi)genetic regulatory features, such as TFs and histone modifications, serve as strong predictors for revealing the function of lncRNAs. Overall, FunlncModel is a strong and stable prediction model for identifying functional lncRNAs in specific cellular contexts. FunlncModel is available as a web server at https://bio.liclab.net/FunlncModel/.
Collapse
Affiliation(s)
- Yan-Yu Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- School of Computer, University of South China, Hengyang, Hunan, 421001, China
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Feng-Cui Qian
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- School of Computer, University of South China, Hengyang, Hunan, 421001, China
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Guo-Rui Zhang
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Xue-Cang Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, 163000, China
| | - Li-Wei Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zheng-Min Yu
- School of Computer, University of South China, Hengyang, Hunan, 421001, China
| | - Wei Liu
- College of Science, Heilongjiang Institute of Technology, Harbin, Heilongjiang, 150000, China
| | - Qiu-Yu Wang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- School of Computer, University of South China, Hengyang, Hunan, 421001, China
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Chun-Quan Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan, 421001, China
- School of Computer, University of South China, Hengyang, Hunan, 421001, China
- Institute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
11
|
Rohban R, Martins CP, Esni F. Advanced therapy to cure diabetes: mission impossible is now possible? Front Cell Dev Biol 2024; 12:1484859. [PMID: 39629270 PMCID: PMC11611888 DOI: 10.3389/fcell.2024.1484859] [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: 08/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Cell and Gene therapy are referred to as advanced therapies that represent overlapping fields of regenerative medicine. They have similar therapeutic goals such as to modify cellular identity, improve cell function, or fight a disease. These two therapeutic avenues, however, possess major differences. While cell therapy involves introduction of new cells, gene therapy entails introduction or modification of genes. Furthermore, the aim of cell therapy is often to replace, or repair damaged tissue, whereas gene therapy is used typically as a preventive approach. Diabetes mellitus severely affects the quality of life of afflicted individuals and has various side effects including cardiovascular, ophthalmic disorders, and neuropathy while putting enormous economic pressure on both the healthcare system and the patient. In recent years, great effort has been made to develop cutting-edge therapeutic interventions for diabetes treatment, among which cell and gene therapies stand out. This review aims to highlight various cell- and gene-based therapeutic approaches leading to the generation of new insulin-producing cells as a topmost "panacea" for treating diabetes, while deliberately avoiding a detailed molecular description of these approaches. By doing so, we aim to target readers who are new to the field and wish to get a broad helicopter overview of the historical and current trends of cell- and gene-based approaches in β-cell regeneration.
Collapse
Affiliation(s)
- Rokhsareh Rohban
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Christina P. Martins
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- McGowan Institute for regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
12
|
Prud’homme GJ, Wang Q. Anti-Inflammatory Role of the Klotho Protein and Relevance to Aging. Cells 2024; 13:1413. [PMID: 39272986 PMCID: PMC11394293 DOI: 10.3390/cells13171413] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The α-Klotho protein (hereafter Klotho) is an obligate coreceptor for fibroblast growth factor 23 (FGF23). It is produced in the kidneys, brain and other sites. Klotho insufficiency causes hyperphosphatemia and other anomalies. Importantly, it is associated with chronic pathologies (often age-related) that have an inflammatory component. This includes atherosclerosis, diabetes and Alzheimer's disease. Its mode of action in these diseases is not well understood, but it inhibits or regulates multiple major pathways. Klotho has a membrane form and a soluble form (s-Klotho). Cytosolic Klotho is postulated but not well characterized. s-Klotho has endocrine properties that are incompletely elucidated. It binds to the FGF receptor 1c (FGFR1c) that is widely expressed (including endothelial cells). It also attaches to soluble FGF23, and FGF23/Klotho binds to FGFRs. Thus, s-Klotho might be a roaming FGF23 coreceptor, but it has other functions. Notably, Klotho (cell-bound or soluble) counteracts inflammation and appears to mitigate related aging (inflammaging). It inhibits NF-κB and the NLRP3 inflammasome. This inflammasome requires priming by NF-κB and produces active IL-1β, membrane pores and cell death (pyroptosis). In accord, Klotho countered inflammation and cell injury induced by toxins, damage-associated molecular patterns (DAMPs), cytokines, and reactive oxygen species (ROS). s-Klotho also blocks the TGF-β receptor and Wnt ligands, which lessens fibrotic disease. Low Klotho is associated with loss of muscle mass (sarcopenia), as occurs in aging and chronic diseases. s-Klotho counters the inhibitory effects of myostatin and TGF-β on muscle, reduces inflammation, and improves muscle repair following injury. The inhibition of TGF-β and other factors may also be protective in diabetic retinopathy and age-related macular degeneration (AMD). This review examines Klotho functions especially as related to inflammation and potential applications.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 220 Walmer Rd, Toronto, ON M5R 3R7, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200030, China
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201318, China
| |
Collapse
|
13
|
Choi H, Oh D, Kim M, Jawad A, Zheng H, Cai L, Lee J, Kim E, Lee G, Jang H, Moon C, Hyun SH. Establishment of porcine embryonic stem cells in simplified serum free media and feeder free expansion. Stem Cell Res Ther 2024; 15:245. [PMID: 39113095 PMCID: PMC11304784 DOI: 10.1186/s13287-024-03858-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND The establishment of stable porcine embryonic stem cells (pESCs) can contribute to basic and biomedical research, including comparative developmental biology, as well as assessing the safety of stem cell-based therapies. Despite these advantages, most pESCs obtained from in vitro blastocysts require complex media and feeder layers, making routine use, genetic modification, and differentiation into specific cell types difficult. We aimed to establish pESCs with a single cell-passage ability, high proliferative potency, and stable in long-term culture from in vitro-derived blastocysts using a simplified serum-free medium. METHODS We evaluated the establishment efficiency of pESCs from in vitro blastocysts using various basal media (DMEM/F10 (1:1), DMEM/F12, and a-MEM) and factors (FGF2, IWR-1, CHIR99021, and WH-4-023). The pluripotency and self-renewal capacity of the established pESCs were analyzed under feeder or feeder-free conditions. Ultimately, we developed a simplified culture medium (FIW) composed of FGF2, IWR-1, and WH-4-023 under serum-free conditions. RESULTS The pESC-FIW lines were capable of single-cell passaging with short cell doubling times and expressed the pluripotency markers POU5F1, SOX2, and NANOG, as well as cell surface markers SSEA1, SSEA4, and TRA-1-60. pESC-FIW showed a stable proliferation rate and normal karyotype, even after 50 passages. Transcriptome analysis revealed that pESC-FIW were similar to reported pESC maintained in complex media and showed gastrulating epiblast cell characteristics. pESC-FIW were maintained for multiple passages under feeder-free conditions on fibronectin-coated plates using mTeSR™, a commercial medium used for feeder-free culture, exhibiting characteristics similar to those observed under feeder conditions. CONCLUSIONS These results indicated that inhibition of WNT and SRC was sufficient to establish pESCs capable of single-cell passaging and feeder-free expansion under serum-free conditions. The easy maintenance of pESCs facilitates their application in gene editing technology for agriculture and biomedicine, as well as lineage commitment studies.
Collapse
Affiliation(s)
- Hyerin Choi
- Veterinary Medical Center, College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Dongjin Oh
- Veterinary Medical Center, College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Mirae Kim
- Veterinary Medical Center, College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Ali Jawad
- Veterinary Medical Center, College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Haomiao Zheng
- Veterinary Medical Center, College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea
| | - Lian Cai
- Veterinary Medical Center, College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, Republic of Korea
| | - Joohyeong Lee
- Department of Companion Animal Industry, Semyung University, Jecheon, 27136, Republic of Korea
| | - Eunhye Kim
- Laboratory of Molecular Diagnostics and Cell Biology, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Gabsang Lee
- Department of Neurology, Institute for Cell Engineering, School of Medicine, Johns Hopkins Medicine, Baltimore, ML, USA
| | - Hyewon Jang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine, BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine, BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Sang-Hwan Hyun
- Veterinary Medical Center, College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, Republic of Korea.
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, Republic of Korea.
- Vet-ICT Convergence Education and Research Center (VICERC), Chungbuk National University, Cheongju, Republic of Korea.
- Chungbuk National University Hospital, Cheongju, Republic of Korea.
| |
Collapse
|
14
|
Li J, Niu C, Ai H, Li X, Zhang L, Lang Y, Wang S, Gao F, Mei X, Yu C, Sun L, Huang Y, Zheng L, Wang G, Sun Y, Yang X, Song Z, Bao Y. TSP50 Attenuates DSS-Induced Colitis by Regulating TGF-β Signaling Mediated Maintenance of Intestinal Mucosal Barrier Integrity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305893. [PMID: 38189580 PMCID: PMC10953580 DOI: 10.1002/advs.202305893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/03/2023] [Indexed: 01/09/2024]
Abstract
The integrity of the intestinal mucosal barrier is crucial for protecting the intestinal epithelium against invasion by commensal bacteria and pathogens, thereby combating colitis. The investigation revealed that the absence of TSP50 compromised the integrity of the intestinal mucosal barrier in murine subjects. This disruption facilitated direct contact between intestinal bacteria and the intestinal epithelium, thereby increasing susceptibility to colitis. Mechanistic analysis indicated that TSP50 deficiency in intestinal stem cells (ISCs) triggered aberrant activation of the TGF-β signaling pathway and impeded the differentiation of goblet cells in mice, leading to impairment of mucosal permeability. By inhibiting the TGF-β pathway, the functionality of the intestinal mucosal barrier is successfully restored and mitigated colitis in TSP50-deficient mice. In conclusion, TSP50 played a crucial role in maintaining the intestinal mucosal barrier function and exhibited the preventive effect against the development of colitis by regulating the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Jiawei Li
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine ProductsNortheast Normal UniversityChangchun130024China
- National Engineering Laboratory for Druggable Gene and Protein ScreeningNortheast Normal UniversityChangchun130117China
| | - Chunxue Niu
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine ProductsNortheast Normal UniversityChangchun130024China
- The Key Laboratory of Molecular Epigenetics of Ministry of EducationNortheast Normal UniversityChangchunJilin130024China
| | - Huihan Ai
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine ProductsNortheast Normal UniversityChangchun130024China
- Department of General SurgeryAffiliated Tumor Hospital of Zhengzhou UniversityZhengzhouHenan450000China
| | - Xiaoli Li
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine ProductsNortheast Normal UniversityChangchun130024China
| | - Linlin Zhang
- National Engineering Laboratory for Druggable Gene and Protein ScreeningNortheast Normal UniversityChangchun130117China
| | - Yan Lang
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine ProductsNortheast Normal UniversityChangchun130024China
| | - Shuyue Wang
- The Key Laboratory of Molecular Epigenetics of Ministry of EducationNortheast Normal UniversityChangchunJilin130024China
| | - Feng Gao
- National Engineering Laboratory for Druggable Gene and Protein ScreeningNortheast Normal UniversityChangchun130117China
| | - Xianglin Mei
- Department of PathologyThe Second Hospital of Jilin UniversityChangchun130041China
| | - Chunlei Yu
- The Key Laboratory of Molecular Epigenetics of Ministry of EducationNortheast Normal UniversityChangchunJilin130024China
| | - Luguo Sun
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine ProductsNortheast Normal UniversityChangchun130024China
| | - Yanxin Huang
- National Engineering Laboratory for Druggable Gene and Protein ScreeningNortheast Normal UniversityChangchun130117China
| | - Lihua Zheng
- National Engineering Laboratory for Druggable Gene and Protein ScreeningNortheast Normal UniversityChangchun130117China
| | - Guannan Wang
- National Engineering Laboratory for Druggable Gene and Protein ScreeningNortheast Normal UniversityChangchun130117China
| | - Ying Sun
- National Engineering Laboratory for Druggable Gene and Protein ScreeningNortheast Normal UniversityChangchun130117China
| | - Xiaoguang Yang
- The Key Laboratory of Molecular Epigenetics of Ministry of EducationNortheast Normal UniversityChangchunJilin130024China
| | - Zhenbo Song
- National Engineering Laboratory for Druggable Gene and Protein ScreeningNortheast Normal UniversityChangchun130117China
| | - Yongli Bao
- NMPA Key Laboratory for Quality Control of Cell and Gene Therapy Medicine ProductsNortheast Normal UniversityChangchun130024China
| |
Collapse
|
15
|
Nong C, Chen Y, Yang H, Chen N, Tian C, Li S, Chen H. Phenotypic sorting of individual male and female intersex Cherax quadricarinatus and analysis of molecular differences in the gonadal transcriptome. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 49:101194. [PMID: 38246110 DOI: 10.1016/j.cbd.2024.101194] [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: 12/05/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024]
Abstract
Cherax quadricarinatus exhibit sexual dimorphism, with males outpacing females in size specification and growth rate. However, there is limited understanding of the molecular mechanisms underlying sex determination and sex differentiation in crustaceans. To study the differences between intersex individuals and normal individuals, this study counted the proportion of intersex individuals in the natural population, collected the proportion of 7 different phenotypes in 200 intersex individuals, and observed the differences in tissue sections. RNA-seq was used to study the different changes in the transcriptome of normal and intersex gonads. The results showed that: the percentage of intersex in the natural population was 1.5 %, and the percentage of different types of intersex ranged from 0.5 % to 22.5 %; the sections revealed that the development of normal ovaries was stagnant at the primary oocyte stage when intersex individuals with ovaries were present; We screened for pathways and genes that may be associated with gonadal development and sex, including ovarian steroid synthesis, estrogen signaling pathway, oocyte meiosis, progesterone-mediated oocyte maturation, etc. Relevant genes including tra2a, dmrta2, ccnb2, foxl2, and smad4. This study provides an important molecular basis for sex determination, sex-controlled breeding, and unisex breeding in red crayfish.
Collapse
Affiliation(s)
- Chuntai Nong
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Yibin Chen
- Guangdong Evergreen Feed Industry Co., Ltd., Evergreen Tower, Zhanjiang, Guangdong, China
| | - Hao Yang
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Nanxiong Chen
- Guangdong Evergreen Feed Industry Co., Ltd., Evergreen Tower, Zhanjiang, Guangdong, China
| | - Changxu Tian
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Sedong Li
- Guangdong Evergreen Feed Industry Co., Ltd., Evergreen Tower, Zhanjiang, Guangdong, China.; Zhanjiang Ocean and Fishery Development Research Center, Zhanjiang, China.
| | - Huapu Chen
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China; Guangdong Havwii agriculture group Co., Ltd, Zhanjiang 524266, China.
| |
Collapse
|
16
|
Chao S, Yan H, Bu P. Asymmetric division of stem cells and its cancer relevance. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:5. [PMID: 38411768 PMCID: PMC10897644 DOI: 10.1186/s13619-024-00188-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/30/2024] [Indexed: 02/28/2024]
Abstract
Asymmetric division is a fundamental process for generating cell diversity and maintaining the stem cell population. During asymmetric division, proteins, organelles, and even RNA are distributed unequally between the two daughter cells, determining their distinct cell fates. The mechanisms orchestrating this process are extremely complex. Dysregulation of asymmetric division can potentially trigger cancer progression. Cancer stem cells, in particular, undergo asymmetric division, leading to intra-tumoral heterogeneity, which contributes to treatment refractoriness. In this review, we delve into the cellular and molecular mechanisms that govern asymmetric division and explore its relevance to tumorigenesis.
Collapse
Affiliation(s)
- Shanshan Chao
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huiwen Yan
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pengcheng Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, 100101, China.
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
17
|
Chaudhary JK, Ahamad N, Rath PC. Mesenchymal stem cells (MSCs) from the mouse bone marrow show differential expression of interferon regulatory factors IRF-1 and IRF-2. Mol Biol Rep 2024; 51:97. [PMID: 38194130 DOI: 10.1007/s11033-023-09025-9] [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: 08/17/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Interferon regulatory factors (IRF-1 and IRF-2) are transcription factors widely implicated in various cellular processes, including regulation of inflammatory responses to pathogens, cell proliferation, oncogenesis, differentiation, autophagy, and apoptosis. METHODS We have studied the expression of IRF-1, IRF-2 mRNAs by RT-PCR, cellular localization of the proteins by immunofluorescence, and expression of mRNAs of genes regulated by IRF-1, IRF-2 by RT-PCR in mouse bone marrow cells (BMCs) and mesenchymal stem cells (MSCs). RESULTS Higher level of IRF-1 mRNA was observed in BMCs and MSCs compared to that of IRF-2. Similarly, differential expression of IRF-1 and IRF-2 proteins was observed in BMCs and MSCs. IRF-1 was predominantly localized in the cytoplasm, whereas IRF-2 was localized in the nuclei of BMCs. MSCs showed nucleo-cytoplasmic distribution of IRF-1 and nuclear localization of IRF-2. Constitutive expression of IRF-1 and IRF-2 target genes: monocyte chemoattractant protein-1 (MCP-1), vascular cell adhesion molecule-1 (VCAM-1), cyclooxygenase-2 (COX-2), matrix metalloproteinase-9 (MMP-9), and caspase-1 was observed in both BMCs and MSCs. MSCs showed constitutive expression of the pluripotency-associated factors, Oct3/4 and Sox-2. Lipopolysaccharide (LPS)-treatment of MSCs induced prominent cellular localization of IRF-1 and IRF-2. CONCLUSIONS Our results suggest that IRF-1 and IRF-2 exhibit differential expression of their mRNAs and subcellular localization of the proteins in BMCs and MSCs. These cells also show differential levels of constitutive expression of IRF-1 and IRF-2 target genes. This may regulate immune-responsive properties of BMCs and MSCs through IRF-1, IRF-2-dependent gene expression and protein-protein interaction. Regulating IRF-1 and IRF-2 may be helpful for immunomodulatory functions of MSCs for cell therapy and regenerative medicine.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Naseem Ahamad
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
18
|
Anvar Z, Chakchouk I, Sharif M, Mahadevan S, Su L, Anikar S, Naini FA, Utama AB, Van den Veyver IB. Comparison of Four Protocols for In Vitro Differentiation of Human Embryonic Stem Cells into Trophoblast Lineages by BMP4 and Dual Inhibition of Activin/Nodal and FGF2 Signaling. Reprod Sci 2024; 31:173-189. [PMID: 37658178 PMCID: PMC10784360 DOI: 10.1007/s43032-023-01334-5] [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: 12/20/2022] [Accepted: 08/16/2023] [Indexed: 09/03/2023]
Abstract
Human embryonic stem cells (hESCs) cultured in media containing bone morphogenic protein 4 (BMP4; B) differentiate into trophoblast-like cells. Supplementing media with inhibitors of activin/nodal signaling (A83-01) and of fibroblast growth factor 2 (PD173074) suppresses mesoderm and endoderm formation and improves specification of trophoblast-like lineages, but with variable effectiveness. We compared differentiation in four BMP4-containing media: mTeSR1-BMP4 only, mTeSR1-BAP, basal medium with BAP (basal-BAP), and a newly defined medium, E7-BAP. These media variably drive early differentiation towards trophoblast-like lineages with upregulation of early trophoblast markers CDX2 and KRT7 and downregulation of pluripotency markers (OCT4 and NANOG). As expected, based on differences between media in FGF2 and its inhibitors, downregulation of mesendoderm marker EOMES was variable between media. By day 7, only hESCs grown in E7-BAP or basal-BAP expressed HLA-G protein, indicating the presence of cells with extravillous trophoblast characteristics. Expression of HLA-G and other differentiation markers (hCG, KRT7, and GCM1) was highest in basal-BAP, suggesting a faster differentiation in this medium, but those cultures were more inhomogeneous and still expressed some endodermal and pluripotency markers. In E7-BAP, HLA-G expression increased later and was lower. There was also a low but maintained expression of some C19MC miRNAs, with more CpG hypomethylation of the ELF5 promoter, suggesting that E7-BAP cultures differentiate slower along the trophoblast lineage. We conclude that while all protocols drive differentiation into trophoblast lineages with varying efficiency, they have advantages and disadvantages that must be considered when selecting a protocol for specific experiments.
Collapse
Affiliation(s)
- Zahra Anvar
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Imen Chakchouk
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Momal Sharif
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Sangeetha Mahadevan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Li Su
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Swathi Anikar
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Fatemeh Alavi Naini
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - Ignatia B Van den Veyver
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA.
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
19
|
Schuhwerk H, Brabletz T. Mutual regulation of TGFβ-induced oncogenic EMT, cell cycle progression and the DDR. Semin Cancer Biol 2023; 97:86-103. [PMID: 38029866 DOI: 10.1016/j.semcancer.2023.11.009] [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: 06/27/2023] [Revised: 10/06/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
TGFβ signaling and the DNA damage response (DDR) are two cellular toolboxes with a strong impact on cancer biology. While TGFβ as a pleiotropic cytokine affects essentially all hallmarks of cancer, the multifunctional DDR mostly orchestrates cell cycle progression, DNA repair, chromatin remodeling and cell death. One oncogenic effect of TGFβ is the partial activation of epithelial-to-mesenchymal transition (EMT), conferring invasiveness, cellular plasticity and resistance to various noxae. Several reports show that both individual networks as well as their interface affect chemo-/radiotherapies. However, the underlying mechanisms remain poorly resolved. EMT often correlates with TGFβ-induced slowing of proliferation, yet numerous studies demonstrate that particularly the co-activated EMT transcription factors counteract anti-proliferative signaling in a partially non-redundant manner. Collectively, evidence piled up over decades underscore a multifaceted, reciprocal inter-connection of TGFβ signaling / EMT with the DDR / cell cycle progression, which we will discuss here. Altogether, we conclude that full cell cycle arrest is barely compatible with the propagation of oncogenic EMT traits and further propose that 'EMT-linked DDR plasticity' is a crucial, yet intricate facet of malignancy, decisively affecting metastasis formation and therapy resistance.
Collapse
Affiliation(s)
- Harald Schuhwerk
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
20
|
Krzysiek-Maczka G, Brzozowski T, Ptak-Belowska A. Helicobacter pylori-activated fibroblasts as a silent partner in gastric cancer development. Cancer Metastasis Rev 2023; 42:1219-1256. [PMID: 37460910 PMCID: PMC10713772 DOI: 10.1007/s10555-023-10122-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/20/2023] [Indexed: 12/18/2023]
Abstract
The discovery of Helicobacter pylori (Hp) infection of gastric mucosa leading to active chronic gastritis, gastroduodenal ulcers, and MALT lymphoma laid the groundwork for understanding of the general relationship between chronic infection, inflammation, and cancer. Nevertheless, this sequence of events is still far from full understanding with new players and mediators being constantly identified. Originally, the Hp virulence factors affecting mainly gastric epithelium were proposed to contribute considerably to gastric inflammation, ulceration, and cancer. Furthermore, it has been shown that Hp possesses the ability to penetrate the mucus layer and directly interact with stroma components including fibroblasts and myofibroblasts. These cells, which are the source of biophysical and biochemical signals providing the proper balance between cell proliferation and differentiation within gastric epithelial stem cell compartment, when exposed to Hp, can convert into cancer-associated fibroblast (CAF) phenotype. The crosstalk between fibroblasts and myofibroblasts with gastric epithelial cells including stem/progenitor cell niche involves several pathways mediated by non-coding RNAs, Wnt, BMP, TGF-β, and Notch signaling ligands. The current review concentrates on the consequences of Hp-induced increase in gastric fibroblast and myofibroblast number, and their activation towards CAFs with the emphasis to the altered communication between mesenchymal and epithelial cell compartment, which may lead to inflammation, epithelial stem cell overproliferation, disturbed differentiation, and gradual gastric cancer development. Thus, Hp-activated fibroblasts may constitute the target for anti-cancer treatment and, importantly, for the pharmacotherapies diminishing their activation particularly at the early stages of Hp infection.
Collapse
Affiliation(s)
- Gracjana Krzysiek-Maczka
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Tomasz Brzozowski
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Agata Ptak-Belowska
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland
| |
Collapse
|
21
|
Cai C, Wan P, Wang H, Cai X, Wang J, Chai Z, Wang J, Wang H, Zhang M, Yang N, Wu Z, Zhu J, Yang X, Li Y, Yue B, Dang R, Zhong J. Transcriptional and open chromatin analysis of bovine skeletal muscle development by single-cell sequencing. Cell Prolif 2023; 56:e13430. [PMID: 36855961 PMCID: PMC10472525 DOI: 10.1111/cpr.13430] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Skeletal muscle is a complex heterogeneous tissue and characterizing its cellular heterogeneity and transcriptional and epigenetic signatures are important for understanding the details of its ontogeny. In our study, we applied scRNA-seq and scATAC-seq to investigate the cell types, molecular features, transcriptional and epigenetic regulation, and patterns of developing bovine skeletal muscle from gestational, lactational and adult stages. Detailed molecular analyses were used to dissect cellular heterogeneity, and we deduced the differentiation trajectory of myogenic cells and uncovered their dynamic gene expression profiles. SCENIC analysis was performed to demonstrate key regulons during cell fate decisions. We explored the future expression states of these heterogeneous cells by RNA velocity analysis and found extensive networks of intercellular communication using the toolkit CellChat. Moreover, the transcriptomic and chromatin accessibility modalities were confirmed to be highly concordant, and integrative analysis of chromatin accessibility and gene expression revealed key transcriptional regulators acting during myogenesis. In bovine skeletal muscle, by scRNA-seq and scATAC-seq analysis, different cell types such as adipocytes, endothelial cells, fibroblasts, lymphocytes, monocytes, pericyte cells and eight skeletal myogenic subpopulations were identified at the three developmental stages. The pseudotime trajectory exhibited a distinct sequential ordering for these myogenic subpopulations and eight distinct gene clusters were observed according to their expression pattern. Moreover, specifically expressed TFs (such as MSC, MYF5, MYOD1, FOXP3, ESRRA, BACH1, SIX2 and ATF4) associated with muscle development were predicted, and likely future transcriptional states of individual cells and the developmental dynamics of differentiation among neighbouring cells were predicted. CellChat analysis on the scRNA-seq data set then classified many ligand-receptor pairs among these cell clusters, which were further categorized into significant signalling pathways, including BMP, IGF, WNT, MSTN, ANGPTL, TGFB, TNF, VEGF and FGF. Finally, scRNA-seq and scATAC-seq results were successfully integrated to reveal a series of specifically expressed TFs that are likely to be candidates for the promotion of cell fate transition during bovine skeletal muscle development. Overall, our results outline a single-cell dynamic chromatin/transcriptional landscape for normal bovine skeletal muscle development; these provide an important resource for understanding the structure and function of mammalian skeletal muscle, which will promote research into its biology.
Collapse
Affiliation(s)
- Cuicui Cai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Guyuan BranchNingxia Academy of Agriculture and Forestry SciencesGuyuanChina
| | - Peng Wan
- Guyuan BranchNingxia Academy of Agriculture and Forestry SciencesGuyuanChina
| | - Hui Wang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Xin Cai
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Jiabo Wang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Zhixin Chai
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Jikun Wang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Haibo Wang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Ming Zhang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Nan Yang
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Zhijuan Wu
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Jiangjiang Zhu
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Xueyao Yang
- Guyuan BranchNingxia Academy of Agriculture and Forestry SciencesGuyuanChina
| | - Yulian Li
- Guyuan BranchNingxia Academy of Agriculture and Forestry SciencesGuyuanChina
| | - Binglin Yue
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| | - Ruihua Dang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Jincheng Zhong
- Key Laboratory of Qinghai‐Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of EducationSouthwest Minzu UniversityChengduChina
| |
Collapse
|
22
|
Mostufi-Zadeh-Haghighi G, Veratti P, Zodel K, Greve G, Waterhouse M, Zeiser R, Cleary ML, Lübbert M, Duque-Afonso J. Functional Characterization of Transforming Growth Factor-β Signaling in Dasatinib Resistance and Pre-BCR + Acute Lymphoblastic Leukemia. Cancers (Basel) 2023; 15:4328. [PMID: 37686604 PMCID: PMC10486903 DOI: 10.3390/cancers15174328] [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: 07/14/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The multi-kinase inhibitor dasatinib has been implicated to be effective in pre-B-cell receptor (pre-BCR)-positive acute lymphoblastic leukemia (ALL) expressing the E2A-PBX1 fusion oncoprotein. The TGFβ signaling pathway is involved in a wide variety of cellular processes, including embryonic development and cell homeostasis, and it can have dual roles in cancer: suppressing tumor growth at early stages and mediating tumor progression at later stages. In this study, we identified the upregulation of the TGFβ signaling pathway in our previously generated human dasatinib-resistant pre-BCR+/E2A-PBX1+ ALL cells using global transcriptomic analysis. We confirm the upregulation of the TGFβ pathway member SMAD3 at the transcriptional and translational levels in dasatinib-resistant pre-BCR+/E2A-PBX1+ ALL cells. Hence, dasatinib blocks, at least partially, TGFβ-induced SMAD3 phosphorylation in several B-cell precursor (BCP) ALL cell lines as well as in dasatinib-resistant pre-BCR+/E2A-PBX1+ ALL cells. Activation of the TGFβ signaling pathway by TGF-β1 leads to growth inhibition by cell cycle arrest at the G0/G1 stage, increase in apoptosis and transcriptional changes of SMAD-targeted genes, e.g. c-MYC downregulation, in pre-BCR+/E2A-PBX1+ ALL cells. These results provide a better understanding about the role that the TGFβ signaling pathway plays in leukemogenesis of BCP-ALL as well as in secondary drug resistance to dasatinib.
Collapse
Affiliation(s)
- Gila Mostufi-Zadeh-Haghighi
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (G.M.-Z.-H.); (P.V.); (K.Z.); (G.G.); (M.W.); (R.Z.); (M.L.)
| | - Pia Veratti
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (G.M.-Z.-H.); (P.V.); (K.Z.); (G.G.); (M.W.); (R.Z.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Freiburg, 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kyra Zodel
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (G.M.-Z.-H.); (P.V.); (K.Z.); (G.G.); (M.W.); (R.Z.); (M.L.)
| | - Gabriele Greve
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (G.M.-Z.-H.); (P.V.); (K.Z.); (G.G.); (M.W.); (R.Z.); (M.L.)
| | - Miguel Waterhouse
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (G.M.-Z.-H.); (P.V.); (K.Z.); (G.G.); (M.W.); (R.Z.); (M.L.)
| | - Robert Zeiser
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (G.M.-Z.-H.); (P.V.); (K.Z.); (G.G.); (M.W.); (R.Z.); (M.L.)
| | - Michael L. Cleary
- Department of Pathology, Stanford University, Stanford, CA 94305, USA;
| | - Michael Lübbert
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (G.M.-Z.-H.); (P.V.); (K.Z.); (G.G.); (M.W.); (R.Z.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Freiburg, 79106 Freiburg, Germany
| | - Jesús Duque-Afonso
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (G.M.-Z.-H.); (P.V.); (K.Z.); (G.G.); (M.W.); (R.Z.); (M.L.)
| |
Collapse
|
23
|
Xu YJ, Dai SK, Duan CH, Zhang ZH, Liu PP, Liu C, Du HZ, Lu XK, Hu S, Li L, Teng ZQ, Liu CM. ASH2L regulates postnatal neurogenesis through Onecut2-mediated inhibition of TGF-β signaling pathway. Cell Death Differ 2023; 30:1943-1956. [PMID: 37433907 PMCID: PMC10406892 DOI: 10.1038/s41418-023-01189-y] [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: 08/20/2021] [Revised: 06/18/2023] [Accepted: 06/29/2023] [Indexed: 07/13/2023] Open
Abstract
The ability of neural stem/progenitor cells (NSPCs) to proliferate and differentiate is required through different stages of neurogenesis. Disturbance in the regulation of neurogenesis causes many neurological diseases, such as intellectual disability, autism, and schizophrenia. However, the intrinsic mechanisms of this regulation in neurogenesis remain poorly understood. Here, we report that Ash2l (Absent, small or homeotic discs-like 2), one core component of a multimeric histone methyltransferase complex, is essential for NSPC fate determination during postnatal neurogenesis. Deletion of Ash2l in NSPCs impairs their capacity for proliferation and differentiation, leading to simplified dendritic arbors in adult-born hippocampal neurons and deficits in cognitive abilities. RNA sequencing data reveal that Ash2l primarily regulates cell fate specification and neuron commitment. Furthermore, we identified Onecut2, a major downstream target of ASH2L characterized by bivalent histone modifications, and demonstrated that constitutive expression of Onecut2 restores defective proliferation and differentiation of NSPCs in adult Ash2l-deficient mice. Importantly, we identified that Onecut2 modulates TGF-β signaling in NSPCs and that treatment with a TGF-β inhibitor rectifies the phenotype of Ash2l-deficient NSPCs. Collectively, our findings reveal the ASH2L-Onecut2-TGF-β signaling axis that mediates postnatal neurogenesis to maintain proper forebrain function.
Collapse
Affiliation(s)
- Ya-Jie Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Shang-Kun Dai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Chun-Hui Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Zi-Han Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Pei-Pei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Cong Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Hong-Zhen Du
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Xu-Kun Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, 215000, Suzhou, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
24
|
Pan W, Wang Y, Zhao C. miR-140-5p attenuates hepatic fibrosis by directly targeting TGFβR1. Scand J Gastroenterol 2023; 58:1335-1343. [PMID: 37313731 DOI: 10.1080/00365521.2023.2223735] [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: 05/08/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To explore the protective effect and related mechanism of miR-140-5p on liver fibrosis by interfering with TGF-β/Smad signaling pathway. METHODS Liver fibrosis mice models were established by intraperitoneal injection of CCL4. Hematoxylin and eosin (HE) staining was used to detect the structural and morphological changes of the liver. Masson staining was used to detect collagen deposition. Human hepatic stellate cells (HSCs, LX-2) were transfected with miR-140-5p mimic or inhibitor then treated with TGF-β1. The qRT-PCR and Western blotting was used to detect the expression of related molecules. The luciferase reporter assay was used to identify the target of miR-140-5p. RESULTS Our results indicated that miR-140-5p expression was downregulated in fibrotic liver tissues of model mice and LX-2 cells treated with TGF-β1. The overexpression of miR-140-5p decreased the expression of collagen1(COL1) and α-smooth muscle actin(α-SMA), inhibited the phosphorylation of Smad-2/3 (pSmad-2/3) in LX-2 cells. Conversely, the knockdown of miR-140-5p upregulated COL1 and α-SMA expression, increased Smad-2/3 phosphorylation. A dual-luciferase reporter assay showed that TGFβR1 was a target gene of miR-140-5p. The overexpression of miR-140-5p suppressed TGFβR1 expression in LX-2 cells. Additionally, knockdown of TGFβR1 decreased the expression of COL1 and α-SMA. Conversely, the overexpression of TGFβR1 reversed the inhibitory effect of miR-140-5p upregulation on expression of COL1 and α-SMA. CONCLUSION miR-140-5p bound to TGFβR1 mRNA 3'-untranslated region(3'UTR) and inhibited the expression of TGFβR1, pSmad-2/3, COL1 and α-SMA, thereby exerting a potential therapeutic effect on hepatic fibrosis.
Collapse
Affiliation(s)
- Wenchao Pan
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yadong Wang
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Caiyan Zhao
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
25
|
De Kleijn KMA, Zuure WA, Straasheijm KR, Martens MB, Avramut MC, Koning RI, Martens GJM. Human cortical spheroids with a high diversity of innately developing brain cell types. Stem Cell Res Ther 2023; 14:50. [PMID: 36959625 PMCID: PMC10035191 DOI: 10.1186/s13287-023-03261-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 02/28/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) human brain spheroids are instrumental to study central nervous system (CNS) development and (dys)function. Yet, in current brain spheroid models the limited variety of cell types hampers an integrated exploration of CNS (disease) mechanisms. METHODS Here we report a 5-month culture protocol that reproducibly generates H9 embryonic stem cell-derived human cortical spheroids (hCSs) with a large cell-type variety. RESULTS We established the presence of not only neuroectoderm-derived neural progenitor populations, mature excitatory and inhibitory neurons, astrocytes and oligodendrocyte (precursor) cells, but also mesoderm-derived microglia and endothelial cell populations in the hCSs via RNA-sequencing, qPCR, immunocytochemistry and transmission electron microscopy. Transcriptomic analysis revealed resemblance between the 5-months-old hCSs and dorsal frontal rather than inferior regions of human fetal brains of 19-26 weeks of gestational age. Pro-inflammatory stimulation of the generated hCSs induced a neuroinflammatory response, offering a proof-of-principle of the applicability of the spheroids. CONCLUSIONS Our protocol provides a 3D human brain cell model containing a wide variety of innately developing neuroectoderm- as well as mesoderm-derived cell types, furnishing a versatile platform for comprehensive examination of intercellular CNS communication and neurological disease mechanisms.
Collapse
Affiliation(s)
- Kim M A De Kleijn
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behavior, Centre for Neuroscience, Faculty of Science, Radboud University, 6525GA, Nijmegen, The Netherlands.
- NeuroDrug Research Ltd, 6525ED, Nijmegen, The Netherlands.
| | - Wieteke A Zuure
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behavior, Centre for Neuroscience, Faculty of Science, Radboud University, 6525GA, Nijmegen, The Netherlands
| | | | | | - M Cristina Avramut
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Roman I Koning
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300RC, Leiden, The Netherlands
| | - Gerard J M Martens
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behavior, Centre for Neuroscience, Faculty of Science, Radboud University, 6525GA, Nijmegen, The Netherlands
- NeuroDrug Research Ltd, 6525ED, Nijmegen, The Netherlands
| |
Collapse
|
26
|
Mohammadi Y, Zangooei M, Zardast M, Mamashli M, Rezaei Farimani A. The effect of crocin and losartan on TGF-β gene expression and histopathology of kidney tissue in a rat model of diabetic nephropathy. AVICENNA JOURNAL OF PHYTOMEDICINE 2023; 13:189-199. [PMID: 37333473 PMCID: PMC10274314 DOI: 10.22038/ajp.2022.21414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 06/20/2023]
Abstract
Objective Diabetic nephropathy is one of the most common microvascular complications of diabetes mellitus that finally leads to complete loss of kidney function. Therefore, this study aimed to evaluate the effect of crocin and losartan on TGF-β gene expression and histopathology of kidney tissue in a rat model of diabetic nephropathy. Materials and Methods Forty male Wistar rats were randomly divided into five groups (n=8): Untreated control, Diabetic (D), D + crocin, D + losartan, and D + losartan + crocin. Induction of diabetes was performed using streptozotocin (50 mg/kg/ Intraperitoneal injection). At the end of the eight-week period, the rats were sacrificed. Spectrophotometry measured serum glucose, urea, creatinine, and uric acid levels. Microalbumin and creatinine levels were measured in 24-hour urine. Real-time PCR was used to determine the relative expression of the TGF-β gene in kidney tissue. Renal tissue histopathology was also examined. Results The results showed that hyperglycemia increased biochemical factors associated with diabetes, TGF-β gene expression, and kidney damage. Separate treatment with crocin and losartan led to a decrease in renal function factors and TGF-β gene expression and improved kidney damage. Conclusion Our results showed that crocin could improve kidney function in diabetic conditions. In addition, we showed that crocin increases the effectiveness of losartan. Consequently, we suggest that crocin in combination with chemical drugs can be a potential therapeutic agent for diabetes and its complications. Nonetheless, human studies are needed to make firm findings.
Collapse
Affiliation(s)
- Yaser Mohammadi
- Qaen School of Nursing and Midwifery, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Zangooei
- Department of Clinical Biochemistry, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahmoud Zardast
- Medical Toxicology and Drug Abuse Research Center, Department of Pathology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Morteza Mamashli
- Department of Clinical Biochemistry, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Azam Rezaei Farimani
- Department of Clinical Biochemistry, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
27
|
Chen Z, Yu H, Chen X, Chen W, Song W, Li Z. Mutual regulation between glycosylation and transforming growth factor-β isoforms signaling pathway. Int J Biol Macromol 2023; 236:123818. [PMID: 36858092 DOI: 10.1016/j.ijbiomac.2023.123818] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/18/2023] [Accepted: 02/19/2023] [Indexed: 03/02/2023]
Abstract
Transforming growth factor-beta (TGF-β) superfamily members orchestrate a wide breadth of biological processes. Through Sma and Mad (Smad)-related dependent or noncanonical pathways, TGF-β members involve in the occurrence and development of many diseases such as cancers, fibrosis, autoimmune diseases, cardiovascular diseases and brain diseases. Glycosylation is one kind of the most common posttranslational modifications on proteins or lipids. Abnormal protein glycosylation can lead to protein malfunction and biological process disorder, thereby causing serious diseases. Previously, researchers commonly make comprehensive systematic overviews on the roles of TGF-β signaling in a specific disease or biological process. In recent years, more and more evidences associate glycosylation modification with TGF-β signaling pathway, and we can no longer disengage and ignore the roles of glycosylation from TGF-β signaling to make investigation. In this review, we provide an overview of current findings involved in glycosylation within TGF-βs and theirs receptors, and the interaction effects between glycosylation and TGF-β subfamily signaling, concluding that there is an intricate mutual regulation between glycosylation and TGF-β signaling, hoping to present the glycosylation regulatory patterns that concealed in TGF-βs signaling pathways.
Collapse
Affiliation(s)
- Zhuo Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Hanjie Yu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Xiangqin Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Wentian Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Wanghua Song
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, PR China.
| |
Collapse
|
28
|
Sibuea S, Ho JK, Pouton CW, Haynes JM. TGFβ3, dibutyryl cAMP and a notch inhibitor modulate phenotype late in stem cell-derived dopaminergic neuron maturation. Front Cell Dev Biol 2023; 11:1111705. [PMID: 36819101 PMCID: PMC9928866 DOI: 10.3389/fcell.2023.1111705] [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: 11/30/2022] [Accepted: 01/19/2023] [Indexed: 02/04/2023] Open
Abstract
The generation of midbrain dopaminergic neurons (mDAs) from pluripotent stem cells (hPSC) holds much promise for both disease modelling studies and as a cell therapy for Parkinson's disease (PD). Generally, dopaminergic neuron differentiation paradigms rely on inhibition of smad signalling for neural induction followed by hedgehog signalling and an elevation of β-catenin to drive dopaminergic differentiation. Post-patterning, differentiating dopaminergic neuron cultures are permitted time for maturation after which the success of these differentiation paradigms is usually defined by expression of tyrosine hydroxylase (TH), the rate limiting enzyme in the synthesis of dopamine. However, during maturation, culture media is often supplemented with additives to promote neuron survival and or promote cell differentiation. These additives include dibutyryl cyclic adenosine monophosphate (dbcAMP), transforming growth factor β3 (TGFβ3) and or the γ-secretase inhibitor (DAPT). While these factors are routinely added to cultures, their impact upon pluripotent stem cell-derived mDA phenotype is largely unclear. In this study, we differentiate pluripotent stem cells toward a dopaminergic phenotype and investigate how the omission of dbcAMP, TGFβ3 or DAPT, late in maturation, affects the regulation of multiple dopaminergic neuron phenotype markers. We now show that the removal of dbcAMP or TGFβ3 significantly and distinctly impacts multiple markers of the mDA phenotype (FOXA2, EN1, EN2, FOXA2, SOX6), while commonly increasing both MSX2 and NEUROD1 and reducing expression of both tyrosine hydroxylase and WNT5A. Removing DAPT significantly impacted MSX2, OTX2, EN1, and KCNJ6. In the absence of any stressful stimuli, we suggest that these culture additives should be viewed as mDA phenotype-modifying, rather than neuroprotective. We also suggest that their addition to cultures is likely to confound the interpretation of both transplantation and disease modelling studies.
Collapse
Affiliation(s)
- Shanti Sibuea
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences Monash University, Parkville, VIC, Australia,National Agency of Drug and Food Control, Jakarta, Indonesia
| | - Joan K. Ho
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences Monash University, Parkville, VIC, Australia
| | - Colin W. Pouton
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences Monash University, Parkville, VIC, Australia
| | - John M. Haynes
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences Monash University, Parkville, VIC, Australia,*Correspondence: John M. Haynes,
| |
Collapse
|
29
|
Liu S, Baeg GH, Yang Y, Goh FG, Bao H, Wagner EJ, Yang X, Cai Y. The Integrator complex desensitizes cellular response to TGF-β/BMP signaling. Cell Rep 2023; 42:112007. [PMID: 36641752 DOI: 10.1016/j.celrep.2023.112007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/12/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Maintenance of stem cells requires the concerted actions of niche-derived signals and stem cell-intrinsic factors. Although Decapentaplegic (Dpp), a Drosophila bone morphogenetic protein (BMP) molecule, can act as a long-range morphogen, its function is spatially limited to the germline stem cell niche in the germarium. We show here that Integrator, a complex known to be involved in RNA polymerase II (RNAPII)-mediated transcriptional regulation in the nucleus, promotes germline differentiation by restricting niche-derived Dpp/BMP activity in the cytoplasm. Further results show that Integrator works in various developmental contexts to desensitize the cellular response to Dpp/BMP signaling during Drosophila development. Mechanistically, our results show that Integrator forms a multi-subunit complex with the type I receptor Thickveins (Tkv) and other Dpp/BMP signaling components and acts in a negative feedback loop to promote Tkv turnover independent of its transcriptional activity. Similarly, human Integrator subunits bind transforming growth factor β (TGF-β)/BMP signaling components and antagonize their activity, suggesting a conserved role of Integrator across metazoans.
Collapse
Affiliation(s)
- Sen Liu
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore
| | - Gyeong Hun Baeg
- Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Ying Yang
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore
| | - Feng Guang Goh
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Hongcun Bao
- The Women's Hospital and Institute of Genetics, School of Medicine, Zhejiang University, Hang Zhou 310058, China
| | - Eric J Wagner
- Department of Biochemistry and Biophysics, Center for RNA Biology, Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, KMRB B.9629, Rochester, NY 14642 USA
| | - Xiaohang Yang
- The Women's Hospital and Institute of Genetics, School of Medicine, Zhejiang University, Hang Zhou 310058, China
| | - Yu Cai
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.
| |
Collapse
|
30
|
Qian Q, Chen Z, Xu J, Zhu Y, Xu W, Gao X, Jiang Q, Zhang X. Pathogenicity of Plesiomonas shigelloides causing mass mortalities of largemouth bass (Micropterus salmoides) and its induced host immune response. FISH & SHELLFISH IMMUNOLOGY 2023; 132:108487. [PMID: 36503060 DOI: 10.1016/j.fsi.2022.108487] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/27/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
The outbreak of mass mortality of M. salmoides occurred in an aquaculture farm in Jiangsu province of China, showing signs of skin ulceration and haemorrhages. The bacteria were isolated from diseased largemouth bass, and identified as Plesiomonas shigelloides based on morphological, physiological and biochemical features, as well as 16S rRNA gene sequence analysis. The pathogenicity of P. shigelloides was determined by challenge experiments, and the median lethal dosage (LD50) of the isolate NJS1 for M. salmoides was calculated as 1.6 × 105 CFU/mL at 7 d post-infection. Histopathological analysis revealed that extensive necrosis, vacuolization and inflammation were presented in the kidney, liver and gill of the diseased fish. Detection of virulence-related genes showed that P. shigelloides NJS1 was positive for astA, astB, astD, astE, actP and 6 ahpA. Additionally, the host defensive response of M. salmoides infected by P. shigelloides was analyzed by quantitive real-time PCR (qRT-PCR), and the results showed that the expression levels of Cas3, Hep1, HIF, IgM, IL15 and TGF were significantly up-regulated in head kidney, liver and spleen in different hours post-infection, which revealed varying expression profiles and clear transcriptional activation of immune related genes. The results suggested that P. shigelloides was an etiological element in the mass mortalities of M. salmoides and this study provided deeper insights for the pathogenesis and host defensive system in P. shigelloides invasion.
Collapse
Affiliation(s)
- Qieqi Qian
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Zhen Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Jingwen Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yujie Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Wenjing Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Xiaojian Gao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Qun Jiang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Xiaojun Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
31
|
Shi S, Xu D, Gu S, Xu N, Xu P, Cao J, Feng XH. Protein tyrosine phosphatase PTPN2 regulates TGF-β signaling through Smad4 dephosphorylation. Am J Cancer Res 2022; 12:5516-5531. [PMID: 36628288 PMCID: PMC9827090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/16/2022] [Indexed: 01/12/2023] Open
Abstract
Transforming Growth Factor beta (TGF-β) is a multifunctional cytokine that regulates cell proliferation, differentiation, and apoptosis. Dysregulation of the TGF-β signaling is one of the major mechanisms underlying tumor progression. We have previously reported that anaplastic lymphoma kinase (ALK) phosphorylates Smad4 at Tyr95, which compromises the DNA-binding ability of Smad4 and thus renders ALK-positive cancer cells resistant to TGF-β tumor-suppressive action. In this study, we demonstrated that tyrosine phosphatase PTPN2 positively regulated TGF-β signaling through dephosphorylating Smad4 at the Tyr95 site. Both in vitro and cell-based assays revealed that PTPN2 bound to and dephosphorylated Smad4, thereby preserving the DNA-binding ability of Smad4. Furthermore, overexpression of PTPN2 restored TGF-β transcriptional and growth inhibitory responses in ALK-positive cancer cells. Consistently, Spermidine, an activator of PTPN2, also promoted TGF-β-induced gene expression, apoptosis, and anti-proliferation effect. Taken together, we revealed that PTPN2 functioned as a tumor suppressor to antagonize the inhibitory effect of tyrosine phosphorylation of Smad4 and to ensure the proper TGF-β growth inhibitory signaling in cancer cells.
Collapse
Affiliation(s)
- Sujing Shi
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China,Center for Life Sciences, Shaoxing Institute, Zhejiang UniversityShaoxing 321000, Zhejiang, China,Cancer Center, Zhejiang UniversityHangzhou 310058, Zhejiang, China
| | - Dewei Xu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China,Center for Life Sciences, Shaoxing Institute, Zhejiang UniversityShaoxing 321000, Zhejiang, China,State Grid Zhejiang Electric Power Co., LtdHangzhou 310007, Zhejiang, China
| | - Shuchen Gu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China,Center for Life Sciences, Shaoxing Institute, Zhejiang UniversityShaoxing 321000, Zhejiang, China,Cancer Center, Zhejiang UniversityHangzhou 310058, Zhejiang, China
| | - Ningyi Xu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China,Center for Life Sciences, Shaoxing Institute, Zhejiang UniversityShaoxing 321000, Zhejiang, China,Westlake UniversityHangzhou 310024, Zhejiang, China
| | - Pinglong Xu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China,Center for Life Sciences, Shaoxing Institute, Zhejiang UniversityShaoxing 321000, Zhejiang, China,Cancer Center, Zhejiang UniversityHangzhou 310058, Zhejiang, China
| | - Jin Cao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China,Center for Life Sciences, Shaoxing Institute, Zhejiang UniversityShaoxing 321000, Zhejiang, China,Cancer Center, Zhejiang UniversityHangzhou 310058, Zhejiang, China
| | - Xin-Hua Feng
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China,Center for Life Sciences, Shaoxing Institute, Zhejiang UniversityShaoxing 321000, Zhejiang, China,Cancer Center, Zhejiang UniversityHangzhou 310058, Zhejiang, China,The Second Affiliated Hospital, Zhejiang UniversityHangzhou 310009, Zhejiang, China
| |
Collapse
|
32
|
Zhang H, Wan GZ, Wang YY, Chen W, Guan JZ. The role of erythrocytes and erythroid progenitor cells in tumors. Open Life Sci 2022; 17:1641-1656. [PMID: 36567722 PMCID: PMC9755711 DOI: 10.1515/biol-2022-0102] [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: 03/01/2022] [Revised: 05/09/2022] [Accepted: 05/30/2022] [Indexed: 12/23/2022] Open
Abstract
In the current research context of precision treatment of malignant tumors, the advantages of immunotherapy are unmatched by conventional antitumor therapy, which can prolong progression-free survival and overall survival. The search for new targets and novel combination therapies can improve the efficacy of immunotherapy and reduce adverse effects. Since current research targets for immunotherapy mainly focus on lymphocytes, little research has been done on erythrocytes. Nucleated erythroid precursor stem cells have been discovered to play an essential role in tumor progression. Researchers are exploring new targets and therapeutic approaches for immunotherapy from the perspective of erythroid progenitor cells (EPCs). Recent studies have shown that different subtypes of EPCs have specific surface markers and distinct biological roles in tumor immunity. CD45+ EPCs are potent myeloid-derived suppressor cell-like immunosuppressants that reduce the patient's antitumor immune response. CD45- EPCs promote tumor invasion and metastasis by secreting artemin. A specific type of EPC also promotes angiogenesis and provides radiation protection. Therefore, EPCs may be involved in tumor growth, infiltration, and metastasis. It may also be an important cause of anti-angiogenesis and immunotherapy resistance. This review summarizes recent research advances in erythropoiesis, EPC features, and their impacts and processes on tumors.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People’s Liberation Army) General Hospital, Beijing 100091, China,Department of Oncology, The Eighth Medical Center, Chinese PLA (People’s Liberation Army) General Hospital, Beijing 100071, China,Postgraduate Department of Hebei North University, Zhangjiakou 075000, China
| | - Guang-zhi Wan
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People’s Liberation Army) General Hospital, Beijing 100071, China
| | - Yu-ying Wang
- Department of Oncology, First Medical Center, Chinese PLA (People’s Liberation Army) General Hospital, Beijing, China
| | - Wen Chen
- Department of Pathology, The Eighth Medical Center, Chinese PLA (People’s Liberation Army) General Hospital, Beijing 100091, China
| | - Jing-Zhi Guan
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People’s Liberation Army) General Hospital, Beijing 100071, China
| |
Collapse
|
33
|
Jhuang YL, Yang CW, Tseng YF, Hsu CL, Li HY, Yuan RH, Jeng YM. SIN3-HDAC complex-associated factor, a chromatin remodelling gene located in the 12p amplicon, is a potential germ cell tumour-specific oncogene. J Pathol 2022; 258:353-365. [PMID: 36056608 DOI: 10.1002/path.6007] [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: 03/30/2022] [Revised: 08/20/2022] [Accepted: 08/30/2022] [Indexed: 01/27/2023]
Abstract
A genetic hallmark of malignant germ cell tumours (GCTs) is isochromosome 12p, but oncogenes located in 12p that are specifically expressed in GCT have not yet been identified. SIN3-HDAC complex-associated factor (SINHCAF) is a subunit of the Sin3/histone deacetylase (HDAC) complex, and it defines a Sin3a-Hdac complex variant that is required for the self-renewal of mouse embryonic stem cells. This study demonstrated that SINHCAF is expressed in a vast majority of malignant GCTs and is rarely expressed in somatic malignancy. Fluorescence in situ hybridisation revealed SINHCAF amplification in malignant GCTs. SINHCAF silencing using shRNA reduced anchorage-dependent cell proliferation and tumoursphere formation and inhibited tumour cell migration and invasion in GCT cell lines. Moreover, in the GCT cell line NTERA2/D1, SINHCAF silencing inhibited the expression of genes associated with embryonic stem cells and induced the expression of genes associated with neuronal and white fat cell differentiation. Compared with somatic cell lines, GCT cell lines were more susceptible to HDAC inhibitor treatment. Thus, we identified SINHCAF to be a potential oncogene located in the amplicon of chromosome 12p and showed that SINHCAF was specifically expressed in malignant GCTs. HDAC inhibitor treatment may counteract the oncogenic activity of SINHCAF and is a promising therapeutic approach for GCTs. © 2022 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yu-Ling Jhuang
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan.,Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Wei Yang
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
| | - Yu-Fen Tseng
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Huei-Ying Li
- Medical Microbiota Center of the First Core Laboratory, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ray-Hwang Yuan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.,Department of Surgery, National Taiwan University Hospital, Hsinchu Branch, Hsinchu, Taiwan
| | - Yung-Ming Jeng
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan.,Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
34
|
Setiawan AM, Kamarudin TA, Abd Ghafar N. The role of BMP4 in adipose-derived stem cell differentiation: A minireview. Front Cell Dev Biol 2022; 10:1045103. [PMID: 36340030 PMCID: PMC9634734 DOI: 10.3389/fcell.2022.1045103] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/11/2022] [Indexed: 12/02/2022] Open
Abstract
Bone morphogenetic protein 4 (BMP4) is a member of the transforming growth factor beta (TGF-β) superfamily of cytokines responsible for stem cells’ commitment to differentiation, proliferation, and maturation. To date, various studies have utilized BMP4 as a chemical inducer for in vitro differentiation of human mesenchymal stem cells (MSCs) based on its potential. BMP4 drives in vitro differentiation of ADSC via TGF-β signaling pathway by interactions with BMP receptors leading to the activation of smad-dependent and smad-independent pathways. The BMP4 signaling pathways are regulated by intracellular and extracellular BMP4 antagonists. Extracellular BMP4 antagonist prevents interaction between BMP4 ligand to its receptors, while intracellular BMP4 antagonist shutdowns the smad-dependent pathways through multiple mechanisms. BMP4 proved as one of the popular differentiation factors to induce ADSC differentiation into cell from mesodermal origin. However, addition of all-trans retinoic acid is also needed in trans-differentiation of ADSC into ectodermal lineage cells. Suggesting that both BMP4 and RA signaling pathways may be necessary to be activated for in vitro trans-differentiation of ADSC.
Collapse
Affiliation(s)
- Abdul Malik Setiawan
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Department of Anatomy, Maulana Malik Ibrahim State Islamic University, Malang, Indonesia
| | - Taty Anna Kamarudin
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- *Correspondence: Taty Anna Kamarudin,
| | - Norzana Abd Ghafar
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
35
|
Venkatesan M, Semper C, Skrivergaard S, Di Leo R, Mesa N, Rasmussen MK, Young JF, Therkildsen M, Stogios PJ, Savchenko A. Recombinant production of growth factors for application in cell culture. iScience 2022; 25:105054. [PMID: 36157583 PMCID: PMC9489951 DOI: 10.1016/j.isci.2022.105054] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/07/2022] [Accepted: 08/26/2022] [Indexed: 11/04/2022] Open
Abstract
Culturing eukaryotic cells has widespread applications in research and industry, including the emerging field of cell-cultured meat production colloquially referred to as “cellular agriculture”. These applications are often restricted by the high cost of growth medium necessary for cell growth. Mitogenic protein growth factors (GFs) are essential components of growth medium and account for upwards of 90% of the total costs. Here, we present a set of expression constructs and a simplified protocol for recombinant production of functionally active GFs, including FGF2, IGF1, PDGF-BB, and TGF-β1 in Escherichia coli. Using this E. coli expression system, we produced soluble GF orthologs from species including bovine, chicken, and salmon. Bioactivity analysis revealed orthologs with improved performance compared to commercially available alternatives. We estimated that the production cost of GFs using our methodology will significantly reduce the cost of cell culture medium, facilitating low-cost protocols tailored for cultured meat production and tissue engineering.
Developed methodology for low-cost production of soluble, bioactive GFs Purified GFs were active on NIH-3T3 and bovine satellite cells Some GF orthologs outperformed commercially sourced GFs Production of GFs using these methods can foster significant cost savings
Collapse
Affiliation(s)
- Meenakshi Venkatesan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada
| | - Cameron Semper
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Rosa Di Leo
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada
| | - Nathalie Mesa
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada
| | | | | | | | - Peter J Stogios
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada
| | - Alexei Savchenko
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
36
|
Duan Z, Lin X, Wang L, Zhen Q, Jiang Y, Chen C, Yang J, Lee CH, Qin Y, Li Y, Zhao B, Wang J, Zhang Z. Specificity of TGF-β1 signal designated by LRRC33 and integrin α Vβ 8. Nat Commun 2022; 13:4988. [PMID: 36008481 PMCID: PMC9411592 DOI: 10.1038/s41467-022-32655-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/03/2022] [Indexed: 12/20/2022] Open
Abstract
Myeloid lineage cells present the latent form of transforming growth factor-β1 (L-TGF-β1) to the membrane using an anchor protein LRRC33. Integrin αVβ8 activates extracellular L-TGF-β1 to trigger the downstream signaling functions. However, the mechanism designating the specificity of TGF-β1 presentation and activation remains incompletely understood. Here, we report cryo-EM structures of human L-TGF-β1/LRRC33 and integrin αVβ8/L-TGF-β1 complexes. Combined with biochemical and cell-based analyses, we demonstrate that LRRC33 only presents L-TGF-β1 but not the -β2 or -β3 isoforms due to difference of key residues on the growth factor domains. Moreover, we reveal a 2:2 binding mode of integrin αVβ8 and L-TGF-β1, which shows higher avidity and more efficient L-TGF-β1 activation than previously reported 1:2 binding mode. We also uncover that the disulfide-linked loop of the integrin subunit β8 determines its exquisite affinity to L-TGF-β1. Together, our findings provide important insights into the specificity of TGF-β1 signaling achieved by LRRC33 and integrin αVβ8.
Collapse
Affiliation(s)
- Zelin Duan
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Xuezhen Lin
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, 518107, Shenzhen, Guangdong, China
| | - Lixia Wang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, 518107, Shenzhen, Guangdong, China
| | - Qiuxin Zhen
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Yuefeng Jiang
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Chuxin Chen
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Chia-Hsueh Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yan Qin
- Parthenon Therapeutics, 40 Guest street, Boston, MA, 02135, USA
| | - Ying Li
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, 518107, Shenzhen, Guangdong, China
| | - Bo Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, 518107, Shenzhen, Guangdong, China.
| | - Jianchuan Wang
- Center for Translational Research, Shenzhen Bay Laboratory, 518007, Shenzhen, Guangdong, China.
| | - Zhe Zhang
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Peking University, 100871, Beijing, China.
| |
Collapse
|
37
|
Takano C, Horie M, Taiko I, Trinh QD, Kanemaru K, Komine-Aizawa S, Hayakawa S, Miki T. Inhibition of Epithelial-Mesenchymal Transition Maintains Stemness in Human Amniotic Epithelial Cells. Stem Cell Rev Rep 2022; 18:3083-3091. [PMID: 35931939 DOI: 10.1007/s12015-022-10420-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 10/15/2022]
Abstract
Human amniotic epithelial cells (hAECs), which are a type of placental stem cell, express stem cell marker genes and are capable of differentiating into all three germ layers under appropriate culture conditions. hAECs are known to undergo TGF-β-dependent epithelial-mesenchymal transition (EMT); however, the impact of EMT on the stemness or differentiation of hAECs has not yet been determined. Here, we first confirmed that hAECs undergo EMT immediately after starting primary culture. Comprehensive transcriptome analysis using RNA-seq revealed that inhibition of TGF-β-dependent EMT maintained the expression of stemness-related genes, including NANOG and POU5F1, in hAECs. Moreover, the maintenance of stemness did not affect the nontumorigenic characteristics of hAECs. We showed for the first time that TGF-β-dependent EMT negatively affected the stemness of hAECs, providing novel insight into cellular processes of placental stem cells.
Collapse
Affiliation(s)
- Chika Takano
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.,Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Masafumi Horie
- Department of Molecular and Cellular Pathology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Isamu Taiko
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Kazunori Kanemaru
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Toshio Miki
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan.
| |
Collapse
|
38
|
Hachana S, Larrivée B. TGF-β Superfamily Signaling in the Eye: Implications for Ocular Pathologies. Cells 2022; 11:2336. [PMID: 35954181 PMCID: PMC9367584 DOI: 10.3390/cells11152336] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/06/2023] Open
Abstract
The TGF-β signaling pathway plays a crucial role in several key aspects of development and tissue homeostasis. TGF-β ligands and their mediators have been shown to be important regulators of ocular physiology and their dysregulation has been described in several eye pathologies. TGF-β signaling participates in regulating several key developmental processes in the eye, including angiogenesis and neurogenesis. Inadequate TGF-β signaling has been associated with defective angiogenesis, vascular barrier function, unfavorable inflammatory responses, and tissue fibrosis. In addition, experimental models of corneal neovascularization, diabetic retinopathy, proliferative vitreoretinopathy, glaucoma, or corneal injury suggest that aberrant TGF-β signaling may contribute to the pathological features of these conditions, showing the potential of modulating TGF-β signaling to treat eye diseases. This review highlights the key roles of TGF-β family members in ocular physiology and in eye diseases, and reviews approaches targeting the TGF-β signaling as potential treatment options.
Collapse
Affiliation(s)
- Soumaya Hachana
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Bruno Larrivée
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
39
|
Prud’homme GJ, Kurt M, Wang Q. Pathobiology of the Klotho Antiaging Protein and Therapeutic Considerations. FRONTIERS IN AGING 2022; 3:931331. [PMID: 35903083 PMCID: PMC9314780 DOI: 10.3389/fragi.2022.931331] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 12/06/2022]
Abstract
The α-Klotho protein (henceforth denoted Klotho) has antiaging properties, as first observed in mice homozygous for a hypomorphic Klotho gene (kl/kl). These mice have a shortened lifespan, stunted growth, renal disease, hyperphosphatemia, hypercalcemia, vascular calcification, cardiac hypertrophy, hypertension, pulmonary disease, cognitive impairment, multi-organ atrophy and fibrosis. Overexpression of Klotho has opposite effects, extending lifespan. In humans, Klotho levels decline with age, chronic kidney disease, diabetes, Alzheimer’s disease and other conditions. Low Klotho levels correlate with an increase in the death rate from all causes. Klotho acts either as an obligate coreceptor for fibroblast growth factor 23 (FGF23), or as a soluble pleiotropic endocrine hormone (s-Klotho). It is mainly produced in the kidneys, but also in the brain, pancreas and other tissues. On renal tubular-cell membranes, it associates with FGF receptors to bind FGF23. Produced in bones, FGF23 regulates renal excretion of phosphate (phosphaturic effect) and vitamin D metabolism. Lack of Klotho or FGF23 results in hyperphosphatemia and hypervitaminosis D. With age, human renal function often deteriorates, lowering Klotho levels. This appears to promote age-related pathology. Remarkably, Klotho inhibits four pathways that have been linked to aging in various ways: Transforming growth factor β (TGF-β), insulin-like growth factor 1 (IGF-1), Wnt and NF-κB. These can induce cellular senescence, apoptosis, inflammation, immune dysfunction, fibrosis and neoplasia. Furthermore, Klotho increases cell-protective antioxidant enzymes through Nrf2 and FoxO. In accord, preclinical Klotho therapy ameliorated renal, cardiovascular, diabetes-related and neurodegenerative diseases, as well as cancer. s-Klotho protein injection was effective, but requires further investigation. Several drugs enhance circulating Klotho levels, and some cross the blood-brain barrier to potentially act in the brain. In clinical trials, increased Klotho was noted with renin-angiotensin system inhibitors (losartan, valsartan), a statin (fluvastatin), mTOR inhibitors (rapamycin, everolimus), vitamin D and pentoxifylline. In preclinical work, antidiabetic drugs (metformin, GLP-1-based, GABA, PPAR-γ agonists) also enhanced Klotho. Several traditional medicines and/or nutraceuticals increased Klotho in rodents, including astaxanthin, curcumin, ginseng, ligustilide and resveratrol. Notably, exercise and sport activity increased Klotho. This review addresses molecular, physiological and therapeutic aspects of Klotho.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
- *Correspondence: Gérald J. Prud’homme,
| | - Mervé Kurt
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Yinuo Pharmaceutical Co., Ltd., Shanghai, China
| |
Collapse
|
40
|
Liu Z, Hu X, Liang Y, Yu J, Li H, Shokhirev MN, Zheng Y. Glucocorticoid signaling and regulatory T cells cooperate to maintain the hair-follicle stem-cell niche. Nat Immunol 2022; 23:1086-1097. [PMID: 35739197 PMCID: PMC9283297 DOI: 10.1038/s41590-022-01244-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/17/2022] [Indexed: 01/03/2023]
Abstract
Maintenance of tissue homeostasis is dependent on the communication between stem cells and supporting cells in the same niche. Regulatory T cells (Treg cells) are emerging as a critical component of the stem-cell niche for supporting their differentiation. How Treg cells sense dynamic signals in this microenvironment and communicate with stem cells is mostly unknown. In the present study, by using hair follicles (HFs) to study Treg cell-stem cell crosstalk, we show an unrecognized function of the steroid hormone glucocorticoid in instructing skin-resident Treg cells to facilitate HF stem-cell (HFSC) activation and HF regeneration. Ablation of the glucocorticoid receptor (GR) in Treg cells blocks hair regeneration without affecting immune homeostasis. Mechanistically, GR and Foxp3 cooperate in Treg cells to induce transforming growth factor β3 (TGF-β3), which activates Smad2/3 in HFSCs and facilitates HFSC proliferation. The present study identifies crosstalk between Treg cells and HFSCs mediated by the GR-TGF-β3 axis, highlighting a possible means of manipulating Treg cells to support tissue regeneration.
Collapse
Affiliation(s)
- Zhi Liu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Xianting Hu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Otolaryngology Head and Neck Surgery, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Yuqiong Liang
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jingting Yu
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Huabin Li
- Department of Otolaryngology Head and Neck Surgery, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ye Zheng
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
41
|
Kim JG, Rim YA, Ju JH. The Role of Transforming Growth Factor Beta in Joint Homeostasis and Cartilage Regeneration. Tissue Eng Part C Methods 2022; 28:570-587. [PMID: 35331016 DOI: 10.1089/ten.tec.2022.0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) is an important regulator of joint homeostasis, of which dysregulation is closely associated with the development of osteoarthritis (OA). In normal conditions, its biological functions in a joint environment are joint protective, but it can be dramatically altered in different contexts, making its therapeutic application a challenge. However, with the deeper insights into the TGF-β functions, it has been proven that TGF-β augments cartilage regeneration by chondrocytes, and differentiates both the precursor cells of chondrocytes and stem cells into cartilage-generating chondrocytes. Following documentation of the therapeutic efficacy of chondrocytes augmented by TGF-β in the last decade, there is an ongoing phase III clinical trial examining the therapeutic efficacy of a mixture of allogeneic chondrocytes and TGF-β-overexpressing cells. To prepare cartilage-restoring chondrocytes from induced pluripotent stem cells (iPSCs), the stem cells are differentiated mainly using TGF-β with some other growth factors. Of note, clinical trials evaluating the therapeutic efficacy of iPSCs for OA are scheduled this year. Mesenchymal stromal stem cells (MSCs) have inherent limitations in that they differentiate into the osteochondral pathway, resulting in the production of poor-quality cartilage. Despite the established essential role of TGF-β in chondrogenic differentiation of MSCs, whether the coordinated use of TGF-β in MSC-based therapy for degenerated cartilage is effective is unknown. We herein reviewed the general characteristics and mechanism of action of TGF-β in a joint environment. Furthermore, we discussed the core interaction of TGF-β with principal cells of OA cell-based therapies, the chondrocytes, MSCs, and iPSCs. Impact Statement Transforming growth factor-beta (TGF-β) has been widely used as a core regulator to improve or formulate therapeutic regenerative cells for degenerative joints. It differentiates stem cells into chondrocytes and improves the chondrogenic potential of differentiated chondrocytes. Herein, we discussed the overall characteristics of TGF-β and reviewed the comprehension and utilization of TGF-β in cell-based therapy for degenerative joint disease.
Collapse
Affiliation(s)
- Jung Gon Kim
- Division of Rheumatology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
42
|
Närvä E, Taskinen ME, Lilla S, Isomursu A, Pietilä M, Weltner J, Isola J, Sihto H, Joensuu H, Zanivan S, Norman J, Ivaska J. MASTL is enriched in cancerous and pluripotent stem cells and influences OCT1/OCT4 levels. iScience 2022; 25:104459. [PMID: 35677646 PMCID: PMC9167974 DOI: 10.1016/j.isci.2022.104459] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/04/2022] [Accepted: 05/13/2022] [Indexed: 11/01/2022] Open
Abstract
MASTL is a mitotic accelerator with an emerging role in breast cancer progression. However, the mechanisms behind its oncogenicity remain largely unknown. Here, we identify a previously unknown role and eminent expression of MASTL in stem cells. MASTL staining from a large breast cancer patient cohort indicated a significant association with β3 integrin, an established mediator of breast cancer stemness. MASTL silencing reduced OCT4 levels in human pluripotent stem cells and OCT1 in breast cancer cells. Analysis of the cell-surface proteome indicated a strong link between MASTL and the regulation of TGF-β receptor II (TGFBR2), a key modulator of TGF-β signaling. Overexpression of wild-type and kinase-dead MASTL in normal mammary epithelial cells elevated TGFBR2 levels. Conversely, MASTL depletion in breast cancer cells attenuated TGFBR2 levels and downstream signaling through SMAD3 and AKT pathways. Taken together, these results indicate that MASTL supports stemness regulators in pluripotent and cancerous stem cells.
Collapse
Affiliation(s)
- Elisa Närvä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Maria E. Taskinen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | | | - Aleksi Isomursu
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Mika Pietilä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Jere Weltner
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Jorma Isola
- Laboratory of Cancer Biology, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Harri Sihto
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| | - Heikki Joensuu
- University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Sara Zanivan
- CRUK Beatson Institute, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Jim Norman
- CRUK Beatson Institute, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
- Department of Life Technologies, University of Turku, 20520 Turku, Finland
- Western Finnish Cancer Center (FICAN West), University of Turku, 20520 Turku, Finland
- Foundation for the Finnish Cancer Institute, Tukholmankatu 8, Helsinki, Finland
| |
Collapse
|
43
|
Fink M, Wrana JL. Regulation of homeostasis and regeneration in the adult intestinal epithelium by the TGF-β superfamily. Dev Dyn 2022; 252:445-462. [PMID: 35611490 DOI: 10.1002/dvdy.500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
The delicate balance between the homeostatic maintenance and regenerative capacity of the intestine makes this a fascinating tissue of study. The intestinal epithelium undergoes continuous homeostatic renewal but is also exposed to a diverse array of stresses that can range from physiological processes such as digestion, to exposure to infectious agents, drugs, radiation therapy, and inflammatory stimuli. The intestinal epithelium has thus evolved to efficiently maintain and reinstate proper barrier function that is essential for intestinal integrity and function. Factors governing homeostatic epithelial turnover are well described, however, the dynamic regenerative mechanisms that occur following injury are the subject of intense ongoing investigations. The TGF-β superfamily is a key regulator of both homeostatic renewal and regenerative processes of the intestine. Here we review the roles of TGF-β and BMP on the adult intestinal epithelium during self-renewal and injury to provide a framework for understanding how this major family of morphogens can tip the scale between intestinal health and disease. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mardi Fink
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey L Wrana
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
44
|
Tsubakihara Y, Ohata Y, Okita Y, Younis S, Eriksson J, Sellin ME, Ren J, Ten Dijke P, Miyazono K, Hikita A, Imamura T, Kato M, Heldin CH, Moustakas A. TGFβ selects for pro-stemness over pro-invasive phenotypes during cancer cell epithelial-mesenchymal transition. Mol Oncol 2022; 16:2330-2354. [PMID: 35348275 PMCID: PMC9208077 DOI: 10.1002/1878-0261.13215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/07/2022] [Accepted: 03/24/2022] [Indexed: 11/08/2022] Open
Abstract
Transforming growth factor β (TGFβ) induces epithelial-mesenchymal transition (EMT), which correlates with stemness and invasiveness. Mesenchymal-epithelial transition (MET) is induced by TGFβ withdrawal and correlates with metastatic colonization. Whether TGFβ promotes stemness and invasiveness simultaneously via EMT remains unclear. We established a breast cancer cell model expressing red fluorescent protein (RFP) under the E-cadherin promoter. In 2D cultures, TGFβ induced EMT, generating RFPlow cells with a mesenchymal transcriptome, and regained RFP, with an epithelial transcriptome, after MET induced by TGFβ withdrawal. RFPlow cells generated robust mammospheres, with epithelio-mesenchymal cell surface features. Mammospheres that were forced to adhere generated migratory cells, devoid of RFP, a phenotype which was inhibited by a TGFβ receptor kinase inhibitor. Further stimulation of RFPlow mammospheres with TGFβ suppressed the generation of motile cells, but enhanced mammosphere growth. Accordingly, mammary fat-pad-transplanted mammospheres, in the absence of exogenous TGFβ treatment, established lung metastases with evident MET (RFPhigh cells). In contrast, TGFβ-treated mammospheres revealed high tumor-initiating capacity, but limited metastatic potential. Thus, the biological context of partial EMT and MET allows TGFβ to differentiate between pro-stemness and pro-invasive phenotypes.
Collapse
Affiliation(s)
- Yutaro Tsubakihara
- Dept. of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden.,Dept. of Experimental Pathology and Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Japan
| | - Yae Ohata
- Dept. of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden
| | - Yukari Okita
- Dept. of Experimental Pathology and Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Japan
| | - Shady Younis
- Dept. of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden.,Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Jens Eriksson
- Dept. of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden
| | - Mikael E Sellin
- Dept. of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden
| | - Jiang Ren
- Dept. of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Dept. of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Kohei Miyazono
- Dept. of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Atsuhiko Hikita
- Div. of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takeshi Imamura
- Dept. of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Mitsuyasu Kato
- Dept. of Experimental Pathology and Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Japan
| | - Carl-Henrik Heldin
- Dept. of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden
| | - Aristidis Moustakas
- Dept. of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden
| |
Collapse
|
45
|
Givelet M, Firlej V, Lassalle B, Gille AS, Lapoujade C, Holtzman I, Jarysta A, Haghighirad F, Dumont F, Jacques S, Letourneur F, Pflumio F, Allemand I, Patrat C, Thiounn N, Wolf JP, Riou L, Barraud-Lange V, Fouchet P. Transcriptional profiling of β-2M -SPα-6 +THY1 + spermatogonial stem cells in human spermatogenesis. Stem Cell Reports 2022; 17:936-952. [PMID: 35334216 PMCID: PMC9023810 DOI: 10.1016/j.stemcr.2022.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022] Open
Abstract
Male infertility is responsible for approximately half of all cases of reproductive issues. Spermatogenesis originates in a small pool of spermatogonial stem cells (SSCs), which are of interest for therapy of infertility but remain not well defined in humans. Using multiparametric analysis of the side population (SP) phenotype and the α-6 integrin, THY1, and β-2 microglobulin cell markers, we identified a population of human primitive undifferentiated spermatogonia with the phenotype β-2 microglobulin (β-2M)−SPα-6+THY1+, which is highly enriched in stem cells. By analyzing the expression signatures of this SSC-enriched population along with other germinal progenitors, we established an exhaustive transcriptome of human spermatogenesis. Transcriptome profiling of the human β-2M−SPα-6+THY1+ population and comparison with the profile of mouse undifferentiated spermatogonia provide insights into the molecular networks and key transcriptional regulators regulating human SSCs, including the basic-helix-loop-helix (bHLH) transcriptional repressor HES1, which we show to be implicated in maintenance of SSCs in vitro.
Human β-2M−SPα-6+THY1+ undifferentiated spermatogonia are enriched in stem cells Comparative transcriptomics analysis of human and murine spermatogonia HES1 is involved in the physiology of SSCs in vitro
Collapse
Affiliation(s)
- Maelle Givelet
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France; Institut Cochin, INSERM U1016, Département de Génétique, Développement et Cancer, Équipe Génomique Epigénétique et Physiopathologie de la Reproduction, 75014 Paris, France
| | - Virginie Firlej
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France; Institut Cochin, INSERM U1016, Département de Génétique, Développement et Cancer, Équipe Génomique Epigénétique et Physiopathologie de la Reproduction, 75014 Paris, France
| | - Bruno Lassalle
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France
| | - Anne Sophie Gille
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France; Institut Cochin, INSERM U1016, Département de Génétique, Développement et Cancer, Équipe Génomique Epigénétique et Physiopathologie de la Reproduction, 75014 Paris, France
| | - Clementine Lapoujade
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France
| | - Isabelle Holtzman
- Institut Cochin, INSERM U1016, Département de Génétique, Développement et Cancer, Équipe Génomique Epigénétique et Physiopathologie de la Reproduction, 75014 Paris, France
| | - Amandine Jarysta
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France
| | - Farahd Haghighirad
- UFR Médecine Paris Centre-Université de Paris, 15 rue de l'école de Médecine, 75006 Paris, France
| | - Florent Dumont
- Université Paris Saclay, UMS IPSIT, 92296 Châtenay-Malabry, France
| | - Sébastien Jacques
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS UMR8104, Plateforme Séquençage et Génomique, 75014 Paris, France
| | - Franck Letourneur
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS UMR8104, Plateforme Séquençage et Génomique, 75014 Paris, France
| | - Françoise Pflumio
- Université de Paris and Université Paris-Saclay, INSERM, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, LSHL, 92265 Fontenay-aux-Roses, France
| | - Isabelle Allemand
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France
| | - Catherine Patrat
- UFR Médecine Paris Centre-Université de Paris, 15 rue de l'école de Médecine, 75006 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, CHU Cochin, Histologie-Embryologie-Biologie de la Reproduction, 75014 Paris, France
| | - Nicolas Thiounn
- Department of urology and transplant surgery, Hôpital européen Georges-Pompidou, AP-HP, Université de Paris, 20 rue Leblanc, 75015 Paris, France
| | - Jean Philippe Wolf
- UFR Médecine Paris Centre-Université de Paris, 15 rue de l'école de Médecine, 75006 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, CHU Cochin, Histologie-Embryologie-Biologie de la Reproduction, 75014 Paris, France
| | - Lydia Riou
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France
| | - Virginie Barraud-Lange
- UFR Médecine Paris Centre-Université de Paris, 15 rue de l'école de Médecine, 75006 Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, CHU Cochin, Histologie-Embryologie-Biologie de la Reproduction, 75014 Paris, France
| | - Pierre Fouchet
- Université de Paris and Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
46
|
Holtzer L, Wesseling-Rozendaal Y, Verhaegh W, van de Stolpe A. Measurement of activity of developmental signal transduction pathways to quantify stem cell pluripotency and phenotypically characterize differentiated cells. Stem Cell Res 2022; 61:102748. [PMID: 35325817 DOI: 10.1016/j.scr.2022.102748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/28/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022] Open
Abstract
Important challenges in stem cell research and regenerative medicine are reliable assessment of pluripotency state and purity of differentiated cell populations. Pluripotency and differentiation are regulated and determined by activity of developmental signal transduction pathways (STPs). To date activity of these STPs could not be directly measured on a cell sample. Here we validate a novel assay platform for measurement of activity of developmental STPs (STP) for use in stem cells and stem cell derivatives. In addition to previously developed STP assays, we report development of an additional STP assay for the MAPK-AP1 pathway. Subsequently, activity of Notch, Hedgehog, TGFβ, Wnt, PI3K, MAPK-AP1, and NFκB signaling pathways was calculated from Affymetrix transcriptome data of human pluripotent embryonic (hES) and iPS cell lines under different culture conditions, organ-derived multipotent stem cells, and differentiated cell types, to generate quantitative STP activity profiles. Results show that the STP assay technology enables reliable and quantitative measurement of multiple STP activities simultaneously on any individual cell sample. Using the technology, we found that culture conditions dominantly influence the pluripotent stem cell STP activity profile, while the origin of the stem cell line was a minor variable. A pluripotency STP activity profile (Pluripotency qPAP) was defined (active PI3K, MAPK, Hedgehog, Notch, TGFβ, and NFκB pathway, inactive Wnt pathway). Differentiation of hES cells to intestinal progenitor cells resulted in an STP activity profile characterized by active PI3K, Wnt and Notch pathways, comparable to the STP activity profile measured on primary intestinal crypt stem cells. Quantitative STP activity measurement is expected to improve experimental reproducibility and standardization of pluripotent and multipotent stem cell culture/differentiation, and enable controlled manipulation of pluripotency/differentiation state using pathway targeting compounds.
Collapse
Affiliation(s)
- Laurent Holtzer
- Molecular Pathway Diagnostics, Philips, Eindhoven, The Netherlands.
| | | | - Wim Verhaegh
- Molecular Pathway Diagnostics, Philips, Eindhoven, The Netherlands.
| | | |
Collapse
|
47
|
Aydin O, Passaro AP, Raman R, Spellicy SE, Weinberg RP, Kamm RD, Sample M, Truskey GA, Zartman J, Dar RD, Palacios S, Wang J, Tordoff J, Montserrat N, Bashir R, Saif MTA, Weiss R. Principles for the design of multicellular engineered living systems. APL Bioeng 2022; 6:010903. [PMID: 35274072 PMCID: PMC8893975 DOI: 10.1063/5.0076635] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/02/2022] [Indexed: 12/14/2022] Open
Abstract
Remarkable progress in bioengineering over the past two decades has enabled the formulation of fundamental design principles for a variety of medical and non-medical applications. These advancements have laid the foundation for building multicellular engineered living systems (M-CELS) from biological parts, forming functional modules integrated into living machines. These cognizant design principles for living systems encompass novel genetic circuit manipulation, self-assembly, cell-cell/matrix communication, and artificial tissues/organs enabled through systems biology, bioinformatics, computational biology, genetic engineering, and microfluidics. Here, we introduce design principles and a blueprint for forward production of robust and standardized M-CELS, which may undergo variable reiterations through the classic design-build-test-debug cycle. This Review provides practical and theoretical frameworks to forward-design, control, and optimize novel M-CELS. Potential applications include biopharmaceuticals, bioreactor factories, biofuels, environmental bioremediation, cellular computing, biohybrid digital technology, and experimental investigations into mechanisms of multicellular organisms normally hidden inside the "black box" of living cells.
Collapse
Affiliation(s)
| | - Austin P. Passaro
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia 30602, USA
| | - Ritu Raman
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | - Robert P. Weinberg
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, Massachusetts 02115, USA
| | | | - Matthew Sample
- Center for Ethics and Law in the Life Sciences, Leibniz Universität Hannover, 30167 Hannover, Germany
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Jeremiah Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Roy D. Dar
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Sebastian Palacios
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Jason Wang
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Jesse Tordoff
- Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Nuria Montserrat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | | | - M. Taher A. Saif
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Ron Weiss
- Author to whom correspondence should be addressed:
| |
Collapse
|
48
|
Assessment of Mineralization, Oxidative Stress, and Inflammation Mechanisms in the Pulp of Primary Teeth. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031554] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Inflammation in primary teeth (PT) is commonly associated with a lower sensibility to painful stimuli, compared to permanent teeth, and usually leads to late presentation for dental treatment. Data obtained on the molecular assessments of dental pulp and clinical examinations could guide practitioners to conduct precise diagnoses and correct treatments. The aim of our pilot study was to assess the levels of several biomarkers (e.g., mineralization, oxidative stress, and inflammation) in primary teeth. The research included 46 dental pulp specimens collected from the primary teeth of children and adolescents between the ages of 6 and 12. The experimental groups consisted of 18 samples collected from primary teeth with acute pulpitis and 15 samples from chronically inflamed pulp tissues. The control group was represented by 13 specimens acquired from clinically healthy primary teeth. The enzyme-linked immunosorbent assay (ELISA) technique was used to determine the protein expression of tumor necrosis factor-α (TNF-α), superoxide dismutase-3 (SOD-3), osteocalcin, and transforming growth factor-β1 (TGF-β1) in the lysates. Our results revealed that all of the studied parameters presented statistically significant (p ≤ 0.05) increased levels in both experimental groups compared to the control samples. Furthermore, osteocalcin presented statistically significant increased concentrations in chronically- versus acute-inflamed pulp samples (p ≤ 0.05). The studied molecules may have an influential role in acute and chronic pulp inflammation in primary teeth.
Collapse
|
49
|
Weng Z, Wang Y, Ouchi T, Liu H, Qiao X, Wu C, Zhao Z, Li L, Li B. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:356-371. [PMID: 35485439 PMCID: PMC9052415 DOI: 10.1093/stcltm/szac004] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/19/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
| | | | - Takehito Ouchi
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xianghe Qiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Chenzhou Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Bo Li
- Corresponding author: Bo Li, DDS, PhD, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No.14, 3rd Section of Ren Min Nan Rd. Chengdu, Sichuan 610041, People’s Republic of China.
| |
Collapse
|
50
|
Connexin 43 Gene Ablation Does Not Alter Human Pluripotent Stem Cell Germ Lineage Specification. Biomolecules 2021; 12:biom12010015. [PMID: 35053163 PMCID: PMC8773696 DOI: 10.3390/biom12010015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/23/2023] Open
Abstract
During embryonic germ layer development, cells communicate with each other and their environment to ensure proper lineage specification and tissue development. Connexin (Cx) proteins facilitate direct cell–cell communication through gap junction channels. While previous reports suggest that gap junctional intercellular communication may contribute to germ layer formation, there have been limited comprehensive expression analyses or genetic ablation studies on Cxs during human pluripotent stem cell (PSC) germ lineage specification. We screened the mRNA profile and protein expression patterns of select human Cx isoforms in undifferentiated human induced pluripotent stem cells (iPSCs), and after directed differentiation into the three embryonic germ lineages: ectoderm, definitive endoderm, and mesoderm. Transcript analyses by qPCR revealed upregulation of Cx45 and Cx62 in iPSC-derived ectoderm; Cx45 in mesoderm; and Cx30.3, Cx31, Cx32, Cx36, Cx37, and Cx40 in endoderm relative to control human iPSCs. Generated Cx43 (GJA1) CRISPR-Cas9 knockout iPSCs successfully differentiated into cells of all three germ layers, suggesting that Cx43 is dispensable during directed iPSC lineage specification. Furthermore, qPCR screening of select Cx transcripts in our GJA1-/- iPSCs showed no significant Cx upregulation in response to the loss of Cx43 protein. Future studies will reveal possible compensation by additional Cxs, suggesting targets for future CRISPR-Cas9 ablation studies in human iPSC lineage specification.
Collapse
|