1
|
Capetini VC, Quintanilha BJ, Garcia BREV, Rogero MM. Dietary modulation of microRNAs in insulin resistance and type 2 diabetes. J Nutr Biochem 2024; 133:109714. [PMID: 39097171 DOI: 10.1016/j.jnutbio.2024.109714] [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: 11/30/2023] [Revised: 07/13/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
The prevalence of type 2 diabetes is increasing worldwide. Various molecular mechanisms have been proposed to interfere with the insulin signaling pathway. Recent advances in proteomics and genomics indicate that one such mechanism involves the post-transcriptional regulation of insulin signaling by microRNA (miRNA). These noncoding RNAs typically induce messenger RNA (mRNA) degradation or translational repression by interacting with the 3' untranslated region (3'UTR) of target mRNA. Dietary components and patterns, which can either enhance or impair the insulin signaling pathway, have been found to regulate miRNA expression in both in vitro and in vivo studies. This review provides an overview of the current knowledge of how dietary components influence the expression of miRNAs related to the control of the insulin signaling pathway and discusses the potential application of these findings in precision nutrition.
Collapse
Affiliation(s)
- Vinícius Cooper Capetini
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil; Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, Institute of Pharmaceutical Science, Department of Pharmacology, King's College London, London, United Kingdom.
| | - Bruna Jardim Quintanilha
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil
| | - Bruna Ruschel Ewald Vega Garcia
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Marcelo Macedo Rogero
- Nutritional Genomics and Inflammation Laboratory (GENUIN), Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), São Paulo Research Foundation (FAPESP), São Paulo, Brazil
| |
Collapse
|
2
|
Li T, Zhang W, Wang J, Liu B, Gao Q, Zhang J, Qian H, Pan J, Liu M, Huang Q, Fang A, Zhang Q, Gong X, Cui R, Liang Y, Lu Q, Wu W, Chi Z. Circulating Small Extracellular Vesicles Involved in Systemic Regulation Respond to RGC Degeneration in Glaucoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309307. [PMID: 38923329 PMCID: PMC11348076 DOI: 10.1002/advs.202309307] [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: 11/30/2023] [Revised: 05/14/2024] [Indexed: 06/28/2024]
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide and is characterized by progressive retinal ganglion cell (RGC) degeneration and vision loss. Since irreversible neurodegeneration occurs before diagnosable, early diagnosis and effective neuroprotection are critical for glaucoma management. Small extracellular vesicles (sEVs) are demonstrated to be potential novel biomarkers and therapeutics for a variety of diseases. In this study, it is found that intravitreal injection of circulating plasma-derived sEVs (PDEV) from glaucoma patients ameliorated retinal degeneration in chronic ocular hypertension (COH) mice. Moreover, it is found that PDEV-miR-29s are significantly upregulated in glaucoma patients and are associated with visual field defects in progressed glaucoma. Subsequently, in vivo and in vitro experiments are conducted to investigate the possible function of miR-29s in RGC pathophysiology. It is showed that the overexpression of miR-29b-3p effectively prevents RGC degeneration in COH mice and promotes the neuronal differentiation of human induced pluripotent stem cells (hiPSCs). Interestingly, engineered sEVs with sufficient miR-29b-3p delivery exhibit more effective RGC protection and neuronal differentiation efficiency. Thus, elevated PDEV-miR-29s may imply systemic regulation to prevent RGC degeneration in glaucoma patients. This study provides new insights into PDEV-based glaucoma diagnosis and therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Tong Li
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Wen‐Meng Zhang
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Jie Wang
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Bai‐Jing Liu
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Qiao Gao
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Jing Zhang
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Hai‐Dong Qian
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Jun‐Yi Pan
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Ming Liu
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Qing Huang
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Ai‐Wu Fang
- National Clinical Research Center for Ocular DiseasesEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Qi Zhang
- National Clinical Research Center for Ocular DiseasesEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian‐Hui Gong
- National Clinical Research Center for Ocular DiseasesEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Ren‐Zhe Cui
- Department of OphthalmologyAffiliated Hospital of Yanbian UniversityYanji136200China
| | - Yuan‐Bo Liang
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
- National Clinical Research Center for Ocular DiseasesEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Qin‐Kang Lu
- Department of OphthalmologyYinzhou People's HospitalMedical School of Ningbo UniversityNingbo315040China
| | - Wen‐Can Wu
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
- National Clinical Research Center for Ocular DiseasesEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Zai‐Long Chi
- State Key Laboratory of OphthalmologyOptometry and Visual ScienceEye Hospital of Wenzhou Medical UniversityWenzhou325027China
- National Clinical Research Center for Ocular DiseasesEye Hospital of Wenzhou Medical UniversityWenzhou325027China
| |
Collapse
|
3
|
Ali A, Mahla SB, Reza V, Hossein A, Bahareh K, Mohammad H, Fatemeh S, Mostafa AB, Leili R. MicroRNAs: Potential prognostic and theranostic biomarkers in chronic lymphocytic leukemia. EJHAEM 2024; 5:191-205. [PMID: 38406506 PMCID: PMC10887358 DOI: 10.1002/jha2.849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 12/13/2023] [Accepted: 12/29/2023] [Indexed: 02/27/2024]
Abstract
Small noncoding ribonucleic acids called microRNAs coordinate numerous critical physiological and biological processes such as cell division, proliferation, and death. These regulatory molecules interfere with the function of many genes by binding the 3'-UTR region of target mRNAs to inhibit their translation or even degrade them. Given that a large proportion of miRNAs behave as either tumor suppressors or oncogenes, any genetic or epigenetic aberration changeing their structure and/or function could initiate tumor formation and development. An example of such cancers is chronic lymphocytic leukemia (CLL), the most prevalent adult leukemia in Western nations, which is caused by unregulated growth and buildup of defective cells in the peripheral blood and lymphoid organs. Genetic alterations at cellular and molecular levels play an important role in the occurrence and development of CLL. In this vein, it was noted that the development of this disease is noticeably affected by changes in the expression and function of miRNAs. Many studies on miRNAs have shown that these molecules are pivotal in the prognosis of different cancers, including CLL, and their epigenetic alterations (e.g., methylation) can predict disease progression and response to treatment. Furthermore, miRNAs are involved in the development of drug resistance in CLL, and targeting these molecules can be considered a new therapeutic approach for the treatment of this disease. MiRNA screening can offer important information on the etiology and development of CLL. Considering the importance of miRNAs in gene expression regulation, their application in the diagnosis, prognosis, and treatment of CLL is reviewed in this paper.
Collapse
Affiliation(s)
- Afgar Ali
- Research Center for Hydatid Disease in IranKerman University of Medical SciencesKermanIran
| | - Sattarzadeh Bardsiri Mahla
- Stem Cells and Regenerative Medicine Innovation CenterKerman University of Medical SciencesKermanIran
- Department of Hematology and Laboratory Sciences, Faculty of Allied Medical SciencesKerman University of Medical SciencesKermanIran
| | - Vahidi Reza
- Research Center for Hydatid Disease in IranKerman University of Medical SciencesKermanIran
| | - Arezoomand Hossein
- Department of Hematology and Laboratory Sciences, Faculty of Allied Medical SciencesKerman University of Medical SciencesKermanIran
| | - Kashani Bahareh
- Department of Medical Genetics, School of MedicineTehran University of Medical SciencesTehranIran
| | - Hosseininaveh Mohammad
- Research Center for Hydatid Disease in IranKerman University of Medical SciencesKermanIran
| | - Sharifi Fatemeh
- Research Center of Tropical and Infectious DiseasesKerman University of Medical SciencesKermanIran
| | - Amopour Bahnamiry Mostafa
- Department of Research and Development, Production and Research ComplexPasteur Institute of IranTehranIran
| | - Rouhi Leili
- Student Research CommitteeKerman University of Medical SciencesKermanIran
| |
Collapse
|
4
|
Jin L, Long Y, Zhang Q, Long J. MiRNAs regulate cell communication in osteogenesis-angiogenesis coupling during bone regeneration. Mol Biol Rep 2023; 50:8715-8728. [PMID: 37642761 DOI: 10.1007/s11033-023-08709-6] [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: 03/24/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023]
Abstract
Bone regeneration is a complex process that requires not only the participation of multiple cell types, but also signal communication between cells. The two basic processes of osteogenesis and angiogenesis are closely related to bone regeneration and bone homeostasis. H-type vessels are a subtype of bone vessels characterized by high expression of CD31 and EMCN. These vessels play a key role in the regulation of bone regeneration and are important mediators of coupling between osteogenesis and angiogenesis. Molecular regulation between different cell types is important for coordination of osteogenesis and angiogenesis that promotes bone regeneration. MiRNAs are small non-coding RNAs that predominantly regulate gene expression at the post-transcriptional level and are closely related to cell communication. Specifically, miRNAs transduce external stimuli through various cell signaling pathways and cause a series of physiological and pathological effects. They are also deeply involved in the bone repair process. This review focuses on three signaling pathways related to osteogenesis-angiogenesis coupling, as well as the miRNAs involved in these pathways. Elucidation of the molecular mechanisms governing osteogenesis and angiogenesis is of great significance for bone regeneration.
Collapse
Affiliation(s)
- Liangyu Jin
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Yifei Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Qiuling Zhang
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Jie Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China.
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China.
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
5
|
Kizub IV. Induced pluripotent stem cells for cardiovascular therapeutics: Progress and perspectives. REGULATORY MECHANISMS IN BIOSYSTEMS 2023; 14:451-468. [DOI: 10.15421/10.15421/022366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
The discovery of methods for reprogramming adult somatic cells into induced pluripotent stem cells (iPSCs) opens up prospects of developing personalized cell-based therapy options for a variety of human diseases as well as disease modeling and new drug discovery. Like embryonic stem cells, iPSCs can give rise to various cell types of the human body and are amenable to genetic correction. This allows usage of iPSCs in the development of modern therapies for many virtually incurable human diseases. The review summarizes progress in iPSC research in the context of application in the cardiovascular field including modeling cardiovascular disease, drug study, tissue engineering, and perspectives for personalized cardiovascular medicine.
Collapse
|
6
|
Pastar I, Marjanovic J, Liang L, Stone RC, Kashpur O, Jozic I, Head CR, Smith A, Gerami-Naini B, Garlick JA, Tomic-Canic M. Cellular reprogramming of diabetic foot ulcer fibroblasts triggers pro-healing miRNA-mediated epigenetic signature. Exp Dermatol 2021; 30:1065-1072. [PMID: 34114688 DOI: 10.1111/exd.14405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/07/2021] [Accepted: 05/26/2021] [Indexed: 12/17/2022]
Abstract
Diabetic foot ulcers (DFUs), a prevalent complication of diabetes, constitute a major medical challenge with a critical need for development of cell-based therapies. We previously generated induced pluripotent stem cells (iPSCs) from dermal fibroblasts derived from the DFU patients, location-matched skin of diabetic patients and normal healthy donors and re-differentiated them into fibroblasts. To assess the epigenetic microRNA (miR) regulated changes triggered by cellular reprogramming, we performed miRs expression profiling. We found let-7c, miR-26b-5p, -29c-3p, -148a-3p, -196a-5p, -199b-5p and -374a-5p suppressed in iPSC-derived fibroblasts in vitro and in 3D dermis-like self-assembly tissue, whereas their corresponding targets involved in cellular migration were upregulated. Moreover, targets involved in organization of extracellular matrix were induced after fibroblast reprogramming. PLAT gene, the crucial fibrinolysis factor, was upregulated in iPSC-derived fibroblasts and was confirmed as a direct target of miR-196a-5p. miR-197-3p and miR-331-3p were found upregulated specifically in iPSC-derived diabetic fibroblasts, while their targets CAV1 and CDKN3 were suppressed. CAV1, an important negative regulator of wound healing, was confirmed as a direct miR-197-3p target. Together, our findings demonstrate that iPSC reprogramming is an effective approach for erasing the diabetic non-healing miR-mediated epigenetic signature and promoting a pro-healing cellular phenotype.
Collapse
Affiliation(s)
- Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jelena Marjanovic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Liang Liang
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rivka C Stone
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Olga Kashpur
- Department of Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Ivan Jozic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Cheyanne R Head
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Avi Smith
- Department of Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Behzad Gerami-Naini
- Department of Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Jonathan A Garlick
- Department of Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
7
|
Fráguas-Eggenschwiler M, Eggenschwiler R, Söllner JH, Cortnumme L, Vondran FWR, Cantz T, Ott M, Niemann H. Direct conversion of porcine primary fibroblasts into hepatocyte-like cells. Sci Rep 2021; 11:9334. [PMID: 33927320 PMCID: PMC8085017 DOI: 10.1038/s41598-021-88727-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
The pig is an important model organism for biomedical research, mainly due to its extensive genetic, physiological and anatomical similarities with humans. Until date, direct conversion of somatic cells into hepatocyte-like cells (iHeps) has only been achieved in rodents and human cells. Here, we employed lentiviral vectors to screen a panel of 12 hepatic transcription factors (TF) for their potential to convert porcine fibroblasts into hepatocyte-like cells. We demonstrate for the first time, hepatic conversion of porcine somatic cells by over-expression of CEBPα, FOXA1 and HNF4α2 (3TF-piHeps). Reprogrammed 3TF-piHeps display a hepatocyte-like morphology and show functional characteristics of hepatic cells, including albumin secretion, Dil-AcLDL uptake, storage of lipids and glycogen and activity of cytochrome P450 enzymes CYP1A2 and CYP2C33 (CYP2C9 in humans). Moreover, we show that markers of mature hepatocytes are highly expressed in 3TF-piHeps, while fibroblastic markers are reduced. We envision piHeps as useful cell sources for future studies on drug metabolism and toxicity as well as in vitro models for investigation of pig-to-human infectious diseases.
Collapse
Affiliation(s)
- Mariane Fráguas-Eggenschwiler
- Gastroenterology, Hepatology and Endocrinology Department, Hannover Medical School, Hannover, Germany. .,Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany.
| | - Reto Eggenschwiler
- Gastroenterology, Hepatology and Endocrinology Department, Hannover Medical School, Hannover, Germany.,Translational Hepatology and Stem Cell Biology, REBIRTH - Research Center for Translational Regenerative Medicine and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jenny-Helena Söllner
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), Mariensee, Neustadt, Germany
| | - Leon Cortnumme
- Translational Hepatology and Stem Cell Biology, REBIRTH - Research Center for Translational Regenerative Medicine and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian W R Vondran
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Tübingen, Germany
| | - Tobias Cantz
- Gastroenterology, Hepatology and Endocrinology Department, Hannover Medical School, Hannover, Germany.,Translational Hepatology and Stem Cell Biology, REBIRTH - Research Center for Translational Regenerative Medicine and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael Ott
- Gastroenterology, Hepatology and Endocrinology Department, Hannover Medical School, Hannover, Germany.,Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Heiner Niemann
- Gastroenterology, Hepatology and Endocrinology Department, Hannover Medical School, Hannover, Germany. .,Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany.
| |
Collapse
|
8
|
Sun Q, Liu S, Feng J, Kang Y, Zhou Y, Guo S. Current Status of MicroRNAs that Target the Wnt Signaling Pathway in Regulation of Osteogenesis and Bone Metabolism: A Review. Med Sci Monit 2021; 27:e929510. [PMID: 33828067 PMCID: PMC8043416 DOI: 10.12659/msm.929510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The directional differentiation of bone mesenchymal stem cells (BMSCs) is regulated by a variety of transcription factors and intracellular signaling pathways. In the past, it was thought that the directional differentiation of BMSCs was related to transforming growth factors, such as bone morphogenetic protein (BMP) and MAPK pathway. However, in recent years, some scholars have pointed out that the Wnt signaling pathway, which is a necessary complex network of protein interactions for biological growth and development, takes a significant role in this process and plays a major part in regulating the development of osteoblasts by exerting signal transduction into cells. Also, they have proved the Wnt protein therapeutic truly have positive effects on the viability and osteogenic capacity of bone graft. Recent studies have shown that microRNAs (miRNAs) play an important regulatory role in this process. MiRNAs such as miRNA-218, miRNA-335, miRNA-29, microRNA-30 and other miRNAs exert negative or positive effects on some crucial molecules in the Wnt/β-catenin pathway, which in turn affect bone metabolism and osteopathy. Thus, miRNAs have been suggested as therapeutic targets for some metabolic bone diseases. This article aims to provide an update on the current status of microRNAs that target the Wnt signaling pathway in the regulation of osteogenesis and bone metabolism and includes a discussion of future areas of research, which can be a theoretical basis for bone metabolism-related diseases.
Collapse
Affiliation(s)
- Qiang Sun
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Siyu Liu
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Jingyi Feng
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yue Kang
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - You Zhou
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Shu Guo
- Department of Plastic Surgery, The First Hopital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
9
|
Pasca S, Jurj A, Zdrenghea M, Tomuleasa C. The Potential Equivalents of TET2 Mutations. Cancers (Basel) 2021; 13:cancers13071499. [PMID: 33805247 PMCID: PMC8036366 DOI: 10.3390/cancers13071499] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/10/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary In acute myeloid leukemia (AML) TET2 mutations have been observed to be mutually exclusive with IDH1, IDH2, and WT1 mutations, all of them showing a similar impact on the transcription profile. Because of this, it is possible that TET2/IDH1/2/WT1 mutated AML could be considered as having similar characteristics between each other. Nonetheless, other genes also interact with TET2 and influence its activity. Because of this, it is possible that other signatures exist that would mimic the effect of TET2 mutations. Thus, in this review, we searched the literature for the genes that were observed to interact with TET2 and classified them in the following manner: transcription alteration, miRs, direct interaction, posttranslational changes, and substrate reduction. Abstract TET2 is a dioxygenase dependent on Fe2+ and α-ketoglutarate which oxidizes 5-methylcytosine (5meC) to 5-hydroxymethylcytosine (5hmeC). TET proteins successively oxidize 5mC to yield 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC). Among these oxidized methylcytosines, 5fC and 5caC are directly excised by thymine DNA glycosylase (TDG) and ultimately replaced with unmethylated cytosine. Mutations in TET2 have been shown to lead to a hypermethylated state of the genome and to be responsible for the initiation of the oncogenetic process, especially in myeloid and lymphoid malignancies. Nonetheless, this was also shown to be the case in other cancers. In AML, TET2 mutations have been observed to be mutually exclusive with IDH1, IDH2, and WT1 mutations, all of them showing a similar impact on the transcription profile of the affected cell. Because of this, it is possible that TET2/IDH1/2/WT1 mutated AML could be considered as having similar characteristics between each other. Nonetheless, other genes also interact with TET2 and influence its effect, thus making it possible that other signatures exist that would mimic the effect of TET2 mutations. Thus, in this review, we searched the literature for the genes that were observed to interact with TET2 and classified them in the following manner: transcription alteration, miRs, direct interaction, posttranslational changes, and substrate reduction. What we propose in the present review is the potential extension of the TET2/IDH1/2/WT1 entity with the addition of certain expression signatures that would be able to induce a similar phenotype with that induced by TET2 mutations. Nonetheless, we recommend that this approach be taken on a disease by disease basis.
Collapse
Affiliation(s)
- Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj Napoca, Romania; (S.P.); (M.Z.); (C.T.)
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400124 Cluj Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Correspondence:
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj Napoca, Romania; (S.P.); (M.Z.); (C.T.)
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400124 Cluj Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj Napoca, Romania; (S.P.); (M.Z.); (C.T.)
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400124 Cluj Napoca, Romania
| |
Collapse
|
10
|
Hrdlicka HC, Pereira RC, Shin B, Yee SP, Deymier AC, Lee SK, Delany AM. Inhibition of miR-29-3p isoforms via tough decoy suppresses osteoblast function in homeostasis but promotes intermittent parathyroid hormone-induced bone anabolism. Bone 2021; 143:115779. [PMID: 33253931 PMCID: PMC7770763 DOI: 10.1016/j.bone.2020.115779] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 01/07/2023]
Abstract
miRNAs play a vital role in post-transcriptional regulation of gene expression in osteoblasts and osteoclasts, and the miR-29 family is expressed in both lineages. Using mice globally expressing a miR-29-3p tough decoy, we demonstrated a modest 30-60% decrease all three miR-29-3p isoforms: miR-29a, miR-29b, and miR-29c. While the miR-29-3p decoy did not impact osteoclast number or function, the tough decoy decreased bone formation in growing mice, which led to decreased trabecular bone volume in mature animals. These data support previous in vitro studies suggesting that miR-29-3p is a positive regulator of osteoblast differentiation. In contrast, when mice were treated with intermittent parathyroid hormone (PTH1-34), inhibition of miR-29-3p augmented the effect of PTH on cortical bone anabolism, increased bone formation rate and osteoblast surface, and increased levels of Ctnnb1/βcatenin mRNA, which is a miR-29 target. These findings highlight differences in the mechanisms controlling basal level bone formation and bone formation induced by intermittent PTH. Overall, the global miR-29-3p tough decoy model represents a modest loss-of-function, which could be a relevant tool for assessing the possible impact of systemically administered miR-29-3p inhibitors. Our studies provide a potential rationale for co-administration of PTH1-34 and miR-29-3p inhibitors, to boost bone formation in severely affected osteoporosis patients, particularly in the cortical compartment.
Collapse
Affiliation(s)
- Henry C Hrdlicka
- Center for Molecular Oncology, UConn Health Center, Farmington, CT, United States of America
| | - Renata C Pereira
- Division of Pediatric Nephrology, David Geffen School of Medicine at University of California, Los Angeles, United States of America
| | - Bongjin Shin
- Center on Aging, UConn Health Center, Farmington, CT, United States of America
| | - Siu-Pok Yee
- Center for Mouse Genome Modification, UConn Health Center, Farmington, CT, United States of America
| | - Alix C Deymier
- Institute of Material Sciences, UConn Health Center, Farmington, CT, United States of America
| | - Sun-Kyeong Lee
- Center on Aging, UConn Health Center, Farmington, CT, United States of America.
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health Center, Farmington, CT, United States of America.
| |
Collapse
|
11
|
Bhattacharya M, Sharma AR, Sharma G, Patra BC, Lee SS, Chakraborty C. Interaction between miRNAs and signaling cascades of Wnt pathway in chronic lymphocytic leukemia. J Cell Biochem 2020; 121:4654-4666. [PMID: 32100920 DOI: 10.1002/jcb.29683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 01/30/2020] [Indexed: 12/11/2022]
Abstract
Chronic lymphocytic leukemia (CLL), a severe problem all over the world and represents around 25% of all total leukemia cases, is generating the need for novel targets against CLL. Wnt signaling cascade regulates cell proliferation, differentiation, and cell death processes. Thus, any alteration of the Wnt signaling pathway protein cascade might develop into various types of cancers, either by upregulation or downregulation of the Wnt signaling pathway protein components. In addition, it is reported that activation of the Wnt signaling pathway is associated with the transcriptional activation of microRNAs (miRNAs) by binding to its promoter region, suggesting feedback regulation. Considering the protein regulatory functions of various miRNAs, they can be approached therapeutically as modulatory targets for protein components of the Wnt signaling pathway. In this article, we have discussed the potential role of miRNAs in the regulation of Wnt signaling pathway proteins related to the pathogenesis of CLL via crosstalk between miRNAs and Wnt signaling pathway proteins. This might provide a clear insight into the Wnt protein regulatory function of various miRNAs and provide a better understanding of developing advanced and promising therapeutic approaches against CLL.
Collapse
Affiliation(s)
- Manojit Bhattacharya
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University Hospital-College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea.,Department of Zoology, Vidyasagar University, Midnapore, West Bengal, India
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University Hospital-College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Garima Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University Hospital-College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | | | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University Hospital-College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, India
| |
Collapse
|
12
|
Rana MA, Ijaz B, Daud M, Tariq S, Nadeem T, Husnain T. Interplay of Wnt β-catenin pathway and miRNAs in HBV pathogenesis leading to HCC. Clin Res Hepatol Gastroenterol 2019; 43:373-386. [PMID: 30377095 DOI: 10.1016/j.clinre.2018.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 09/05/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023]
Abstract
The prevalence of Hepatocellular carcinoma (HCC) has been identified world-wide. Plethora of factors including chronic infection of HBV/HCV has been characterized for the development of HCC. Although the onset and progression of HCC has been linked with awry of various signaling pathways but precise mechanism, still lies under the multitude layers of curiosity. HBV is spreading with insane speed throughout the world and has been found a main culprit in HCC development after regulating the several cellular pathways including Wnt/β-catenin, Raf/MAPK, Akt and affecting cell multiplication to genomic instability. The role of Wnt/FZD/β-catenin signaling pathway is centralized in liver functions and its anomalous activation leads to HCC development. β-catenin mainly plays a pivotal role in canonical pathway of the system. Altered mainly overexpression of β-catenin along its nuclear localization tunes the aberrations in liver functions and set disease progression. In the development of HCC, modulation of Wnt/FZD/β-catenin signaling pathway by HBV has been established. As HBV infects the cell it affects the miRNAs, the master regulators of cell. Previous studies showed the connection between HBV and cellular miRNAs. In the present review, we unveiled how HBV is deciphering the cellular miRNAs like miR-26a, miR-15a, miR-16-1, miR-148a, miR-132, miR-122, miR-34a, miR-21, miR-29a, miR-222 and miR-199a/b-3p to modulate the Wnt/FZD/β-catenin signaling pathway and develop HCC. These HBV mediated miRNAs may prove future therapeutic options to treat HBV-Wnt/FZD/β-catenin associated HCC.
Collapse
Affiliation(s)
- Muhammad Adeel Rana
- Department of microbiology, Quaid-i-Azam University, Islamabad, Pakistan; Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Bushra Ijaz
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan.
| | - Muhammad Daud
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Sommyya Tariq
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Tariq Nadeem
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Tayyab Husnain
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore Pakistan
| |
Collapse
|
13
|
de Souza Lima IM, Schiavinato JLDS, Paulino Leite SB, Sastre D, Bezerra HLDO, Sangiorgi B, Corveloni AC, Thomé CH, Faça VM, Covas DT, Zago MA, Giacca M, Mano M, Panepucci RA. High-content screen in human pluripotent cells identifies miRNA-regulated pathways controlling pluripotency and differentiation. Stem Cell Res Ther 2019; 10:202. [PMID: 31287022 PMCID: PMC6615276 DOI: 10.1186/s13287-019-1318-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 06/11/2019] [Accepted: 06/30/2019] [Indexed: 01/13/2023] Open
Abstract
Background By post-transcriptionally regulating multiple target transcripts, microRNAs (miRNAs or miR) play important biological functions. H1 embryonic stem cells (hESCs) and NTera-2 embryonal carcinoma cells (ECCs) are two of the most widely used human pluripotent model cell lines, sharing several characteristics, including the expression of miRNAs associated to the pluripotent state or with differentiation. However, how each of these miRNAs functionally impacts the biological properties of these cells has not been systematically evaluated. Methods We investigated the effects of 31 miRNAs on NTera-2 and H1 hESCs, by transfecting miRNA mimics. Following 3–4 days of culture, cells were stained for the pluripotency marker OCT4 and the G2 cell-cycle marker Cyclin B1, and nuclei and cytoplasm were co-stained with Hoechst and Cell Mask Blue, respectively. By using automated quantitative fluorescence microscopy (i.e., high-content screening (HCS)), we obtained several morphological and marker intensity measurements, in both cell compartments, allowing the generation of a multiparametric miR-induced phenotypic profile describing changes related to proliferation, cell cycle, pluripotency, and differentiation. Results Despite the overall similarities between both cell types, some miRNAs elicited cell-specific effects, while some related miRNAs induced contrasting effects in the same cell. By identifying transcripts predicted to be commonly targeted by miRNAs inducing similar effects (profiles grouped by hierarchical clustering), we were able to uncover potentially modulated signaling pathways and biological processes, likely mediating the effects of the microRNAs on the distinct groups identified. Specifically, we show that miR-363 contributes to pluripotency maintenance, at least in part, by targeting NOTCH1 and PSEN1 and inhibiting Notch-induced differentiation, a mechanism that could be implicated in naïve and primed pluripotent states. Conclusions We present the first multiparametric high-content microRNA functional screening in human pluripotent cells. Integration of this type of data with similar data obtained from siRNA screenings (using the same HCS assay) could provide a large-scale functional approach to identify and validate microRNA-mediated regulatory mechanisms controlling pluripotency and differentiation. Electronic supplementary material The online version of this article (10.1186/s13287-019-1318-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ildercílio Mota de Souza Lima
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Josiane Lilian Dos Santos Schiavinato
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Sarah Blima Paulino Leite
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Danuta Sastre
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil
| | - Hudson Lenormando de Oliveira Bezerra
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Bruno Sangiorgi
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Amanda Cristina Corveloni
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Carolina Hassibe Thomé
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Marco Antônio Zago
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil.,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic and Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Miguel Mano
- Molecular Medicine Laboratory, International Centre for Genetic and Engineering and Biotechnology (ICGEB), Trieste, Italy.,Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Rodrigo Alexandre Panepucci
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP, CEP: 14051-140, Brazil. .,Department of Genetics and Internal Medicine, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
14
|
Gao Y, Qiao H, Lu Z, Hou Y. miR‑29 promotes the proliferation of cultured rat neural stem/progenitor cells via the PTEN/AKT signaling pathway. Mol Med Rep 2019; 20:2111-2118. [PMID: 31257508 PMCID: PMC6691275 DOI: 10.3892/mmr.2019.10445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/26/2019] [Indexed: 12/19/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) are self-renewing, multipotent cells and remain in the human brain throughout an individual's lifetime. NSPCs are activated by brain damage and contribute towards repair and motor function recovery in the central nervous system (CNS). It was previously reported that miR-29 was involved in regulating proliferation, differentiation and survival in hepatocellular carcinoma, and osteoblast and mantle cell lymphoma; however, the effects of miR-29 on NSPCs remain unclear. In the present study, it was demonstrated via Cell Counting Kit-8 assays that overexpression of miR-29 promoted the viability of NPSCs, and downregulated the expression of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) protein. Additionally, treatment with a PTEN-specific inhibitor (VO-OHpic trihydrate) abolished the effects of the miR-29 inhibitor on PTEN expression, as determined via western blotting. Flow cytometry and 5-bromo-2-deoxyuridine (BrdU) staining revealed that overexpression of miR-29 further promoted the proliferation of NSPCs; however, knocking down miR-29 inhibited cell proliferation. VO-OHpic trihydrate reversed the effects of miR-29 knockdown on cell proliferation. Furthermore, it was observed that overexpression of miR-29 increased the phosphorylation levels of AKT. Collectively, the results indicated that overexpression of miR-29 promoted the proliferation of cultured rat NSPCs and decreased the expression of PTEN protein, and that the activation of Akt may be a potential underlying mechanism. The present findings may provide novel insight for the development of strategies for stem cell-mediated treatment of CNS diseases.
Collapse
Affiliation(s)
- Yunan Gao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Hu Qiao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zhen Lu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yuxia Hou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
15
|
Smieszek A, Kornicka K, Szłapka-Kosarzewska J, Androvic P, Valihrach L, Langerova L, Rohlova E, Kubista M, Marycz K. Metformin Increases Proliferative Activity and Viability of Multipotent Stromal Stem Cells Isolated from Adipose Tissue Derived from Horses with Equine Metabolic Syndrome. Cells 2019; 8:E80. [PMID: 30678275 PMCID: PMC6406832 DOI: 10.3390/cells8020080] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
In this study, we investigated the influence of metformin (MF) on proliferation and viability of adipose-derived stromal cells isolated from horses (EqASCs). We determined the effect of metformin on cell metabolism in terms of mitochondrial metabolism and oxidative status. Our purpose was to evaluate the metformin effect on cells derived from healthy horses (EqASCHE) and individuals affected by equine metabolic syndrome (EqASCEMS). The cells were treated with 0.5 μM MF for 72 h. The proliferative activity was evaluated based on the measurement of BrdU incorporation during DNA synthesis, as well as population doubling time rate (PDT) and distribution of EqASCs in the cell cycle. The influence of metformin on EqASC viability was determined in relation to apoptosis profile, mitochondrial membrane potential, oxidative stress markers and BAX/BCL-2 mRNA ratio. Further, we were interested in possibility of metformin affecting the Wnt3a signalling pathway and, thus, we determined mRNA and protein level of WNT3A and β-catenin. Finally, using a two-tailed RT-qPCR method, we investigated the expression of miR-16-5p, miR-21-5p, miR-29a-3p, miR-140-3p and miR-145-5p. Obtained results indicate pro-proliferative and anti-apoptotic effects of metformin on EqASCs. In this study, MF significantly improved proliferation of EqASCs, which manifested in increased synthesis of DNA and lowered PDT value. Additionally, metformin improved metabolism and viability of cells, which correlated with higher mitochondrial membrane potential, reduced apoptosis and increased WNT3A/β-catenin expression. Metformin modulates the miRNA expression differently in EqASCHE and EqASCEMS. Metformin may be used as a preconditioning agent which stimulates proliferative activity and viability of EqASCs.
Collapse
Affiliation(s)
- Agnieszka Smieszek
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Katarzyna Kornicka
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Jolanta Szłapka-Kosarzewska
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Peter Androvic
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- Laboratory of Growth Regulators, Faculty of Science, Palacky University, 78371 Olomouc, Czech Republic.
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
| | - Lucie Langerova
- Gene Core BIOCEV, Průmyslová 595, Vestec 252 50, Czech Republic.
| | - Eva Rohlova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, 128 43 Prague, Czech Republic.
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- TATAA Biocenter AB, 411 03 Gothenburg, Sweden.
| | - Krzysztof Marycz
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392 Giessen, Germany.
| |
Collapse
|
16
|
Panepucci RA, de Souza Lima IM. Arrayed functional genetic screenings in pluripotency reprogramming and differentiation. Stem Cell Res Ther 2019; 10:24. [PMID: 30635073 PMCID: PMC6330485 DOI: 10.1186/s13287-018-1124-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Thoroughly understanding the molecular mechanisms responsible for the biological properties of pluripotent stem cells, as well as for the processes involved in reprograming, differentiation, and transition between Naïve and Primed pluripotent states, is of great interest in basic and applied research. Although pluripotent cells have been extensively characterized in terms of their transcriptome and miRNome, a comprehensive understanding of how these gene products specifically impact their biology, depends on gain- or loss-of-function experimental approaches capable to systematically interrogate their function. We review all studies carried up to date that used arrayed screening approaches to explore the function of these genetic elements on those biological contexts, using focused or genome-wide genetic libraries. We further discuss the limitations and advantages of approaches based on assays with population-level primary readouts, derived from single-parameter plate readers, or cell-level primary readouts, obtained using multiparametric flow cytometry or quantitative fluorescence microscopy (i.e., high-content screening). Finally, we discuss technical limitation and future perspectives, highlighting how the integration of screening data may lead to major advances in the field of stem cell research and therapy.
Collapse
Affiliation(s)
- Rodrigo Alexandre Panepucci
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP CEP: 14051-140 Brazil
- Department of Genetics, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP Brazil
| | - Ildercílio Mota de Souza Lima
- Laboratory of Functional Biology (LFBio), Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, SP CEP: 14051-140 Brazil
- Department of Genetics, Ribeirao Preto Medical School, University of São Paulo (FMRP-USP), Ribeirão Preto, SP Brazil
| |
Collapse
|
17
|
Abstract
Research on stem cells is one of the fastest growing areas of regenerative medicine that paves the way for a comprehensive solution to cell therapy. Today, stem cells are precious assets for generating different types of cells derived from either natural embryonic stem (ES) cells or induced pluripotent stem (iPS) cells. The iPS technology can revolutionize the future of clinics by offering personalized medicine, which will provide the future treatment for curing untreatable diseases. Although iPS cell therapy is now at its infancy, promising research has motivated scientists to pursue this therapeutic approach. In this article, we provide information regarding similarities and differences between ES and iPS cells, and focus on the non-integrating methods of iPS generation via RNA molecules, especially microRNAs with an emphasis on the elucidation of their role and importance in pluripotency.
Collapse
Affiliation(s)
- Abbas Beh-Pajooh
- REBIRTH-Group Translational Hepatology and Stem Cell Biology, Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Tobias Cantz
- REBIRTH-Group Translational Hepatology and Stem Cell Biology, Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
18
|
Brumbaugh J, Di Stefano B, Wang X, Borkent M, Forouzmand E, Clowers KJ, Ji F, Schwarz BA, Kalocsay M, Elledge SJ, Chen Y, Sadreyev RI, Gygi SP, Hu G, Shi Y, Hochedlinger K. Nudt21 Controls Cell Fate by Connecting Alternative Polyadenylation to Chromatin Signaling. Cell 2018; 172:106-120.e21. [PMID: 29249356 PMCID: PMC5766360 DOI: 10.1016/j.cell.2017.11.023] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/08/2017] [Accepted: 11/10/2017] [Indexed: 10/18/2022]
Abstract
Cell fate transitions involve rapid gene expression changes and global chromatin remodeling, yet the underlying regulatory pathways remain incompletely understood. Here, we identified the RNA-processing factor Nudt21 as a novel regulator of cell fate change using transcription-factor-induced reprogramming as a screening assay. Suppression of Nudt21 enhanced the generation of induced pluripotent stem cells, facilitated transdifferentiation into trophoblast stem cells, and impaired differentiation of myeloid precursors and embryonic stem cells, suggesting a broader role for Nudt21 in cell fate change. We show that Nudt21 directs differential polyadenylation of over 1,500 transcripts in cells acquiring pluripotency, although only a fraction changed protein levels. Remarkably, these proteins were strongly enriched for chromatin regulators, and their suppression neutralized the effect of Nudt21 during reprogramming. Collectively, our data uncover Nudt21 as a novel post-transcriptional regulator of cell fate and establish a direct, previously unappreciated link between alternative polyadenylation and chromatin signaling.
Collapse
Affiliation(s)
- Justin Brumbaugh
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Bruno Di Stefano
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Xiuye Wang
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Marti Borkent
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Elmira Forouzmand
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Katie J Clowers
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Benjamin A Schwarz
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Marian Kalocsay
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen J Elledge
- Howard Hughes Medical Institute, Brigham and Women's Hospital and Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yue Chen
- Department of Biochemistry, Molecular Biology, and Biophysics, College of Biological Sciences, University of Minnesota, Saint Paul, MN 55018, USA
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Yongsheng Shi
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA.
| | - Konrad Hochedlinger
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
19
|
Ferrosenescence: The iron age of neurodegeneration? Mech Ageing Dev 2017; 174:63-75. [PMID: 29180225 DOI: 10.1016/j.mad.2017.11.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Abstract
Aging has been associated with iron retention in many cell types, including the neurons, promoting neurodegeneration by ferroptosis. Excess intracellular iron accelerates aging by damaging the DNA and blocking genomic repair systems, a process we define as ferrosenescence. Novel neuroimaging and proteomic techniques have pinpointed indicators of both iron retention and ferrosenescence, allowing for their early correction, potentially bringing prevention of neurodegenerative disorders within reach. In this review, we take a closer look at the early markers of iron dyshomeostasis in neurodegenerative disorders, focusing on preventive strategies based on nutritional and microbiome manipulations.
Collapse
|