1
|
Vasetska A, Packeiser EM, Körber H, Aslan S, Ay S, Findik M, Binli F, Selçuk M, Speiser-Fontaine C, Goericke-Pesch S. Molecular response of canine testis to GnRH agonist: Insights into AR, HIF-1α, and HSPs expression during arrest and recovery of spermatogenesis. Cell Stress Chaperones 2025; 30:9-21. [PMID: 39631561 PMCID: PMC11719361 DOI: 10.1016/j.cstres.2024.11.007] [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: 11/08/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024] Open
Abstract
Slow-release gonadotropin-releasing hormone (GnRH) agonist implants are frequently used for contraception in male dogs. Although the effects are fully reversible, there is still concern about the safety of the implant's mode of action. Addressing this, we investigated cellular stress and androgen receptor (AR) signaling during downregulation and recovery. Testicular tissues were sampled from dogs castrated at different time points after GnRH implant removal and compared with untreated controls. AR, hypoxia-inducible factor 1 (HIF1A), heat shock proteins heat shock protein 72 (HSP72), heat shock protein 73 (heat shock cognate, HSPA8) (HSP73), heat shock protein A2 (HSPA2), heat shock protein 90 alpha (inducible isoform) (HSP90AA1), and heat shock protein 90 beta (constitutive isoform) (HSP90AB1) were investigated by quantitative real-time polymerase chain reaction and AR, HSP72, HSP73, and HSP90 immunohistochemically. While AR, HIF1A, and HSP70 were upregulated at gene expression level, HSPA8, HSPA2, and HSP90AA1 expression were downregulated during spermatogenic arrest; HSP90AB1 expression did not change. Immunohistochemistry verified AR-expression in Sertoli, peritubular, and Leydig cells, occasionally also in spermatogonia. Stress-inducible HSP72 was occasionally detected, while constitutive HSP73 and HSP90 were abundantly expressed by germ cells. Our results were similar to studies on seasonal breeders such as pine voles, geese, fish, and soft-shelled turtles. Accordingly, GnRH implants did not impose additional cellular stress on testicular cells when compared with natural recrudescence. Since comparative data on HIF1α are scarce, we cannot draw conclusions about hypoxic conditions.
Collapse
Affiliation(s)
- Anastasiia Vasetska
- Unit for Reproductive Medicine - Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Eva-Maria Packeiser
- Unit for Reproductive Medicine - Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Hanna Körber
- Unit for Reproductive Medicine - Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Selim Aslan
- Department of Obstetrics and Gynaecology - Faculty of Veterinary Medicine, Near East University, Nicosia, Cyprus
| | - Serhan Ay
- Department of Obstetrics and Gynaecology - Faculty of Veterinary Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Murat Findik
- Department of Obstetrics and Gynaecology - Faculty of Veterinary Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Firdevs Binli
- Department of Obstetrics and Gynaecology - Faculty of Veterinary Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Murat Selçuk
- Department of Reproduction and Artificial Insemination - Faculty of Veterinary Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | | | - Sandra Goericke-Pesch
- Unit for Reproductive Medicine - Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany.
| |
Collapse
|
2
|
Olenic M, Deelkens C, Heyman E, De Vlieghere E, Zheng X, van Hengel J, De Schauwer C, Devriendt B, De Smet S, Thorrez L. Review: Livestock cell types with myogenic differentiation potential: Considerations for the development of cultured meat. Animal 2025; 19 Suppl 1:101242. [PMID: 39097434 DOI: 10.1016/j.animal.2024.101242] [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: 12/08/2023] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 08/05/2024] Open
Abstract
With the current environmental impact of large-scale animal production and societal concerns about the welfare of farm animals, researchers are questioning whether we can cultivate animal cells for the purpose of food production. This review focuses on a pivotal aspect of the cellular agriculture domain: cells. We summarised information on the various cell types from farm animals currently used for the development of cultured meat, including mesenchymal stromal cells, myoblasts, and pluripotent stem cells. The review delves into the advantages and limitations of each cell type and considers factors like the selection of the appropriate cell source, as well as cell culture conditions that influence cell performance. As current research in cultured meat seeks to create muscle fibers to mimic the texture and nutritional profile of meat, we focused on the myogenic differentiation capacity of the cells. The most commonly used cell type for this purpose are myoblasts or satellite cells, but given their limited proliferation capacity, efforts are underway to formulate myogenic differentiation protocols for mesenchymal stromal cells and pluripotent stem cells. The multipotent character of the latter cell types might enable the creation of other tissues found in meat, such as adipose and connective tissues. This review can help guiding the selection of a cell type or culture conditions in the context of cultured meat development.
Collapse
Affiliation(s)
- M Olenic
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - C Deelkens
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Medical Cell Biology, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - E Heyman
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - E De Vlieghere
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Belgium
| | - X Zheng
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium
| | - J van Hengel
- Medical Cell Biology, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - C De Schauwer
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - B Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - S De Smet
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - L Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium.
| |
Collapse
|
3
|
Wang Y, Wang L, Jiang Z, Qu M, Meng Z, Sun Q, Du Y, Wang Y. Non-dietary exposure to phthalates in primary school children: Risk and correlation with anthropometric indices, cardiovascular and respiratory diseases. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117203. [PMID: 39423508 DOI: 10.1016/j.ecoenv.2024.117203] [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: 06/18/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Phthalates are endocrine disruptors of increasing concern for human health; however, previous studies have only assessed the association between internal exposure and human health. We aimed to assess the non-carcinogenic and carcinogenic risks of non-dietary exposure to phthalates in indoor environments among primary school children and their correlations with health indicators. A study involving 54 children was conducted in Jinan, Shandong Province, China. Questionnaires and health examinations were conducted, dust in hard-to-clean corners of students' classrooms and homes was collected, and airborne phthalates in the middle of classrooms and family living rooms were sampled. The gas-phase phthalate concentrations, individual exposure, and non-carcinogenic and carcinogenic risks were calculated. Associations were estimated using linear mixed models. The findings revealed that phthalates posed a non-carcinogenic risk to 7.4 % of the children and a moderate carcinogenic risk to 27.8 % of the children, with higher non-carcinogenic and carcinogenic risks to girls than to boys. Five phthalates were negatively correlated with body mass index, dimethyl phthalate and diethyl phthalate (DEP) were significantly correlated with waist circumference, and di-iso-butyl phthalate (DiBP) was negatively correlated with hip circumference. DiBP, di-n-butyl phthalate, and DEP, were significantly correlated with cardiovascular disease, DEP and di (2-n-butoxyethyl) phthalate were correlated with decreased lung function, and di-n-octyl phthalate influenced airway inflammation. The findings indicated that phthalate exposure may negatively impact children's health, thereby warranting further comprehensive research on the health effects of these chemicals.
Collapse
Affiliation(s)
- Yuchen Wang
- School of Environment and Energy Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Lixin Wang
- School of Environment and Energy Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China.
| | - Zhiyu Jiang
- School of Environment and Energy Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Meinan Qu
- School of Environment and Energy Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Ziyan Meng
- School of Environment and Energy Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Qinghua Sun
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yanjun Du
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yanwen Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
4
|
Lin YC, Wuputra K, Kato K, Ku CC, Saito S, Noguchi M, Nakamura Y, Hsiao M, Lin CS, Wu DC, Kawaguchi A, Yu HS, Yokoyama KK. Di-n-butyl phthalate promotes the neural differentiation of mouse embryonic stem cells through neurogenic differentiation 1. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123722. [PMID: 38460589 DOI: 10.1016/j.envpol.2024.123722] [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: 09/13/2023] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
An understanding of the risk of gene deletion and mutation posed by endocrine-disrupting chemicals (EDCs) is necessary for the identification of etiological reagents for many human diseases. Therefore, the characterization of the genetic traits caused by developmental exposure to EDCs is an important research subject. A new regenerative approach using embryonic stem cells (ESCs) holds promise for the development of stem-cell-based therapies and the identification of novel therapeutic agents against human diseases. Here, we focused on the characterization of the genetic traits and alterations in pluripotency/stemness triggered by phthalate ester derivatives. Regarding their in vitro effects, we reported the abilities of ESCs regarding proliferation, cell-cycle control, and neural ectoderm differentiation. The expression of their stemness-related genes and their genetic changes toward neural differentiation were examined, which led to the observation that the tumor suppressor gene product p53/retinoblastoma protein 1 and its related cascades play critical functions in cell-cycle progression, cell death, and neural differentiation. In addition, the expression of neurogenic differentiation 1 was affected by exposure to di-n-butyl phthalate in the context of cell differentiation into neural lineages. The nervous system is one of the most sensitive tissues to exposure to phthalate ester derivatives. The present screening system provides a good tool for studying the mechanisms underlying the effects of EDCs on the developmental regulation of humans and rodents, especially on the neuronal development of ESCs.
Collapse
Affiliation(s)
- Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, 807, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan; Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Kohsuke Kato
- Department of Infection Biology, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 305-8577, Japan
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, 807, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan; Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita, Tochigi, 329-1571, Japan
| | - Michiya Noguchi
- Cell Engineering Division, BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Yukio Nakamura
- Cell Engineering Division, BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Michael Hsiao
- Genome Research Center, Academia Sinica, Nangan, Taipei, 115, Taiwan
| | - Chang-Shen Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, 807, Taiwan; Department of Biological Sciences, National Sun Yan-Sen University, Kaohsiung, 80424, Taiwan
| | - Deng-Chyang Wu
- Graduate Institute of Medicine, Kaohsiung Medical University, 807, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan; Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan; Department of Gastroenterology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Atsushi Kawaguchi
- Department of Infection Biology, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 305-8577, Japan
| | - Hsin-Su Yu
- Emeritus Professor in College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Kazunari K Yokoyama
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, 807, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
5
|
Chen L, Tang B, Xie G, Yang R, Zhang B, Wang Y, Zhang Y, Jiang D, Zhang X. Bovine Pluripotent Stem Cells: Current Status and Prospects. Int J Mol Sci 2024; 25:2120. [PMID: 38396797 PMCID: PMC10889747 DOI: 10.3390/ijms25042120] [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: 12/11/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Pluripotent stem cells (PSCs) can differentiate into three germ layers and diverse autologous cell lines. Since cattle are the most commonly used large domesticated animals, an important food source, and bioreactors, great efforts have been made to establish bovine PSCs (bPSCs). bPSCs have great potential in bovine breeding and reproduction, modeling in vitro differentiation, imitating cancer development, and modeling diseases. Currently, bPSCs mainly include bovine embryonic stem cells (bESCs), bovine induced pluripotent stem cells (biPSCs), and bovine expanded potential stem cells (bEPSCs). Establishing stable bPSCs in vitro is a critical scientific challenge, and researchers have made numerous efforts to this end. In this review, the category of PSC pluripotency; the establishment of bESCs, biPSCs, and bEPSCs and its challenges; and the application outlook of bPSCs are discussed, aiming to provide references for future research.
Collapse
Affiliation(s)
- Lanxin Chen
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Bo Tang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guanghong Xie
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Rui Yang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Boyang Zhang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yueqi Wang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yan Zhang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Daozhen Jiang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xueming Zhang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
6
|
Weeratunga P, Harman RM, Van de Walle GR. Induced pluripotent stem cells from domesticated ruminants and their potential for enhancing livestock production. Front Vet Sci 2023; 10:1129287. [PMID: 36891466 PMCID: PMC9986305 DOI: 10.3389/fvets.2023.1129287] [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: 12/21/2022] [Accepted: 01/31/2023] [Indexed: 02/22/2023] Open
Abstract
Ruminant livestock, including cattle, sheep, goat, and buffalo, are essential for global food security and serve valuable roles in sustainable agricultural systems. With the limited availability of embryonic stem cells (ESCs) from these species, ruminant induced pluripotent stem cells (iPSCs) and iPSC-like cells provide a valuable research tool for agricultural, veterinary, biomedical, and pharmaceutical applications, as well as for the prospect of translation to human medicine. iPSCs are generated by reprogramming of adult or fetal cells to an ESC-like state by ectopic expression of defined transcription factors. Despite the slow pace the field has evolved in livestock species compared to mice and humans, significant progress has been made over the past 15 years in using different cell sources and reprogramming protocols to generate iPSCs/iPSC-like cells from ruminants. This mini review summarizes the current literature related to the derivation of iPSCs/iPSC-like cells from domesticated ruminants with a focus on reprogramming protocols, characterization, associated limitations, and potential applications in ruminant basic science research and production.
Collapse
Affiliation(s)
- Prasanna Weeratunga
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
7
|
Begum TF, Carpenter D. Health effects associated with phthalate activity on nuclear receptors. REVIEWS ON ENVIRONMENTAL HEALTH 2022; 37:567-583. [PMID: 34592072 DOI: 10.1515/reveh-2020-0162] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 09/09/2021] [Indexed: 06/13/2023]
Abstract
Phthalates are endocrine disruptors, widely used as plasticizers to impart flexibility in plastics, and as solvents in personal care products. Due to their nearly ubiquitous use in consumer products, most humans are exposed to phthalates daily. There has been extensive research on the reproductive health effects associated with phthalate exposure, but less attention has been paid to other actions. This review aims to summarize the known action of phthalates on different nuclear receptors. Some phthalates bind to and activate the estrogen receptor, making them weakly estrogenic. However, other phthalates antagonize androgen receptors. Some high molecular weight phthalates antagonize thyroid receptors, affecting metabolism. Several phthalates activate and interfere with the normal function of different peroxisome proliferator-activated receptors (PPARs), receptors that have critical roles in lipid metabolism and energy homeostasis. Some phthalates activate the aryl hydrocarbon receptor, which is critical for xenobiotic metabolism. Although phthalates have a short half-life in vivo, because people are continuously exposed, studies should examine the health effects of phthalates associated with long-term exposure. There is limited research on the effects of phthalates on health outcomes aside from reproductive function, particularly concerning are childhood adiposity, behavior, and learning. There is also limited information on actions of phthalates not mediated via nuclear receptors. Humans are exposed to multiple chemicals simultaneously, and how chemical mixtures act on nuclear receptor activity needs study. Although we know a great deal about phthalates, there is still much that remains uncertain. Future studies need to further examine their other potential health effects.
Collapse
Affiliation(s)
- Thoin Farzana Begum
- Department of Environmental Health Sciences, School of Public Health, University at Albany, Rensselaer, NY, USA
| | - David Carpenter
- Department of Environmental Health Sciences, School of Public Health, University at Albany, Rensselaer, NY, USA
- Institute for Health and the Environment, University at Albany, Rensselaer, NY, USA
| |
Collapse
|
8
|
Berlina AN, Ragozina MY, Komova NS, Serebrennikova KV, Zherdev AV, Dzantiev BB. Development of Lateral Flow Test-System for the Immunoassay of Dibutyl Phthalate in Natural Waters. BIOSENSORS 2022; 12:1002. [PMID: 36354511 PMCID: PMC9688391 DOI: 10.3390/bios12111002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
The use of a large amount of toxic synthetic materials leads to an increase in the pollution of environmental objects. Phthalates are compounds structurally related to esters of phthalic acid that are widely used in the manufacturing of synthetic packaging materials as plasticizers. Their danger is conditioned by leaching into the environment and penetrating into living organisms with negative consequences and effects on various organs and tissues. This work presents the first development of lateral flow immunoassay to detect dibutyl phthalate, one of the most common representatives of the phthalates group. To form a test zone, a hapten-protein conjugate was synthesized, and gold nanoparticles conjugated with antibodies to dibutyl phthalate were used as a detecting conjugate. The work includes the preparation of immunoreagents, selectivity investigation, and the study of the characteristics of the medium providing a reliable optical signal. Under the selected conditions for the analysis, the detection limit was 33.4 ng/mL, and the working range of the determined concentrations was from 42.4 to 1500 ng/mL. Time of the assay-15 min. The developed technique was successfully applied to detect dibutyl phthalate in natural waters with recovery rates from 75 to 115%.
Collapse
|
9
|
Recchia K, Pessôa LVDF, Pieri NCG, Pires PRL, Bressan FF. Influence of Cell Type in In Vitro Induced Reprogramming in Cattle. Life (Basel) 2022; 12:1139. [PMID: 36013318 PMCID: PMC9409886 DOI: 10.3390/life12081139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/16/2022] [Accepted: 07/24/2022] [Indexed: 06/15/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have been considered an essential tool in stem cell research due to their potential to develop new therapies and technologies and answer essential questions about mammalian early development. An important step in generating iPSCs is selecting their precursor cell type, influencing the reprogramming efficiency and maintenance in culture. In this study, we aim to characterize bovine mesenchymal cells from adipose tissue (bAdMSCs) and fetal fibroblasts (bFFs) and to compare the reprogramming efficiency of these cells when induced to pluripotency. The cells were characterized by immunostaining (CD90, SSEA1, SSEA3, and SSEA4), induced differentiation in vitro, proliferation rates, and were subjected to cell reprogramming using the murine OSKM transcription factors. The bFFs presented morphological changes resembling pluripotent cells after reprogramming and culture with different supplementation, and putative iPSCs were characterized by immunostaining (OCT4, SOX2, NANOG, and AP). In the present study, we demonstrated that cell line origin and cellular proliferation rate are determining factors for reprogramming cells into pluripotency. The generation of biPSCs is a valuable tool to improve both translational medicine and animal production and to study the different supplements required to maintain the pluripotency of bovine cells in vitro.
Collapse
Affiliation(s)
- Kaiana Recchia
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo 05508-270, SP, Brazil
| | - Laís Vicari de Figueiredo Pessôa
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (L.V.d.F.P.); (N.C.G.P.); (P.R.L.P.)
| | - Naira Caroline Godoy Pieri
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (L.V.d.F.P.); (N.C.G.P.); (P.R.L.P.)
| | - Pedro Ratto Lisboa Pires
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (L.V.d.F.P.); (N.C.G.P.); (P.R.L.P.)
| | - Fabiana Fernandes Bressan
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo 05508-270, SP, Brazil
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (L.V.d.F.P.); (N.C.G.P.); (P.R.L.P.)
| |
Collapse
|
10
|
Pillai VV, Koganti PP, Kei TG, Gurung S, Butler WR, Selvaraj V. Efficient induction and sustenance of pluripotent stem cells from bovine somatic cells. Biol Open 2021; 10:272681. [PMID: 34719702 PMCID: PMC8565620 DOI: 10.1242/bio.058756] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022] Open
Abstract
Although derivation of naïve bovine embryonic stem cells is unachieved, the possibility for generation of bovine induced pluripotent stem cells (biPSCs) has been generally reported. However, attempts to sustain biPSCs by promoting self-renewal have not been successful. Methods established for maintaining murine and human induced pluripotent stem cells (iPSCs) do not support self-renewal of iPSCs for any bovid species. In this study, we examined methods to enhance complete reprogramming and concurrently investigated signaling relevant to pluripotency of the bovine blastocyst inner cell mass (ICM). First, we identified that forced expression of SV40 large T antigen together with the reprogramming genes (OCT4, SOX2, KLF4 and MYC) substantially enhanced the reprogramming efficacy of bovine fibroblasts to biPSCs. Second, we uncovered that TGFβ signaling is actively perturbed in the ICM. Inhibition of ALK4/5/7 to block TGFβ/activin/nodal signaling together with GSK3β and MEK1/2 supported robust in vitro self-renewal of naïve biPSCs with unvarying colony morphology, steady expansion, expected pluripotency gene expression and committed differentiation plasticity. Core similarities between biPSCs and stem cells of the 16-cell-stage bovine embryo indicated a stable ground state of pluripotency; this allowed us to reliably gain predictive understanding of signaling in bovine pluripotency using systems biology approaches. Beyond defining a high-fidelity platform for advancing biPSC-based biotechnologies that have not been previously practicable, these findings also represent a significant step towards understanding corollaries and divergent aspects of bovine pluripotency. This article has an associated First Person interview with the joint first authors of the paper. Summary: Pluripotency reprogramming by overcoming the stable epigenome of bovine cells, and uncovering precise early embryo self-renewal mechanisms enables sustenance and expansion of authentic induced pluripotent stem cells in vitro.
Collapse
Affiliation(s)
- Viju Vijayan Pillai
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - Prasanthi P Koganti
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - Tiffany G Kei
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - Shailesh Gurung
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - W Ronald Butler
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| |
Collapse
|
11
|
Sundaravadivelu PK, Raina K, Thool M, Ray A, Joshi JM, Kaveeshwar V, Sudhagar S, Lenka N, Thummer RP. Tissue-Restricted Stem Cells as Starting Cell Source for Efficient Generation of Pluripotent Stem Cells: An Overview. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1376:151-180. [PMID: 34611861 DOI: 10.1007/5584_2021_660] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Induced pluripotent stem cells (iPSCs) have vast biomedical potential concerning disease modeling, drug screening and discovery, cell therapy, tissue engineering, and understanding organismal development. In the year 2006, a groundbreaking study reported the generation of iPSCs from mouse embryonic fibroblasts by viral transduction of four transcription factors, namely, Oct4, Sox2, Klf4, and c-Myc. Subsequently, human iPSCs were generated by reprogramming fibroblasts as a starting cell source using two reprogramming factor cocktails [(i) OCT4, SOX2, KLF4, and c-MYC, and (ii) OCT4, SOX2, NANOG, and LIN28]. The wide range of applications of these human iPSCs in research, therapeutics, and personalized medicine has driven the scientific community to optimize and understand this reprogramming process to achieve quality iPSCs with higher efficiency and faster kinetics. One of the essential criteria to address this is by identifying an ideal cell source in which pluripotency can be induced efficiently to give rise to high-quality iPSCs. Therefore, various cell types have been studied for their ability to generate iPSCs efficiently. Cell sources that can be easily reverted to a pluripotent state are tissue-restricted stem cells present in the fetus and adult tissues. Tissue-restricted stem cells can be isolated from fetal, cord blood, bone marrow, and other adult tissues or can be obtained by differentiation of embryonic stem cells or trans-differentiation of other tissue-restricted stem cells. Since these cells are undifferentiated cells with self-renewal potential, they are much easier to reprogram due to the inherent characteristic of having an endogenous expression of few pluripotency-inducing factors. This review presents an overview of promising tissue-restricted stem cells that can be isolated from different sources, namely, neural stem cells, hematopoietic stem cells, mesenchymal stem cells, limbal epithelial stem cells, and spermatogonial stem cells, and their reprogramming efficacy. This insight will pave the way for developing safe and efficient reprogramming strategies and generating patient-specific iPSCs from tissue-restricted stem cells derived from various fetal and adult tissues.
Collapse
Affiliation(s)
- Pradeep Kumar Sundaravadivelu
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Khyati Raina
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Madhuri Thool
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.,Department of Biotechnology, National Institute of Pharmaceutical Education and Research Guwahati, Changsari, Guwahati, Assam, India
| | - Arnab Ray
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Jahnavy Madhukar Joshi
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, India
| | - Vishwas Kaveeshwar
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, India
| | - S Sudhagar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Guwahati, Changsari, Guwahati, Assam, India
| | - Nibedita Lenka
- National Centre for Cell Science, S. P. Pune University Campus, Ganeshkhind, Pune, Maharashtra, India.
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| |
Collapse
|
12
|
Su Y, Wang L, Fan Z, Liu Y, Zhu J, Kaback D, Oudiz J, Patrick T, Yee SP, Tian X(C, Polejaeva I, Tang Y. Establishment of Bovine-Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms221910489. [PMID: 34638830 PMCID: PMC8508593 DOI: 10.3390/ijms221910489] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/04/2021] [Accepted: 09/24/2021] [Indexed: 11/22/2022] Open
Abstract
Pluripotent stem cells (PSCs) have been successfully developed in many species. However, the establishment of bovine-induced pluripotent stem cells (biPSCs) has been challenging. Here we report the generation of biPSCs from bovine mesenchymal stem cells (bMSCs) by overexpression of lysine-specific demethylase 4A (KDM4A) and the other reprogramming factors OCT4, SOX2, KLF4, cMYC, LIN28, and NANOG (KdOSKMLN). These biPSCs exhibited silenced transgene expression at passage 10, and had prolonged self-renewal capacity for over 70 passages. The biPSCs have flat, primed-like PSC colony morphology in combined media of knockout serum replacement (KSR) and mTeSR, but switched to dome-shaped, naïve-like PSC colony morphology in mTeSR medium and 2i/LIF with single cell colonization capacity. These cells have comparable proliferation rate to the reported primed- or naïve-state human PSCs, with three-germ layer differentiation capacity and normal karyotype. Transcriptome analysis revealed a high similarity of biPSCs to reported bovine embryonic stem cells (ESCs) and embryos. The naïve-like biPSCs can be incorporated into mouse embryos, with the extended capacity of integration into extra-embryonic tissues. Finally, at least 24.5% cloning efficiency could be obtained in nuclear transfer (NT) experiment using late passage biPSCs as nuclear donors. Our report represents a significant advance in the establishment of bovine PSCs.
Collapse
Affiliation(s)
- Yue Su
- Department of Animal Science, Institute of Systems Genetics, University of Connecticut, Storrs, CT 06268, USA; (Y.S.); (L.W.); (J.Z.); (J.O.); (X.T.)
| | - Ling Wang
- Department of Animal Science, Institute of Systems Genetics, University of Connecticut, Storrs, CT 06268, USA; (Y.S.); (L.W.); (J.Z.); (J.O.); (X.T.)
| | - Zhiqiang Fan
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (Z.F.); (Y.L.); (T.P.)
| | - Ying Liu
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (Z.F.); (Y.L.); (T.P.)
| | - Jiaqi Zhu
- Department of Animal Science, Institute of Systems Genetics, University of Connecticut, Storrs, CT 06268, USA; (Y.S.); (L.W.); (J.Z.); (J.O.); (X.T.)
| | - Deborah Kaback
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA; (D.K.); (S.P.Y.)
| | - Julia Oudiz
- Department of Animal Science, Institute of Systems Genetics, University of Connecticut, Storrs, CT 06268, USA; (Y.S.); (L.W.); (J.Z.); (J.O.); (X.T.)
| | - Tayler Patrick
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (Z.F.); (Y.L.); (T.P.)
| | - Siu Pok Yee
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA; (D.K.); (S.P.Y.)
| | - Xiuchun (Cindy) Tian
- Department of Animal Science, Institute of Systems Genetics, University of Connecticut, Storrs, CT 06268, USA; (Y.S.); (L.W.); (J.Z.); (J.O.); (X.T.)
| | - Irina Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (Z.F.); (Y.L.); (T.P.)
- Correspondence: (I.P.); (Y.T.)
| | - Young Tang
- Department of Animal Science, Institute of Systems Genetics, University of Connecticut, Storrs, CT 06268, USA; (Y.S.); (L.W.); (J.Z.); (J.O.); (X.T.)
- Correspondence: (I.P.); (Y.T.)
| |
Collapse
|
13
|
Mohammadi H, Ashari S. Mechanistic insight into toxicity of phthalates, the involved receptors, and the role of Nrf2, NF-κB, and PI3K/AKT signaling pathways. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:35488-35527. [PMID: 34024001 DOI: 10.1007/s11356-021-14466-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 05/14/2021] [Indexed: 06/12/2023]
Abstract
The wide use of phthalates, as phthalates are used in the manufacturing of not only plastics but also many others goods, has become a main concern in the current century because of their potency to induce deleterious effects on organism health. The toxic effects of phthalates such as reproductive toxicity, cardiotoxicity, hepatotoxicity, nephrotoxicity, teratogenicity, and tumor development have been widely indicated by previous experimental studies. Some of the important mechanisms of toxicity by phthalates are the induction and promotion of inflammation, oxidative stress, and apoptosis. Awareness of the involved molecular pathways of these mechanisms will permit the detection of exact molecular targets of phthalates to protect or treat their toxicity. Up to now, various transcription factors and signaling pathways have been associated with phthalate-induced toxicity which by influencing on nuclear surface and the expression of different genes can alter cell hemostasis. In different studies, the role of nuclear factor erythroid 2-related factor 2 (Nrf2), nuclear factor-κB (NF-κB), and phosphatidylinositol-3-kinase (PI3K)/AKT signaling pathways in processes of oxidative stress, inflammation, apoptosis, and cancer has been shown following exposure to phthalates. In the present review, we aim to survey experimental studies (in vitro and in vivo) in order to show firstly the most involved receptors and also the importance and the role of the mentioned signaling pathways in phthalate-induced toxicity, and with considering this point, the future studies can focus on these molecular targets as a strategic method to reduce environmental chemicals-induced toxicity especially phthalates toxic effects.
Collapse
Affiliation(s)
- Hamidreza Mohammadi
- Pharmaceutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Toxicology/Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sorour Ashari
- Department of Toxicology/Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
14
|
Strategy to Establish Embryo-Derived Pluripotent Stem Cells in Cattle. Int J Mol Sci 2021; 22:ijms22095011. [PMID: 34065074 PMCID: PMC8125899 DOI: 10.3390/ijms22095011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 12/29/2022] Open
Abstract
Stem cell research is essential not only for the research and treatment of human diseases, but also for the genetic preservation and improvement of animals. Since embryonic stem cells (ESCs) were established in mice, substantial efforts have been made to establish true ESCs in many species. Although various culture conditions were used to establish ESCs in cattle, the capturing of true bovine ESCs (bESCs) has not been achieved. In this review, the difficulty of establishing bESCs with various culture conditions is described, and the characteristics of proprietary induced pluripotent stem cells and extended pluripotent stem cells are introduced. We conclude with a suggestion of a strategy for establishing true bESCs.
Collapse
|
15
|
Baralić K, Jorgovanović D, Živančević K, Buha Djordjević A, Antonijević Miljaković E, Miljković M, Kotur-Stevuljević J, Antonijević B, Đukić-Ćosić D. Combining in vivo pathohistological and redox status analysis with in silico toxicogenomic study to explore the phthalates and bisphenol A mixture-induced testicular toxicity. CHEMOSPHERE 2021; 267:129296. [PMID: 33348264 DOI: 10.1016/j.chemosphere.2020.129296] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 06/12/2023]
Abstract
The aim of this study was to: (i) determine and compare the capacity of bis (2 -ethylhexyl) phthalate (DEHP), dibutyl phthalate (DBP), bisphenol A (BPA), and their mixture to produce testicular toxicity after the subacute exposure; (ii) explore the mechanisms behind the observed changes using in silico toxicogenomic approach. Male rats were randomly split into groups (n = 6): (1) Control (corn oil); (2) DEHP (50 mg/kg b.w./day); (3) DBP (50 mg/kg b.w./day); (4) BPA (25 mg/kg b.w./day); and (5) MIX (50 mg/kg b.w./day DEHP + 50 mg/kg b.w/day DBP + 25 mg/kg b.w./day BPA). Animals were sacrificed after 28 days of oral exposure, testes were extracted and prepared for histological assessments under the light microscope (haematoxylin and eosin staining) and redox status analysis. The Comparative Toxicogenomics Database (CTD; http://CTD.mdibl.org), Cytoscape software (https://cytoscape.org) and ToppGene Suite (https://toppgene.cchmc.org) were used for data-mining. Present pathohistological study has demonstrated more pronounced testicular toxicity of the MIX group (desquamated germinal epithelium cells, enlarged cells with hyperchromatic nuclei, multinucleated cell forms and intracytoplasmic vacuoles) in comparison with the single substances, while effects on redox status parameters were either more prominent, or present only in the MIX group. In silico investigation revealed 20 genes linked to male reproductive disorders, affected by all three investigated substances. Effects on metabolism, AhR pathway, apoptosis and oxidative stress could be singled out as the most probable mechanisms involved in the subacute DEHP, DBP and BPA mixture testicular toxicity, while the effect on oxidative stress parameters was confirmed by in vivo experiment.
Collapse
Affiliation(s)
- Katarina Baralić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia.
| | - Dragica Jorgovanović
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Katarina Živančević
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Aleksandra Buha Djordjević
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Evica Antonijević Miljaković
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Milica Miljković
- Department of Biochemistry, University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Jelena Kotur-Stevuljević
- Department of Biochemistry, University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Biljana Antonijević
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| | - Danijela Đukić-Ćosić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221, Belgrade, Serbia
| |
Collapse
|
16
|
Xiang J, Wang H, Zhang Y, Wang J, Liu F, Han X, Lu Z, Li C, Li Z, Gao Y, Tian Y, Wang Y, Li X. LCDM medium supports the derivation of bovine extended pluripotent stem cells with embryonic and extraembryonic potency in bovine-mouse chimeras from iPSCs and bovine fetal fibroblasts. FEBS J 2021; 288:4394-4411. [PMID: 33524211 DOI: 10.1111/febs.15744] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 12/19/2020] [Accepted: 01/29/2021] [Indexed: 12/15/2022]
Abstract
Cattle have emerged as one of the most important domestic animals widely used for meat, milk, and fur. Derivation of bovine pluripotent stem cells (PSCs) can be applied in drug selecting and human disease modeling and facilitated agriculture-related applications such as production of genetically excellent cattle by gene editing. Extended PSCs (EPSCs), capable of differentiating into embryonic and extraembryonic parts, have been generated in mouse, human, and pig. Whether bovine EPSCs could be generated, and their chimeric competency remains unclear. This study focused on derivation of bovine EPSCs using LCDM medium and exploring the characteristics of EPSCs among different species, including bovine, mouse, and human EPSCs. Here, using LCDM medium (consisting of hLIF, CHIR99021, (S)-(+)-dimethindene maleate, and minocycline hydrochloride) enables the derivation of bovine EPSCs from induced PSCs (iPSCs) and bovine fetal fibroblasts (BFF) with stable morphology, pluripotent marker expression, and in vitro differentiation ability. Notably, bovine EPSCs exhibited interspecies chimeric contribution to embryonic and extraembryonic tissues in pre-implantation blastocysts and postimplantation bovine-mouse chimeras. Transcriptome analysis revealed the unique molecular characteristics of bovine EPSCs compared with iPSCs. The similarities and differences in molecular features across bovine, human, and mouse EPSCs were also described by transcriptome analysis. Taken together, the LCDM culture system containing chemical cocktails can be used for the establishment and long-term passaging of bovine EPSCs with embryonic and extraembryonic potency in bovine-mouse chimeras. Our findings lay the foundation of generating PSCs in domestic animals and open avenues for basic and applied research in biology, medicine, and agriculture. DATABASE: Gene expression data of bovine EPSCs and bovine iPSCs are available in the GEO databases under the accession number PRJNA693452.
Collapse
Affiliation(s)
- Jinzhu Xiang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Hanning Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yuanyuan Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jing Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Fang Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xuejie Han
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Zhenyu Lu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Chen Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Zihong Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yanru Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yujing Tian
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yingjie Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xueling Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
17
|
Su Y, Zhu J, Salman S, Tang Y. Induced pluripotent stem cells from farm animals. J Anim Sci 2021; 98:5937369. [PMID: 33098420 DOI: 10.1093/jas/skaa343] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
The development of the induced pluripotent stem cells (iPSCs) technology has revolutionized the world on the establishment of pluripotent stem cells (PSCs) across a great variety of animal species. Generation of iPSCs from domesticated animals would provide unrestricted cell resources for the study of embryonic development and cell differentiation of these species, for screening and establishing desired traits for sustainable agricultural production, and as veterinary and preclinical therapeutic tools for animal and human diseases. Induced PSCs from domesticated animals thus harbor enormous scientific, economical, and societal values. Although much progress has been made toward the generation of PSCs from these species, major obstacles remain precluding the exclamation of the establishment of bona fide iPSCs. The most prominent of them remain the inability of these cells to silence exogenous reprogramming factors, the obvious reliance on exogenous factors for their self-renewal, and the restricted development potential in vivo. In this review, we summarize the history and current progress in domestic farm animal iPSC generation, with a focus on swine, ruminants (cattle, ovine, and caprine), horses, and avian species (quails and chickens). We also discuss the problems associated with the farm animal iPSCs and potential future directions toward the complete reprogramming of somatic cells from farm animals.
Collapse
Affiliation(s)
- Yue Su
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Jiaqi Zhu
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Saleh Salman
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Young Tang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT
| |
Collapse
|
18
|
Amir S, Shah STA, Mamoulakis C, Docea AO, Kalantzi OI, Zachariou A, Calina D, Carvalho F, Sofikitis N, Makrigiannakis A, Tsatsakis A. Endocrine Disruptors Acting on Estrogen and Androgen Pathways Cause Reproductive Disorders through Multiple Mechanisms: A Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:1464. [PMID: 33557243 PMCID: PMC7913912 DOI: 10.3390/ijerph18041464] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Increasing contamination of the environment by toxic compounds such as endocrine disrupting chemicals (EDCs) is one of the major causes of reproductive defects in both sexes. Estrogen/androgen pathways are of utmost importance in gonadal development, determination of secondary sex characteristics and gametogenesis. Most of the EDCs mediate their action through respective receptors and/or downstream signaling. The purpose of this review is to highlight the mechanism by which EDCs can trigger antagonistic or agonistic response, acting through estrogen/androgen receptors causing reproductive defects that lead to infertility. In vitro, in vivo and in silico studies focusing on the impact of EDCs on estrogen/androgen pathways and related proteins published in the last decade were considered for the review. PUBMED and PUBCHEM were used for literature search. EDCs can bind to estrogen receptors (ERα and ERβ) and androgen receptors or activate alternative receptors such as G protein-coupled receptors (GPCR), GPR30, estrogen-related receptor (ERRγ) to activate estrogen signaling via downstream kinases. Bisphenol A, dichlorodiphenyltrichloroethane, dichlorodiphenyldichloroethylene, polychlorinated biphenyls and phthalates are major toxicants that interfere with the normal estrogen/androgen pathways leading to infertility in both sexes through many ways, including DNA damage in spermatozoids, altered methylation pattern, histone modifications and miRNA expression.
Collapse
Affiliation(s)
- Saira Amir
- Department of Biosciences, COMSATS University Islamabad, Islamabad 44000, Pakistan; (S.A.); (S.T.A.S.)
| | - Syed Tahir Abbas Shah
- Department of Biosciences, COMSATS University Islamabad, Islamabad 44000, Pakistan; (S.A.); (S.T.A.S.)
| | - Charalampos Mamoulakis
- Department of Urology, University General Hospital of Heraklion, Medical School, University of Crete, 700 13 Heraklion, Greece
| | - Anca Oana Docea
- Department of Toxicology, Faculty of Pharmacy, University of Medicine and Pharmacy, Petru Rares, 200349 Craiova, Romania
| | - Olga-Ioanna Kalantzi
- Department of Environment, University of Aegean, University Hill, 81100 Mytilini, Greece;
| | - Athanasios Zachariou
- Department of Urology, Ioannina University School of Medicine, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Felix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
| | - Nikolaos Sofikitis
- Department of Urology, Ioannina University School of Medicine, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Antonios Makrigiannakis
- Department of Obstetrics and Gynecology, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
19
|
Scarfone RA, Pena SM, Russell KA, Betts DH, Koch TG. The use of induced pluripotent stem cells in domestic animals: a narrative review. BMC Vet Res 2020; 16:477. [PMID: 33292200 PMCID: PMC7722595 DOI: 10.1186/s12917-020-02696-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are undifferentiated stem cells characterized by the ability to differentiate into any cell type in the body. iPSCs are a relatively new and rapidly developing technology in many fields of biology, including developmental anatomy and physiology, pathology, and toxicology. These cells have great potential in research as they are self-renewing and pluripotent with minimal ethical concerns. Protocols for their production have been developed for many domestic animal species, which have since been used to further our knowledge in the progression and treatment of diseases. This research is valuable both for veterinary medicine as well as for the prospect of translation to human medicine. Safety, cost, and feasibility are potential barriers for this technology that must be considered before widespread clinical adoption. This review will analyze the literature pertaining to iPSCs derived from various domestic species with a focus on iPSC production and characterization, applications for tissue and disease research, and applications for disease treatment.
Collapse
Affiliation(s)
- Rachel A Scarfone
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Samantha M Pena
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Keith A Russell
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Dean H Betts
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Thomas G Koch
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada.
| |
Collapse
|
20
|
Companion animals get close to the toxic aspects of antropogenic world: cytotoxicity of phthalates and bisphenol A on dog testicular primary cells. Cytotechnology 2020; 72:629-638. [PMID: 32435861 DOI: 10.1007/s10616-020-00401-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/11/2020] [Indexed: 10/24/2022] Open
Abstract
Phthalates, which are among the most abundant plasticizers, have detrimental effects on the reproductive system. Similar to human, dogs are prominently exposed to phthalates in daily routines at low concentrations; while toys, training devices and commercial dog foods are considered as the primary sources of exposure. This study aimed to reveal and compare the cytotoxic effects of selected phthalates (Benzyl butyl phthalate (BBP), Diethyl phthalate (DEP), Bis (2-ethylhexyl) phthalate (DEHP), Di-'isobutyl' phthalate (DIBP), Di-'isodecyl' phthalate (-DIDP) Di-'isononyl' phthalate (DINP), Dimethyl phthalate (DMP), Di-n-octyl phthalate (DNOP)), and Bisphenol A (BPA) following 24 h exposure on primary testicular parenchymal cells of dog in vitro at concentrations between 0.001 and 2.5 nM. According to cytotoxicity results, DEHP was found to be the most toxic phthalate with IC50 at 22.53 µM; while DMP was the least (169.17 nM). IC50 of BPA was 161.81 nM, less than the average (61.95 nM) of phthalates. In addition, dog primary testicular cells were found more susceptible to the high molecular weight phthalates (DNOP, DEHP, DINP, DIDP) than low molecular weight phthalates (DMP, DEP, DIBP, BBP). Further studies should focus on morphological, physiological and molecular differences to comprehend the mechanisms involved as well as decreasing the risk for impaired spermatogenesis caused by environmental toxicants in companion animal medicine.
Collapse
|
21
|
Jiang Y, An XL, Yu H, Cai NN, Zhai YH, Li Q, Cheng H, Zhang S, Tang B, Li ZY, Zhang XM. Transcriptome profile of bovine iPSCs derived from Sertoli Cells. Theriogenology 2020; 146:120-132. [DOI: 10.1016/j.theriogenology.2019.11.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/16/2019] [Accepted: 11/17/2019] [Indexed: 12/18/2022]
|
22
|
Guvvala PR, Ravindra JP, Selvaraju S. Impact of environmental contaminants on reproductive health of male domestic ruminants: a review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:3819-3836. [PMID: 31845245 DOI: 10.1007/s11356-019-06980-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 11/06/2019] [Indexed: 06/10/2023]
Abstract
Environmental contaminants are gaining more attention in the livestock sector lately due to their harmful effects on productivity and fertility of livestock. Recent research indicates that many domestic ruminants are becoming subfertile/infertile due to confounding reasons associated with management. Contaminants like metals, metalloids, herbicides, pesticides, insecticides, chemicals, or natural contaminants are present everywhere in day to day life and are becoming a threat to the livestock. Studies on a broad-spectrum of animals suggest that high doses of acute or low doses of chronic exposure to the contaminants lead to disruption of multi-organs/systems including reproductive function. The lowered reproductive efficiency in animals is attributed to the endocrine disruptor activities of the environmental contaminants on the gonads, affecting gametogenesis and steroidogenesis. In vitro studies on testicular cells and the semen suggest that spermatozoa are more susceptible to damage by environmental contaminants. The quality of the semen happens to be a critical factor in the livestock industry. Contaminants affecting gametogenesis and steroidogenesis may lead to devastating consequences to the livestock reproduction, and thus the production. However, there is a lack of collective data on the effect of such environmental contaminants on the fertility of male domestic ruminants. This review discusses the studies related to the impact of environmental contaminants on male fertility in large (bull and buffalo) and small (sheep and goat) ruminants by focusing on the underlying molecular interactions between the contaminants and gonads.
Collapse
Affiliation(s)
- Pushpa Rani Guvvala
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Adugodi, Bengaluru, 560030, India.
| | - Janivara Parameswaraiah Ravindra
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Adugodi, Bengaluru, 560030, India
| | - Sellappan Selvaraju
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Adugodi, Bengaluru, 560030, India
- ICAR-National Fellow, ICAR-National Institute of Animal Nutrition and Physiology, Adugodi, Bengaluru, 560030, India
| |
Collapse
|
23
|
Glycerin Monostearate Aggravates Male Reproductive Toxicity Caused by Di(2-ethylhexyl) Phthalate in Rats. Curr Med Sci 2019; 39:1003-1008. [DOI: 10.1007/s11596-019-2135-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/17/2019] [Indexed: 11/27/2022]
|
24
|
Pessôa LVDF, Bressan FF, Freude KK. Induced pluripotent stem cells throughout the animal kingdom: Availability and applications. World J Stem Cells 2019; 11:491-505. [PMID: 31523369 PMCID: PMC6716087 DOI: 10.4252/wjsc.v11.i8.491] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023] Open
Abstract
Up until the mid 2000s, the capacity to generate every cell of an organism was exclusive to embryonic stem cells. In 2006, researchers Takahashi and Yamanaka developed an alternative method of generating embryonic-like stem cells from adult cells, which they coined induced pluripotent stem cells (iPSCs). Such iPSCs possess most of the advantages of embryonic stem cells without the ethical stigma associated with derivation of the latter. The possibility of generating “custom-made” pluripotent cells, ideal for patient-specific disease models, alongside their possible applications in regenerative medicine and reproduction, has drawn a lot of attention to the field with numbers of iPSC studies published growing exponentially. IPSCs have now been generated for a wide variety of species, including but not limited to, mouse, human, primate, wild felines, bovines, equines, birds and rodents, some of which still lack well-established embryonic stem cell lines. The paucity of robust characterization of some of these iPSC lines as well as the residual expression of transgenes involved in the reprogramming process still hampers the use of such cells in species preservation or medical research, underscoring the requirement for further investigations. Here, we provide an extensive overview of iPSC generated from a broad range of animal species including their potential applications and limitations.
Collapse
Affiliation(s)
- Laís Vicari de Figueiredo Pessôa
- Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Fabiana Fernandes Bressan
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-000, São Paulo, Brazil
| | - Kristine Karla Freude
- Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| |
Collapse
|
25
|
Baken KA, Lambrechts N, Remy S, Mustieles V, Rodríguez-Carrillo A, Neophytou CM, Olea N, Schoeters G. A strategy to validate a selection of human effect biomarkers using adverse outcome pathways: Proof of concept for phthalates and reproductive effects. ENVIRONMENTAL RESEARCH 2019; 175:235-256. [PMID: 31146096 DOI: 10.1016/j.envres.2019.05.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 05/21/2023]
Abstract
Human biomonitoring measures the concentrations of environmental chemicals or their metabolites in body fluids or tissues. Complementing exposure biomarkers with mechanistically based effect biomarkers may further elucidate causal pathways between chemical exposure and adverse health outcomes. We combined information on effect biomarkers previously implemented in human observational studies with mechanisms of action reported in experimental studies and with information from published Adverse Outcome Pathways (AOPs), focusing on adverse reproductive effects of phthalate exposure. Phthalates constitute a group of chemicals that are ubiquitous in consumer products and have been related to a wide range of adverse health effects. As a result of a comprehensive literature search, we present an overview of effect biomarkers for reproductive toxicity that are substantiated by mechanistic information. The activation of several receptors, such as PPARα, PPARγ, and GR, may initiate events leading to impaired male and female fertility as well as other adverse effects of phthalate exposure. Therefore, these receptors appear as promising targets for the development of novel effect biomarkers. The proposed strategy connects the fields of epidemiology and toxicology and may strengthen the weight of evidence in observational studies that link chemical exposures to health outcomes.
Collapse
Affiliation(s)
- Kirsten A Baken
- Unit Health, Flemish Institute for Technological Research (VITO NV), Mol, Belgium.
| | - Nathalie Lambrechts
- Unit Health, Flemish Institute for Technological Research (VITO NV), Mol, Belgium
| | - Sylvie Remy
- Unit Health, Flemish Institute for Technological Research (VITO NV), Mol, Belgium; Department of Epidemiology and Social Medicine, University of Antwerp, Antwerp, Belgium
| | - Vicente Mustieles
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, Granada, Spain; Center for Biomedical Research (CIBM), University of Granada, Granada, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain
| | | | - Christiana M Neophytou
- Department of Biological Sciences, School of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus
| | - Nicolas Olea
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, Granada, Spain; Center for Biomedical Research (CIBM), University of Granada, Granada, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain
| | - Greet Schoeters
- Unit Health, Flemish Institute for Technological Research (VITO NV), Mol, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Department of Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
26
|
Pillai VV, Kei TG, Reddy SE, Das M, Abratte C, Cheong SH, Selvaraj V. Induced pluripotent stem cell generation from bovine somatic cells indicates unmet needs for pluripotency sustenance. Anim Sci J 2019; 90:1149-1160. [PMID: 31322312 DOI: 10.1111/asj.13272] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/10/2019] [Accepted: 06/17/2019] [Indexed: 12/16/2022]
Abstract
Mechanisms that direct reprogramming of differentiated somatic cells to induced pluripotent stem cells (iPSCs), albeit incomplete in understanding, are highly conserved across all mammalian species studied. Equally, proof of principle that iPSCs can be derived from domestic cattle has been reported in several publications. In our efforts to derive and study bovine iPSCs, we encountered inadequacy of methods to generate, sustain, and characterize these cells. Our results suggest that iPSC protocols optimized for mouse and human somatic cells do not effectively translate to bovine somatic cells, which show some refractoriness to reprogramming that also affects sustenance. Moreover, methods that enhance reprogramming efficiency in mouse and human cells had no effect on improving bovine cell reprogramming. Although use of retroviral vectors coding for bovine OCT4, SOX2, KLF4, cMYC, and NANOG appeared to produce consistent iPSC-like cells from both fibroblasts and cells from the Wharton's jelly, these colonies could not be sustained. Use of bovine genes could successfully reprogram both mouse and human cells. These findings indicated either incomplete reprogramming and/or discordant/inadequate culture conditions for bovine pluripotent stem cells. Therefore, additional studies that advance core knowledge of bovine pluripotency are necessary before any anticipated iPSC-driven bovine technologies can be realized.
Collapse
Affiliation(s)
- Viju V Pillai
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Tiffany G Kei
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Shannon E Reddy
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Moubani Das
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Christian Abratte
- iPSC Core Laboratory, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Soon H Cheong
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| |
Collapse
|
27
|
Wang C, Zheng P, Adeniran SO, Ma M, Huang F, Adegoke EO, Zhang G. Thyroid hormone (T 3) is involved in inhibiting the proliferation of newborn calf Sertoli cells via the PI3K/Akt signaling pathway in vitro. Theriogenology 2019; 133:1-9. [PMID: 31051388 DOI: 10.1016/j.theriogenology.2019.04.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 04/19/2019] [Accepted: 04/19/2019] [Indexed: 11/30/2022]
Abstract
The experiment was designed to study the effects of Thyroid hormone (T3) on the proliferation and differentiation of newborn calf Sertoli cells (SCs) to provide a theoretical and practical basis for increased testicular semen production. In this experiment, the cck8 method was used to detect the effects of different concentrations of T3 on the proliferation rate of newborn calf SCs. qPCR and Western Blot methods were used to explore the effects of T3 on the proliferation and differentiation of calves SCs and whether T3 through Wnt/β-catenin and PI3K/Akt pathways can regulate the proliferation and differentiation of SCs. We found that dosage (T3) and time correlated with proliferation inhibition of SC. T3 inhibited the proliferation of SC by down-regulating cyclinD1, upregulating p21Cip, p27Kip1, and other cell-cycle factors. By up-regulating AR and down-regulating KRT-18, T3 promoted the maturated differentiation of SC. T3 could not affect the expression of β-catenin in SC of newborn calf, indicating that T3 may not regulate SCs proliferation through the Wnt pathway. T3 also negatively regulated the gene expression and protein levels of some genes in the PI3K/Akt signaling pathway. We concluded that T3 inhibited newborn calf SCs proliferation through the PI3K/Akt signaling pathway and possibly promoted their differentiation.
Collapse
Affiliation(s)
- Chen Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Peng Zheng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - S O Adeniran
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Mingjun Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Fushuo Huang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - E O Adegoke
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Guixue Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
28
|
Pawłowski K, Pires JAA, Faulconnier Y, Chambon C, Germon P, Boby C, Leroux C. Mammary Gland Transcriptome and Proteome Modifications by Nutrient Restriction in Early Lactation Holstein Cows Challenged with Intra-Mammary Lipopolysaccharide. Int J Mol Sci 2019; 20:E1156. [PMID: 30845783 PMCID: PMC6429198 DOI: 10.3390/ijms20051156] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/22/2019] [Accepted: 02/27/2019] [Indexed: 11/27/2022] Open
Abstract
: The objective is to study the effects of nutrient restrictions, which induce a metabolic imbalance on the inflammatory response of the mammary gland in early lactation cows. The aim is to decipher the molecular mechanisms involved, by comparing a control, with a restriction group, a transcriptome and proteome, after an intra-mammary lipopolysaccharide challenge. Multi-parous cows were either allowed ad libitum intake of a lactation diet (n = 8), or a ration containing low nutrient density (n = 8; 48% barley straw and dry matter basis) for four days starting at 24 ± 3 days in milk. Three days after the initiation of their treatments, one healthy rear mammary quarter of 12 lactating cows was challenged with 50 µg of lipopolysaccharide (LPS). Transcriptomic and proteomic analyses were performed on mammary biopsies obtained 24 h after the LPS challenge, using bovine 44K microarrays, and nano-LC-MS/MS, respectively. Restriction-induced deficits in energy, led to a marked negative energy balance (41 versus 97 ± 15% of Net Energy for Lactation (NEL) requirements) and metabolic imbalance. A microarray analyses identified 25 differentially expressed genes in response to restriction, suggesting that restriction had modified mammary metabolism, specifically β-oxidation process. Proteomic analyses identified 53 differentially expressed proteins, which suggests that the modification of protein synthesis from mRNA splicing to folding. Under-nutrition influenced mammary gland expression of the genes involved in metabolism, thereby increasing β-oxidation and altering protein synthesis, which may affect the response to inflammation.
Collapse
Affiliation(s)
- Karol Pawłowski
- Université Clermont Auvergne,INRA, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France.
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences,02-776 Warsaw, Poland.
| | - José A A Pires
- Université Clermont Auvergne,INRA, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France.
| | - Yannick Faulconnier
- Université Clermont Auvergne,INRA, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France.
| | - Christophe Chambon
- INRA, INRA, Plateforme d'Exploration du Métabolisme, composante protéomique PFEMcp), F-63122 Saint-Genès Champanelle, France.
| | - Pierre Germon
- INRA Val de Loire, UMR ISP, F-37380 Nouzilly, France.
| | - Céline Boby
- Université Clermont Auvergne,INRA, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France.
| | - Christine Leroux
- Université Clermont Auvergne,INRA, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France.
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
29
|
Yimpring N, Teankum K, Srisuwatanasagul S, Kunnasut N, Am-In N, Suriyaphol G. Alteration of androgen receptor expression, apoptosis and cell proliferation in cryptorchid suckling, nursery and growing-finishing pigs. Theriogenology 2019; 127:49-55. [PMID: 30665073 DOI: 10.1016/j.theriogenology.2019.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 01/03/2019] [Accepted: 01/10/2019] [Indexed: 12/18/2022]
Abstract
Cryptorchidism, a condition of one or two undescended testicles, is a common congenital disease in pigs, causing loss in the pig industry. One of the major factors affecting testicular descent is the androgen receptor (AR), which binds to androgen and then regulates the expression of androgen-responsive genes in the inguinoscrotal phase of testicular descent. AR expression has been reported to regulate apoptosis in testicular stem cells. The present study aimed to immunohistochemically examine AR and Ki-67 protein expression and apoptosis detection in unilateral undescended testicles (UDT) and descended testicles in cryptorchid pigs (DT) of suckling (aged 1-2 weeks), nursery (aged 6 weeks) and growing-finishing pigs (aged 12, 15 and 20 weeks) and in normal testicles (NT) at 1-2 and 12 weeks of age. At 1-2 weeks, decreased expression of AR was observed in UDT and DT compared with NT and was lower than that at 6-20 weeks. The expression of Ki-67, a marker of cell proliferation, in UDT and DT at 12 weeks was lower than that in NT at the same age. In addition, Ki-67 expression in UDT at 6 and 12 weeks was lower than that in UDT at 1-2 and 15-20 weeks. More testicular apoptosis was revealed in UDT at 1-2 weeks than in DT and NT at the same age. At 15-20 weeks, more apoptosis was detected in UDT than in DT. Positive correlation of AR expression in DT at 6 and 12 weeks was also noted, in addition to the association of the expression of AR and Ki-67 in NT at 12 weeks. Taken together, this study unveiled the low expression of AR and high apoptosis detection in UDT, whereas low expression of AR and low apoptosis detection were noted in DT in suckling piglets. Diminished cell proliferation was shown in UDT at 6-12 weeks, whereas high apoptosis was observed in UDT at 15-20 weeks. High expression of AR was shown only in nursery pigs. Distinct expression of AR in DT and NT at 1-2 and 12 weeks indicated that both conditions were not interchangeable.
Collapse
Affiliation(s)
- Nathamon Yimpring
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Komkrich Teankum
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sayamon Srisuwatanasagul
- Department of Anatomy, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Nanthida Kunnasut
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Nutthee Am-In
- Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Gunnaporn Suriyaphol
- Biochemistry Unit, Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
30
|
Saito S, Lin YC, Nakamura Y, Eckner R, Wuputra K, Kuo KK, Lin CS, Yokoyama KK. Potential application of cell reprogramming techniques for cancer research. Cell Mol Life Sci 2019; 76:45-65. [PMID: 30283976 PMCID: PMC6326983 DOI: 10.1007/s00018-018-2924-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 09/15/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023]
Abstract
The ability to control the transition from an undifferentiated stem cell to a specific cell fate is one of the key techniques that are required for the application of interventional technologies to regenerative medicine and the treatment of tumors and metastases and of neurodegenerative diseases. Reprogramming technologies, which include somatic cell nuclear transfer, induced pluripotent stem cells, and the direct reprogramming of specific cell lineages, have the potential to alter cell plasticity in translational medicine for cancer treatment. The characterization of cancer stem cells (CSCs), the identification of oncogene and tumor suppressor genes for CSCs, and the epigenetic study of CSCs and their microenvironments are important topics. This review summarizes the application of cell reprogramming technologies to cancer modeling and treatment and discusses possible obstacles, such as genetic and epigenetic alterations in cancer cells, as well as the strategies that can be used to overcome these obstacles to cancer research.
Collapse
Affiliation(s)
- Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita, Tochigi, 329-1571, Japan
- College of Engineering, Nihon University, Koriyama, Fukushima, 963-8642, Japan
| | - Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Richard Eckner
- Department of Biochemistry and Molecular Biology, Rutgers, New Jersey Medical School-Rutgers, The State University of New Jersey, Newark, NJ, 07101, USA
| | - Kenly Wuputra
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Kung-Kai Kuo
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Chang-Shen Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.
| | - Kazunari K Yokoyama
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Faculty of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
31
|
Wu W, Wu P, Yang F, Sun DL, Zhang DX, Zhou YK. Association of phthalate exposure with anthropometric indices and blood pressure in first-grade children. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:23125-23134. [PMID: 29860692 DOI: 10.1007/s11356-018-2447-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 05/29/2018] [Indexed: 06/08/2023]
Abstract
We aimed to assess the relationship of urine phthalate metabolite concentrations with anthropometric indices, and blood pressure in first-grade children. We detected 11 phthalate metabolites in urine and estimated anthropometric indices, including skinfold measurements, waist circumference (WC), and body mass index (BMI) in 276 children aged 6-8 years. Multivariate linear regression models were used to assess the associations between urinary phthalate metabolite levels, and anthropometric and blood pressure indices in a gender-specific manner. In boys, a 1-ng/mL increase in monobenzyl phthalate (MBzP) concentration was associated with a 0.027-cm decrease in the skinfold measurement (95% confidence interval [CI], - 0.053 to 0.001), whereas a 1-ng/mL increase in mono-ethyl-phthalate (MEP) concentration was associated with a 0.016-mm Hg decrease in systolic blood pressure (95% CI, - 0.031 to 0.001). MBzP, mono-(2-ethyl-5-carboxypentyl) phthalate (MECPP), and MEOHP concentrations were also inversely associated with WC. However, in girls, MEP concentrations were positively associated with chest measurements, but were inversely associated with WC. A 1-ng/mL increase in monomethyl phthalate concentrations was associated with a 0.039-cm increase in skinfold measurements (95% CI, 0.002 to 0.076), whereas a 1-ng/mL increase in MECPP concentrations was associated with a 0.050 cm decrease in skinfold measurements (95% CI, - 0.095 to - 0.005). In this exploratory, cross-sectional analysis, we identified various interesting associations between different phthalate metabolite levels and anthropometric indices, which suggest that some of phthalate metabolite should be considered in addition to the prevalence rates of overweight and obesity.
Collapse
Affiliation(s)
- Wei Wu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan, 430065, China.
| | - Ping Wu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan, 430065, China
| | - Fang Yang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan, 430065, China
| | - Dan-Ling Sun
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan, 430065, China
| | - De-Xing Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan, 430065, China
| | - Yi-Kai Zhou
- MOE Key Laboratory of Environment & Health, Institute of Environmental Medicine, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
32
|
Zhao S, Li D, Bei XY, Zhu YP, Sun WL, Shen C, Wood K, Han BM, Jiang JT. Maternal exposure to di-n-butyl phthalate (DBP) promotes epithelial-mesenchymal transition via regulation of autophagy in uroepithelial cell. Toxicology 2018; 406-407:114-122. [PMID: 30053495 DOI: 10.1016/j.tox.2018.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/07/2018] [Accepted: 07/23/2018] [Indexed: 01/09/2023]
Abstract
Maternal exposure to di-n-butyl phthalate (DBP) induces hypospadias, but the underlying mechanisms remain elusive. Here we hypothesize that aberrant activation of autophagy and epithelial-mesenchymal transition (EMT) are the leading cause of DBP-related hypospadias. Pregnant rats received DBP orally at a dose of 750 mg/kg/day during gestational days 14-18. In DBP-induced hypospadiac male offspring, immunohistochemistry (IHC) staining and Western blot showed increased expression of autophagy and EMT markers in genital tubercle (GT) tissue compared to the control. In addition, lower testosterone levels and androgen receptor (AR) expression in GT tissue were detected. In vitro studies revealed that impaired AR signaling was involved in DBP-induced autophagy and autophagy activation furthermore promoted EMT in urethral epithelial cells. DBP combined with chloroquine, an autophagy inhibitor, reduced the expression of EMT markers compared with DBP treatment alone, while DBP combined with the autophagy inducer rapamycin elevated the expression of EMT markers. The autophagy-lysosomal pathway inhibitor CQ but not proteasome inhibitor MG-132 rescued the decrease of E-cadherin after DBP treatment, which indicated autophagy-induced E-cadherin degradation contributes to DBP-related EMT. Taken together, our findings show that prenatal exposure to DBP induces abnormal autophagy and EMT that may play important roles in hypospadias development.
Collapse
Affiliation(s)
- Sheng Zhao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Deng Li
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiao-Yu Bei
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yi-Ping Zhu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wen-Lan Sun
- Department of Geriatrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Cheng Shen
- The First Affiliated Yijishan Hospital of Wannan Medical College, China
| | - Kristofer Wood
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ 85013, United States
| | - Bang-Min Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Jun-Tao Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
33
|
Oncogenic function of the homeobox A13-long noncoding RNA HOTTIP-insulin growth factor-binding protein 3 axis in human gastric cancer. Oncotarget 2017; 7:36049-36064. [PMID: 27144338 PMCID: PMC5094982 DOI: 10.18632/oncotarget.9102] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 04/11/2016] [Indexed: 12/11/2022] Open
Abstract
To study the mechanisms of gastric tumorigenesis, we have established CSN cell line from human normal gastric mucosa, and CS12, a tumorigenic and invasive gastric cancer cell line from CSN passages. Many stem cell markers were expressed in both CSN and CS12 cells, but LGR5 and NANOG were expressed only in CS12 cells. Increased expression of homeobox A13 (HoxA13) and its downstream cascades was significant for the tumorigenic activity of CS12 cells, and was associated with recruitment of E2F-1 to HoxA13 promoter accompanied with increased trimethylation of histone H3 lysine 4 (H3K4me3) at the hypomethylated E2F motifs. Knockdown of HoxA13 caused the downregulation of long non-coding RNA HOTTIP and insulin growth factor-binding protein 3 (IGFBP-3) genes, indicating that both were targets of HoxA13. Concurrent regulation of HoxA13-HOTTIP was mediated by the mixed lineage leukemia-WD repeat domain 5 complex, which caused the trimethylation of H3K4 and then stimulated cell proliferation. HoxA13 transactivated the IGFBP-3 promoter through the HOX-binding site. Activation of IGFBP-3 stimulated the oncogenic potential and invasion activity. Increased expression of HoxA13 (63.2%) and IGFBP-3 (28.6%) was detected in human gastric cancer tissues and was found in the gastric cancer data of The Cancer Genome Atlas. Taken together, the HoxA13–HOTTIP–IGFBP-3 cascade is critical for the carcinogenic characteristics of CS12 cells.
Collapse
|
34
|
Ma T, Yin X, Han R, Ding J, Zhang H, Han X, Li D. Effects of In Utero Exposure to Di-n-Butyl Phthalate on Testicular Development in Rat. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14101284. [PMID: 29064414 PMCID: PMC5664784 DOI: 10.3390/ijerph14101284] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 10/20/2017] [Accepted: 10/20/2017] [Indexed: 11/16/2022]
Abstract
Humans are inevitably exposed to ubiquitous phthalate esters (PAEs). In utero exposure to di-n-butyl phthalate (DBP) induces abnormal development of the testis and reproductive tract in male offspring, which correspond closely with the human condition of testicular dysgenesis syndrome (TDS)-like syndrome. However, the underlying mechanisms have not been elucidated in detail. In this study, pregnant rats were orally exposed to either corn oil (controls) or DBP at three different doses by gavage during Gestational Days 12.5-21.5. Pathological examinations were performed for toxicity evaluation. Proliferation and apoptosis related proteins (ras related dexamethasone induced 1 (Rasd1), mitogen-activated protein kinase kinases1/2 (MEK1/2), Bcl-2, and Bax) were measured for mechanisms exploration. The results showed that different doses of DBP caused male developmental and reproductive toxicity in rats, including the decrease of anogenital distance (AGD), the histological damage of testis, and apoptosis of seminiferous tubule cells. Our data suggested that DBP played chronic and continuous toxic roles on male reproductive system by disrupting expression of Rasd1 and MEK1/2 as well as Bcl-2/Bax ratio. Further research is warranted.
Collapse
Affiliation(s)
- Tan Ma
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Xiaoqin Yin
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Ruitong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Jie Ding
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Huan Zhang
- Department of Clinical and Experimental Medicine, Linköping University, SE-581 83 Linköping, Sweden.
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Dongmei Li
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
35
|
Wu DC, Wang SSW, Liu CJ, Wuputra K, Kato K, Lee YL, Lin YC, Tsai MH, Ku CC, Lin WH, Wang SW, Kishikawa S, Noguchi M, Wu CC, Chen YT, Chai CY, Lin CLS, Kuo KK, Yang YH, Miyoshi H, Nakamura Y, Saito S, Nagata K, Lin CS, Yokoyama KK. Reprogramming Antagonizes the Oncogenicity of HOXA13-Long Noncoding RNA HOTTIP Axis in Gastric Cancer Cells. Stem Cells 2017; 35:2115-2128. [PMID: 28782268 DOI: 10.1002/stem.2674] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 06/26/2017] [Accepted: 07/15/2017] [Indexed: 12/26/2022]
Abstract
Reprogramming of cancer cells into induced pluripotent stem cells (iPSCs) is a compelling idea for inhibiting oncogenesis, especially through modulation of homeobox proteins in this reprogramming process. We examined the role of various long noncoding RNAs (lncRNAs)-homeobox protein HOXA13 axis on the switching of the oncogenic function of bone morphogenetic protein 7 (BMP7), which is significantly lost in the gastric cancer cell derived iPS-like cells (iPSLCs). BMP7 promoter activation occurred through the corecruitment of HOXA13, mixed-lineage leukemia 1 lysine N-methyltransferase, WD repeat-containing protein 5, and lncRNA HoxA transcript at the distal tip (HOTTIP) to commit the epigenetic changes to the trimethylation of lysine 4 on histone H3 in cancer cells. By contrast, HOXA13 inhibited BMP7 expression in iPSLCs via the corecruitment of HOXA13, enhancer of zeste homolog 2, Jumonji and AT rich interactive domain 2, and lncRNA HoxA transcript antisense RNA (HOTAIR) to various cis-element of the BMP7 promoter. Knockdown experiments demonstrated that HOTTIP contributed positively, but HOTAIR regulated negatively to HOXA13-mediated BMP7 expression in cancer cells and iPSLCs, respectively. These findings indicate that the recruitment of HOXA13-HOTTIP and HOXA13-HOTAIR to different sites in the BMP7 promoter is crucial for the oncogenic fate of human gastric cells. Reprogramming with octamer-binding protein 4 and Jun dimerization protein 2 can inhibit tumorigenesis by switching off BMP7. Stem Cells 2017;35:2115-2128.
Collapse
Affiliation(s)
- Deng-Chyang Wu
- Division of Gastroenterology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Stem Cell Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Sophie S W Wang
- Division of Gastroenterology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Stem Cell Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Jung Liu
- Division of Gastroenterology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Stem Cell Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kohsuke Kato
- Department of Infection Biology, Graduate School of Comprehensive Human Sciences, the University of Tsukuba, Tsukuba, Japan
| | | | - Ying-Chu Lin
- School of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Ho Tsai
- Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Hsin Lin
- Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shin-Wei Wang
- Division of Gastroenterology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Stem Cell Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shotaro Kishikawa
- Gene Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Michiya Noguchi
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Chu-Chieh Wu
- Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ting Chen
- Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chen-Lung Steve Lin
- Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kung-Kai Kuo
- Center for Stem Cell Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ya-Han Yang
- Center for Stem Cell Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hiroyuki Miyoshi
- Department of Physiology, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Shigeo Saito
- School of Science and Engineering, Teikyo University, Utsunomia, Tochigi, Japan.,Saito Laboratory of Cell Technology, Yaita, Tochigi, Japan
| | - Kyosuke Nagata
- Department of Infection Biology, Graduate School of Comprehensive Human Sciences, the University of Tsukuba, Tsukuba, Japan
| | - Chang-Shen Lin
- Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Kazunari K Yokoyama
- Center for Stem Cell Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Infection Biology, Graduate School of Comprehensive Human Sciences, the University of Tsukuba, Tsukuba, Japan.,Department of Molecular Preventive Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
36
|
Characterization of the single-cell derived bovine induced pluripotent stem cells. Tissue Cell 2017; 49:521-527. [PMID: 28720304 DOI: 10.1016/j.tice.2017.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/19/2017] [Accepted: 05/20/2017] [Indexed: 12/12/2022]
Abstract
Single-cell derived bovine induced pluripotent stem cells (iPSCs) were generated by the introduction of piggyBac transposons with CAG promoting transcription factors (Oct3/4, Sox2, Klf4 and cMyc). In the study, the bovine iPSCs colony from single cell could passage more than 50 passages after enzymatic dissociation into single cells. These bovine iPSCs cells kept the normal karyotype and displayed dome shaped clones similar to mouse embryonic stem cells. They showed pluripotency in many ways, including their expression of pluripotency markers, such as OCT3/4, NANOG, SOX2, SSEA1, SSEA4, and AP in immunofluorescence assay, Oct4, Nanog, Sox2, Klf4 and cMyc in RT-PCR. Additionally, single-cell derived bovine iPSCs formed embryoid bodies and teratomas that all subsequently gave rise to differentiated cells from all three embryonic germ layers. The results showed that our reprogramming method could obtain high efficiency single-cell cloning bovine iPSCs, and the efficiency of single cell cloning is 40%.
Collapse
|
37
|
Kim D, Jung YG, Roh S. Microarray analysis of embryo-derived bovine pluripotent cells: The vulnerable state of bovine embryonic stem cells. PLoS One 2017; 12:e0173278. [PMID: 28257460 PMCID: PMC5336296 DOI: 10.1371/journal.pone.0173278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/17/2017] [Indexed: 12/20/2022] Open
Abstract
Although there are many studies about pluripotent stem cells, little is known about pluripotent pathways and the difficulties of maintaining the pluripotency of bovine cells in vitro. Here, we investigated differently expressed genes (DEG) in bovine embryo-derived stem-like cells (eSLCs) from various origins to validate their distinct characteristics of pluripotency and differentiation. We identified core pluripotency markers and additional markers which were not determined as pluripotency markers yet in bovine eSLCs. Using the KEGG database, TGFβ, WNT, and LIF signaling were related to the maintenance of pluripotency. In contrast, some DEGs related to the LIF pathway were down-regulated, suggesting that reactivation of the pathway may be required for the establishment of true bovine embryonic stem cells (ESCs). Interestingly, oncogenes were co-down-regulated, while tumor suppressor genes were co-up-regulated in eSLCs, implying that this pattern may induce abnormal teratomas. These data analyses of signaling pathways provide essential information on authentic ESCs in addition to providing evidence for pluripotency in bovine eSLCs.
Collapse
Affiliation(s)
- Daehwan Kim
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | | | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
38
|
Gao HT, Xu R, Cao WX, Qian LL, Wang M, Lu L, Xu Q, Yu SQ. Effects of six priority controlled phthalate esters with long-term low-dose integrated exposure on male reproductive toxicity in rats. Food Chem Toxicol 2017; 101:94-104. [PMID: 28089693 DOI: 10.1016/j.fct.2017.01.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 01/16/2023]
Abstract
Human beings are inevitably exposed to ubiquitous phthalate esters (PEs) surroundings. The purposes of this study were to investigate the effects of long-term low-dose exposure to the mixture of six priority controlled phthalate esters (MIXPs): dimethyl phthalate (DMP), diethyl phthalate (DEP), di(n-butyl) phthalate (DBP), butyl benzyl phthalate (BBP), di(2-ethyhexyl) phthalate (DEHP) and di-n-octyl phthalate (DNOP), on male rat reproductive system and further to explore the underlying mechanisms of the reproductive toxicity. The male rats were orally exposed to either sodium carboxymethyl cellulose as controls or MIXPs at three different low-doses by gavage for 15 weeks. Testosterone and luteinizing hormone (LH) in serum were analyzed, and pathological examinations were performed for toxicity evaluation. Steroidogenic proteins (StAR, P450scc, CYP17A1 and 17β-HSD), cell cycle and apoptosis-related proteins (p53, Chk1, Cdc2, CDK6, Bcl-2 and Bax) were measured for mechanisms exploration. MIXPs with long-term low-dose exposure could cause male reproductive toxicity to the rats, including the decrease of both serum and testicular testosterone, and the constructional damage of testis. These effects were related to down-regulated steroidogenic proteins, arresting cell cycle progression and promoting apoptosis in rat testicular cells. The results indicate that MIXPs with long-term low-dose exposure may pose male reproductive toxicity in human.
Collapse
Affiliation(s)
- Hai-Tao Gao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Run Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Wei-Xin Cao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Liang-Liang Qian
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Min Wang
- Zibo Municipal Center for Disease Control and Prevention, Zibo 255026, China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Yale University, New Haven, CT 06520-8034, USA
| | - Qian Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Yale University, New Haven, CT 06520-8034, USA.
| | - Shu-Qin Yu
- Jiangsu Key Laboratory for Supramolecular Medicinal Material and Applications, College of Life Sciences, Nanjing Normal University, Nanjing 210046, China; Jiangsu Province Key Laboratory for Molecular and Medicinal Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, China.
| |
Collapse
|
39
|
Ling J, Lopez-Dee ZP, Cottell C, Wolfe L, Nye D. Regulation of mRNA Translation Is a Novel Mechanism for Phthalate Toxicity. PLoS One 2016; 11:e0167914. [PMID: 27992464 PMCID: PMC5167351 DOI: 10.1371/journal.pone.0167914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/22/2016] [Indexed: 12/29/2022] Open
Abstract
Phthalates are a group of plasticizers that are widely used in many consumer products and medical devices, thus generating a huge burden to human health. Phthalates have been known to cause a number of developmental and reproductive disorders functioning as endocrine modulators. They are also involved in carcinogenesis with mechanisms less understood. To further understand the molecular mechanisms of phthalate toxicity, in this study we reported a new effect of phthalates on mRNA translation/protein synthesis, a key regulatory step of gene expression. Butyl benzyl phthalate (BBP) was found to directly inhibit mRNA translation in vitro but showed a complicated pattern of affecting mRNA translation in cells. In human kidney embryonic cell (HEK-293T), BBP increased cap-dependent mRNA translation at lower concentrations but showed inhibitory effect at higher concentrations. Cap-independent translation was not affected. On the other hand, mono (2-ethylhexyl) phthalate (MEHP) as a major metabolite of another important phthalate di (2-ethylhexyl) phthalate (DEHP) inhibited both can-dependent and -independent mRNA translation in vivo. In contrast, BBP and MEHP exhibited an overall promoting effect on mRNA translation in cancer cells. Mechanistic studies identified that the level and phosphorylation of eIF4E-BP (eIF4E binding protein) and the amount of eIF4GI in eIF4F complex were altered in accordance with the effect of BBP on translation. BBP was also identified to directly bind to eIF4E, providing a further mechanism underlying the regulation of mRNA by phthalate. At the cellular level BBP inhibited normal cell growth but slightly promoted cancer cells (HT29) growth. Overall, this study provides the first evidence that phthalates can directly regulate mRNA translation as a novel mechanism to mediate their biological toxicities.
Collapse
Affiliation(s)
- Jun Ling
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
- * E-mail:
| | - Zenaida P. Lopez-Dee
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
| | - Colby Cottell
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
- Department of Sciences, Marywood University, Scranton, Pennsylvania, United States of America
| | - Laura Wolfe
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
- Department of Sciences, Marywood University, Scranton, Pennsylvania, United States of America
| | - Derek Nye
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania, United States of America
- Department of Biology, Wilkes University, Wilkes-Barre, Pennsylvania, United States of America
| |
Collapse
|
40
|
Shen C, Wu S, Chen H, Rashid S, Wen Y. Phthalate degradation by glow discharge plasma enhanced with pyrite in aqueous solution. WATER SCIENCE AND TECHNOLOGY : A JOURNAL OF THE INTERNATIONAL ASSOCIATION ON WATER POLLUTION RESEARCH 2016; 74:1365-1375. [PMID: 27685966 DOI: 10.2166/wst.2016.316] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In order to prevent health risk from potential exposures to phthalates, a glow discharge plasma (GDP) process was applied for phthalate degradation in aqueous solution. The results revealed that the phthalate derivatives 4-hydroxyphthalic acid, 4-methylphthalic acid and 4-tert-butylphthalic anhydride could be degraded efficiently in GDP process (498 V, 0.2 A) with high removal efficiencies of over 99% in 60 minutes. Additionally, pyrite as a promising heterogeneous iron source in the Fenton reaction was found to be favorable for GDP process. The phthalate degradation reaction could be significantly enhanced by the continuous formation of •OH and the inhibition of the quenching reaction in the pyrite Fenton system due to the constant dissolution of Fe(II) from pyrite surface. Meanwhile, the initial pH value showed little impact on the degradation of phthalates and the energy efficiency of GDP system for phthalate degradation ranged between 0.280 × 10-9 and 1.210 × 10-9 mol/J, which is similar to the GDP system with phenol, bisphenol A and methyl tert-butyl ether as the substrates. Further, the X-ray diffraction and scanning electron microscopy with energy dispersive X-ray spectroscopy analyses indicated that the pyrite was relatively stable in GDP system and there was no obvious polymeric compound formed on the catalyst surface. Overall, this GDP process offers high removal efficiency, simple technology, considerable energy efficiency and the applicability to salt-containing phthalate wastewater.
Collapse
Affiliation(s)
- Chensi Shen
- College of Environmental Science and Engineering, Donghua University, Shanghai 201620, China
| | - Shaoshuai Wu
- Sanqishi Township Government of Yuyao City, Yuyao 315412, China
| | - Hui Chen
- Zhejiang Avocado Biotechnologies Co., Ltd, Yuyao 315412, China
| | - Sadia Rashid
- College of Environmental Science and Engineering, Donghua University, Shanghai 201620, China
| | - Yuezhong Wen
- Institute of Environmental Science, Zhejiang University, Hangzhou 310029, China E-mail:
| |
Collapse
|
41
|
Kuo KK, Lee KT, Chen KK, Yang YH, Lin YC, Tsai MH, Wuputra K, Lee YL, Ku CC, Miyoshi H, Nakamura Y, Saito S, Wu CC, Chai CY, Eckner R, Steve Lin CL, Wang SSW, Wu DC, Lin CS, Yokoyama KK. Positive Feedback Loop of OCT4 and c-JUN Expedites Cancer Stemness in Liver Cancer. Stem Cells 2016; 34:2613-2624. [PMID: 27341307 DOI: 10.1002/stem.2447] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 05/09/2016] [Accepted: 06/06/2016] [Indexed: 12/12/2022]
Abstract
The network of stemness genes and oncogenes in human patient-specific reprogrammed cancer stem cells (CSCs) remains elusive, especially in liver cancer. HepG2-derived induced pluripotent stem cell-like cells (HepG2-iPS-like cells) were generated by introducing Yamanaka factors and the knockdown vector shTP53. They exhibited features of stemness and a higher tumorigenesis after xenograft transplantation compared with HepG2 cells. The cancerous mass of severe combined immunodeficiency (SCID) mice derived from one colony was dissected and cultured to establish reprogrammed HepG2-derived CSC-like cells (designated rG2-DC-1C). A single colony exhibited 42% occurrence of tumors with higher proliferation capacities. rG2-DC-1C showed continuous expression of the OCT4 stemness gene and of representative tumor markers, potentiated chemoresistance characteristics, and invasion activities. The sphere-colony formation ability and the invasion activity of rG2-DC-1C were also higher than those of HepG2 cells. Moreover, the expression of the OCT4 gene and the c-JUN oncogene, but not of c-MYC, was significantly elevated in rG2-DC-1C, whereas no c-JUN expression was observed in HepG2 cells. The positive-feedback regulation via OCT4-mediated transactivation of the c-JUN promoter and the c-JUN-mediated transactivation of the OCT4 promoter were crucial for promoting cancer development and maintaining cancer stemness in rG2-DC-1C. Increased expression of OCT4 and c-JUN was detected in the early stage of human liver cancer. Therefore, the positive feedback regulation of OCT4 and c-JUN, resulting in the continuous expression of oncogenes such as c-JUN, seems to play a critical role in the determination of the cell fate decision from iPS cells to CSCs in liver cancer. Stem Cells 2016;34:2613-2624.
Collapse
Affiliation(s)
- Kung-Kai Kuo
- Department of Surgery.,Center of Stem Cell Research
| | | | | | - Ya-Han Yang
- Department of Surgery.,Graduate Institute of Medicine.,Center of Stem Cell Research
| | | | | | | | | | | | - Hiroyuki Miyoshi
- Department of Physiology, Keio University School of Medicine, Shinanomachi, Tokyo, 160-8582, Japan
| | | | - Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita, Tochigi, 329-1571, Japan
| | | | | | - Richard Eckner
- Department of Biochemistry and Molecular Biology, Rutgers New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, New Jersey, USA
| | | | - Sophie S-W Wang
- Department of Gastroenterology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.,Center of Stem Cell Research
| | - Deng-Chyang Wu
- Department of Gastroenterology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.,Center of Stem Cell Research.,Center of Infectious Diseases and Cancer Research.,Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, 807, Taiwan
| | - Chang-Shen Lin
- Graduate Institute of Medicine.,Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
| | - Kazunari K Yokoyama
- Graduate Institute of Medicine.,Center of Stem Cell Research.,Center of Infectious Diseases and Cancer Research.,Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.,Faculty of Science and Engineering, Tokushima Bunri University, Sanuki, 763-2193, Japan.,Department of Molecular Preventive Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
42
|
Cravero D, Martignani E, Miretti S, Accornero P, Pauciullo A, Sharma R, Donadeu FX, Baratta M. Generation of Induced Pluripotent Stem Cells from Bovine Epithelial Cells and Partial Redirection Toward a Mammary Phenotype In Vitro. Cell Reprogram 2016; 17:211-20. [PMID: 26053520 DOI: 10.1089/cell.2014.0087] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In contrast to adult stem cells, induced pluripotent stem cells (iPSCs) can be grown robustly in vitro and differentiated into virtually any tissue, thus providing an attractive alternative for biomedical applications. Although iPSC technology is already being used in human biomedicine, its potential in animal production has not been investigated. Herein, we investigated the potential application of iPSCs in dairy production by generating bovine iPSCs and establishing their ability to generate mammary epithelial tissue. iPSCs were derived by retrovirus-mediated expression of murine Oct4, Sox2, Klf4, and c-Myc in mammary epithelium and dermal fibroblasts. The resulting reprogrammed cells stained positive for alkaline phosphatase and showed renewed expression of pluripotency genes, including Lin28, Rex1, Oct4, Sox2, and Nanog. In addition, injection of epithelial- or fibroblast-derived reprogrammed cells into nonobese diabetic (NOD/NOD) mice resulted in the formation of teratomas containing differentiated derivatives of the three germ layers, including cartilage, membranous ossification, stratified squamous epithelial tissue, hair follicles, neural pinwheels, and different types of glandular tissue. Finally, mammary epithelium-derived iPSCs could be induced to differentiate back to a mammary phenotype characterized by epithelial cells expressing cytokeratin 14 (CK14), CK18, and smooth muscle actin (SMA) as a result of treatment with 10 nM progesterone. This study reports for the first time the generation of iPSCs from bovine epithelial cells and demonstrates the potential of using iPSCs technology for generating bovine mammary tissue in vitro.
Collapse
Affiliation(s)
- Diego Cravero
- 1 Department of Veterinary Science, University of Torino , 10095 Grugliasco (TO), Italy
| | - Eugenio Martignani
- 1 Department of Veterinary Science, University of Torino , 10095 Grugliasco (TO), Italy
| | - Silvia Miretti
- 1 Department of Veterinary Science, University of Torino , 10095 Grugliasco (TO), Italy
| | - Paulo Accornero
- 1 Department of Veterinary Science, University of Torino , 10095 Grugliasco (TO), Italy
| | - Alfredo Pauciullo
- 2 Department Agricultural, Forest and Food Sciences, University of Torino , Grugliasco (TO), Italy
| | - Ruchi Sharma
- 3 The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh , Midlothian, United Kingdom
| | - Francesco Xavier Donadeu
- 3 The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh , Midlothian, United Kingdom
| | - Mario Baratta
- 1 Department of Veterinary Science, University of Torino , 10095 Grugliasco (TO), Italy
| |
Collapse
|
43
|
Abstract
This review deals with the latest advances in the study of embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) from domesticated species, with a focus on pigs, cattle, sheep, goats, horses, cats, and dogs. Whereas the derivation of fully pluripotent ESC from these species has proved slow, reprogramming of somatic cells to iPSC has been more straightforward. However, most of these iPSC depend on the continued expression of the introduced transgenes, a major drawback to their utility. The persistent failure in generating ESC and the dependency of iPSC on ectopic genes probably stem from an inability to maintain the stability of the endogenous gene networks necessary to maintain pluripotency. Based on work in humans and rodents, achievement of full pluripotency will likely require fine adjustments in the growth factors and signaling inhibitors provided to the cells. Finally, we discuss the future utility of these cells for biomedical and agricultural purposes.
Collapse
Affiliation(s)
- Toshihiko Ezashi
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211; , ,
| | - Ye Yuan
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211; , ,
| | - R Michael Roberts
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211; , ,
| |
Collapse
|
44
|
Saito S, Lin YC, Murayama Y, Nakamura Y, Eckner R, Niemann H, Yokoyama KK. Retracted article: In vitro derivation of mammalian germ cells from stem cells and their potential therapeutic application. Cell Mol Life Sci 2015; 72:4545-60. [PMID: 26439925 PMCID: PMC4628088 DOI: 10.1007/s00018-015-2020-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 07/27/2015] [Accepted: 08/07/2015] [Indexed: 01/12/2023]
Abstract
Pluripotent stem cells (PSCs) are a unique type of cells because they
exhibit the characteristics of self-renewal and pluripotency. PSCs may be induced to
differentiate into any cell type, even male and female germ cells, suggesting their
potential as novel cell-based therapeutic treatment for infertility problems.
Spermatogenesis is an intricate biological process that starts from self-renewal of
spermatogonial stem cells (SSCs) and leads to differentiated haploid spermatozoa.
Errors at any stage in spermatogenesis may result in male infertility. During the
past decade, much progress has been made in the derivation of male germ cells from
various types of progenitor stem cells. Currently, there are two main approaches for
the derivation of functional germ cells from PSCs, either the induction of in vitro
differentiation to produce haploid cell products, or combination of in vitro
differentiation and in vivo transplantation. The production of mature and fertile
spermatozoa from stem cells might provide an unlimited source of autologous gametes
for treatment of male infertility. Here, we discuss the current state of the art
regarding the differentiation potential of SSCs, embryonic stem cells, and induced
pluripotent stem cells to produce functional male germ cells. We also discuss the
possible use of livestock-derived PSCs as a novel option for animal reproduction and
infertility treatment.
Collapse
Affiliation(s)
- Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita, Tochigi, 329-1571, Japan. .,SPK Co., Ltd., Aizuwakamatsu, Fukushima, 965-0025, Japan. .,College of Engineering, Nihon University, Koriyama, Fukushima, 963-8642, Japan.
| | - Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaoshiung Medical University, 100 Shin-Chuan 1st Road, Kaohsiung, 807, Taiwan
| | - Yoshinobu Murayama
- College of Engineering, Nihon University, Koriyama, Fukushima, 963-8642, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, 3050074, Japan
| | - Richard Eckner
- Department of Biochemistry and Molecular Biology, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07101, USA
| | - Heiner Niemann
- Institute of Farm Animal Genetics, Friedrich-Löffler-Institut, Mariensee, 31535, Neustadt, Germany.
| | - Kazunari K Yokoyama
- Graduate Institute of Medicine, Center of Stem Cell Research, Center of Environmental Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd, San Ming District, Kaohsiung, 807, Taiwan. .,Faculty of Science and Engineering, Tokushima Bunri University, Sanuki, 763-2193, Japan. .,Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
45
|
Thompson PA, Khatami M, Baglole CJ, Sun J, Harris SA, Moon EY, Al-Mulla F, Al-Temaimi R, Brown DG, Colacci A, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Amedei A, Hamid RA, Lowe L, Guarnieri T, Bisson WH. Environmental immune disruptors, inflammation and cancer risk. Carcinogenesis 2015; 36 Suppl 1:S232-53. [PMID: 26106141 DOI: 10.1093/carcin/bgv038] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
An emerging area in environmental toxicology is the role that chemicals and chemical mixtures have on the cells of the human immune system. This is an important area of research that has been most widely pursued in relation to autoimmune diseases and allergy/asthma as opposed to cancer causation. This is despite the well-recognized role that innate and adaptive immunity play as essential factors in tumorigenesis. Here, we review the role that the innate immune cells of inflammatory responses play in tumorigenesis. Focus is placed on the molecules and pathways that have been mechanistically linked with tumor-associated inflammation. Within the context of chemically induced disturbances in immune function as co-factors in carcinogenesis, the evidence linking environmental toxicant exposures with perturbation in the balance between pro- and anti-inflammatory responses is reviewed. Reported effects of bisphenol A, atrazine, phthalates and other common toxicants on molecular and cellular targets involved in tumor-associated inflammation (e.g. cyclooxygenase/prostaglandin E2, nuclear factor kappa B, nitric oxide synthesis, cytokines and chemokines) are presented as example chemically mediated target molecule perturbations relevant to cancer. Commentary on areas of additional research including the need for innovation and integration of systems biology approaches to the study of environmental exposures and cancer causation are presented.
Collapse
Affiliation(s)
- Patricia A Thompson
- Department of Pathology, Stony Brook Medical School, Stony Brook, NY 11794, USA, Inflammation and Cancer Research, National Cancer Institute (NCI) (Retired), NIH, Bethesda, MD 20817, USA, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada, Department of Biochemistry, Rush University, Chicago, IL 60612, USA, Prevention and Cancer Control, Cancer Care Ontario, 620 University Avenue, Toronto, Ontario M5G 2L3, Canada, Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of South Korea, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy, The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA, Advanced Molecular Science Research Centre, King George's Medical University, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy, Faculty of Medicine and Health Sciences, Universiti Putra, Malaysia, Serdang, Selangor 43400, Malaysia, Getting to Know Cancer, Room 229A, 36 Arthur St, Truro, Nova Scotia B2N 1X5, Canada Department of Biology, Geology and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi, 3, 40126 Bologna, Italy Center for Appl
| | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (NCI) (Retired), NIH, Bethesda, MD 20817, USA
| | - Carolyn J Baglole
- Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, IL 60612, USA
| | - Shelley A Harris
- Prevention and Cancer Control, Cancer Care Ontario, 620 University Avenue, Toronto, Ontario M5G 2L3, Canada
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of South Korea
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Chiara Mondello
- The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - A Ivana Scovassi
- The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Hosni K Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra, Malaysia, Serdang, Selangor 43400, Malaysia
| | - Leroy Lowe
- Getting to Know Cancer, Room 229A, 36 Arthur St, Truro, Nova Scotia B2N 1X5, Canada
| | - Tiziana Guarnieri
- Department of Biology, Geology and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi, 3, 40126 Bologna, Italy Center for Applied Biomedical Research, S. Orsola-Malpighi University Hospital, Via Massarenti, 9, 40126 Bologna, Italy, National Institute of Biostructures and Biosystems, Viale Medaglie d' Oro, 305, 00136 Roma, Italy and
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97331, USA
| |
Collapse
|
46
|
Talluri TR, Kumar D, Glage S, Garrels W, Ivics Z, Debowski K, Behr R, Niemann H, Kues WA. Derivation and characterization of bovine induced pluripotent stem cells by transposon-mediated reprogramming. Cell Reprogram 2015; 17:131-40. [PMID: 25826726 DOI: 10.1089/cell.2014.0080] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are a seminal breakthrough in stem cell research and are promising tools for advanced regenerative therapies in humans and reproductive biotechnology in farm animals. iPSCs are particularly valuable in species in which authentic embryonic stem cell (ESC) lines are yet not available. Here, we describe a nonviral method for the derivation of bovine iPSCs employing Sleeping Beauty (SB) and piggyBac (PB) transposon systems encoding different combinations of reprogramming factors, each separated by self-cleaving peptide sequences and driven by the chimeric CAGGS promoter. One bovine iPSC line (biPS-1) generated by a PB vector containing six reprogramming genes was analyzed in detail, including morphology, alkaline phosphatase expression, and typical hallmarks of pluripotency, such as expression of pluripotency markers and formation of mature teratomas in immunodeficient mice. Moreover, the biPS-1 line allowed a second round of SB transposon-mediated gene transfer. These results are promising for derivation of germ line-competent bovine iPSCs and will facilitate genetic modification of the bovine genome.
Collapse
Affiliation(s)
- Thirumala R Talluri
- 1 Institut für Nutztiergenetik, Friedrich-Loeffler-Institut , Mariensee, 31535 Neustadt, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Bovine induced pluripotent stem cells are more resistant to apoptosis than testicular cells in response to mono-(2-ethylhexyl) phthalate. Int J Mol Sci 2014; 15:5011-31. [PMID: 24658443 PMCID: PMC3975437 DOI: 10.3390/ijms15035011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 03/04/2014] [Accepted: 03/06/2014] [Indexed: 12/27/2022] Open
Abstract
Although the androgen receptor (AR) has been implicated in the promotion of apoptosis in testicular cells (TSCs), the molecular pathway underlying AR-mediated apoptosis and its sensitivity to environmental hormones in TSCs and induced pluripotent stem cells (iPSCs) remain unclear. We generated the iPSCs from bovine TSCs via the electroporation of OCT4. The established iPSCs were supplemented with leukemia inhibitory factor and bone morphogenetic protein 4 to maintain and stabilize the expression of stemness genes and their pluripotency. Apoptosis signaling was assessed after exposure to mono-(2-ethylhexyl) phthalate (MEHP), the active metabolite of di-(2-ethylhexyl) phthalate. Here, we report that iPSCs were more resistant to MEHP-induced apoptosis than were original TSCs. MEHP also repressed the expression of AR and inactivated WNT signaling, and then led to the commitment of cells to apoptosis via the cyclin dependent kinase inhibitor p21CIP1. The loss of the frizzed receptor 7 and the gain of p21CIP were responsible for the stimulatory effect of MEHP on AR-mediated apoptosis. Our results suggest that testicular iPSCs can be used to study the signaling pathways involved in the response to environmental disruptors, and to assess the toxicity of environmental endocrine disruptors in terms of the maintenance of stemness and pluripotency.
Collapse
|