1
|
Mattei DN, Harman RM, Van de Walle GR, Smith R, Grivel JC, Abdelalim EM, Vinardell T. Effect of pregnancy on isolation efficiency and in vitro proliferation of equine peripheral-blood derived mesenchymal stromal cells. Theriogenology 2024; 224:107-118. [PMID: 38761667 DOI: 10.1016/j.theriogenology.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Mesenchymal stromal cells (MSCs) have regenerative and immunomodulatory potential and may be used to treat injured tissues. Pregnancy has been associated with increased MSCs in the peripheral circulation in multiple species, but to date, there are no reports on this matter in horses. This study aimed to evaluate the effect of pregnancy on isolation efficiency and proliferation capacity of equine MSCs derived from the peripheral blood (PB) of mares. Venous blood samples were collected at the 11th month of gestation and 1 month after delivery from clinically healthy Arabian mares that presented normal pregnancies. Blood samples were processed for in vitro cellular culture and hormonal and metabolic profiles. MSCs were isolated and characterized by trilineage differentiation potential, immunophenotyping, analyzed by gene sequencing and proliferation assays. The isolation of peripheral blood mononuclear cells (PBMCs) of pregnant mares were associated with higher isolation efficiency and proliferative capacity of MSCs derived from peripheral blood (PB-MSCs) recovered pre-partum than those isolated post-partum. Although fetal gender, parity, 5α-reduced pregnanes, insulin, and cortisol were shown to affect cellular proliferation, individual factors and the small population studied must be considered. This study suggests that PB-MSCs from pregnant mares could be a valuable alternative source of MSCs for therapeutic purposes.
Collapse
Affiliation(s)
- Debora N Mattei
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Equine Veterinary Medical Center, Member of Qatar Foundation, P.O. Box 5825, Doha, Qatar
| | - Rebecca M Harman
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Rd, Ithaca, NY 14850, USA
| | - Gerlinde R Van de Walle
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Rd, Ithaca, NY 14850, USA
| | - Roger Smith
- Department of Clinical Science and Services, The Royal Veterinary College, Hawkshead Lane, Hatfield, Hertfordshire, AL9 7TA, United Kingdom
| | - Jean Charles Grivel
- Deep Phenotyping Core, Sidra Medicine, PO Box 26999, Al Garrafa St, Ar-Rayyan, Doha, Qatar
| | - Essam M Abdelalim
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Division, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar; Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar
| | - Tatiana Vinardell
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Member of Qatar Foundation, PO Box 34110, Education City, Doha, Qatar; Equine Veterinary Medical Center, Member of Qatar Foundation, P.O. Box 5825, Doha, Qatar.
| |
Collapse
|
2
|
Gorsek Sparovec T, Markert UR, Reif P, Schoell W, Moser G, Feichtinger J, Mihalic ZN, Kargl J, Gargett CE, Gold D. The fate of human SUSD2+ endometrial mesenchymal stem cells during decidualization. Stem Cell Res 2022; 60:102671. [PMID: 35093718 DOI: 10.1016/j.scr.2022.102671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 12/30/2021] [Accepted: 01/12/2022] [Indexed: 12/26/2022] Open
Abstract
Regeneration of the endometrial stromal compartment in premenopausal women is likely maintained by the perivascular endometrial mesenchymal stem/stromal cells (eMSC) expressing sushi domain containing 2 (SUSD2). The fate of SUSD2+ eMSC during pregnancy and their role in decidualization is not fully known. The aim of our study was to determine the effect of progesterone on the stemness of the SUSD2+ eMSC isolated from non-pregnant uterine samples. Secondary objectives were to characterize the functional capacity including differentiation and clonogenicity assays of SUSD2+ eMSC isolated from decidua at full term and compare it to the capacity of those isolated from non-pregnant uterine samples. Progesterone treatment induced changes in the decidual gene expression profile in non-pregnant SUSD2+ eMSC. Data analysis of a publicly available single cell RNA-seq data set revealed differential expression of several mesenchymal and epithelial signature genes between the SUSD2+ eMSC and the decidual stromal cells, suggesting mesenchymal-to-epithelial transition occurs during decidualization. Histological analysis revealed a significantly lower abundance of SUSD2+ eMSC in 1st trimester and full term samples compared to non-pregnant samples, p = 0.0296 and 0.005, respectively. The differentiation and the colony forming capacity did not differ significantly between the cells isolated from non-pregnant and pregnant uterine samples. Our results suggest that SUSD2+ eMSC undergo decidualization in vitro, while maintaining MSC plasma membrane phenotype. Human eMSC seem to play an important role in the course of endometrial decidualization and embryo implantation. Pregnancy reduced the abundance of SUSD2+ eMSC, however eMSC function remains intact.
Collapse
Affiliation(s)
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany.
| | - Philipp Reif
- Department of Obstetrics and Gynaecology, Medical University of Graz, Austria.
| | - Wolfgang Schoell
- Department of Obstetrics and Gynaecology, Medical University of Graz, Austria.
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria.
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria.
| | - Zala Nikita Mihalic
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria.
| | - Julia Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria.
| | - Caroline E Gargett
- Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Australia.
| | - Daniela Gold
- Department of Obstetrics and Gynaecology, Medical University of Graz, Austria.
| |
Collapse
|
3
|
Tan L, Liu X, Dou H, Hou Y. Characteristics and regulation of mesenchymal stem cell plasticity by the microenvironment — specific factors involved in the regulation of MSC plasticity. Genes Dis 2022; 9:296-309. [PMID: 35224147 PMCID: PMC8843883 DOI: 10.1016/j.gendis.2020.10.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/05/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs), multipotent stromal cells, have attracted extensive attention in the field of regenerative medicine and cell therapy due to the capacity of self-renewal, multilineage differentiation, and immune regulation. MSCs have different cellular effects in different diseases, and even have markedly different curative effects with different tissue sources, indicating the plasticity of MSCs. The phenotypes, secreted factors, and proliferative, migratory, differentiating, and immunomodulatory effects of MSCs depend on certain mediators present in their microenvironment. Understanding microenvironmental factors and their internal mechanisms in MSC responses may help in subsequent prediction and improvement of clinical benefits. This review highlighted the recent advances in MSC plasticity in the physiological and pathological microenvironment and multiple microenvironmental factors regulating MSC plasticity. It also highlighted some progress in the underlying molecular mechanisms of MSC remodeling in the microenvironment. It might provide references for the improvement in vitro culture of MSCs, clinical application, and in vivo induction.
Collapse
|
4
|
Sanie-Jahromi F, NejatyJahromy Y, Jahromi RR. A Review on the Role of Stem Cells against SARS-CoV-2 in Children and Pregnant Women. Int J Mol Sci 2021; 22:11787. [PMID: 34769218 PMCID: PMC8584228 DOI: 10.3390/ijms222111787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/10/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022] Open
Abstract
Since the COVID-19 outbreak was acknowledged by the WHO on 30 January 2020, much research has been conducted to unveil various features of the responsible SARS-CoV-2 virus. Different rates of contagion in adults, children, and pregnant women may guide us to understand the underlying infection conditions of COVID-19. In this study, we first provide a review of recent reports of COVID-19 clinical outcomes in children and pregnant women. We then suggest a mechanism that explains the curious case of COVID-19 in children/pregnant women. The unique stem cell molecular signature, as well as the very low expression of angiotensin-converting enzyme 2 and the lower ACE/ACE2 ratio in stem cells of children/pregnant women compared to adults might be the cause of milder symptoms of COVID-19 in them. This study provides the main molecular keys on how stem cells can function properly and exert their immunomodulatory and regenerative effects in COVID-19-infected children/pregnant women, while failing to replicate their role in adults. This can lay the groundwork for both predicting the pattern of spread and severity of the symptoms in a population and designing novel stem cell-based treatment and prevention strategies for COVID-19.
Collapse
Affiliation(s)
- Fatemeh Sanie-Jahromi
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz 7134997446, Iran;
| | - Yaser NejatyJahromy
- Institut für Physikalische und Theoretische Chemie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53012 Bonn, Germany
| | - Rahim Raoofi Jahromi
- Department of Infectious Disease, Peymanieh Hospital, Jahrom University of Medical Science, Jahrom 7414846199, Iran
| |
Collapse
|
5
|
Towards Physiologic Culture Approaches to Improve Standard Cultivation of Mesenchymal Stem Cells. Cells 2021; 10:cells10040886. [PMID: 33924517 PMCID: PMC8069108 DOI: 10.3390/cells10040886] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest for their use in cell-based therapies due to their multipotent differentiation and immunomodulatory capacities. In consequence of limited numbers following their isolation from the donor tissue, MSCs require extensive expansion performed in traditional 2D cell culture setups to reach adequate amounts for therapeutic use. However, prolonged culture of MSCs in vitro has been shown to decrease their differentiation potential and alter their immunomodulatory properties. For that reason, preservation of these physiological characteristics of MSCs throughout their in vitro culture is essential for improving the efficiency of therapeutic and in vitro modeling applications. With this objective in mind, many studies already investigated certain parameters for enhancing current standard MSC culture protocols with regard to the effects of specific culture media components or culture conditions. Although there is a lot of diversity in the final therapeutic uses of the cells, the primary stage of standard isolation and expansion is imperative. Therefore, we want to review on approaches for optimizing standard MSC culture protocols during this essential primary step of in vitro expansion. The reviewed studies investigate and suggest improvements focused on culture media components (amino acids, ascorbic acid, glucose level, growth factors, lipids, platelet lysate, trace elements, serum, and xenogeneic components) as well as culture conditions and processes (hypoxia, cell seeding, and dissociation during passaging), in order to preserve the MSC phenotype and functionality during the primary phase of in vitro culture.
Collapse
|
6
|
Martínez-Razo LD, Martínez-Ibarra A, Vázquez-Martínez ER, Cerbón M. The impact of Di-(2-ethylhexyl) Phthalate and Mono(2-ethylhexyl) Phthalate in placental development, function, and pathophysiology. ENVIRONMENT INTERNATIONAL 2021; 146:106228. [PMID: 33157377 DOI: 10.1016/j.envint.2020.106228] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/11/2020] [Accepted: 10/19/2020] [Indexed: 05/21/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a chemical widely distributed in the environment as is extensively used in the plastic industry. DEHP is considered an endocrine disruptor chemical (EDC) and humans are inevitably and unintentionally exposed to this EDC through several sources including food, beverages, cosmetics, medical devices, among others. DEHP exposure has been associated and may be involved in the development of various pathologies; importantly, pregnant women are a particular risk group considering that endocrine alterations during gestation may impact fetal programming leading to the development of several chronic diseases in adulthood. Recent studies have indicated that exposure to DEHP and its metabolite Mono(2-ethylhexyl) phthalate (MEHP) may impair placental development and function, which in turn would have a negative impact on fetal growth. Studies performed in several trophoblastic and placental models have shown the negative impact of DEHP and MEHP in key processes related to placental development such as implantation, differentiation, invasion and angiogenesis. In addition, many alterations in placental functions like hormone signaling, metabolism, transfer of nutrients, immunomodulation and oxidative stress response have been reported. Moreover, clinical-epidemiological evidence supports the association between DEHP exposure and adverse pregnancy outcomes and pathologies. In this review, we aim to summarize for the first time current knowledge about the impact of DEHP and MEHP exposure on placental development and pathophysiology, as well as the mechanisms involved. We also remark the importance of exploring DEHP and MEHP effects in different trophoblast cell populations and discuss new perspectives regarding this topic.
Collapse
Affiliation(s)
- Luis Daniel Martínez-Razo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, Mexico
| | - Alejandra Martínez-Ibarra
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, Mexico; Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico
| | - Edgar Ricardo Vázquez-Martínez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, Mexico
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología "Isidro Espinosa de los Reyes" - Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 11000, Mexico.
| |
Collapse
|
7
|
Luo T, Liu Q, Tan A, Duan L, Jia Y, Nong L, Tang J, Zhou W, Xie W, Lu Y, Yu Q, Liu Y. Mesenchymal Stem Cell-Secreted Exosome Promotes Chemoresistance in Breast Cancer via Enhancing miR-21-5p-Mediated S100A6 Expression. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:283-293. [PMID: 33294586 PMCID: PMC7689030 DOI: 10.1016/j.omto.2020.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022]
Abstract
Emerging evidence has shown the role of mesenchymal stem cell-derived exosome (MSC-exo) in inducing resistance of cancer cells to chemotherapy. However, it remains unclear whether the change of MSC-exo in response to chemotherapy also contributes to chemoresistance. In this study, we investigated the effect of a standard-of-care chemotherapeutic agent, doxorubicin (Dox), on MSC-exo and its contribution to the development of Dox resistance in breast cancer cells (BCs). We found that the exosome secreted by Dox-treated MSCs (Dt-MSC-exo) induced a higher degree of Dox resistance in BCs when compared with non-treated MSC-exo. By analysis of the MSC-exo-induced transcriptome change in BCs, we identified S100A6, a chemoresistant gene, as a top-ranked gene induced by MSC-exo in BCs, which was further enhanced by Dt-MSC-exo. Furthermore, we found that Dox induced the expression of miR-21-5p in MSCs and MSC-exo, which was required for the expression of S100A6 in BCs. Importantly, silencing of miR-21-5p expression in MSCs and MSC-exo abolished the resistance of BCs to Dox, indicating an exosomal miR-21-5p-regulated S100A6 in chemoresistance. Our study thus uncovered a novel mechanistic insight into the role of MSC-secreted exosome in the development of chemoresistance in the tumor microenvironment.
Collapse
Affiliation(s)
- Tao Luo
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Qiaoyuan Liu
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Aihua Tan
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Lixia Duan
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Yuxian Jia
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Li Nong
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Jing Tang
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Wenxian Zhou
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Weimin Xie
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Yongkui Lu
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| | - Qiang Yu
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Yan Liu
- The Fifth Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, P.R. China
| |
Collapse
|
8
|
A systematic study on chitosan-liposome based systems for biomedical applications. Int J Biol Macromol 2020; 160:470-481. [DOI: 10.1016/j.ijbiomac.2020.05.192] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/14/2020] [Accepted: 05/22/2020] [Indexed: 12/24/2022]
|
9
|
Cruz T, López-Giraldo A, Noell G, Guirao A, Casas-Recasens S, Garcia T, Saco A, Sellares J, Agustí A, Faner R. Smoking Impairs the Immunomodulatory Capacity of Lung-Resident Mesenchymal Stem Cells in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2019; 61:575-583. [DOI: 10.1165/rcmb.2018-0351oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Tamara Cruz
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
| | - Alejandra López-Giraldo
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Guillaume Noell
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Angela Guirao
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | | | - Tamara Garcia
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
| | - Adela Saco
- Department of Pathology, Hospital Clinic, Barcelona, Spain
| | - Jacobo Sellares
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Alvar Agustí
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Rosa Faner
- Centro Investigación Biomédica en Red Enfermedades Respiratorias, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| |
Collapse
|
10
|
Shah NM, Lai PF, Imami N, Johnson MR. Progesterone-Related Immune Modulation of Pregnancy and Labor. Front Endocrinol (Lausanne) 2019; 10:198. [PMID: 30984115 PMCID: PMC6449726 DOI: 10.3389/fendo.2019.00198] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/11/2019] [Indexed: 12/17/2022] Open
Abstract
Pregnancy involves a complex interplay between maternal neuroendocrine and immunological systems in order to establish and sustain a growing fetus. It is thought that the uterus at pregnancy transitions from quiescent to laboring state in response to interactions between maternal and fetal systems at least partly via altered neuroendocrine signaling. Progesterone (P4) is a vital hormone in maternal reproductive tissues and immune cells during pregnancy. As such, P4 is widely used in clinical interventions to improve the chance of embryo implantation, as well as reduce the risk of miscarriage and premature labor. Here we review research to date that focus on the pathways through which P4 mediates its actions on both the maternal reproductive and immune system. We will dissect the role of P4 as a modulator of inflammation, both systemic and intrinsic to the uterus, during human pregnancy and labor.
Collapse
Affiliation(s)
- Nishel M. Shah
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Pei F. Lai
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Nesrina Imami
- Department of Medicine, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Mark R. Johnson
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Pulmonary group 2 innate lymphoid cells: surprises and challenges. Mucosal Immunol 2019; 12:299-311. [PMID: 30664706 PMCID: PMC6436699 DOI: 10.1038/s41385-018-0130-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 02/04/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) are a recently described subset of innate lymphocytes with important immune and homeostatic functions at multiple tissue sites, especially the lung. These cells expand locally after birth and during postnatal lung maturation and are present in the lung and other peripheral organs. They are modified by a variety of processes and mediate inflammatory responses to respiratory pathogens, inhaled allergens and noxious particles. Here, we review the emerging roles of ILC2s in pulmonary homeostasis and discuss recent and surprising advances in our understanding of how hormones, age, neurotransmitters, environmental challenges, and infection influence ILC2s. We also review how these responses may underpin the development, progression and severity of pulmonary inflammation and chronic lung diseases and highlight some of the remaining challenges for ILC2 biology.
Collapse
|
12
|
Sheller-Miller S, Richardson L, Martin L, Jin J, Menon R. Systematic review of p38 mitogen-activated kinase and its functional role in reproductive tissues. Am J Reprod Immunol 2018; 80:e13047. [PMID: 30178469 PMCID: PMC6261682 DOI: 10.1111/aji.13047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress (OS) plays a role in uterine tissue remodeling during pregnancy and parturition. While p38 MAPK is an OS-response kinase, a precise functional role is unknown. Therefore, we conducted a systematic review of literature on p38 MAPK expression, activation, and function in reproductive tissues throughout pregnancy and parturition, published between January 1980 and August 2017, using four electronic databases (Web of Science, PubMed, Medline, and CoCHRANE). We identified 418 reports; 108 were selected for full-text evaluation and 74 were included in final review. p38 MAPK was investigated using feto-maternal primary or immortalized cells, tissue explants, and animal models. Western blot was most commonly used to report phosphorylated (active) p38 MAPK. Human placenta (27), chorioamniotic membranes (14), myometrium (13), decidua (8), and cervix (1) were the studied tissues. p38 MAPK's functions were tissue and gestational age dependent. Isoform specificity was hardly reported. p38 MAPK activity was induced by ROS or proinflammatory cytokines to promote cell signaling linked to cell fate, primed uterus, ripened cervix, and proinflammatory cytokine/chemokine production. In 35 years, reports on p38 MAPK's role during pregnancy and parturition are scarce and current literature is insufficient to provide a comprehensive description of p38 MAPK's mechanistic role during pregnancy and parturition.
Collapse
Affiliation(s)
- Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Lauren Richardson
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
- Department of Neuroscience & Cell Biology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Laura Martin
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | - Jin Jin
- Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| |
Collapse
|
13
|
Sexual dimorphism in hepatitis B and C and hepatocellular carcinoma. Semin Immunopathol 2018; 41:203-211. [PMID: 30498927 DOI: 10.1007/s00281-018-0727-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/04/2018] [Indexed: 12/14/2022]
Abstract
The incidence of viral hepatitis B or C (HBV/HCV) infection and hepatocellular carcinoma is higher in male compared to female populations, showing a faster disease progression and results in a worse overall survival. Indeed, women are in general better protected from viral infections and show a lower risk of death from malignant cancer in comparison to men. Females mount stronger innate and adaptive immune responses than males, and therefore, most of the autoimmune diseases occur predominantly in females. Next to occupational and/or behavioral factors, cellular and molecular differences between the two sexes contribute to this observation. In this review, we will discuss underlying mechanisms that are important for the observed sex-related differences in liver diseases. A better appreciation of these differences between the two sexes might be of value for better and gender-specific treatment options.
Collapse
|
14
|
Knockdown of TGF-β1 expression in human umbilical cord mesenchymal stem cells reverts their exosome-mediated EMT promoting effect on lung cancer cells. Cancer Lett 2018; 428:34-44. [DOI: 10.1016/j.canlet.2018.04.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/13/2018] [Accepted: 04/18/2018] [Indexed: 02/08/2023]
|
15
|
Abboud R, Akil M, Charcosset C, Greige-Gerges H. Interaction of glucocorticoids and progesterone derivatives with human serum albumin. Chem Phys Lipids 2017; 207:271-278. [PMID: 28435101 DOI: 10.1016/j.chemphyslip.2017.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 04/11/2017] [Accepted: 04/17/2017] [Indexed: 01/09/2023]
Affiliation(s)
- Rola Abboud
- Bioactive Molecules Research Group, PRASE, Doctoral School of Sciences and Technologies, Faculty of Sciences, Lebanese University, Lebanon; Laboratoire d'Automatique et de Génie des Procédés (LAGEP), UMR-CNRS 5007, Université Claude Bernard Lyon 1, CPE Lyon, Bat 308G, 43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne Cedex, France
| | - Mohammad Akil
- Kalma laboratory, Faculty of Sciences, Lebanese University, Lebanon
| | - Catherine Charcosset
- Laboratoire d'Automatique et de Génie des Procédés (LAGEP), UMR-CNRS 5007, Université Claude Bernard Lyon 1, CPE Lyon, Bat 308G, 43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne Cedex, France
| | - Hélène Greige-Gerges
- Bioactive Molecules Research Group, PRASE, Doctoral School of Sciences and Technologies, Faculty of Sciences, Lebanese University, Lebanon.
| |
Collapse
|
16
|
Ghosh S, Klein RS. Sex Drives Dimorphic Immune Responses to Viral Infections. THE JOURNAL OF IMMUNOLOGY 2017; 198:1782-1790. [PMID: 28223406 DOI: 10.4049/jimmunol.1601166] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/24/2016] [Indexed: 02/07/2023]
Abstract
New attention to sexual dimorphism in normal mammalian physiology and disease has uncovered a previously unappreciated breadth of mechanisms by which females and males differentially exhibit quantitative phenotypes. Thus, in addition to the established modifying effects of hormones, which prenatally and postpubertally pattern cells and tissues in a sexually dimorphic fashion, sex differences are caused by extragonadal and dosage effects of genes encoded on sex chromosomes. Sex differences in immune responses, especially during autoimmunity, have been studied predominantly within the context of sex hormone effects. More recently, immune response genes have been localized to sex chromosomes themselves or found to be regulated by sex chromosome genes. Thus, understanding how sex impacts immunity requires the elucidation of complex interactions among sex hormones, sex chromosomes, and immune response genes. In this Brief Review, we discuss current knowledge and new insights into these intricate relationships in the context of viral infections.
Collapse
Affiliation(s)
- Soumitra Ghosh
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Robyn S Klein
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110; .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and.,Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
17
|
The novel α-glucan YCP improves the survival rates and symptoms in septic mice by regulating myeloid-derived suppressor cells. Acta Pharmacol Sin 2017. [PMID: 28649127 DOI: 10.1038/aps.2017.27] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a life-threatening health condition that is initially characterized by uncontrolled inflammation, followed by the development of persistent immunosuppression. YCP is a novel α-glucan purified from the mycelium of the marine fungus Phoma herbarum YS4108, which has displayed strong antitumor activity via enhancing host immune responses. In this study, we investigated whether YCP could influence the development of sepsis in a mouse model. Caecal ligation and puncture (CLP)-induced sepsis was established in mice that were treated with YCP (20 mg/kg, ip or iv) 2 h before, 4 and 24 h after the CLP procedure, and then every other day. YCP administration greatly improved the survival rate (from 39% to 72% on d 10 post-CLP) and ameliorated disease symptoms in the septic mice. Furthermore, YCP administration significantly decreased the percentage of myeloid-derived suppressor cells (MDSCs) in the lungs and livers, which were dramatically elevated during sepsis. In cultured BM-derived cells, addition of YCP (30, 100 μg/mL) significantly decreased the expansion of MDSCs; YCP dose-dependently decreased the phosphorylation of STAT3 and increased the expression of interferon regulatory factor-8 (IRF-8). When BM-derived MDSCs were co-cultured with T cells, YCP dose-dependently increased the production of arginase-1 (Arg-1) and inducible nitric oxide synthase (iNOS), and activated the NF-κB pathway. In addition, the effects of YCP on MDSCs appeared to be dependent on toll-like receptor (TLR) 4. These results reveal that YCP inhibits the expansion of MDSCs via STAT3 while enhancing their immunosuppressive function, partially through NF-κB. Our findings suggest that YCP protects mice against sepsis by regulating MDSCs. Thus, YCP may be a potential therapeutic agent for sepsis.
Collapse
|
18
|
Wang M, Liu M, Xie T, Zhang BF, Gao XL. Chitosan-modified cholesterol-free liposomes for improving the oral bioavailability of progesterone. Colloids Surf B Biointerfaces 2017; 159:580-585. [PMID: 28854414 DOI: 10.1016/j.colsurfb.2017.08.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 08/07/2017] [Accepted: 08/17/2017] [Indexed: 01/11/2023]
Abstract
Based on the structurally similar properties of progesterone and cholesterol, chitosan-coated cholesterol-free liposomes (CS-Lipo/Prog) were formulated. CS-Lipo/Prog are spherical and uniform in size (662.1±19.3nm) with positive potential (28.19±1.97mV). The average drug entrapment efficiency (EE) is approximately 80%. The in vitro release profile of CS-Lipo/Prog shows sustained release. The in vitro stability evaluation demonstrated that CS-Lipo/Prog can efficiently shield Prog from degradation in the gastrointestinal tract. CS-Lipo/Prog showed a longer MRT and higher AUC0-infinite after oral administration to mice than in the control group (progesterone-free). The relative bioavailability of CS-Lipo/Prog was higher than that of progesterone soft capsules (QINING®) and Lipo/Prog. Collectively, these findings suggest that cholesterol-free chitosan-coated liposomes are a promising alternative for improving the oral bioavailability of progesterone.
Collapse
Affiliation(s)
- Mei Wang
- Department of Pharmaceutics, College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Meng Liu
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Tingting Xie
- Department of Pharmaceutical Care, PLA General Hospital, Beijing, China
| | - Bing-Feng Zhang
- College of Chemistry and Bio-engineering, Yichun University, Yichun, China.
| | - Xiao-Li Gao
- Department of Pharmaceutics, College of Pharmacy, Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
19
|
Moslehi A, Hashemi-Beni B, Moslehi A, Akbari MA, Adib M. The effect of progesterone and 17-β estradiol on membrane-bound HLA-G in adipose derived stem cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:341-6. [PMID: 27382350 PMCID: PMC4930902 DOI: 10.4196/kjpp.2016.20.4.341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/22/2015] [Accepted: 03/17/2015] [Indexed: 11/18/2022]
Abstract
Membrane-bound HLA-G (mHLA-G) discovery on adipose derived stem cells (ADSCs) as a tolerogenic and immunosuppressive molecule was very important. Many documents have shown that HLA-G expression can be controlled via some hormones such as progesterone (P4) and estradiol (E2). Therefore, this study was designed to evaluate progesterone and estradiol effects on mHLA-G in ADSCs at restricted and combination concentrations. Three independent cell lines were cultured in complete free phenol red DMEM and subcultured to achieve suffi cient cells. These cells were treated with P4, E2 and P4 plus E2 at physiologic and pregnancy concentrations for 3 days in cell culture conditions. The HLA-G positive ADSCs was measured via monoclonal anti HLA-G-FITC/MEMG-09 by means of flow cytometry in nine groups. Data were analyzed by one way ANOVA and Tukey's post hoc tests. There were no signifi cant values of the mean percentage of HLA-G positive cells in E2-treated and the combination of P4 plus E2-treated ADSCs compared to control cells (p value>0.05) but P4 had a signifi cant increase on mHLA-G in ADSCs (p value<0.05). High P4 concentration increased mHLA-G but E2 and the combination of P4 plus E2 could not change mHLA-G on ADSCs.
Collapse
Affiliation(s)
- Akram Moslehi
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Batool Hashemi-Beni
- Department of Anatomical Science and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Azam Moslehi
- Department of Physiology & Pharmacology, Medical School, Qom University of Medical Sciences, Qom 3713649373, Iran
| | - Maryam Ali Akbari
- Department of Anatomical Science and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Minoo Adib
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| |
Collapse
|
20
|
Phungphong S, Kijtawornrat A, Wattanapermpool J, Bupha-Intr T. Regular exercise modulates cardiac mast cell activation in ovariectomized rats. J Physiol Sci 2016; 66:165-73. [PMID: 26467449 PMCID: PMC10717377 DOI: 10.1007/s12576-015-0409-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/25/2015] [Indexed: 12/14/2022]
Abstract
It is well accepted that regular exercise is a significant factor in the prevention of cardiac dysfunction; however, the cardioprotective mechanism is as yet not well defined. We have examined whether regular exercise can modulate the activity of cardiac mast cells (CMC) after deprivation of female sex hormones, as well as the density and percentage degranulation of mast cells, in ventricular tissue of ovariectomized (OVX) rats after an 11-week running program. A significant increase in CMC density with a greater percentage degranulation was induced after ovarian sex hormone deprivation. Increased CMC density was prevented by estrogen supplements, but not by regular training. To the contrary, increased CMC degranulation in the OVX rat heart was attenuated by exercise training, but not by estrogen supplement. These findings indicate a significant correlation between the degree of CMC degranulation and myocyte cross-section area. However, no change in the expression of inflammatory mediators, including chymase, interleukin-6, and interleukin-10, was detected. Taken together, these results clearly indicate one of the cardioprotective mechanisms of regular aerobic exercise is the modulation of CMC activation.
Collapse
Affiliation(s)
- Sukanya Phungphong
- Department of Physiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand
| | - Anusak Kijtawornrat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Jonggonnee Wattanapermpool
- Department of Physiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand
| | - Tepmanas Bupha-Intr
- Department of Physiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| |
Collapse
|
21
|
Zheng K, Wu W, Yang S, Huang L, Chen J, Gong C, Fu Z, Zhang L, Tan J. Bone marrow mesenchymal stem cell implantation for the treatment of radioactivity‑induced acute skin damage in rats. Mol Med Rep 2015; 12:7065-71. [PMID: 26323987 DOI: 10.3892/mmr.2015.4270] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 06/22/2015] [Indexed: 12/21/2022] Open
Abstract
The present study aimed to observe the role of mesenchymal stem cells (MSCs) in the repair of acute skin damage caused by radiation. Rat bone marrow MSCs (BMSCs) were isolated and cultured in vitro. A rat model of radiation‑induced acute skin damage was established by irradiation of the hind legs of Sprague-Dawley rats using a linear accelerator (45 Gy). After irradiation, rats were randomly divided into two groups: BMSC group and control group. Rats in the BMSC group were treated with a tail vein injection of 2x106 BMSCs (1 ml) immediately after irradiation and a local multipoint injection of 2x106 BMSCs at the injured area two weeks later. Then the wound healing of each rat was observed. The expression of transforming growth factor (TGF)‑β1, stromal cell‑derived factor-1 (SDF‑1) and prostaglandin E2 (PGE2) in the wounded tissues was determined by immunohistochemistry. The results demonstrated that skin damage was milder in the BMSC group than in the control group. Moreover, the speed of healing in the BMSC group was better than that in the control group. In addition, the wound score, it was significantly lower in the BMSC group than in the control group (P<0.05). The expression of PGE2 and TGF‑β1 in the BMSC group was also significantly lower than that in the control group (P<0.05), whereas the SDF‑1 expression was significantly higher in the BMSC group than that in the control group (P<0.05). BMSCs can effectively reduce inflammation and fibrosis in the wounded skin and promote the repair of acute radioactive skin injury. Thus, may be developed as a novel treatment for wound healing.
Collapse
Affiliation(s)
- Kai Zheng
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Weizhen Wu
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Shunliang Yang
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Lianghu Huang
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Jin Chen
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Chungui Gong
- Radiotherapy Center, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Zhichao Fu
- Radiotherapy Center, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Linlin Zhang
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Jianming Tan
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
22
|
Sung JH, An HS, Jeong JH, Shin S, Song SY. Megestrol Acetate Increases the Proliferation, Migration, and Adipogenic Differentiation of Adipose-Derived Stem Cells via Glucocorticoid Receptor. Stem Cells Transl Med 2015; 4:789-99. [PMID: 25972147 DOI: 10.5966/sctm.2015-0009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/08/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED : Because adipose-derived stem cells (ASCs) are usually expanded to acquire large numbers of cells for therapeutic applications, it is important to increase the production yield and regenerative potential during expansion. Therefore, a tremendous need exists for alternative ASC stimuli during cultivation to increase the proliferation and adipogenic differentiation of ASCs. The present study primarily investigated the involvement of megestrol acetate (MA), a progesterone analog, in the stimulation of ASCs, and identifies the target receptors underlying stimulation. Mitogenic and adipogenic effects of MA were investigated in vitro, and pharmacological inhibition and small interfering (si) RNA techniques were used to identify the molecular mechanisms involved in the MA-induced stimulation of ASCs. MA significantly increased the proliferation, migration, and adipogenic differentiation of ASCs in a dose-dependent manner. Glucocorticoid receptor (GR) is highly expressed compared with other nuclear receptors in ASCs, and this receptor is phosphorylated after MA treatment. MA also upregulated genes downstream of GR in ASCs, including ANGPTL4, DUSP1, ERRF11, FKBP5, GLUL, and TSC22D3. RU486, a pharmacological inhibitor of GR, and transfection of siGR significantly attenuated MA-induced proliferation, migration, and adipogenic differentiation of ASCs. Although the adipogenic differentiation potential of MA was inferior to that of dexamethasone, MA had mitogenic effects in ASCs. Collectively, these results indicate that MA increases the proliferation, migration, and adipogenic differentiation of ASCs via GR phosphorylation. SIGNIFICANCE Magestrol acetate (MA) increases the proliferation, migration, and adipogenic differentiation of adipose-derived stem cells (ASCs) via glucocorticoid receptor phosphorylation. Therefore, MA can be applied to increase the production yield during expansion and can be used to facilitate adipogenic differentiation of ASCs.
Collapse
Affiliation(s)
- Jong-Hyuk Sung
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo-Sun An
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Hyun Jeong
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soyoung Shin
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Yong Song
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea; STEMORE Co. Ltd., Incheon, Republic of Korea; College of Pharmacy, Wonkwang University, Iksan, Republic of Korea; Institute for Human Tissue Restoration, Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
|
24
|
Effect of Progesterone, Its Hydroxylated and Methylated Derivatives, and Dydrogesterone on Lipid Bilayer Membranes. J Membr Biol 2015; 248:811-24. [DOI: 10.1007/s00232-015-9803-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/12/2015] [Indexed: 10/23/2022]
|
25
|
Tang RJ, Shen SN, Zhao XY, Nie YZ, Xu YJ, Ren J, Lv MM, Hou YY, Wang TT. Mesenchymal stem cells-regulated Treg cells suppress colitis-associated colorectal cancer. Stem Cell Res Ther 2015; 6:71. [PMID: 25889203 PMCID: PMC4414289 DOI: 10.1186/s13287-015-0055-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 08/30/2014] [Accepted: 03/18/2015] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Previous studies have produced controversial results regarding whether mesenchymal stem cells (MSCs) promote or inhibit tumor development. Given the dual role of MSCs in inflammation and cancer, in this study the colitis-associated colorectal cancer (CAC) model was used to examine whether umbilical cord tissue-derived MSCs could prevent neoplasm by inhibiting chronic inflammation. METHODS MSCs were obtained and identified using flow cytometry. Colitis-associated colorectal cancer model was induced using azoxymethane (AOM) and dextran sulfate sodium (DSS) and MSCs were injected intravenously twice. Levels of immune cells in mesenteric lymph node including regulatory T (Treg) cells were detected using flow cytometry. Naïve T cells and Jurkat cells were co-cultured with MSCs and the effect of MSCs on Treg cells differentiation was evaluated. RESULTS After injection through tail vein, MSCs could migrate to colon and suppress colitis-related neoplasm. This tumor suppressive effect was characterized by longer colon length, decreased tumor numbers and decreased expression of Ki-67. Moreover, MSCs alleviated the pathology of inflammation in the colitis stage of CAC model and inhibited inflammation cytokines both in colon and serum. Furthermore, Treg cells were accumulated in mesenteric lymph node of MSCs-treated mice while the percentage of T helper cells 2 (Th2) and Th17 were not changed. Of note, MSCs secreted transforming growth factor-β (TGF-β) enhanced the induction of Treg cells from naïve T cells. The conditioned medium of MSCs also activated Smad2 signaling, which has been reported to regulate Treg cells. CONCLUSIONS These results proved that MSCs could migrate to colon tissues and induce the differentiation of Treg cells via Smad2 as so to inhibit the colitis and suppress the development of CAC.
Collapse
Affiliation(s)
- Rui-jing Tang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Su-nan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| | - Xiao-yin Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yun-zhong Nie
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yu-jun Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Jing Ren
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Ming-ming Lv
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Ya-yi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| | - Ting-ting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, China.
| |
Collapse
|
26
|
Zhao X, Liu D, Gong W, Zhao G, Liu L, Yang L, Hou Y. The toll-like receptor 3 ligand, poly(I:C), improves immunosuppressive function and therapeutic effect of mesenchymal stem cells on sepsis via inhibiting MiR-143. Stem Cells 2014; 32:521-33. [PMID: 24105952 DOI: 10.1002/stem.1543] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 08/02/2013] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are attractive candidates for clinical therapeutic applications. Recent studies indicate MSCs express active Toll-like receptors (TLRs), but their effect on MSCs and the underlying mechanisms remain unclear. In this study, we found that, after treating human umbilical cord MSCs with various TLR ligands, only TLR3 ligand, poly(I:C), could significantly increase the expression of cyclooxygenase-2 (COX-2). Furthermore, poly(I:C) could enhance MSCs' anti-inflammatory effect on macrophages. Next, we focused on the regulatory roles of microRNAs (miRNAs) in the process of poly(I:C) activating MSCs. Our experiments indicated that miR-143 expression was significantly decreased in MSCs with poly(I:C) treatment, and the expression level of miR-143 could regulate the effect of poly(I:C) on MSCs' immunosuppressive function. Subsequent results showed that the reporter genes with putative miR-143 binding sites from the transforming growth factor-β-activated kinase-1 (TAK1) and COX-2 3' untranslated regions were downregulated in the presence of miR-143. In addition, mRNA and protein expression of TAK1 and COX-2 in MSCs was also downregulated with miR-143 overexpression, suggesting that TAK1 and COX-2 are target genes of miR-143 in MSCs. Consistent with miR-143 overexpression, TAK1 interference also attenuated MSCs' immunosuppressive function enhanced by poly(I:C). Additionally, it was shown that TLR3-activated MSCs could improve survival in cecal ligation and puncture (CLP)-induced sepsis, while miR-143 overexpression reduced the effectiveness of this therapy. These results proved that poly(I:C) improved the immunosuppressive abilities of MSCs, revealed the regulatory role of miRNAs in the process, and may provide an opportunity for potential novel therapies for sepsis.
Collapse
Affiliation(s)
- Xiaoyin Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Female sex hormones ameliorate arthritis in SKG mice. Biochem Biophys Res Commun 2013; 434:740-5. [DOI: 10.1016/j.bbrc.2013.03.111] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 02/07/2023]
|
28
|
Laroni A, Novi G, Kerlero de Rosbo N, Uccelli A. Towards clinical application of mesenchymal stem cells for treatment of neurological diseases of the central nervous system. J Neuroimmune Pharmacol 2013; 8:1062-76. [PMID: 23579931 DOI: 10.1007/s11481-013-9456-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 03/31/2013] [Indexed: 12/13/2022]
Abstract
The diagnosis of a neurological disease of the central nervous system (CNS) is often associated with the anticipation of an irreversible and untreatable disability. This is the case also of multiple sclerosis (MS) where approved treatments effectively modulate the autoimmune attack to myelin antigens, but poorly affect neurodegeneration and do not promote tissue repair. Thus, stem cell-based therapies are increasingly being considered a possible strategy for diseases of the CNS. Mesenchymal stem cells (MSC), the safety of which has been demonstrated in the last 20 years through clinical trials and case studies, are of particular interest in view not only of their neuroprotective, but also of their immunomodulatory properties. Here, we review the therapeutic features of MSC that make them relevant in the treatment of CNS illnesses and discuss the pioneer clinical experience with MSC-based therapy in neurological diseases.
Collapse
Affiliation(s)
- Alice Laroni
- Department of Neurosciences Ophthalmology, Genetics, Rehabilitation and Child Health, University of Genoa, Genoa, Italy
| | | | | | | |
Collapse
|