1
|
Asharaf S, Chakraborty K, Paulose SK, Dhara S, Chakraborty RD, Varghese C. Photoprotective sulfated mannogalactan from heterotrophic Bacillus velezensis blocks UV-A mediated matrix metalloproteinase expression and nuclear DNA damage in human dermal fibroblast. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 260:113022. [PMID: 39288553 DOI: 10.1016/j.jphotobiol.2024.113022] [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: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
Prolonged exposure of human dermal fibroblasts (HDF) to ultraviolet (UV) radiation triggers the production of reactive oxygen species by upregulating the expression of matrix metalloproteinases (MMPs), causing type-I collagen degradation and photoaging. A sulfated (1 → 3)/(1 → 4) mannogalactan exopolysaccharide (BVP-2) characterized as [→3)-α-Galp-{(1 → 4)-α-6-O-SO3-Manp}-(1 → 3)-α-6-O-SO3-Galp-(1→] was isolated from seaweed-associated heterotrophic bacterium Bacillus velezensis MTCC13097. Whole genome analysis of B. velezensis MTCC13097 (Accession number JAKYLL000000000) revealed saccharine biosynthetic gene clusters for exopolysaccharide production. BVP-2 administered cells showed noteworthy reduction in mitochondrial superoxide (∼85 %, p < 0.05) and ROS production (62 %) than those exhibited by UV-A irradiated HDF cells. Oxidative imbalance in HDF cells (after UV-A exposure) was recovered with BVP-2 treatment by significantly downregulating nitric oxide (NO) production (98.6 μM/mL, 1.9-fold) and DNA damage (⁓67 %) in comparison with UV-A induced cells (191.8 μM/mL and 98.7 %, respectively). UV-irradiated HDF cells showed a ∼30-50 % downregulation in the expression of MMPs (1, 2, and 9) following treatment with BVP-2. Considerable amount of sulfation (18 %) along with (1 → 3)/(1 → 4) glycosidic linkages in BVP-2 could be pivotal factors for down-regulation of the intracellular MMP-1, which was further supported by molecular docking and structure-activity studies. The (1 → 3)/(1 → 4)-linked bacterial exopolysaccharide (BVP-2) might be used as prospective natural lead to attenuate and mitigate UV-A-induced photoaging.
Collapse
Affiliation(s)
- Sumayya Asharaf
- Marine Biotechnology Fish Nutrition and Health Division, Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala, India; Faculty of Marine Sciences, Lakeside Campus, Cochin University of Science and Technology, Cochin, Kerala, India
| | - Kajal Chakraborty
- Marine Biotechnology Fish Nutrition and Health Division, Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala, India.
| | - Silpa Kunnappilly Paulose
- Marine Biotechnology Fish Nutrition and Health Division, Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala, India; Department of Chemistry, Mangalore University, Mangalagangothri 574199, Karnataka, India
| | - Shubhajit Dhara
- Marine Biotechnology Fish Nutrition and Health Division, Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala, India; Department of Chemistry, Mangalore University, Mangalagangothri 574199, Karnataka, India
| | - Rekha Devi Chakraborty
- Shellfish Fisheries Division, Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin 682018, Kerala, India
| | - Chesvin Varghese
- Marine Biotechnology Fish Nutrition and Health Division, Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala, India
| |
Collapse
|
2
|
Chen YH, Hung YP, Chen CY, Chen YT, Tsai TC, Yang JJ, Wu CC. ELIXCYTE ®, an Allogenic Adipose-Derived Stem Cell Product, Mitigates Osteoarthritis by Reducing Inflammation and Preventing Cartilage Degradation In Vitro. Curr Issues Mol Biol 2024; 46:8395-8406. [PMID: 39194712 DOI: 10.3390/cimb46080495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) comprise a promising therapy for osteoarthritis (OA). The therapeutic potential of ELIXCYTE®, an allogeneic human ADSC (hADSC) product, was demonstrated in a phase I/II OA clinical trial. However, the exact mechanism underlying such effects is not clear. Moreover, studies suggest that interleukin-11 (IL-11) has anti-inflammatory, tissue-regenerative, and immune-regulatory functions. Our aim was to unravel the mechanism associated with the therapeutic effects of ELIXCYTE® on OA and its relationship with IL-11. We cocultured ELIXCYTE® with normal human articular chondrocytes (NHACs) in synovial fluid obtained from individuals with OA (OA-SF) to investigate its effect on chondrocyte matrix synthesis and degradation and inflammation by assessing gene expression and cytokine levels. NHACs exposed to OA-SF exhibited increased MMP13 expression. However, coculturing ELIXCYTE® with chondrocytes in OA-SF reduced MMP13 expression in chondrocytes and downregulated PTGS2 and FGF2 expression in ELIXCYTE®. ELIXCYTE® treatment elevated anti-inflammatory cytokine (IL-1RA, IL-10, and IL-13) levels, and the reduction in MMP13 was positively correlated with IL-11 concentrations in OA-SF. These findings indicate that IL-11 in OA-SF might serve as a predictive biomarker for the ELIXCYTE® treatment response in OA, emphasizing the therapeutic potential of ELIXCYTE® to mitigate OA progression and provide insights into its immunomodulatory effects.
Collapse
Affiliation(s)
- Yu-Hsiu Chen
- Rheumatology/Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Yi-Pei Hung
- UnicoCell Biomed Co., Ltd., Taipei 11494, Taiwan
| | | | - Yi-Ting Chen
- UnicoCell Biomed Co., Ltd., Taipei 11494, Taiwan
| | | | - Jui-Jung Yang
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Chia-Chun Wu
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| |
Collapse
|
3
|
Swain HN, Boyce PD, Bromet BA, Barozinksy K, Hance L, Shields D, Olbricht GR, Semon JA. Mesenchymal stem cells in autoimmune disease: A systematic review and meta-analysis of pre-clinical studies. Biochimie 2024; 223:54-73. [PMID: 38657832 DOI: 10.1016/j.biochi.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/08/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Mesenchymal Stem Cells (MSCs) are of interest in the clinic because of their immunomodulation capabilities, capacity to act upstream of inflammation, and ability to sense metabolic environments. In standard physiologic conditions, they play a role in maintaining the homeostasis of tissues and organs; however, there is evidence that they can contribute to some autoimmune diseases. Gaining a deeper understanding of the factors that transition MSCs from their physiological function to a pathological role in their native environment, and elucidating mechanisms that reduce their therapeutic relevance in regenerative medicine, is essential. We conducted a Systematic Review and Meta-Analysis of human MSCs in preclinical studies of autoimmune disease, evaluating 60 studies that included 845 patient samples and 571 control samples. MSCs from any tissue source were included, and the study was limited to four autoimmune diseases: multiple sclerosis, rheumatoid arthritis, systemic sclerosis, and lupus. We developed a novel Risk of Bias tool to determine study quality for in vitro studies. Using the International Society for Cell & Gene Therapy's criteria to define an MSC, most studies reported no difference in morphology, adhesion, cell surface markers, or differentiation into bone, fat, or cartilage when comparing control and autoimmune MSCs. However, there were reported differences in proliferation. Additionally, 308 biomolecules were differentially expressed, and the abilities to migrate, invade, and form capillaries were decreased. The findings from this study could help to explain the pathogenic mechanisms of autoimmune disease and potentially lead to improved MSC-based therapeutic applications.
Collapse
Affiliation(s)
- Hailey N Swain
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Parker D Boyce
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Bradley A Bromet
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Kaiden Barozinksy
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Lacy Hance
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Dakota Shields
- Department of Mathematics and Statistics, Missouri University of Science and Technology, USA
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, USA.
| |
Collapse
|
4
|
Kaszubowska L, Kaczor JJ, Karnia MJ, Foerster J, Kmieć Z. Expression of a stress-inducible heme oxygenase-1 in NK cells is maintained in the process of human aging. Front Immunol 2024; 15:1398468. [PMID: 39100660 PMCID: PMC11294084 DOI: 10.3389/fimmu.2024.1398468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction Heme oxygenase-1 (HO-1) is a stress-inducible heat shock protein (HSP32) that exerts cytoprotective effects against oxidative stress and inflammation, and is involved in the maintenance of cellular homeostasis. This study aimed to evaluate the expression of HO-1 in natural killer (NK) cells from individuals of different age groups after stimulation with various factors, and to analyze the relationships between the concentration of this cytoprotective protein and parameters corresponding to oxidative stress and inflammation, that is, NOD-like receptor protein 3 (NLRP3), glutathione (GSH), GSH disulfide (GSSG), and interleukin 6 (IL-6). Methods The study population comprised three age groups: young adults (age range, 19-23 years), older adults aged under 85 years (age range, 73-84 years), and older adults aged over 85 years (age range, 85-92 years). NLRP3, GSH, and GSSG concentrations were measured in serum, whereas the HO-1 concentration and IL-6 expression were studied in NK cells cultivated for 48 h and stimulated with IL-2, lipopolysaccharide (LPS), or phorbol 12-myristate 13-acetate (PMA) with ionomycin. Results The analysis of serum NLRP3, GSH, and GSSG concentrations revealed no statistically significant differences among the studied age groups. However, some typical trends of aging were observed, such as a decrease in GSH concentration and an increase in both GSSG level, and GSSG/GSH ratio. The highest basal expression of IL-6 and lowest basal content of HO-1 were found in NK cells of adults over 85 years of age. The NK cells in this age group also showed the highest sensitivity to stimulation with the applied factors. Moreover, statistically significant negative correlations were observed between HO-1 and IL-6 expression levels in the studied NK cells. Conclusions These results showed that NK cells can express HO-1 at a basal level, which was significantly increased in activated cells, even in the oldest group of adults. The reciprocal relationship between HO-1 and IL-6 expression suggests a negative feedback loop between these parameters.
Collapse
Affiliation(s)
| | - Jan Jacek Kaczor
- Department of Animal and Human Physiology, University of Gdańsk, Gdańsk, Poland
| | | | - Jerzy Foerster
- Department of Social and Clinical Gerontology, Medical University of Gdańsk, Gdańsk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
5
|
Donnelly H, Ross E, Xiao Y, Hermantara R, Taqi AF, Doherty-Boyd WS, Cassels J, Tsimbouri PM, Dunn KM, Hay J, Cheng A, Meek RMD, Jain N, West C, Wheadon H, Michie AM, Peault B, West AG, Salmeron-Sanchez M, Dalby MJ. Bioengineered niches that recreate physiological extracellular matrix organisation to support long-term haematopoietic stem cells. Nat Commun 2024; 15:5791. [PMID: 38987295 PMCID: PMC11237034 DOI: 10.1038/s41467-024-50054-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 06/27/2024] [Indexed: 07/12/2024] Open
Abstract
Long-term reconstituting haematopoietic stem cells (LT-HSCs) are used to treat blood disorders via stem cell transplantation. The very low abundance of LT-HSCs and their rapid differentiation during in vitro culture hinders their clinical utility. Previous developments using stromal feeder layers, defined media cocktails, and bioengineering have enabled HSC expansion in culture, but of mostly short-term HSCs and progenitor populations at the expense of naive LT-HSCs. Here, we report the creation of a bioengineered LT-HSC maintenance niche that recreates physiological extracellular matrix organisation, using soft collagen type-I hydrogels to drive nestin expression in perivascular stromal cells (PerSCs). We demonstrate that nestin, which is expressed by HSC-supportive bone marrow stromal cells, is cytoprotective and, via regulation of metabolism, is important for HIF-1α expression in PerSCs. When CD34+ve HSCs were added to the bioengineered niches comprising nestin/HIF-1α expressing PerSCs, LT-HSC numbers were maintained with normal clonal and in vivo reconstitution potential, without media supplementation. We provide proof-of-concept that our bioengineered niches can support the survival of CRISPR edited HSCs. Successful editing of LT-HSCs ex vivo can have potential impact on the treatment of blood disorders.
Collapse
Affiliation(s)
- Hannah Donnelly
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Ewan Ross
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Yinbo Xiao
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Rio Hermantara
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, United Kingdom
| | - Aqeel F Taqi
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, United Kingdom
| | - W Sebastian Doherty-Boyd
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Jennifer Cassels
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Penelope M Tsimbouri
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Karen M Dunn
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Jodie Hay
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Annie Cheng
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - R M Dominic Meek
- Department of Trauma and Orthopaedics, Queen Elizabeth University Hospital, Glasgow, G51 4TF, United Kingdom
| | - Nikhil Jain
- Institute of Inflammation and Ageing, University of Birmingham, Queen Elizabeth Hospital, Birmingham, B15 2WB, United Kingdom
| | - Christopher West
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Helen Wheadon
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Alison M Michie
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Bruno Peault
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Adam G West
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, United Kingdom
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom.
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom.
| |
Collapse
|
6
|
Sun L, Rao S, Kerim K, Lu J, Li H, Zhao S, Shen P, Sun W. A chemically adjustable BMP6-IL6 axis in mesenchymal stem cells drives acute myeloid leukemia cell differentiation. Biochem Pharmacol 2024; 225:116262. [PMID: 38705535 DOI: 10.1016/j.bcp.2024.116262] [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: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Chemotherapy alone or in combination with allogeneic stem cell transplantation has been the standard of care for acute myeloid leukemia (AML) for decades. Leukemia relapse with limited treatment options remains the main cause of treatment failure. Therefore, an effective and safe approach to improve treatment outcomes is urgently needed for most AML patients. Mesenchymal stem cells (MSCs) have been reported to efficiently induce apoptosis and shape the fate of acute myeloid leukemia cells. Here, we identified LG190155 as a potent compound that enhances the antileukemia efficiency of MSCs. Pretreatment of MSCs with LG190155 significantly provoked differentiation in both AML patient-derived primary leukemia cells and AML cell lines and reduced the tumor burden in the AML mouse model. Using the quantitative proteomic technique, we discovered a pivotal mechanism that mediates AML cell differentiation, in which autocrine bone morphogenetic protein 6 (BMP6) in MSCs boosted IL-6 secretion and further acted on leukemic cells to trigger differentiation. Furthermore, the activity of the BMP6-IL6 axis was dramatically enhanced by activating vitamin D receptor (VDR) in MSCs. Our data illustrated an effective preactivated approach to reinforcing the antileukemia effect of MSCs, which could serve as an effective therapeutic strategy for AML.
Collapse
Affiliation(s)
- Luchen Sun
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Shangrui Rao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Kamran Kerim
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jianhua Lu
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hongzheng Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Shengsheng Zhao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Pingping Shen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Shenzhen Research Institute of NanJing University, Shenzhen 518000, China.
| | - Weijian Sun
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
7
|
Hirakawa T, Nakabayashi K, Ito N, Hata K, Imi S, Shibata M, Urushiyama D, Miyata K, Yotsumoto F, Yasunaga S, Baba T, Miyamoto S. Transwell Culture with Adipose Tissue-Derived Stem Cells and Fertilized Eggs Mimics the In Vivo Development of Fertilized Eggs to Blastocysts in the Fallopian Tube: An Animal Study. Antioxidants (Basel) 2024; 13:704. [PMID: 38929143 PMCID: PMC11200376 DOI: 10.3390/antiox13060704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Many countries, including Japan, are experiencing declining birth rates. Assisted reproductive technologies have consistently demonstrated good results in resolving infertility. Although the development of fertilized eggs into blastocysts has been recognized as a crucial step in assisted reproductive technologies, the involved mechanisms are currently unclear. Here, we established a new culture system for the in vitro development of fertilized eggs into blastocysts. In the Transwell culture system, the rate of blastocysts hatching from fertilized eggs cultured with adipose-derived stem cells (ASCs) was significantly higher than that of blastocysts cultured only with fertilized eggs. Gene ontology analysis revealed that the developed blastocysts displayed essential gene expression patterns in mature blastocysts. Additionally, when cultured with 3rd-passage ASCs, the developed blastocysts expressed the core genes for blastocyst maturation and antioxidant properties compared to those cultured only with fertilized eggs or cultured with 20th-passage ASCs. These results suggest that the Transwell culture system may imitate the in vivo tubal culture state for fertilized eggs. Exosomes derived from stem cells with stemness potential play a powerful role in the development of blastocysts from fertilized eggs. Additionally, the exosomes expressed specific microRNAs; therefore, the Transwell culture system resulted in a higher rate of pregnancy. In future, the extraction of their own extracellular vesicles from the culture medium might contribute to the development of novel assisted reproductive technologies.
Collapse
Affiliation(s)
- Toyofumi Hirakawa
- Department of Obstetrics & Gynecology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; (T.H.); (S.I.); (M.S.); (D.U.); (K.M.); (F.Y.)
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.N.); (N.I.); (K.H.)
| | - Noriko Ito
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.N.); (N.I.); (K.H.)
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.N.); (N.I.); (K.H.)
| | - Shiori Imi
- Department of Obstetrics & Gynecology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; (T.H.); (S.I.); (M.S.); (D.U.); (K.M.); (F.Y.)
| | - Mami Shibata
- Department of Obstetrics & Gynecology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; (T.H.); (S.I.); (M.S.); (D.U.); (K.M.); (F.Y.)
| | - Daichi Urushiyama
- Department of Obstetrics & Gynecology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; (T.H.); (S.I.); (M.S.); (D.U.); (K.M.); (F.Y.)
| | - Kohei Miyata
- Department of Obstetrics & Gynecology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; (T.H.); (S.I.); (M.S.); (D.U.); (K.M.); (F.Y.)
| | - Fusanori Yotsumoto
- Department of Obstetrics & Gynecology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; (T.H.); (S.I.); (M.S.); (D.U.); (K.M.); (F.Y.)
| | - Shin’ichiro Yasunaga
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan;
| | - Tsukasa Baba
- Department of Obstetrics & Gynecology, School of Medicine, Iwate Medical University, Morioka 028-3694, Japan;
| | - Shingo Miyamoto
- Department of Obstetrics & Gynecology, School of Medicine, Iwate Medical University, Morioka 028-3694, Japan;
- Cybele Corporation Limited, 2-128-14 Sugukita, Kasugashi 816-0864, Japan
| |
Collapse
|
8
|
Li T, Li RHW, Ng EHY, Yeung WSB, Chiu PCN, Chan RWS. Interleukin 6 at menstruation promotes the proliferation and self-renewal of endometrial mesenchymal stromal/stem cells through the WNT/β-catenin signaling pathway. Front Immunol 2024; 15:1378863. [PMID: 38765018 PMCID: PMC11099287 DOI: 10.3389/fimmu.2024.1378863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/15/2024] [Indexed: 05/21/2024] Open
Abstract
Background At menstruation, the functional layer of the human endometrium sheds off due to the trigger of the release of inflammatory factors, including interleukin 6 (IL-6), as a result of a sharp decline in progesterone levels, leading to tissue breakdown and bleeding. The endometrial mesenchymal stem-like cells (CD140b+CD146+ eMSC) located in the basalis are responsible for the cyclical regeneration of the endometrium after menstruation. Endometrial cells from the menstruation phase have been proven to secrete a higher amount of IL-6 and further enhance the self-renewal and clonogenic activity of eMSC. However, the IL-6-responsive mechanism remains unknown. Thus, we hypothesized that IL-6 secreted from niche cells during menstruation regulates the proliferation and self-renewal of eMSC through the WNT/β-catenin signaling pathway. Methods In this study, the content of IL-6 across the menstrual phases was first evaluated. Coexpression of stem cell markers (CD140b and CD146) with interleukin 6 receptor (IL-6R) was confirmed by immunofluorescent staining. In vitro functional assays were conducted to investigate the effect of IL-6 on the cell activities of eMSC, and the therapeutic role of these IL-6- and WNT5A-pretreated eMSC on the repair of injured endometrium was observed using an established mouse model. Results The endometrial cells secrete a high amount of IL-6 under hypoxic conditions, which mimic the physiological microenvironment in the menstruation phase. Also, the expression of IL-6 receptors was confirmed in our eMSC, indicating their capacity to respond to IL-6 in the microenvironment. Exogenous IL-6 can significantly enhance the self-renewal, proliferation, and migrating capacity of eMSC. Activation of the WNT/β-catenin signaling pathway was observed upon IL-6 treatment, while suppression of the WNT/β-catenin signaling impaired the stimulatory role of IL-6 on eMSC activities. IL-6- and WNT5A-pretreated eMSC showed better performance during the regeneration of the injured mouse endometrium. Conclusion We demonstrate that the high level of IL-6 produced by endometrial cells at menstruation can induce the stem cells in the human endometrium to proliferate and migrate through the activation of the WNT/β-catenin pathway. Treatment of eMSC with IL-6 and WNT5A might enhance their therapeutic potential in the regeneration of injured endometrium.
Collapse
Affiliation(s)
- Tianqi Li
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Raymond H. W. Li
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Ernest H. Y. Ng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Rachel W. S. Chan
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
9
|
Sirpilla O, Sakemura RL, Hefazi M, Huynh TN, Can I, Girsch JH, Tapper EE, Cox MJ, Schick KJ, Manriquez-Roman C, Yun K, Stewart CM, Ogbodo EJ, Kimball BL, Mai LK, Gutierrez-Ruiz OL, Rodriguez ML, Gluscevic M, Larson DP, Abel AM, Wierson WA, Olivier G, Siegler EL, Kenderian SS. Mesenchymal stromal cells with chimaeric antigen receptors for enhanced immunosuppression. Nat Biomed Eng 2024; 8:443-460. [PMID: 38561490 DOI: 10.1038/s41551-024-01195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
Allogeneic mesenchymal stromal cells (MSCs) are a safe treatment option for many disorders of the immune system. However, clinical trials using MSCs have shown inconsistent therapeutic efficacy, mostly owing to MSCs providing insufficient immunosuppression in target tissues. Here we show that antigen-specific immunosuppression can be enhanced by genetically modifying MSCs with chimaeric antigen receptors (CARs), as we show for E-cadherin-targeted CAR-MSCs for the treatment of graft-versus-host disease in mice. CAR-MSCs led to superior T-cell suppression and localization to E-cadherin+ colonic cells, ameliorating the animals' symptoms and survival rates. On antigen-specific stimulation, CAR-MSCs upregulated the expression of immunosuppressive genes and receptors for T-cell inhibition as well as the production of immunosuppressive cytokines while maintaining their stem cell phenotype and safety profile in the animal models. CAR-MSCs may represent a widely applicable therapeutic technology for enhancing immunosuppression.
Collapse
Affiliation(s)
- Olivia Sirpilla
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - R Leo Sakemura
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Mehrdad Hefazi
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Truc N Huynh
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Ismail Can
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - James H Girsch
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Erin E Tapper
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Michelle J Cox
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Kendall J Schick
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Claudia Manriquez-Roman
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kun Yun
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Carli M Stewart
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Ekene J Ogbodo
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Brooke L Kimball
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Long K Mai
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Omar L Gutierrez-Ruiz
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Makena L Rodriguez
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Martina Gluscevic
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Daniel P Larson
- Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | - Alex M Abel
- LifEngine Animal Health Laboratories Incorporated, Rochester, MN, USA
| | - Wesley A Wierson
- LifEngine Animal Health Laboratories Incorporated, Rochester, MN, USA
| | - Gloria Olivier
- Department of Business Development, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
10
|
Poblano-Pérez LI, Castro-Manrreza ME, González-Alva P, Fajardo-Orduña GR, Montesinos JJ. Mesenchymal Stromal Cells Derived from Dental Tissues: Immunomodulatory Properties and Clinical Potential. Int J Mol Sci 2024; 25:1986. [PMID: 38396665 PMCID: PMC10888494 DOI: 10.3390/ijms25041986] [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: 12/30/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent cells located in different areas of the human body. The oral cavity is considered a potential source of MSCs because they have been identified in several dental tissues (D-MSCs). Clinical trials in which cells from these sources were used have shown that they are effective and safe as treatments for tissue regeneration. Importantly, immunoregulatory capacity has been observed in all of these populations; however, this function may vary among the different types of MSCs. Since this property is of clinical interest for cell therapy protocols, it is relevant to analyze the differences in immunoregulatory capacity, as well as the mechanisms used by each type of MSC. Interestingly, D-MSCs are the most suitable source for regenerating mineralized tissues in the oral region. Furthermore, the clinical potential of D-MSCs is supported due to their adequate capacity for proliferation, migration, and differentiation. There is also evidence for their potential application in protocols against autoimmune diseases and other inflammatory conditions due to their immunosuppressive capacity. Therefore, in this review, the immunoregulatory mechanisms identified at the preclinical level in combination with the different types of MSCs found in dental tissues are described, in addition to a description of the clinical trials in which MSCs from these sources have been applied.
Collapse
Affiliation(s)
- Luis Ignacio Poblano-Pérez
- Mesenchymal Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center (IMSS), Mexico City 06720, Mexico; (L.I.P.-P.); (G.R.F.-O.)
| | - Marta Elena Castro-Manrreza
- Immunology and Stem Cells Laboratory, FES Zaragoza, National Autonomous University of Mexico (UNAM), Mexico City 09230, Mexico;
| | - Patricia González-Alva
- Tissue Bioengineering Laboratory, Postgraduate Studies, Research Division, Faculty of Dentistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico;
| | - Guadalupe R. Fajardo-Orduña
- Mesenchymal Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center (IMSS), Mexico City 06720, Mexico; (L.I.P.-P.); (G.R.F.-O.)
| | - Juan José Montesinos
- Mesenchymal Stem Cell Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center (IMSS), Mexico City 06720, Mexico; (L.I.P.-P.); (G.R.F.-O.)
| |
Collapse
|
11
|
Sharma D, Sharma A, Hu L, Chen TA, Voon S, Bayless KJ, Goldman J, Walsh AJ, Zhao F. Perfusability and immunogenicity of implantable pre-vascularized tissues recapitulating features of native capillary network. Bioact Mater 2023; 30:184-199. [PMID: 37589031 PMCID: PMC10425689 DOI: 10.1016/j.bioactmat.2023.07.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
Vascularization is a key pre-requisite to engineered anatomical scale three dimensional (3-D) constructs to ensure their nutrient and oxygen supply upon implantation. Presently, engineered pre-vascularized 3-D tissues are limited to only micro-scale hydrogels, which meet neither the anatomical scale needs nor the complexity of natural extracellular matrix (ECM) environments. Anatomical scale perfusable constructs are critically needed for translational applications. To overcome this challenge, we previously developed pre-vascularized ECM sheets with long and oriented dense microvascular networks. The present study further evaluated the patency, perfusability and innate immune response toward these pre-vascularized constructs. Macrophage-co-cultured pre-vascularized constructs were evaluated in vitro to confirm micro-vessel patency and perturbations in macrophage metabolism. Subcutaneously implanted pre-vascularized constructs remained viable and formed a functional anastomosis with host vasculature within 3 days of implantation. This completely biological pre-vascularized construct holds great potential as a building block to engineer perfusable anatomical scale tissues.
Collapse
Affiliation(s)
- Dhavan Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Archita Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Linghao Hu
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Te-An Chen
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Sarah Voon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Kayla J. Bayless
- School of Medicine, Texas A&M University, College Station, TX, United States
| | - Jeremy Goldman
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, United States
| | - Alex J. Walsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| |
Collapse
|
12
|
Lee J, Jang J, Cha SR, Lee SB, Hong SH, Bae HS, Lee YJ, Yang SR. Recombinant Human Bone Morphogenetic Protein-2 Priming of Mesenchymal Stem Cells Ameliorate Acute Lung Injury by Inducing Regulatory T Cells. Immune Netw 2023; 23:e48. [PMID: 38188599 PMCID: PMC10767548 DOI: 10.4110/in.2023.23.e48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) possess immunoregulatory properties and their regulatory functions represent a potential therapy for acute lung injury (ALI). However, uncertainties remain with respect to defining MSCs-derived immunomodulatory pathways. Therefore, this study aimed to investigate the mechanism underlying the enhanced effect of human recombinant bone morphogenic protein-2 (rhBMP-2) primed ES-MSCs (MSCBMP2) in promoting Tregs in ALI mice. MSC were preconditioned with 100 ng/ml rhBMP-2 for 24 h, and then administrated to mice by intravenous injection after intratracheal injection of 1 mg/kg LPS. Treating MSCs with rhBMP-2 significantly increased cellular proliferation and migration, and cytokines array reveled that cytokines release by MSCBMP2 were associated with migration and growth. MSCBMP2 ameliorated LPS induced lung injury and reduced myeloperoxidase activity and permeability in mice exposed to LPS. Levels of inducible nitric oxide synthase were decreased while levels of total glutathione and superoxide dismutase activity were further increased via inhibition of phosphorylated STAT1 in ALI mice treated with MSCBMP2. MSCBMP2 treatment increased the protein level of IDO1, indicating an increase in Treg cells, and Foxp3+CD25+ Treg of CD4+ cells were further increased in ALI mice treated with MSCBMP2. In co-culture assays with MSCs and RAW264.7 cells, the protein level of IDO1 was further induced in MSCBMP2. Additionally, cytokine release of IL-10 was enhanced while both IL-6 and TNF-α were further inhibited. In conclusion, these findings suggest that MSCBMP2 has therapeutic potential to reduce massive inflammation of respiratory diseases by promoting Treg cells.
Collapse
Affiliation(s)
- Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Sang-Ryul Cha
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Se Bi Lee
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Han-Sol Bae
- Cellular Therapeutics Team, Daewoong Pharmaceutical, Yongin 17028, Korea
| | - Young Jin Lee
- Cellular Therapeutics Team, Daewoong Pharmaceutical, Yongin 17028, Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
13
|
Doron G, Wood LB, Guldberg RE, Temenoff JS. Poly(ethylene glycol)-Based Hydrogel Microcarriers Alter Secretory Activity of Genetically Modified Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2023; 9:6282-6292. [PMID: 37906515 PMCID: PMC10646834 DOI: 10.1021/acsbiomaterials.3c00954] [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: 07/13/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023]
Abstract
In order to scale up culture therapeutic cells, such as mesenchymal stromal cells (MSCs), culture in suspension bioreactors using microcarriers (μCs) is preferred. However, the impact of microcarrier type on the resulting MSC secretory activity has not been investigated. In this study, two poly(ethylene glycol) hydrogel formulations with different swelling ratios (named "stiffer" and "softer") were fabricated as μC substrates to culture MSCs and MSCs genetically modified to express the interleukin-1 receptor antagonist (IL-1Ra-MSCs). Changes in cell number, secretory and angiogenic activity, and changes in MAPK signaling were evaluated when cultured on hydrogel μCs, as well as on tissue culture plastic-based Synthemax μCs. We demonstrated that culture on stiffer μCs increased secretion of IL-1Ra compared to culture on Synthemax μCs by IL-1Ra-MSCs by 1.2- to 1.6-fold, as well as their in vitro angiogenic activity, compared to culture on Synthemax μCs, while culture on both stiffer and softer μCs altered the secretion of several other factors compared to culture on Synthemax μCs. Changes in angiogenic activity corresponded with increased gene expression and secretion of hepatocyte growth factor by MSCs cultured on softer μCs by 2.5- to 6-fold compared to MSCs cultured on Synthemax μCs. Quantification of phosphoprotein signaling with the MAPK pathway revealed broad reduction of pathway activation by IL-1Ra-MSCs cultured on both stiffer and softer μCs compared to Synthemax, where phosphorylated c-Jun, ATF2, and MEK1 were reduced specifically on softer μCs. Overall, this study showed that μC surfaces can influence the secretory activity of genetically modified MSCs and identified associated changes in MAPK pathway signaling, which is a known central regulator of cytokine secretion.
Collapse
Affiliation(s)
- Gilad Doron
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| | - Levi B. Wood
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. NW, Atlanta, Georgia 30318, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| | - Robert E. Guldberg
- Knight
Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, Oregon 97403, United States
| | - Johnna S. Temenoff
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| |
Collapse
|
14
|
Bajetto A, Pattarozzi A, Sirito R, Barbieri F, Florio T. Metformin potentiates immunosuppressant activity and adipogenic differentiation of human umbilical cord-mesenchymal stem cells. Int Immunopharmacol 2023; 124:111078. [PMID: 37844465 DOI: 10.1016/j.intimp.2023.111078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023]
Abstract
Metformin, a first-line drug for type-2 diabetes, displays pleiotropic effects on inflammation, aging, and cancer. Obesity triggers a low-grade chronic inflammation leading to insulin resistance, characterized by increased pro-inflammatory cytokines produced by adipocytes and infiltrated immune cells, which contributes to metabolic syndrome. We investigated metformin's differentiation and immunoregulatory properties of human umbilical cord-mesenchymal stem cells (UC-MSC), as cellular basis of its beneficial role in metabolic dysfunctions. Isolation, characterization and multilineage differentiation of UC-MSC were performed using standard protocols and flow-cytometry. Metformin effects on UC-MSC growth was assessed by colony formation and MTT assay, gene and protein expression by qRT-PCR, and western blot analysis. Proliferation of peripheral blood mononuclear cells (PBMCs) co-cultured with metformin-treated UC-MSC-conditioned media was evaluated by dye dilution assay. We show that metformin decreases proliferation and colony formation of UC-MSCs and enhances their adipogenic lineage commitment. Metformin (3 mM) increases PPARγ and downregulates FABP4 mRNA both in basal and in adipogenic culture conditions; however, the modulation of PPARγ expression is unrelated to the antiproliferative effects. Moreover, metformin inhibits UC-MSC inflammatory activity reducing the expression of IL-6, MCP-1, and COX-2. Conditioned media, collected from metformin-treated UC-MSCs, down-regulate CD3+ T lymphocyte growth in stimulated PBMCs and, in particular, reduce the CD8+ T cell population. These results indicate that metformin may favor new adipocyte formation and potentiate immune suppressive properties of UC-MSCs. Thus, adipose tissue regeneration and anti-inflammatory activity may represent possible mechanisms by which metformin exerts its positive effect on lipid metabolism.
Collapse
Affiliation(s)
- Adriana Bajetto
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Alessandra Pattarozzi
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Rodolfo Sirito
- Section of Obstetrics and Gynaecology, International Evangelical Hospital, 16122 Genova, Italy
| | - Federica Barbieri
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; IRCCS, Ospedale Policlinico San Martino, 16132 Genova, Italy.
| |
Collapse
|
15
|
Rosado-Galindo H, Domenech M. Substrate topographies modulate the secretory activity of human bone marrow mesenchymal stem cells. Stem Cell Res Ther 2023; 14:208. [PMID: 37605275 PMCID: PMC10441765 DOI: 10.1186/s13287-023-03450-0] [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: 09/15/2022] [Accepted: 08/11/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) secrete a diversity of factors with broad therapeutic potential, yet current culture methods limit potency outcomes. In this study, we used topographical cues on polystyrene films to investigate their impact on the secretory profile and potency of bone marrow-derived MSCs (hBM-MSCs). hBM-MSCs from four donors were cultured on topographic substrates depicting defined roughness, curvature, grooves and various levels of wettability. METHODS The topographical PS-based array was developed using razor printing, polishing and plasma treatment methods. hBM-MSCs from four donors were purchased from RoosterBio and used in co-culture with peripheral blood mononuclear cells (PBMCs) from Cell Applications Inc. in an immunopotency assay to measure immunosuppressive capacity. Cells were cultured on low serum (2%) for 24-48 h prior to analysis. Image-based analysis was used for cell quantification and morphology assessment. Metabolic activity of BM-hMSCs was measured as the mitochondrial oxygen consumption rate using an extracellular flux analyzer. Conditioned media samples of BM-hMSCs were used to quantify secreted factors, and the data were analyzed using R statistics. Enriched bioprocesses were identify using the Gene Ontology tool enrichGO from the clusterprofiler. One-way and two-way ANOVAs were carried out to identify significant changes between the conditions. Results were deemed statistically significant for combined P < 0.05 for at least three independent experiments. RESULTS Cell viability was not significantly affected in the topographical substrates, and cell elongation was enhanced at least twofold in microgrooves and surfaces with a low contact angle. Increased cell elongation correlated with a metabolic shift from oxidative phosphorylation to a glycolytic state which is indicative of a high-energy state. Differential protein expression and gene ontology analyses identified bioprocesses enriched across donors associated with immune modulation and tissue regeneration. The growth of peripheral blood mononuclear cells (PBMCs) was suppressed in hBM-MSCs co-cultures, confirming enhanced immunosuppressive potency. YAP/TAZ levels were found to be reduced on these topographies confirming a mechanosensing effect on cells and suggesting a potential role in the immunomodulatory function of hMSCs. CONCLUSIONS This work demonstrates the potential of topographical cues as a culture strategy to improve the secretory capacity and enrich for an immunomodulatory phenotype in hBM-MSCs.
Collapse
Affiliation(s)
- Heizel Rosado-Galindo
- Bioengineering Program, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA
| | - Maribella Domenech
- Bioengineering Program, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA.
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Road 108, KM 1.1., Mayagüez, PR, 00680, USA.
| |
Collapse
|
16
|
Jakl V, Popp T, Haupt J, Port M, Roesler R, Wiese S, Friemert B, Rojewski MT, Schrezenmeier H. Effect of Expansion Media on Functional Characteristics of Bone Marrow-Derived Mesenchymal Stromal Cells. Cells 2023; 12:2105. [PMID: 37626914 PMCID: PMC10453497 DOI: 10.3390/cells12162105] [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: 07/19/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The therapeutic efficacy of mesenchymal stromal cells (MSCs) has been shown to rely on their immunomodulatory and regenerative properties. In order to obtain sufficient numbers of cells for clinical applications, MSCs have to be expanded ex vivo. Expansion media with xenogeneic-free (XF) growth-promoting supplements like human platelet lysate (PL) or serum- and xenogeneic-free (SF/XF) formulations have been established as safe and efficient, and both groups provide different beneficial qualities. In this study, MSCs were expanded in XF or SF/XF media as well as in mixtures thereof. MSCs cultured in these media were analyzed for phenotypic and functional properties. MSC expansion was optimal with SF/XF conditions when PL was present. Metabolic patterns, consumption of growth factors, and secretome of MSCs differed depending on the type and concentration of supplement. The lactate per glucose yield increased along with a higher proportion of PL. Many factors in the supernatant of cultured MSCs showed distinct patterns depending on the supplement (e.g., FGF-2, TGFβ, and insulin only in PL-expanded MSC, and leptin, sCD40L PDGF-AA only in SF/XF-expanded MSC). This also resulted in changes in cell characteristics like migratory potential. These findings support current approaches where growth media may be utilized for priming MSCs for specific therapeutic applications.
Collapse
Affiliation(s)
- Viktoria Jakl
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
| | - Tanja Popp
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
| | - Julian Haupt
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, 89081 Ulm, Germany
| | - Matthias Port
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
| | - Reinhild Roesler
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany; (R.R.); (S.W.)
| | - Sebastian Wiese
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany; (R.R.); (S.W.)
| | - Benedikt Friemert
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, 89081 Ulm, Germany
| | - Markus T. Rojewski
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, 89081 Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
17
|
Rivera KR, Bliton RJ, Burclaff J, Czerwinski MJ, Liu J, Trueblood JM, Hinesley CM, Breau KA, Deal HE, Joshi S, Pozdin VA, Yao M, Ziegler AL, Blikslager AT, Daniele MA, Magness ST. Hypoxia Primes Human ISCs for Interleukin-Dependent Rescue of Stem Cell Activity. Cell Mol Gastroenterol Hepatol 2023; 16:823-846. [PMID: 37562653 PMCID: PMC10520368 DOI: 10.1016/j.jcmgh.2023.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND AND AIMS Hypoxia in the intestinal epithelium can be caused by acute ischemic events or chronic inflammation in which immune cell infiltration produces inflammatory hypoxia starving the mucosa of oxygen. The epithelium has the capacity to regenerate after some ischemic and inflammatory conditions suggesting that intestinal stem cells (ISCs) are highly tolerant to acute and chronic hypoxia; however, the impact of hypoxia on human ISC (hISC) function has not been reported. Here we present a new microphysiological system (MPS) to investigate how hypoxia affects hISCs from healthy donors and test the hypothesis that prolonged hypoxia modulates how hISCs respond to inflammation-associated interleukins (ILs). METHODS hISCs were exposed to <1.0% oxygen in the MPS for 6, 24, 48, and 72 hours. Viability, hypoxia-inducible factor 1a (HIF1a) response, transcriptomics, cell cycle dynamics, and response to cytokines were evaluated in hISCs under hypoxia. HIF stabilizers and inhibitors were screened to evaluate HIF-dependent responses. RESULTS The MPS enables precise, real-time control and monitoring of oxygen levels at the cell surface. Under hypoxia, hISCs maintain viability until 72 hours and exhibit peak HIF1a at 24 hours. hISC activity was reduced at 24 hours but recovered at 48 hours. Hypoxia induced increases in the proportion of hISCs in G1 and expression changes in 16 IL receptors. Prolyl hydroxylase inhibition failed to reproduce hypoxia-dependent IL-receptor expression patterns. hISC activity increased when treated IL1β, IL2, IL4, IL6, IL10, IL13, and IL25 and rescued hISC activity caused by 24 hours of hypoxia. CONCLUSIONS Hypoxia pushes hISCs into a dormant but reversible proliferative state and primes hISCs to respond to a subset of ILs that preserves hISC activity. These findings have important implications for understanding intestinal epithelial regeneration mechanisms caused by inflammatory hypoxia.
Collapse
Affiliation(s)
- Kristina R Rivera
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina
| | - R Jarrett Bliton
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina
| | - Joseph Burclaff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina
| | - Michael J Czerwinski
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jintong Liu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jessica M Trueblood
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Caroline M Hinesley
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Keith A Breau
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Halston E Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina
| | - Shlok Joshi
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Vladimir A Pozdin
- Department of Electrical and Computer Engineering, North Carolina State University, Raleigh, North Carolina
| | - Ming Yao
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina
| | - Amanda L Ziegler
- Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Anthony T Blikslager
- Comparative Medicine Institute, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina; Department of Electrical and Computer Engineering, North Carolina State University, Raleigh, North Carolina
| | - Scott T Magness
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
18
|
Herd CL, Mellet J, Mashingaidze T, Durandt C, Pepper MS. Consequences of HIV infection in the bone marrow niche. Front Immunol 2023; 14:1163012. [PMID: 37497228 PMCID: PMC10366613 DOI: 10.3389/fimmu.2023.1163012] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023] Open
Abstract
Dysregulation of the bone marrow niche resulting from the direct and indirect effects of HIV infection contributes to haematological abnormalities observed in HIV patients. The bone marrow niche is a complex, multicellular environment which functions primarily in the maintenance of haematopoietic stem/progenitor cells (HSPCs). These adult stem cells are responsible for replacing blood and immune cells over the course of a lifetime. Cells of the bone marrow niche support HSPCs and help to orchestrate the quiescence, self-renewal and differentiation of HSPCs through chemical and molecular signals and cell-cell interactions. This narrative review discusses the HIV-associated dysregulation of the bone marrow niche, as well as the susceptibility of HSPCs to infection by HIV.
Collapse
|
19
|
Sengun E, Wolfs TGAM, van Bruggen VLE, van Cranenbroek B, Simonetti ER, Ophelders D, de Jonge MI, Joosten I, van der Molen RG. Umbilical cord-mesenchymal stem cells induce a memory phenotype in CD4 + T cells. Front Immunol 2023; 14:1128359. [PMID: 37409122 PMCID: PMC10318901 DOI: 10.3389/fimmu.2023.1128359] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/01/2023] [Indexed: 07/07/2023] Open
Abstract
Inflammation is a physiological state where immune cells evoke a response against detrimental insults. Finding a safe and effective treatment for inflammation associated diseases has been a challenge. In this regard, human mesenchymal stem cells (hMSC), exert immunomodulatory effects and have regenerative capacity making it a promising therapeutic option for resolution of acute and chronic inflammation. T cells play a critical role in inflammation and depending on their phenotype, they can stimulate or suppress inflammatory responses. However, the regulatory effects of hMSC on T cells and the underlying mechanisms are not fully elucidated. Most studies focused on activation, proliferation, and differentiation of T cells. Here, we further investigated memory formation and responsiveness of CD4+ T cells and their dynamics by immune-profiling and cytokine secretion analysis. Umbilical cord mesenchymal stem cells (UC-MSC) were co-cultured with either αCD3/CD28 beads, activated peripheral blood mononuclear cells (PBMC) or magnetically sorted CD4+ T cells. The mechanism of immune modulation of UC-MSC were investigated by comparing different modes of action; transwell, direct cell-cell contact, addition of UC-MSC conditioned medium or blockade of paracrine factor production by UC-MSC. We observed a differential effect of UC-MSC on CD4+ T cell activation and proliferation using PBMC or purified CD4+ T cell co-cultures. UC-MSC skewed the effector memory T cells into a central memory phenotype in both co-culture conditions. This effect on central memory formation was reversible, since UC-MSC primed central memory cells were still responsive after a second encounter with the same stimuli. The presence of both cell-cell contact and paracrine factors were necessary for the most pronounced immunomodulatory effect of UC-MSC on T cells. We found suggestive evidence for a partial role of IL-6 and TGFβ in the UC-MSC derived immunomodulatory function. Collectively, our data show that UC-MSCs clearly affect T cell activation, proliferation and maturation, depending on co-culture conditions for which both cell-cell contact and paracrine factors are needed.
Collapse
Affiliation(s)
- Ezgi Sengun
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Tim G. A. M. Wolfs
- Department of Pediatrics and GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, Netherlands
| | - Valéry L. E. van Bruggen
- Department of Pediatrics and GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, Netherlands
| | - Bram van Cranenbroek
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Elles R. Simonetti
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Daan Ophelders
- Department of Pediatrics and GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, Netherlands
| | - Marien I. de Jonge
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Irma Joosten
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Renate G. van der Molen
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| |
Collapse
|
20
|
Kaszubowska L, Foerster J, Kaczor JJ, Karnia MJ, Kmieć Z. Anti-Inflammatory Klotho Protein Serum Concentration Correlates with Interferon Gamma Expression Related to the Cellular Activity of Both NKT-like and T Cells in the Process of Human Aging. Int J Mol Sci 2023; 24:ijms24098393. [PMID: 37176100 PMCID: PMC10179552 DOI: 10.3390/ijms24098393] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Klotho is a beta-glucuronidase that reveals both anti-inflammatory and anti-oxidative properties that have been associated with mechanisms of aging. The study aimed to analyze the relationships between the serum concentration of soluble α-Klotho and cellular activity of two populations of lymphocytes; T and NKT-like cells corresponding to the level of cytokine secretion; i.e., IFN-γ, TNF-α, and IL-6. The studied population comprised three age groups: young individuals ('young'), seniors aged under 85 ('old'), and seniors aged over 85 ('oldest'). Both NKT-like and T cells were either non-cultured or cultured for 48 h and stimulated appropriately with IL-2, LPS or PMA with ionomycin to compare with unstimulated control cells. In all studied age groups non-cultured or cultured NKT-like cells revealed higher expressions of TNF-α, IL-6, and IFN-γ than T cells. α-Klotho concentration in serum decreased significantly in the process of aging. Intriguingly, only IFN-γ expression revealed a positive correlation with α-Klotho protein serum concentration in both non-cultured and cultured T and NKT-like cells. Since IFN-γ is engaged in the maintenance of immune homeostasis, the observed relationships may indicate the involvement of α-Klotho and cellular IFN-γ expression in the network of adaptive mechanisms developed during the process of human aging.
Collapse
Affiliation(s)
- Lucyna Kaszubowska
- Department of Histology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| | - Jerzy Foerster
- Department of Social and Clinical Gerontology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| | - Jan Jacek Kaczor
- Department of Animal and Human Physiology, University of Gdańsk, J. Bażyńskiego 8 Street, 80-308 Gdańsk, Poland
| | - Mateusz Jakub Karnia
- Department of Animal and Human Physiology, University of Gdańsk, J. Bażyńskiego 8 Street, 80-308 Gdańsk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| |
Collapse
|
21
|
Chakraborty K, Thambi A, Dhara S. Sulfated polygalactofucan from triangular sea bell Turbinaria decurrens attenuates inflammatory cytokines on THP-1 human monocytic macrophages. Int J Biol Macromol 2023; 231:123220. [PMID: 36634794 DOI: 10.1016/j.ijbiomac.2023.123220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/31/2022] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
Inflammation is one of the most significant causes of several chronic diseases, which includes the expression of cytokines activating immune cells to up-regulate the inflammatory cascade. Polysaccharides from marine macroalgae are promising anti-inflammatory agents because of their potential to attenuate inflammatory cytokines. The triangular sea bell Turbinaria decurrens (Sargassaceae) among marine macroalgae is ubiquitous in oceanic waters, and a sulfated polygalactofucan SPTd-2 [→3-(α-L-fucp-(2-OSO3-)-(1 → 4)-α-L-fucp-(3-OAc)-(1 → 4)-β-D-galp-(1→] was purified from the species. The studied polygalactofucan SPTd-2 exhibited anti-inflammatory activities against cyclooxygenase-2 (IC50 10.56 μM) and 5-lipoxygenase (IC50 3.36 μM) with a greater selectivity index (2.35) than ibuprofen (0.44), besides attenuating pro-inflammatory cytokine production, including tumor necrosis factor-α, transforming growth factor-β, interleukin-2, 1β, and interferon-γ. Quantitative real-time polymerase chain reaction displayed that SPTd-2 blocked the mRNA of interferon-γ and interleukin-2, in the human monocytic cell line THP-1. The results showed the potential of SPTd-2 to attenuate inflammation-associated disorders.
Collapse
Affiliation(s)
- Kajal Chakraborty
- Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin, India.
| | - Anjaly Thambi
- Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin, India; Department of Applied Chemistry, Cochin University of Science and Technology, South Kalamassery, Kochi 682022, Kerala State, India
| | - Shubhajit Dhara
- Central Marine Fisheries Research Institute, Ernakulam North, P.B. No. 1603, Cochin, India; Department of Chemistry, Mangalore University, Mangalagangothri 574199, Karnataka State, India
| |
Collapse
|
22
|
Phipps J, Haseli M, Pinzon-Herrera L, Wilson B, Corbitt J, Servoss S, Almodovar J. Delivery of Immobilized IFN-γ With PCN-333 and Its Effect on Human Mesenchymal Stem Cells. ACS Biomater Sci Eng 2023; 9:671-679. [PMID: 36598843 DOI: 10.1021/acsbiomaterials.2c01038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Interferon-gamma (IFN-γ) plays a vital role in modulating the immunosuppressive properties of human mesenchymal stem/stromal cells (hMSCs) used in cell therapies. However, IFN-γ suffers from low bioavailability and degrades in media, creating a challenge when using IFN-γ during the manufacturing of hMSCs. Metal-organic frameworks (MOFs), with their porous interiors, biocompatibility, high loading capacity, and ability to be functionalized for targeting, have become an increasingly suitable platform for protein delivery. In this work, we synthesize the MOF PCN-333(Fe) and show that it can be utilized to immobilize and deliver IFN-γ to the local extracellular environment of hMSCs. In doing so, the cells proliferate and differentiate appropriately with no observed side effects. We demonstrate that PCN-333(Fe) MOFs containing IFN-γ are not cytotoxic to hMSCs, can promote the expression of proteins that play a role in immune response, and are capable of inducing indoleamine 2,3-dioxygenase (IDO) production similar to that of soluble IFN-γ at lower concentrations. Overall, using MOFs to deliver IFN-γ may be leveraged in the future in the manufacturing of therapeutically relevant hMSCs.
Collapse
Affiliation(s)
- Josh Phipps
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Mahsa Haseli
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Luis Pinzon-Herrera
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Ben Wilson
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Joshua Corbitt
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Shannon Servoss
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Jorge Almodovar
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas 72701, United States.,Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| |
Collapse
|
23
|
Rivera KR, Bliton RJ, Burclaff J, Czerwinski MJ, Liu J, Trueblood JM, Hinesley CM, Breau KA, Joshi S, Pozdin VA, Yao M, Ziegler AL, Blikslager AT, Daniele MA, Magness ST. A new microphysiological system shows hypoxia primes human ISCs for interleukin-dependent rescue of stem cell activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.524747. [PMID: 36778265 PMCID: PMC9915581 DOI: 10.1101/2023.01.31.524747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background & Aims Hypoxia in the intestinal epithelium can be caused by acute ischemic events or conditions like Inflammatory Bowel Disease (IBD) where immune cell infiltration produces 'inflammatory hypoxia', a chronic condition that starves the mucosa of oxygen. Epithelial regeneration after ischemia and IBD suggests intestinal stem cells (ISCs) are highly tolerant to acute and chronic hypoxia; however, the impact of acute and chronic hypoxia on human ISC (hISC) properties have not been reported. Here we present a new microphysiological system (MPS) to investigate how hypoxia affects hISCs isolated from healthy human tissues. We then test the hypothesis that some inflammation-associated interleukins protect hISCs during prolonged hypoxia. Methods hISCs were exposed to <1.0% oxygen in the MPS for 6-, 24-, 48- & 72hrs. Viability, HIF1α response, transcriptomics, cell cycle dynamics, and hISC response to cytokines were evaluated. Results The novel MPS enables precise, real-time control and monitoring of oxygen levels at the cell surface. Under hypoxia, hISCs remain viable until 72hrs and exhibit peak HIF1α at 24hrs. hISCs lose stem cell activity at 24hrs that recovers at 48hrs of hypoxia. Hypoxia increases the proportion of hISCs in G1 and regulates hISC capacity to respond to multiple inflammatory signals. Hypoxia induces hISCs to upregulate many interleukin receptors and hISCs demonstrate hypoxia-dependent cell cycle regulation and increased organoid forming efficiency when treated with specific interleukins. Conclusions Hypoxia primes hISCs to respond differently to interleukins than hISCs in normoxia through a transcriptional response. hISCs slow cell cycle progression and increase hISC activity when treated with hypoxia and specific interleukins. These findings have important implications for epithelial regeneration in the gut during inflammatory events.
Collapse
Affiliation(s)
- Kristina R. Rivera
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
| | - R. Jarrett Bliton
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
| | - Joseph Burclaff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
| | - Michael J. Czerwinski
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| | - Jintong Liu
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| | - Jessica M. Trueblood
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Caroline M. Hinesley
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Keith A Breau
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| | - Shlok Joshi
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| | - Vladimir A. Pozdin
- Department of Electrical & Computer Engineering, North Carolina State University, Raleigh, NC, 27695 (USA)
| | - Ming Yao
- Department of Mechanical & Aerospace Engineering, North Carolina State University, Raleigh, NC 27695 (USA)
| | - Amanda L. Ziegler
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Anthony T. Blikslager
- Center for Gastrointestinal Biology and Disease, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Michael A. Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
- Department of Electrical & Computer Engineering, North Carolina State University, Raleigh, NC, 27695 (USA)
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill 911 Oval Dr., Raleigh, NC, 27695 (USA)
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599 (USA)
| |
Collapse
|
24
|
CD146+ Endometrial-Derived Mesenchymal Stem/Stromal Cell Subpopulation Possesses Exosomal Secretomes with Strong Immunomodulatory miRNA Attributes. Cells 2022; 11:cells11244002. [PMID: 36552765 PMCID: PMC9777070 DOI: 10.3390/cells11244002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/28/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
The perivascular localization of endometrial mesenchymal stem/stromal cells (eMSC) allows them to sense local and distant tissue damage, promoting tissue repair and healing. Our hypothesis is that eMSC therapeutic effects are largely exerted via their exosomal secretome (eMSC EXOs) by targeting the immune system and angiogenic modulation. For this purpose, EXOs isolated from Crude and CD146+ eMSC populations were compared for their miRNA therapeutic signatures and immunomodulatory functionality under inflammatory conditions. eMSC EXOs profiling revealed 121 in Crude and 88 in CD146+ miRNAs, with 82 commonly present in both populations. Reactome and KEGG analysis of miRNAs highly present in eMSC EXOs indicated their involvement among others in immune system regulation. From the commonly present miRNAs, four miRNAs (hsa-miR-320e, hsa-miR-182-3p, hsa-miR-378g, hsa-let-7e-5p) were more enriched in CD146+ eMSC EXOs. These miRNAs are involved in macrophage polarization, T cell activation, and regulation of inflammatory cytokine transcription (i.e., TNF-α, IL-1β, and IL-6). Functionally, stimulated macrophages exposed to eMSC EXOs demonstrated a switch towards an alternate M2 status and reduced phagocytic capacity compared to stimulated alone. However, eMSC EXOs did not suppress stimulated human peripheral blood mononuclear cell proliferation, but significantly reduced secretion of 13 pro-inflammatory molecules compared to stimulated alone. In parallel, two anti-inflammatory proteins, IL-10 and IL-13, showed higher secretion, especially upon CD146+ eMSC EXO exposure. Our study suggests that eMSC, and even more, the CD146+ subpopulation, possess exosomal secretomes with strong immunomodulatory miRNA attributes. The resulting evidence could serve as a foundation for eMSC EXO-based therapeutics for the resolution of detrimental aspects of tissue inflammation.
Collapse
|
25
|
Li Y, Yu H, Lv M, Li Q, Zou K, Lv S. Combination therapy with budesonide and N-acetylcysteine ameliorates LPS-induced ALI by attenuating neutrophil recruitment through the miR-196b-5p/Socs3 molecular axis. BMC Pulm Med 2022; 22:388. [PMID: 36289489 PMCID: PMC9608916 DOI: 10.1186/s12890-022-02185-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 09/26/2022] [Indexed: 11/24/2022] Open
Abstract
Background Neutrophil infiltration accelerates the inflammatory response and is highly correlated to the development of acute lung injury (ALI). Budesonide (BUD) and N-acetylcysteine (NAC) both inhibit the inflammatory response to alleviate ALI, so we further investigated whether their combination is better for ALI. Methods In this study, we investigated the effect and mechanism of Combined BUD and NAC therapy on LPS-induced ALI. Rat ALI model and neutrophil abnormal activation model were established by lipopolysaccharide (LPS). BUD and NAC were treated alone or in combination, or cells were transfected with miR-196b-5p mimic or si-Socs3 to evaluate the efficacy and mechanism of BUD and NAC alone or in combination. Histopathological observation of lungs was performed by Hematoxylin Eosin (HE) staining. The quantity of neutrophils and inflammatory factors level in bronchoalveolar lavage fluid (BALF) were determined by Richter-Gimza complex stain and Enzyme-Linked Immunosorbnent Assay (ELISA), respectively. ReverseTranscription-PolymeraseChainReaction (RT–qPCR) was utilized to assess miR-196b-5p and inflammatory factor mRNA levels. The expression level of Socs3 was detected by immunohistochemistry or Western Blot. Results BUD and NAC combined treatment had a better effect on neutrophil recruitment and inflammatory response in LPS-induced ALI than did BUD and NAC alone. Transfection of the miR-196b-5p mimic reversed the effect of combined BUD and NAC. In conclusion, the combination of BUD and NAC is a better treatment for ALI. Conclusions Combination therapy with BUD and NAC ameliorates LPS-induced ALI by attenuating neutrophil recruitment through the miR-196b-5p/Socs3 molecular axis. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-02185-7.
Collapse
Affiliation(s)
- Yang Li
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, 655000 Yunnan China
| | - Huimin Yu
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, 655000 Yunnan China
| | - Meifen Lv
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, 655000 Yunnan China
| | - Qiaofen Li
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, 655000 Yunnan China
| | - Kaiwen Zou
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, 655000 Yunnan China
| | - Shaokun Lv
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, 655000 Yunnan China
| |
Collapse
|
26
|
Dey M, Kim MH, Nagamine M, Karhan E, Kozhaya L, Dogan M, Unutmaz D, Ozbolat IT. Biofabrication of 3D breast cancer models for dissecting the cytotoxic response of human T cells expressing engineered MAIT cell receptors. Biofabrication 2022; 14:10.1088/1758-5090/ac925a. [PMID: 36108605 PMCID: PMC9556424 DOI: 10.1088/1758-5090/ac925a] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/15/2022] [Indexed: 11/12/2022]
Abstract
Immunotherapy has revolutionized cancer treatment with the advent of advanced cell engineering techniques aimed at targeted therapy with reduced systemic toxicity. However, understanding the underlying immune-cancer interactions require development of advanced three-dimensional (3D) models of human tissues. In this study, we fabricated 3D tumor models with increasing complexity to study the cytotoxic responses of CD8+T cells, genetically engineered to express mucosal-associated invariant T (MAIT) cell receptors, towards MDA-MB-231 breast cancer cells. Homotypic MDA-MB-231 and heterotypic MDA-MB-231/human dermal fibroblast tumor spheroids were primed with precursor MAIT cell ligand 5-amino-6-D-ribitylaminouracil (5-ARU). Engineered T cells effectively eliminated tumors after a 3 d culture period, demonstrating that the engineered T cell receptor recognized major histocompatibility complex class I-related (MR1) protein expressing tumor cells in the presence of 5-ARU. Tumor cell killing efficiency of engineered T cells were also assessed by encapsulating these cells in fibrin, mimicking a tumor extracellular matrix microenvironment. Expression of proinflammatory cytokines such as interferon gamma, interleukin-13, CCL-3 indicated immune cell activation in all tumor models, post immunotherapy. Further, in corroborating the cytotoxic activity, we found that granzymes A and B were also upregulated, in homotypic as well as heterotypic tumors. Finally, a 3D bioprinted tumor model was employed to study the effect of localization of T cells with respect to tumors. T cells bioprinted proximal to the tumor had reduced invasion index and increased cytokine secretion, which indicated a paracrine mode of immune-cancer interaction. Development of 3D tumor-T cell platforms may enable studying the complex immune-cancer interactions and engineering MAIT cells for cell-based cancer immunotherapies.
Collapse
Affiliation(s)
- Madhuri Dey
- Department of Chemistry, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA 16802, USA
| | - Myong Hwan Kim
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA 16802, USA
- Biomedical Engineering Department, Penn State University; University Park, PA 16802, USA
| | - Momoka Nagamine
- Department of Chemistry, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA 16802, USA
| | - Ece Karhan
- The Jackson Laboratory for Genomic Medicine; Farmington, CT 06032, USA
| | - Lina Kozhaya
- The Jackson Laboratory for Genomic Medicine; Farmington, CT 06032, USA
| | - Mikail Dogan
- The Jackson Laboratory for Genomic Medicine; Farmington, CT 06032, USA
| | - Derya Unutmaz
- The Jackson Laboratory for Genomic Medicine; Farmington, CT 06032, USA
- University of Connecticut Health Center; Farmington, CT 06032, USA
| | - Ibrahim T. Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA 16802, USA
- Biomedical Engineering Department, Penn State University; University Park, PA 16802, USA
- Engineering Science and Mechanics Department, Penn State University; University Park, PA 16802, USA
- Materials Research Institute, Penn State University; University Park, PA 16802, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA 17033, USA
- Penn State Cancer Institute, Penn State University, Hershey, PA 17033, USA
| |
Collapse
|
27
|
Even KM, Gaesser AM, Ciamillo SA, Linardi RL, Ortved KF. Comparing the immunomodulatory properties of equine BM-MSCs culture expanded in autologous platelet lysate, pooled platelet lysate, equine serum and fetal bovine serum supplemented culture media. Front Vet Sci 2022; 9:958724. [PMID: 36090170 PMCID: PMC9453159 DOI: 10.3389/fvets.2022.958724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Joint injury often leads to cartilage damage and posttraumatic inflammation, which drives continued extracellular matrix degradation culminating in osteoarthritis. Mesenchymal stem cells (MSCs) have been proposed as a biotherapeutic to modulate inflammation within the joint. However, concerns have been raised regarding the immunogenicity of MSCs cultured in traditional fetal bovine serum (FBS) containing media, and the potential of xenogenic antigens to activate the immune system causing rejection and destruction of the MSCs. Xenogen-free alternatives to FBS have been proposed to decrease MSC immunogenicity, including platelet lysate (PL) and equine serum. The objective of this study was to compare the immunomodulatory properties of BM-MSCs culture-expanded in media supplemented with autologous PL (APL), pooled PL (PPL), equine serum (ES) or FBS. We hypothesized that BM-MSCs culture expanded in media with xenogen-free supplements would exhibit superior immunomodulatory properties to those cultured in FBS containing media. Bone marrow-derived MSCs (BM-MSCs) were isolated from six horses and culture expanded in each media type. Blood was collected from each horse to isolate platelet lysate. The immunomodulatory function of the BM-MSCs was assessed via a T cell proliferation assay and through multiplex immunoassay quantification of cytokines, including IL-1β, IL-6, IL-8, IL-10, and TNFα, following preconditioning of BM-MSCs with IL-1β. The concentration of platelet-derived growth factor BB (PDGF-BB), IL-10, and transforming growth factor-β (TGF-β) in each media was measured via immunoassay. BM-MSCs cultured in ES resulted in significant suppression of T cell proliferation (p = 0.02). Cell culture supernatant from preconditioned BM-MSCs cultured in ES had significantly higher levels of IL-6. PDGF-BB was significantly higher in APL media compared to FBS media (p = 0.016), while IL-10 was significantly higher in PPL media than ES and FBS (p = 0.04). TGF-β was highest in APL media, with a significant difference in comparison to ES media (p = 0.03). In conclusion, expansion of equine BM-MSCs in ES may enhance their immunomodulatory abilities, while PL containing media may have some inherent therapeutic potential associated with higher concentrations of growth factors. Further studies are needed to elucidate which xenogen-free supplement optimizes BM-MSC performance.
Collapse
Affiliation(s)
| | | | | | | | - Kyla F. Ortved
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, United States
| |
Collapse
|
28
|
Gonzaga VF, Wenceslau CV, Vieira DP, Policiquio BDO, Khalil C, Araldi RP, Kerkis I. Therapeutic Potential of Human Immature Dental Pulp Stem Cells Observed in Mouse Model for Acquired Aplastic Anemia. Cells 2022; 11:2252. [PMID: 35883695 PMCID: PMC9315564 DOI: 10.3390/cells11142252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/10/2022] [Accepted: 06/23/2022] [Indexed: 11/17/2022] Open
Abstract
Aplastic anemia (AA) is a rare and serious disorder of hematopoietic stem cells (HSCs) that results in the loss of blood cells due to the failure of the bone marrow (BM). Although BM transplantation is used to treat AA, its use is limited by donor availability. In this sense, mesenchymal stem cells (MSCs) can offer a novel therapeutic approach for AA. This is because the MSCs contribute to the hematopoietic niche organization through their repopulating. In our study, we used the human immature dental pulp stem cell (hIDPSC), an MSC-like cell, to explore an alternative therapeutic approach for AA. For this, isogenic C57BL/6 mice were exposed to total body irradiation (TBI) to induce the AA. After 48 h of TBI, the mice were intraperitoneally treated with hIDPSC. The immunohistochemistry analyses confirmed that the hIDPSCs migrated and grafted in the mouse bone marrow (BM) and spleen, providing rapid support to hematopoiesis recovery compared to the group exposed to radiation, but not to those treated with the cells as well as the hematological parameters. Six months after the last hIDPSC transplantation, the BM showed long-term stable hematopoiesis. Our data highlight the therapeutic plasticity and hematoprotective role of hIDPSC for AA and potentially for other hematopoietic failures.
Collapse
Affiliation(s)
- Vivian Fonseca Gonzaga
- Genetics Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil; (V.F.G.); (B.d.O.P.); (R.P.A.)
- Cellavita Pesquisas Científicas Ltda., Valinhos 13271-650, Brazil;
| | | | | | - Bruna de Oliveira Policiquio
- Genetics Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil; (V.F.G.); (B.d.O.P.); (R.P.A.)
- Cellavita Pesquisas Científicas Ltda., Valinhos 13271-650, Brazil;
| | - Charbel Khalil
- Reviva Stem Cell Platform for Research and Applications Center, Bsalim 17-5208, Lebanon;
- Faculty of Pharmacy, Saint Joseph University, Beirut 17-5208, Lebanon
| | - Rodrigo Pinheiro Araldi
- Genetics Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil; (V.F.G.); (B.d.O.P.); (R.P.A.)
- Cellavita Pesquisas Científicas Ltda., Valinhos 13271-650, Brazil;
| | - Irina Kerkis
- Genetics Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil; (V.F.G.); (B.d.O.P.); (R.P.A.)
| |
Collapse
|
29
|
Human umbilical cord blood-derived MSCs trans-differentiate into endometrial cells and regulate Th17/Treg balance through NF-κB signaling in rabbit intrauterine adhesions endometrium. Stem Cell Res Ther 2022; 13:301. [PMID: 35841027 PMCID: PMC9284747 DOI: 10.1186/s13287-022-02990-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/21/2022] [Indexed: 12/03/2022] Open
Abstract
Purpose The fundamental cause of intrauterine adhesions (IUAs) is the destruction and reduction in stem cells in endometrial basal layer, resulting in endometrial reconstruction very difficult. The purpose of this study was to investigate the effects and underlying mechanism of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) on the endometrial reconstruction after transplantation. Methods hUCB-MSCs were isolated and identified by flow cytometry, osteogenic, adipogenic and chondrogenic differentiation assays. The rabbit IUA models were established and set five groups (control, 14/28th day after surgery, estrogen and hUCB-MSCs treatment). The number of endometrial glands and the fibrosis rate were evaluated using HE and Masson staining, respectively. Endometrial proliferation, angiogenesis and inflammation were evaluated by immunohistochemical staining of ER, Ki-67and TGF-β1, respectively. Single-cell RNA sequencing (scRNA-seq) was applied to explore the cell differentiation trajectory after hUCB-MSCs transplanted into IUA endometrium. Finally, molecular mechanism of hUCB-MSCs repairing damaged endometrium was investigated by RNA sequencing, qRT-PCR and Western blot assays. Results After transplantation of the hUCB-MSCs, the increase in endometrial gland number, estrogen receptor (ER) and Ki-67 expression, and the decrease in fibrosis rate and TGF-β expression (P < 0.05), suggested the endometrial repair, angiogenesis and inflammatory suppression. The therapeutic effect of hUCB-MSCs was significantly improved compared with 28th day after surgery and estrogen group. ScRNA-seq demonstrated that the transplanted hUCB-MSCs can trans-differentiate into endometrial cells: epithelial, fibroblast and macrophage. RNA sequencing of six IUA samples combined with qRT-PCR and Western blot assays further revealed that hUCB-MSCs may regulate Th17/Treg balance through NF-κB signaling, thus inhibiting the immune response of damaged endometrium. Conclusions Our study demonstrated that hUCB-MSCs can repair damaged endometrium through trans-differentiation, immunomodulatory capacities and NF-κB signaling, suggesting the treatment value of hUCB-MSCs in IUA. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02990-1.
Collapse
|
30
|
Chouw A, Sartika CR, Milanda T, Faried A. Interleukins Profiling in Umbilical Cord Mesenchymal Stem Cell-Derived Secretome. Stem Cells Cloning 2022; 15:1-9. [PMID: 35444427 PMCID: PMC9015105 DOI: 10.2147/sccaa.s356763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/01/2022] [Indexed: 11/26/2022] Open
Abstract
Background Umbilical cord mesenchymal stem cells (UC-MSCs)-derived secretome is currently used in regenerative therapy. MSCs are believed to secrete a wide spectrum of bioactive molecules which give paracrine effects in immunomodulation and regenerative capacities. One group that was found in secretome is interleukins (ILs), a cytokine that plays an essential role in the process of proliferation, differentiation, maturation, migration, and adhesion of immune cells. However, as there are many types of ILs, the profile of ILs in the UC-MSCs-derived secretome has been limitedly reported. Therefore, in this study, we would like to profile and detect the interleukin concentration secreted by UC-MSCs. Methods UC-MSCs-derived secretome was collected from UC-MSCs passage 5 after 24- and 48-hour incubation (n=9). Secretome was filtered using 0.2 µm and stored at -80°C for further detection. All samples were normalized before the interleukin (IL-2, IL-4, IL-6, IL-9, IL-10, IL-12, IL-17A) detection using a MACSPlex Cytokine Kit. Results The IL-6 has the highest concentration among other interleukins in both groups and increases significantly (p<0.003) after incubation for 48 hours. The pro-inflammatory factors are decreasing while anti-inflammatory factors are increasing after 48-hour incubation. Discussion Our studies show that the UC-MSCs secrete pro- and anti-inflammatory interleukins. The concentration of anti-inflammatory interleukins shows to be increasing, while the pro-inflammatory interleukins are decreasing within the longer incubation time, but this not be applicable for IL-10 and IL-6. IL-6 has the highest concentration among other ILs. These results may provide important clues regarding when is the right time for secretome to be used in therapy patients, because all the molecules in the secretome can lead to many clinical manifestations.
Collapse
Affiliation(s)
- Angliana Chouw
- Doctoral Program, Faculty of Pharmacy Universitas Padjadjaran, Jatinangor, West Java, Indonesia
- Research and Development, Prodia StemCell Indonesia, Jakarta, DKI, Jakarta, Indonesia
| | - Cynthia Retna Sartika
- Research and Development, Prodia StemCell Indonesia, Jakarta, DKI, Jakarta, Indonesia
| | - Tiana Milanda
- Department of Biological Pharmacy, Faculty of Pharmacy Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Ahmad Faried
- Stem Cell Working Group, Faculty of Medicine Universitas Padjadjaran, Dr. Hasan Sadikin Hospital, Bandung, West Java, Indonesia
| |
Collapse
|
31
|
Mallis P, Chatzistamatiou T, Dimou Z, Sarri EF, Georgiou E, Salagianni M, Triantafyllia V, Andreakos E, Stavropoulos-Giokas C, Michalopoulos E. Mesenchymal stromal cell delivery as a potential therapeutic strategy against COVID-19: Promising evidence from in vitro results. World J Biol Chem 2022; 13:47-65. [PMID: 35432769 PMCID: PMC8966500 DOI: 10.4331/wjbc.v13.i2.47] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/28/2021] [Accepted: 03/06/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19) pandemic, which was initiated in December 2019. COVID-19 is characterized by a low mortality rate (< 6%); however, this percentage is higher in elderly people and patients with underlying disorders. COVID-19 is characterized by mild to severe outcomes. Currently, several therapeutic strategies are evaluated, such as the use of anti-viral drugs, prophylactic treatment, monoclonal antibodies, and vaccination. Advanced cellular therapies are also investigated, thus representing an additional therapeutic tool for clinicians. Mesenchymal stromal cells (MSCs), which are known for their immunoregulatory properties, may halt the induced cytokine release syndrome mediated by SARS-CoV-2, and can be considered as a potential stem cell therapy.
AIM To evaluate the immunoregulatory properties of MSCs, upon stimulation with COVID-19 patient serum.
METHODS MSCs derived from the human Wharton’s Jelly (WJ) tissue and bone marrow (BM) were isolated, cryopreserved, expanded, and defined according to the criteria outlined by the International Society for Cellular Therapies. Then, WJ and BM-MSCs were stimulated with a culture medium containing 15% COVID-19 patient serum, 1% penicillin-streptomycin, and 1% L-glutamine for 48 h. The quantification of interleukin (IL)-1 receptor a (Ra), IL-6, IL-10, IL-13, transforming growth factor (TGF)-β1, vascular endothelial growth factor (VEGF)-a, fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and indoleamine-2,3-dioxygenase (IDO) was performed using commercial ELISA kits. The expression of HLA-G1, G5, and G7 was evaluated in unstimulated and stimulated WJ and BM-MSCs. Finally, the interactions between MSCs and patients’ macrophages were established using co-culture experiments.
RESULTS Thawed WJ and BM-MSCs exhibited a spindle-shaped morphology, successfully differentiated to “osteocytes”, “adipocytes”, and “chondrocytes”, and in flow cytometric analysis were characterized by positivity for CD73, CD90, and CD105 (> 95%) and negativity for CD34, CD45, and HLA-DR (< 2%). Moreover, stimulated WJ and BM-MSCs were characterized by increased cytoplasmic granulation, in comparison to unstimulated cells. The HLA-G isoforms (G1, G5, and G7) were successfully expressed by the unstimulated and stimulated WJ-MSCs. On the other hand, only weak expression of HLA-G1 was identified in BM-MSCs. Stimulated MSCs secreted high levels of IL-1Ra, IL-6, IL-10, IL-13, TGF-β1, FGF, VEGF, PDGF, and IDO in comparison to unstimulated cells (P < 0.05) after 12 and 24 h. Finally, macrophages derived from COVID-19 patients successfully adapted the M2 phenotype after co-culturing with stimulated WJ and BM-MSCs.
CONCLUSION WJ and BM-MSCs successfully produced high levels of immunoregulatory agents, which may efficiently modulate the over-activated immune responses of critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | | | - Zetta Dimou
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Eirini-Faidra Sarri
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Eleni Georgiou
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Maria Salagianni
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Vasiliki Triantafyllia
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | | | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| |
Collapse
|
32
|
Horcharoensuk P, Yang-en S, Narkwichean A, Rungsiwiwut R. Proline-based solution maintains cell viability and stemness of canine adipose-derived mesenchymal stem cells after hypothermic storage. PLoS One 2022; 17:e0264773. [PMID: 35231072 PMCID: PMC8887718 DOI: 10.1371/journal.pone.0264773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/17/2022] [Indexed: 11/18/2022] Open
Abstract
Transportation of mesenchymal stem cells (MSCs) under hypothermic conditions in 0.9% normal saline solution (NSS) might increase cell death and alter the stemness of MSCs. The present study aimed to evaluate the effect of proline-based solution (PL-BS) on cell viability and the stemness of newly established canine adipose-derived mesenchymal stem cells (cAD-MSCs) under hypothermic conditions. Characterized cAD-MSCs were stored in 1, 10, and 100 mM PL-BS or NSS at 4°C for 6, 9, and 12 hours prior to an evaluation. The results demonstrated that storage in 1 mM PL-BS for 6 hours decreased cell apoptosis and proliferation ability, but improved cell viability and mitochondrial membrane potential. cAD-MSCs maintained their high expression of CD44 and CD90, but had a low expression of CD34 and MHC class II. Trilineage differentiation ability of cAD-MSCs was not affected by storage in 1 mM PL-BS. Gene expression analysis demonstrated that immunomodulatory genes, including IDO, HGF, PGE-2, and IL-6, were upregulated in cAD-MSCs stored in 1 mM PL-BS. In conclusion, PL-BS can be effectively applied for storing cAD-MSCs under hypothermic conditions. These findings provide a new solution for effective handling of cAD-MSCs which might be promising for clinical applications.
Collapse
Affiliation(s)
| | - Sunantha Yang-en
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | - Amarin Narkwichean
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | - Ruttachuk Rungsiwiwut
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
33
|
Abbasi A, Imaichi S, Ling V, Shukla A. Mesenchymal Stem Cell Behavior on Soft Hydrogels with Aligned Surface Topographies. ACS APPLIED BIO MATERIALS 2022; 5:1890-1900. [PMID: 35199983 DOI: 10.1021/acsabm.1c01260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human mesenchymal stem cells (HMSCs) are important for cell-based therapies. However, the success of HMSC therapy requires large-scale in vitro expansion of these multipotent cells. The traditional expansion of HMSCs on tissue-culture-treated stiff polystyrene induces significant changes in their shape, multipotency, and secretome, leading to early senescence and subdued paracrine activity. To enhance their therapeutic potential, here, we have developed two-dimensional soft hydrogels with imprinted microscale aligned grooves for use as HMSC culture substrates. We showed that, depending on the dimensions of the topographical features, these substrates led to lower cellular spreading and cytoskeletal tension, maintaining multipotency and osteogenic and adipogenic differentiate potential, while lowering cellular senescence. We also observed a greater capacity of HMSCs to produce anti-inflammatory cytokines after short-term priming on these hydrogel substrates. Overall, these soft hydrogels with unique surface topography have shown great promise as in vitro culture substrates to maximize the therapeutic potential of HMSCs.
Collapse
Affiliation(s)
- Akram Abbasi
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Sachiko Imaichi
- Takeda Pharmaceuticals, Cambridge, Massachusetts 02139, United States
| | - Vincent Ling
- Takeda Pharmaceuticals, Cambridge, Massachusetts 02139, United States
| | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
34
|
Floriano JF, Emanueli C, Vega S, Barbosa AMP, Oliveira RGD, Floriano EAF, Graeff CFDO, Abbade JF, Herculano RD, Sobrevia L, Rudge MVC. Pro-angiogenic approach for skeletal muscle regeneration. Biochim Biophys Acta Gen Subj 2022; 1866:130059. [PMID: 34793875 DOI: 10.1016/j.bbagen.2021.130059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
The angiogenesis process is a phenomenon in which numerous molecules participate in the stimulation of the new vessels' formation from pre-existing vessels. Angiogenesis is a crucial step in tissue regeneration and recovery of organ and tissue function. Muscle diseases affect millions of people worldwide overcome the ability of skeletal muscle to self-repair. Pro-angiogenic therapies are key in skeletal muscle regeneration where both myogenesis and angiogenesis occur. These therapies have been based on mesenchymal stem cells (MSCs), exosomes, microRNAs (miRs) and delivery of biological factors. The use of different calls of biomaterials is another approach, including ceramics, composites, and polymers. Natural polymers are use due its bioactivity and biocompatibility in addition to its use as scaffolds and in drug delivery systems. One of these polymers is the natural rubber latex (NRL) which is biocompatible, bioactive, versatile, low-costing, and capable of promoting tissue regeneration and angiogenesis. In this review, the advances in the field of pro-angiogenic therapies are discussed.
Collapse
Affiliation(s)
- Juliana Ferreira Floriano
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sofia Vega
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | | | | | | | | | - Joelcio Francisco Abbade
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil
| | | | - Luis Sobrevia
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland, Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD, 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands.
| | | |
Collapse
|
35
|
A Molecular Analysis of Cytokine Content across Extracellular Vesicles, Secretions, and Intracellular Space from Different Site-Specific Adipose-Derived Stem Cells. Int J Mol Sci 2021; 23:ijms23010397. [PMID: 35008824 PMCID: PMC8745205 DOI: 10.3390/ijms23010397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Cytokines are multifunctional small proteins that have a vital influence on inflammatory states of tissues and play a role in signalling and cellular control mechanisms. Cytokine expression has primarily been viewed as a form of direct secretion of molecules through an active transportation; however, other forms of active transport such as extracellular vesicles are at play. This is particularly important in stem cells where signalling molecules are key to communication managing the levels of proliferation, migration, and differentiation into mature cells. This study investigated cytokines from intracellular content, direct cellular secretions, and extracellular vesicles from adult adipose-derived stem cells isolated from three distinct anatomical locations: abdomen, thigh, and chin. The cells were cultured investigated using live cell microscopy, cytokine assays, and bioinformatics analysis. The cytokines quantified and examined from each sample type showed a distinct difference between niche areas and sample types. The varying levels of TNF-alpha, IL-6 and IL-8 cytokines were shown to play a crucial role in signalling pathways such as MAPK, ERK1/2 and JAK-STAT in cells. On the other hand, the chemotactic cytokines IL-1rn, Eotaxin, IP-10 and MCP-1 showed the most prominent changes across extracellular vesicles with roles in noncanonical signalling. By examining the local and tangential roles of cytokines in stem cells, their roles in signalling and in regenerative mechanisms may be further understood.
Collapse
|
36
|
Özdemir AT, Nalbantsoy A, Özgül Özdemir RB, Berdeli A. Effects of 15-Lipoxygenase Overexpressing Adipose Tissue Mesenchymal Stem Cells on The Th17 / Treg Plasticity. Prostaglandins Other Lipid Mediat 2021; 159:106610. [PMID: 34963632 DOI: 10.1016/j.prostaglandins.2021.106610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022]
Abstract
15-lipoxygenase (15-LOX) is a critical enzyme that allows the direction of arachidonic acid metabolism to change from inflammation into the resolution. This study aims to reveal how the immunomodulation properties of mesenchymal stem cells (MSC) alter by the 15-LOX overexpression. For this purpose, peripheral blood mononuclear cells (PBMCs) isolated from seven healthy volunteers, and both MSCs and 15-LOX overexpressing MSCs (15-LOXMSCs) were co-cultured at different cell ratios (1/1, 1/5 and 1/10). Alterations of CD4+Tbet+, CD4+Gata3+, CD4+RoRC2+, and CD4+FoxP3+ lymphocyte frequencies were detected by flow cytometry, and IFN-γ, IL-4, IL-6, IL-10, IL-17a, TGF-β and LXA4 levels of medium supernatants were measured by ELISA method. According to our findings, MSC and 15-LOXMSCs have a suppressive effect on PHA activated PBMCs. However, as the ratio of PBMCs increased, the effects of 15-LOXMSCs increased significantly, while the effects of MSCs decreased. The most notable effect of the 15-LOX modification was the significant reduction in IL-6, IL-10 and IL-17a expression and the accompanying increase in TGF-β and LXA4 levels. We also observed a similar situation between CD4+RoRC2+ and CD4+FoxP3+ cell frequencies. These data suggested that the effects of MSCs on the balance of Th17 / Treg could change by the 15-LOX overexpression, and this might be in favor of the Treg cells.
Collapse
Affiliation(s)
- Alper Tunga Özdemir
- Ege University, Institute of Health Sciences, Department of Stem Cell, Izmir, Turkey.
| | - Ayşe Nalbantsoy
- Manisa City Hospital, Allergy and Clinical Immunology Clinic, Manisa, Turkey
| | | | - Afig Berdeli
- Ege University, Institute of Health Sciences, Department of Stem Cell, Izmir, Turkey
| |
Collapse
|
37
|
Relevance of Cellular Redox Homeostasis for Vital Functions of Human Dental Pulp Cells. Antioxidants (Basel) 2021; 11:antiox11010023. [PMID: 35052527 PMCID: PMC8772760 DOI: 10.3390/antiox11010023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 01/22/2023] Open
Abstract
Odontogenic MSCs are vulnerable to LPS-triggered bacterial infections, and they respond by secreting inflammatory mediators, such as IL-6, and with mineralization. Since both processes might be prone to a disturbance of the redox homeostasis, the oxidative stress influence on vital functions of human dental pulp cells (HPCs) was investigated. With these aims, a model of LPS-stimulated primary HPCs was established, and anti- and pro-oxidant substances were administered up to 21 days to measure inflammation and mineralization parameters. LPS-stimulated HPCs retained mineralization potential, which was decreased with the antioxidants NAC and fisetin and the pro-oxidant BSO. The expression of surface markers related to odontogenic commitment was influenced accordingly but counteracted by the enhanced expression of BMP2 and ALP at the transcriptional level. LPS triggers an early IL-6 production in non-odontogenic conditions, while it can be measured only after 15 days in the presence of the differentiation medium. The present study shows that HPCs functions causally depend on a tightly regulated cellular redox balance. Our data demonstrate a redox control of pulp MSC odontogenic commitment along with a potential association between an IL-6 late secretion and mineralization. These findings lay the groundwork for investigations on the molecular role of IL-6 in dental hard tissue metabolism.
Collapse
|
38
|
Valsecchi C, Croce S, Maltese A, Montagna L, Lenta E, Nevone A, Girelli M, Milani P, Bosoni T, Massa M, Abbà C, Campanelli R, Ripepi J, De Silvestri A, Carolei A, Palladini G, Zecca M, Nuvolone M, Avanzini MA. Bone Marrow Microenvironment in Light-Chain Amyloidosis: In Vitro Expansion and Characterization of Mesenchymal Stromal Cells. Biomedicines 2021; 9:biomedicines9111523. [PMID: 34829752 PMCID: PMC8614719 DOI: 10.3390/biomedicines9111523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022] Open
Abstract
Immunoglobulin light-chain amyloidosis (AL) is caused by misfolded light chains produced by a small B cell clone. Mesenchymal stromal cells (MSCs) have been reported to affect plasma cell behavior. We aimed to characterize bone marrow (BM)-MSCs from AL patients, considering functional aspects, such as proliferation, differentiation, and immunomodulatory capacities. MSCs were in vitro expanded from the BM of 57 AL patients and 14 healthy donors (HDs). MSC surface markers were analyzed by flow cytometry, osteogenic and adipogenic differentiation capacities were in vitro evaluated, and co-culture experiments were performed in order to investigate MSC immunomodulatory properties towards the ALMC-2 cell line and HD peripheral blood mononuclear cells (PBMCs). AL-MSCs were comparable to HD-MSCs for morphology, immune-phenotype, and differentiation capacities. AL-MSCs showed a reduced proliferation rate, entering senescence at earlier passages than HD-MSCs. The AL-MSC modulatory effect on the plasma-cell line or circulating plasma cells was comparable to that of HD-MSCs. To our knowledge, this is the first study providing a comprehensive characterization of AL-MSCs. It remains to be defined if the observed abnormalities are the consequence of or are involved in the disease pathogenesis. BM microenvironment components in AL may represent the targets for the prevention/treatment of the disease in personalized therapies.
Collapse
Affiliation(s)
- Chiara Valsecchi
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
| | - Stefania Croce
- General Surgery Department, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Alice Maltese
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
| | - Lorenza Montagna
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Elisa Lenta
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
| | - Alice Nevone
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maria Girelli
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Paolo Milani
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Tiziana Bosoni
- Clinical Chemistry Laboratory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Margherita Massa
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Carlotta Abbà
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Rita Campanelli
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Jessica Ripepi
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Annalisa De Silvestri
- Clinical Epidemiology and Biometry Unit, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Adriana Carolei
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Giovanni Palladini
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Marco Zecca
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
| | - Mario Nuvolone
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maria Antonietta Avanzini
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
- Correspondence: ; Tel.: +39-0382-502715
| |
Collapse
|
39
|
Maughon TS, Shen X, Huang D, Michael AOA, Shockey WA, Andrews SH, McRae JM, Platt MO, Fernández FM, Edison AS, Stice SL, Marklein RA. Metabolomics and cytokine profiling of mesenchymal stromal cells identify markers predictive of T-cell suppression. Cytotherapy 2021; 24:137-148. [PMID: 34696960 DOI: 10.1016/j.jcyt.2021.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/02/2021] [Accepted: 08/17/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) have shown great promise in the field of regenerative medicine, as many studies have shown that MSCs possess immunomodulatory function. Despite this promise, no MSC therapies have been licensed by the Food and Drug Administration. This lack of successful clinical translation is due in part to MSC heterogeneity and a lack of critical quality attributes. Although MSC indoleamine 2,3-dioxygnease (IDO) activity has been shown to correlate with MSC function, multiple predictive markers may be needed to better predict MSC function. METHODS Three MSC lines (two bone marrow-derived, one induced pluripotent stem cell-derived) were expanded to three passages. At the time of harvest for each passage, cell pellets were collected for nuclear magnetic resonance (NMR) and ultra-performance liquid chromatography mass spectrometry (MS), and media were collected for cytokine profiling. Harvested cells were also cryopreserved for assessing function using T-cell proliferation and IDO activity assays. Linear regression was performed on functional data against NMR, MS and cytokines to reduce the number of important features, and partial least squares regression (PLSR) was used to obtain predictive markers of T-cell suppression based on variable importance in projection scores. RESULTS Significant functional heterogeneity (in terms of T-cell suppression and IDO activity) was observed between the three MSC lines, as were donor-dependent differences based on passage. Omics characterization revealed distinct differences between cell lines using principal component analysis. Cell lines separated along principal component one based on tissue source (bone marrow-derived versus induced pluripotent stem cell-derived) for NMR, MS and cytokine profiles. PLSR modeling of important features predicted MSC functional capacity with NMR (R2 = 0.86), MS (R2 = 0.83), cytokines (R2 = 0.70) and a combination of all features (R2 = 0.88). CONCLUSIONS The work described here provides a platform for identifying markers for predicting MSC functional capacity using PLSR modeling that could be used as release criteria and guide future manufacturing strategies for MSCs and other cell therapies.
Collapse
Affiliation(s)
- Ty S Maughon
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia, USA; Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Xunan Shen
- Complex Carbohydrate Research Center and Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| | - Danning Huang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Adeola O Adebayo Michael
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - W Andrew Shockey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Seth H Andrews
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia, USA; Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Jon M McRae
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Manu O Platt
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Arthur S Edison
- Complex Carbohydrate Research Center and Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| | - Steven L Stice
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA; Department of Animal and Dairy Sciences, University of Georgia, Athens, Georgia, USA.
| | - Ross A Marklein
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia, USA; Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
40
|
Li A, Guo F, Pan Q, Chen S, Chen J, Liu HF, Pan Q. Mesenchymal Stem Cell Therapy: Hope for Patients With Systemic Lupus Erythematosus. Front Immunol 2021; 12:728190. [PMID: 34659214 PMCID: PMC8516390 DOI: 10.3389/fimmu.2021.728190] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. Although previous studies have demonstrated that SLE is related to the imbalance of cells in the immune system, including B cells, T cells, and dendritic cells, etc., the mechanisms underlying SLE pathogenesis remain unclear. Therefore, effective and low side-effect therapies for SLE are lacking. Recently, mesenchymal stem cell (MSC) therapy for autoimmune diseases, particularly SLE, has gained increasing attention. This therapy can improve the signs and symptoms of refractory SLE by promoting the proliferation of Th2 and Treg cells and inhibiting the activity of Th1, Th17, and B cells, etc. However, MSC therapy is also reported ineffective in some patients with SLE, which may be related to MSC- or patient-derived factors. Therefore, the therapeutic effects of MSCs should be further confirmed. This review summarizes the status of MSC therapy in refractory SLE treatment and potential reasons for the ineffectiveness of MSC therapy from three perspectives. We propose various MSC modification methods that may be beneficial in enhancing the immunosuppression of MSCs in SLE. However, their safety and protective effects in patients with SLE still need to be confirmed by further experimental and clinical evidence.
Collapse
Affiliation(s)
- Aifen Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Quanren Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuxian Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaxuan Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
41
|
TNF-α and IFN-γ Participate in Improving the Immunoregulatory Capacity of Mesenchymal Stem/Stromal Cells: Importance of Cell-Cell Contact and Extracellular Vesicles. Int J Mol Sci 2021; 22:ijms22179531. [PMID: 34502453 PMCID: PMC8431422 DOI: 10.3390/ijms22179531] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) have an immunoregulatory capacity and have been used in different clinical protocols requiring control of the immune response. However, variable results have been obtained, mainly due to the effect of the microenvironment on the induction, increase, and maintenance of MSC immunoregulatory mechanisms. In addition, the importance of cell–cell contact for MSCs to efficiently modulate the immune response has recently been highlighted. Because these interactions would be difficult to achieve in the physiological context, the release of extracellular vesicles (EVs) and their participation as intermediaries of communication between MSCs and immune cells becomes relevant. Therefore, this article focuses on analyzing immunoregulatory mechanisms mediated by cell contact, highlighting the importance of intercellular adhesion molecule-1 (ICAM-1) and the participation of EVs. Moreover, the effects of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ), the main cytokines involved in MSC activation, are examined. These cytokines, when used at the appropriate concentrations and times, would promote increases in the expression of immunoregulatory molecules in the cell and allow the acquisition of EVs enriched with these molecules. The establishment of certain in vitro activation guidelines will facilitate the design of conditioning protocols to obtain functional MSCs or EVs in different pathophysiological conditions.
Collapse
|
42
|
The Impact of Sedentary Lifestyle, High-fat Diet, Tobacco Smoke, and Alcohol Intake on the Hematopoietic Stem Cell Niches. Hemasphere 2021; 5:e615. [PMID: 34291194 PMCID: PMC8288907 DOI: 10.1097/hs9.0000000000000615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/07/2021] [Indexed: 11/25/2022] Open
Abstract
Hematopoietic stem and progenitor cells maintain hematopoiesis throughout life by generating all major blood cell lineages through the process of self-renewal and differentiation. In adult mammals, hematopoietic stem cells (HSCs) primarily reside in the bone marrow (BM) at special microenvironments called “niches.” Niches are thought to extrinsically orchestrate the HSC fate including their quiescence and proliferation. Insight into the HSC niches mainly comes from studies in mice using surface marker identification and imaging to visualize HSC localization and association with niche cells. The advantage of mouse models is the possibility to study the 3-dimensional BM architecture and cell interactions in an intact traceable system. However, this may not be directly translational to human BM. Sedentary lifestyle, unhealthy diet, excessive alcohol intake, and smoking are all known risk factors for various diseases including hematological disorders and cancer, but how do lifestyle factors impact hematopoiesis and the associated niches? Here, we review current knowledge about the HSC niches and how unhealthy lifestyle may affect it. In addition, we summarize epidemiological data concerning the influence of lifestyle factors on hematological disorders and malignancies.
Collapse
|
43
|
Fundamental changes in endogenous bone marrow mesenchymal stromal cells during Type I Diabetes is a pre-neuropathy event. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166187. [PMID: 34102256 DOI: 10.1016/j.bbadis.2021.166187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022]
Abstract
Deficiency of angiogenic and neurotrophic factors under long term diabetes is known to lead to Schwann cell degeneration, clinically manifested as Diabetic Neuropathy (DN). While the transplantation of exogenous allogenic Mesenchymal Stromal Cells (MSCs) has shown amelioration of DN through paracrine action, it is not known what functional changes occur in endogenous bone-marrow MSCs under chronic diabetes in terms of homing, migration and/or paracrine signalling with reference to the end-point clinical manifestation of Diabetic Neuropathy. We thus aimed at determining the changes in BM-MSCs under Type 1 Diabetes with respect to survival, self-renewal, oxidative status, paracrine activity, intracellular Ca2+ response and migration in response to pathological cytokine/chemokine, in reference to the time-point of decline in Nerve Conduction Velocity (NCV) in a rat model. Within one week of diabetes induction, BM-MSCs underwent apoptosis, and compromised their self-renewal capacity, antioxidant defence mechanism and migration toward cytokine/chemokine; whereas epineurial blood vessel thickening and demyelination resulting in NCV decline were observed only after three weeks. By two- and three-weeks post diabetes induction, BM-MSC apoptosis reduced and proliferative ability was restored; however, their self-renewal, migration and intracellular Ca2+ response toward pathological cytokine/chemokine remained impaired. These results indicate that T1D induced intrinsic functional impairments in endogenous BM-MSCs occur before neuropathy onset. This timeline of functional alterations in BM-MSCs also suggest that treatment strategies that target the bone marrow niche early on may help to modulate BM-MSC functional impairments and thus slow down the progression of neuropathy.
Collapse
|