1
|
Mitra S, Tati V, Das P, Joseph J, Bagga B, Shukla S. Mesenchymal stem cell-based adjunctive therapy for Pseudomonas aeruginosa-induced keratitis: A proof-of-concept in-vitro study. Exp Eye Res 2024; 242:109863. [PMID: 38494102 DOI: 10.1016/j.exer.2024.109863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/25/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
PURPOSE Pseudomonas aeruginosa-induced keratitis is one of the most severe and challenging forms of corneal infection, owing to its associated intense inflammatory reactions leading to corneal necrosis and dense corneal scar with loss of vision. Since mesenchymal stem cells (MSCs) are reported to possess antimicrobial and immunomodulatory properties, they can be tested as an adjuvant treatment along with the antibiotics which are the current standard of care. This study aims to investigate the anti-bacterial and immunomodulatory roles of human bone marrow MSC-derived conditioned medium (MSC-CM) in P. aeruginosa-infected human corneal epithelial cells (HCECs) in vitro. METHODS The effect of MSC-CM on the growth of clinical isolates of P. aeruginosa was evaluated by colony-forming unit assay. The expression of inflammatory cytokines (IL-6 and TNF-α) and an antimicrobial peptide (Lipocalin 2) in lipopolysaccharide-treated MSCs and HCECs was analyzed through ELISA. Corneal epithelial repair following infection with P. aeruginosa was studied through scratch assay. RESULTS Compared to control (P. aeruginosa (5*105) incubated in DMEM (1 ml) at 37 °C for 16 h), MSC-CM significantly: i) inhibits the growth of P. aeruginosa (159*109 vs. 104*109 CFU/ml), ii) accelerates corneal epithelial repair following infection with P. aeruginosa (9% vs. 24% closure of the wounded area after 12 h of infection), and iii) downregulates the lipopolysaccharide-induced expression of IL-6, TNF-α and Lipocalin 2 in HCECs. A combination of MSC-CM with an antibiotic, Ciprofloxacin moderately regulated the expression of IL-6, TNF-α, and Lipocalin 2. CONCLUSION MSC-CM holds promise as an adjunctive therapeutic approach for P. aeruginosa-induced corneal epithelial damage.
Collapse
Affiliation(s)
- Sreya Mitra
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L V Prasad Eye Institute, Hyderabad, 500034, India; Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, 500034, India
| | - Vasudeva Tati
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L V Prasad Eye Institute, Hyderabad, 500034, India; Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, 500034, India
| | - Prabhudatta Das
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L V Prasad Eye Institute, Hyderabad, 500034, India; Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, 500034, India
| | - Joveeta Joseph
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L V Prasad Eye Institute, Hyderabad, 500034, India; Jhaveri Microbiology Centre, L V Prasad Eye Institute, Hyderabad, 500034, India.
| | - Bhupesh Bagga
- The Ramoji Foundation Centre for Ocular Infections, L V Prasad Eye Institute, Hyderabad, 500034, India; Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, 500034, India.
| | - Sachin Shukla
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L V Prasad Eye Institute, Hyderabad, 500034, India; Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, 500034, India.
| |
Collapse
|
2
|
Chiu H, Chau Fang A, Chen YH, Koi RX, Yu KC, Hsieh LH, Shyu YM, Amer TA, Hsueh YJ, Tsao YT, Shen YJ, Wang YM, Chen HC, Lu YJ, Huang CC, Lu TT. Mechanistic and Kinetic Insights into Cellular Uptake of Biomimetic Dinitrosyl Iron Complexes and Intracellular Delivery of NO for Activation of Cytoprotective HO-1. JACS AU 2024; 4:1550-1569. [PMID: 38665642 PMCID: PMC11040670 DOI: 10.1021/jacsau.4c00064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024]
Abstract
Dinitrosyl iron unit (DNIU), [Fe(NO)2], is a natural metallocofactor for biological storage, delivery, and metabolism of nitric oxide (NO). In the attempt to gain a biomimetic insight into the natural DNIU under biological system, in this study, synthetic dinitrosyl iron complexes (DNICs) [(NO)2Fe(μ-SCH2CH2COOH)2Fe(NO)2] (DNIC-COOH) and [(NO)2Fe(μ-SCH2CH2COOCH3)2Fe(NO)2] (DNIC-COOMe) were employed to investigate the structure-reactivity relationship of mechanism and kinetics for cellular uptake of DNICs, intracellular delivery of NO, and activation of cytoprotective heme oxygenase (HO)-1. After rapid cellular uptake of dinuclear DNIC-COOMe through a thiol-mediated pathway (tmax = 0.5 h), intracellular assembly of mononuclear DNIC [(NO)2Fe(SR)(SCys)]n-/[(NO)2Fe(SR)(SCys-protein)]n- occurred, followed by O2-induced release of free NO (tmax = 1-2 h) or direct transfer of NO to soluble guanylate cyclase, which triggered the downstream HO-1. In contrast, steady kinetics for cellular uptake of DNIC-COOH via endocytosis (tmax = 2-8 h) and for intracellular release of NO (tmax = 4-6 h) reflected on the elevated activation of cytoprotective HO-1 (∼50-150-fold change at t = 3-10 h) and on the improved survival of DNIC-COOH-primed mesenchymal stem cell (MSC)/human corneal endothelial cell (HCEC) under stressed conditions. Consequently, this study unravels the bridging thiolate ligands in dinuclear DNIC-COOH/DNIC-COOMe as a switch to control the mechanism, kinetics, and efficacy for cellular uptake of DNICs, intracellular delivery of NO, and activation of cytoprotective HO-1, which poses an implication on enhanced survival of postengrafted MSC for advancing the MSC-based regenerative medicine.
Collapse
Affiliation(s)
- Han Chiu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Anyelina Chau Fang
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Yi-Hong Chen
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Ru Xin Koi
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Kai-Ching Yu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Li-Hung Hsieh
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Yueh-Ming Shyu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Tarik Abdelkareem
Mostafa Amer
- Department
of Biological Science and Technology, Institute of Molecular Medicine
and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yi-Jen Hsueh
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yu-Ting Tsao
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yang-Jin Shen
- College
of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department
of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yun-Ming Wang
- Department
of Biological Science and Technology, Institute of Molecular Medicine
and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Hung-Chi Chen
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- College
of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Yu-Jen Lu
- College
of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department
of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chieh-Cheng Huang
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Tsai-Te Lu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
- Department
of Chemistry, National Tsing Hua University, Hsinchu 30013 Taiwan
- Department
of Chemistry, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
3
|
Mehta KJ. Iron-Related Genes and Proteins in Mesenchymal Stem Cell Detection and Therapy. Stem Cell Rev Rep 2023; 19:1773-1784. [PMID: 37269528 PMCID: PMC10238768 DOI: 10.1007/s12015-023-10569-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 06/05/2023]
Abstract
Mesenchymal stem cells (MSCs) are located in various tissues of the body. These cells exhibit regenerative and reparative properties, which makes them highly valuable for cell-based therapy. Despite this, majority of MSC-related studies remain to be translated for regular clinical use. This is partly because there are methodical challenges in pre-administration MSC labelling, post-administration detection and tracking of cells, and in retention of maximal therapeutic potential in-vivo. This calls for exploration of alternative or adjunctive approaches that would enable better detection of transplanted MSCs via non-invasive methods and enhance MSC therapeutic potential in-vivo. Interestingly, these attributes have been demonstrated by some iron-related genes and proteins.Accordingly, this unique forward-looking article integrates the apparently distinct fields of iron metabolism and MSC biology, and reviews the utility of iron-related genes and iron-related proteins in facilitating MSC detection and therapy, respectively. Effects of genetic overexpression of the iron-related proteins ferritin, transferrin receptor-1 and MagA in MSCs and their utilisation as reporter genes for improving MSC detection in-vivo are critically evaluated. In addition, the beneficial effects of the iron chelator deferoxamine and the iron-related proteins haem oxygenase-1, lipocalin-2, lactoferrin, bone morphogenetic protein-2 and hepcidin in enhancing MSC therapeutics are highlighted with the consequent intracellular alterations in MSCs. This review aims to inform both regenerative and translational medicine. It can aid in formulating better methodical approaches that will improve, complement, or provide alternatives to the current pre-transplantation MSC labelling procedures, and enhance MSC detection or augment the post-transplantation MSC therapeutic potential.
Collapse
Affiliation(s)
- Kosha J Mehta
- Centre for Education, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
4
|
Zheng XQ, Lin JL, Huang J, Wu T, Song CL. Targeting aging with the healthy skeletal system: The endocrine role of bone. Rev Endocr Metab Disord 2023; 24:695-711. [PMID: 37402956 DOI: 10.1007/s11154-023-09812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 07/06/2023]
Abstract
Aging is an inevitable biological process, and longevity may be related to bone health. Maintaining strong bone health can extend one's lifespan, but the exact mechanism is unclear. Bone and extraosseous organs, including the heart and brain, have complex and precise communication mechanisms. In addition to its load bearing capacity, the skeletal system secretes cytokines, which play a role in bone regulation of extraosseous organs. FGF23, OCN, and LCN2 are three representative bone-derived cytokines involved in energy metabolism, endocrine homeostasis and systemic chronic inflammation levels. Today, advanced research methods provide new understandings of bone as a crucial endocrine organ. For example, gene editing technology enables bone-specific conditional gene knockout models, which allows the study of bone-derived cytokines to be more precise. We systematically evaluated the various effects of bone-derived cytokines on extraosseous organs and their possible antiaging mechanism. Targeting aging with the current knowledge of the healthy skeletal system is a potential therapeutic strategy. Therefore, we present a comprehensive review that summarizes the current knowledge and provides insights for futures studies.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jia-Liang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jie Huang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Tong Wu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China.
| |
Collapse
|
5
|
Yu C, Zhang F, Zhang L, Li J, Tang S, Li X, Peng M, Zhao Q, Zhu X. A bioinformatics approach to identifying the biomarkers and pathogenesis of major depressive disorder combined with acute myocardial infarction. Am J Transl Res 2023; 15:932-948. [PMID: 36915729 PMCID: PMC10006793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/17/2023] [Indexed: 03/16/2023]
Abstract
This study investigated the pathogenesis of major depressive disorder (MDD) and acute myocardial infarction (AMI) using bioinformatics. We analyzed MDD and AMI (MDD-AMI) datasets provided by the Gene Expression Omnibus (GEO) database for genes common to MDD and AMI using GEO2R and weighted gene co-expression network analysis (WGCNA). We also performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and we used Disease Ontology (DO) analysis to identify a) the pathways through which genes function and b) comorbidities. We also created a protein-protein interaction (PPI) network using the STRING database to identify the hub genes and biomarkers. NetworkAnalyst 3.0 was used to construct a transcription factor (TF) gene regulatory network. We also identified relevant complications and potential drug candidates. The 27 genes common to MDD and AMI were enriched in the pathways regulating TFs and mediating immunity and inflammation. The hub genes in the PPI network included TLR2, HP, ICAM1, LCN2, LTF, VCAN, S100A9 and NFKBIA. Key TFs were KLF9, KLF11, ZNF24, and ZNF580. Cardiovascular, pancreatic, and skeletal diseases were common complications. Hydrocortisone, simvastatin, and estradiol were candidate treatment drugs. Identification of these genes and their pathways may provide new targets for further research on the pathogenesis, biomarkers, and treatment of MDD-AMI. Together our results suggested that TLR2 and VCAN might be the key genes associated with MDD complicated by AMI.
Collapse
Affiliation(s)
- Cheng Yu
- Department of Traditional Chinese Medicine Classics, Shandong University of Traditional Chinese Medicine Affiliated HospitalJinan, Shandong, China
| | - Fengjun Zhang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese MedicineJinan, Shandong, China
| | - Lili Zhang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese MedicineJinan, Shandong, China
| | - Jiajing Li
- Department of Traditional Chinese Medicine Classics, Shandong University of Traditional Chinese Medicine Affiliated HospitalJinan, Shandong, China
| | - Saixue Tang
- First Clinical School of Medicine, Shandong University of Traditional Chinese MedicineJinan, Shandong, China
| | - Xuejun Li
- Department of Traditional Chinese Medicine Classics, Shandong University of Traditional Chinese Medicine Affiliated HospitalJinan, Shandong, China
| | - Min Peng
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
| | - Qiong Zhao
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
| | - Xiuli Zhu
- Department of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinan, Shandong, China
| |
Collapse
|
6
|
Dufrusine B, Valentinuzzi S, Bibbò S, Damiani V, Lanuti P, Pieragostino D, Del Boccio P, D’Alessandro E, Rabottini A, Berghella A, Allocati N, Falasca K, Ucciferri C, Mucedola F, Di Perna M, Martino L, Vecchiet J, De Laurenzi V, Dainese E. Iron Dyshomeostasis in COVID-19: Biomarkers Reveal a Functional Link to 5-Lipoxygenase Activation. Int J Mol Sci 2022; 24:15. [PMID: 36613462 PMCID: PMC9819889 DOI: 10.3390/ijms24010015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is characterized by a broad spectrum of clinical symptoms. After acute infection, some subjects develop a post-COVID-19 syndrome known as long-COVID. This study aims to recognize the molecular and functional mechanisms that occur in COVID-19 and long-COVID patients and identify useful biomarkers for the management of patients with COVID-19 and long-COVID. Here, we profiled the response to COVID-19 by performing a proteomic analysis of lymphocytes isolated from patients. We identified significant changes in proteins involved in iron metabolism using different biochemical analyses, considering ceruloplasmin (Cp), transferrin (Tf), hemopexin (HPX), lipocalin 2 (LCN2), and superoxide dismutase 1 (SOD1). Moreover, our results show an activation of 5-lipoxygenase (5-LOX) in COVID-19 and in long-COVID possibly through an iron-dependent post-translational mechanism. Furthermore, this work defines leukotriene B4 (LTB4) and lipocalin 2 (LCN2) as possible markers of COVID-19 and long-COVID and suggests novel opportunities for prevention and treatment.
Collapse
Affiliation(s)
- Beatrice Dufrusine
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Silvia Valentinuzzi
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Sandra Bibbò
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Verena Damiani
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Ersilia D’Alessandro
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Alberto Rabottini
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Alessandro Berghella
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Nerino Allocati
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Katia Falasca
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Clinic of Infectious Diseases, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Claudio Ucciferri
- Clinic of Infectious Diseases, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Francesco Mucedola
- Clinic of Infectious Diseases, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Marco Di Perna
- Pneumology Department, “SS Annunziata” Hospital, 66100 Chieti, Italy
| | - Laura Martino
- Pneumology Department, “SS Annunziata” Hospital, 66100 Chieti, Italy
| | - Jacopo Vecchiet
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Clinic of Infectious Diseases, S.S. Annunziata Hospital, 66100 Chieti, Italy
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Enrico Dainese
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
7
|
Abedi M, Rahgozar S. Puzzling Out Iron Complications in Cancer Drug Resistance. Crit Rev Oncol Hematol 2022; 178:103772. [PMID: 35914667 DOI: 10.1016/j.critrevonc.2022.103772] [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: 05/28/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 12/09/2022] Open
Abstract
Iron metabolism are frequently disrupted in cancer. Patients with cancer are prone to anemia and receive transfusions frequently; the condition which results in iron overload, contributing to serious therapeutic complications. Iron is introduced as a carcinogen that may increase tumor growth. However, investigations regarding its impact on response to chemotherapy, particularly the induction of drug resistance are still limited. Here, iron contribution to cell signaling and various molecular mechanisms underlying iron-mediated drug resistance are described. A dual role of this vital element in cancer treatment is also addressed. On one hand, the need to administer iron chelators to surmount iron overload and improve the sensitivity of tumor cells to chemotherapy is discussed. On the other hand, the necessary application of iron as a therapeutic option by iron-oxide nanoparticles or ferroptosis inducers is explained. Authors hope that this paper can help unravel the clinical complications related to iron in cancer therapy.
Collapse
Affiliation(s)
- Marjan Abedi
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Soheila Rahgozar
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
8
|
Gao S, Chen T, Wang Z, Ji P, Xu L, Cui W, Wang Y. Immuno-activated mesenchymal stem cell living electrospun nanofibers for promoting diabetic wound repair. J Nanobiotechnology 2022; 20:294. [PMID: 35729570 PMCID: PMC9210587 DOI: 10.1186/s12951-022-01503-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/06/2022] [Indexed: 11/27/2022] Open
Abstract
Diabetic wound is the leading cause of non-traumatic amputations in which oxidative stress and chronic inflammation are main factors affecting wound healing. Although mesenchymal stem cells (MSCs) as living materials can promote skin regeneration, they are still vulnerable to oxidative stress which limits their clinical applications. Herein, we have prepared (polylactic-co-glycolic acid) (PLGA) nanofibers electrospun with LPS/IFN-γ activated macrophage cell membrane. After defining physicochemical properties of the nanofibers modified by LPS/IFN-γ activated mouse RAW264.7 cell derived membrane (RCM-fibers), we demonstrated that the RCM-fibers improved BMMSC proliferation and keratinocyte migration upon oxidative stress in vitro. Moreover, bone marrow derived MSCs (BMMSCs)-loaded RCM-fibers (RCM-fiber-BMMSCs) accelerated wound closure accompanied by rapid re-epithelialization, collagen remodeling, antioxidant stress and angiogenesis in experimental diabetic wound healing in vivo. Transcriptome analysis revealed the upregulation of genes related to wound healing in BMMSCs when co-cultured with the RCM-fibers. Enhanced healing capacity of RCM-fiber-BMMSCs living material was partially mediated through CD200-CD200R interaction. Similarly, LPS/IFN-γ activated THP-1 cell membrane coated nanofibers (TCM-fibers) exhibited similar improvement of human BMMSCs (hBMMSCs) on diabetic wound healing in vivo. Our results thus demonstrate that LPS/IFN-γ activated macrophage cell membrane-modified nanofibers can in situ immunostimulate the biofunctions of BMMSCs, making this novel living material promising in wound repair of human diabetes.
Collapse
Affiliation(s)
- Shaoying Gao
- Department of Burn and Plastic surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China. .,Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Tao Chen
- Department of Burn and Plastic surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zhen Wang
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, People's Republic of China
| | - Ping Ji
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lin Xu
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi, 563000, China.
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, People's Republic of China.
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
9
|
Alshareef GH, Mohammed AE, Abumaree M, Basmaeil YS. Phenotypic and Functional Responses of Human Decidua Basalis Mesenchymal Stem/Stromal Cells to Lipopolysaccharide of Gram-Negative Bacteria. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2021; 14:51-69. [PMID: 34754198 PMCID: PMC8572118 DOI: 10.2147/sccaa.s332952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022]
Abstract
Introduction Human decidua basalis mesenchymal stem cells (DBMSCs) are potential therapeutics for the medication to cure inflammatory diseases, like atherosclerosis. The current study investigates the capacity of DBMSCs to stay alive and function in a harmful inflammatory environment induced by high levels of lipopolysaccharide (LPS). Methods DBMSCs were exposed to different levels of LPS, and their viability and functional responses (proliferation, adhesion, and migration) were examined. Furthermore, DBMSCs’ expression of 84 genes associated with their functional activities in the presence of LPS was investigated. Results Results indicated that LPS had no significant effect on DBMSCs’ adhesion, migration, and proliferation (24 h and 72 h) (p > 0.05). However, DBMSCs’ proliferation was significantly reduced at 10 µg/mL of LPS at 48 h (p < 0.05). In addition, inflammatory cytokines and receptors related to adhesion, proliferation, migration, and differentiation were significantly overexpressed when DBMSCs were treated with 10 µg/mL of LPS (p < 0.05). Conclusion These results indicated that DBMSCs maintained their functional activities (proliferation, adhesion, and migration) in the presence of LPS as there was no variation between the treated DBMSCs and the control group. This study will lay the foundation for future preclinical and clinical studies to confirm the appropriateness of DBMSCs as a potential medication to cure inflammatory diseases, like atherosclerosis.
Collapse
Affiliation(s)
- Ghofran Hasan Alshareef
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, 84428, Saudi Arabia
| | - Afrah E Mohammed
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, 84428, Saudi Arabia
| | - Mohammed Abumaree
- Stem Cell & Regenerative Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Yasser S Basmaeil
- Stem Cell & Regenerative Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Woods K, Guezguez B. Dynamic Changes of the Bone Marrow Niche: Mesenchymal Stromal Cells and Their Progeny During Aging and Leukemia. Front Cell Dev Biol 2021; 9:714716. [PMID: 34447754 PMCID: PMC8383146 DOI: 10.3389/fcell.2021.714716] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are a heterogenous cell population found in a wide range of tissues in the body, known for their nutrient-producing and immunomodulatory functions. In the bone marrow (BM), these MSCs are critical for the regulation of hematopoietic stem cells (HSC) that are responsible for daily blood production and functional immunity throughout an entire organism's lifespan. Alongside other stromal cells, MSCs form a specialized microenvironment BM tissue called "niche" that tightly controls HSC self-renewal and differentiation. In addition, MSCs are crucial players in maintaining bone integrity and supply of hormonal nutrients due to their capacity to differentiate into osteoblasts and adipocytes which also contribute to cellular composition of the BM niche. However, MSCs are known to encompass a large heterogenous cell population that remains elusive and poorly defined. In this review, we focus on deciphering the BM-MSC biology through recent advances in single-cell identification of hierarchical subsets with distinct functionalities and transcriptional profiles. We also discuss the contribution of MSCs and their osteo-adipo progeny in modulating the complex direct cell-to-cell or indirect soluble factors-mediated interactions of the BM HSC niche during homeostasis, aging and myeloid malignancies. Lastly, we examine the therapeutic potential of MSCs for rejuvenation and anti-tumor remedy in clinical settings.
Collapse
Affiliation(s)
- Kevin Woods
- German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Borhane Guezguez
- German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Alijani-Ghazyani Z, Sabzevari R, Roushandeh AM, Jahanian-Najafabadi A, Amiri F, Roudkenar MH. Transplantation of Umbilical Cord-Derived Mesenchymal Stem Cells Overexpressing Lipocalin 2 Ameliorates Ischemia-Induced Injury and Reduces Apoptotic Death in a Rat Acute Myocardial Infarction Model. Stem Cell Rev Rep 2021; 16:968-978. [PMID: 32656623 DOI: 10.1007/s12015-020-10007-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Myocardial infarction (MI) is a leading cause of death worldwide and requires development of efficient therapeutic strategies . Mesenchymal stem cells (MSCs) -based therapy of MI has been promising but inefficient due to undesirable microenvironment of the infarct tissue. Hence, the current study was conducted to fortify MSCs against the unfavorable microenvironment of infarct tissue via overexpression of Lipocalin 2 (Lcn2) as a cytoprotective factor. The engineered cells (Lcn2-MSCs) were transplanted to infarcted heart of a rat model of MI. According to our findings, Lcn2 overexpression resulted in increased MSCs survival in the MI tissue (p < 0.05) compared to non-engineered cells. Furthermore, the infusion of Lcn2-MSCs mitigated Left ventricle (LV) remodeling, decreased fibrosis (p < 0.0001), and reduced apoptotic death of the LVs' cells (p < 0.0001) compared to the control. Our findings suggest a potential novel therapeutic strategy for MI, however, further investigations such as safety and efficacy assessments in large animals followed by clinical trials are required.
Collapse
Affiliation(s)
- Zahra Alijani-Ghazyani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reza Sabzevari
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amaneh Mohammadi Roushandeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.,Anatomical Sciences Department, Medicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Amiri
- Department of Medical Laboratory Science, Paramedicine Faculty, Hamadan University of Medical Science, Hamadan, Iran
| | - Mehryar Habibi Roudkenar
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
12
|
Saadaoui I, Cherif M, Rasheed R, Bounnit T, Al Jabri H, Sayadi S, Hamadou RB, Manning SR. Mychonastes homosphaera (Chlorophyceae): A promising feedstock for high quality feed production in the arid environment. ALGAL RES 2020. [DOI: 10.1016/j.algal.2020.102021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
13
|
Panahi M, Rahimi B, Rahimi G, Yew Low T, Saraygord-Afshari N, Alizadeh E. Cytoprotective effects of antioxidant supplementation on mesenchymal stem cell therapy. J Cell Physiol 2020; 235:6462-6495. [PMID: 32239727 DOI: 10.1002/jcp.29660] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/15/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) are earmarked as perfect candidates for cell therapy and tissue engineering due to their capacity to differentiate into different cell types. However, their potential for application in regenerative medicine declines when the levels of the reactive oxygen and nitrogen species (RONS) increase from the physiological levels, a phenomenon which is at least inevitable in ex vivo cultures and air-exposed damaged tissues. Increased levels of RONS can alter the patterns of osteogenic and adipogenic differentiation and inhibit proliferation, as well. Besides, oxidative stress enhances senescence and cell death, thus lowering the success rates of the MSC engraftment. Hence, in this review, we have selected some representatives of antioxidants and newly emerged nano antioxidants in three main categories, including chemical compounds, biometabolites, and protein precursors/proteins, which are proved to be effective in the treatment of MSCs. We will focus on how antioxidants can be applied to optimize the clinical usage of the MSCs and their associated signaling pathways. We have also reviewed several paralleled properties of some antioxidants and nano antioxidants which can be simultaneously used in real-time imaging, scaffolding techniques, and other applications in addition to their primary antioxidative function.
Collapse
Affiliation(s)
- Mohammad Panahi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahareh Rahimi
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Golbarg Rahimi
- Department of Cellular and Molecular Biology, University of Esfahan, Esfahan, Iran
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Neda Saraygord-Afshari
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Effat Alizadeh
- Drug Applied Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Implication and role of neutrophil gelatinase-associated lipocalin in cancer: lipocalin-2 as a potential novel emerging comprehensive therapeutic target for a variety of cancer types. Mol Biol Rep 2020; 47:2327-2346. [PMID: 31970626 DOI: 10.1007/s11033-020-05261-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
Cancer is a leading cause of mortalities worldwide. Over the past few decades, exploration of molecular mechanisms behind cancer initiation and progression has been of great interest in the viewpoint of both basic and clinical scientists. It is generally believed that identification of key molecules implicated in cancer pathology not only improves our understanding of the disease, but also could result in introduction of novel therapeutic strategies. Neutrophil gelatinase-associated lipocalin (NGAL)/lipocalin-2 (LCN2) is a member of lipocalin superfamily with a variety of functions. Although the main function of LCN2 is still unknown, many studies confirmed its significant role in the initiation, progression, and metastasis of various types of cancer. Furthermore, aberrant expression of LCN2 is also concerned with the chemo- and radio-resistant phenotypes of tumors. Here, we will review the contribution of known functions of LCN2 to the pathophysiology of cancer. We also highlight how the deregulated expression of LCN2 is associated with a variety of fatal types of cancer for which there are no effective therapeutic modalities. The unique and multiple functions of LCN2 and its widespread expression in different types of cancer prompted us to suggest LCN2 could be considered either as a valuable diagnostic and prognostic biomarker or as a potential novel therapeutic target.
Collapse
|
15
|
Dietrich J, Ott L, Roth M, Witt J, Geerling G, Mertsch S, Schrader S. MSC Transplantation Improves Lacrimal Gland Regeneration after Surgically Induced Dry Eye Disease in Mice. Sci Rep 2019; 9:18299. [PMID: 31797895 PMCID: PMC6892942 DOI: 10.1038/s41598-019-54840-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022] Open
Abstract
Dry eye disease (DED) is a multifactorial disease characterized by a disrupted tear film homeostasis and inflammation leading to visual impairments and pain in patients. Aqueous-deficient dry eye (ADDE) causes the most severe progressions and depends mainly on the loss of functional lacrimal gland (LG) tissue. Despite a high prevalence, therapies remain palliative. Therefore, it is of great interest to develop new approaches to curatively treat ADDE. Mesenchymal stem/stromal cells (MSC) have been shown to induce tissue regeneration and cease inflammation. Moreover, an increasing amount of MSC was found in the regenerating LG of mice. Therefore, this study investigated the therapeutic effect of MSC transplantation on damaged LGs using duct ligation induced ADDE in mice. Due to the transplantation of sex-mismatched and eGFP-expressing MSC, MSC could be identified and detected until day 21. MSC transplantation significantly improved LG regeneration, as the amount of vital acinar structures was significantly increased above the intrinsic regeneration capacity of control. Additionally, MSC transplantation modulated the immune reaction as macrophage infiltration was delayed and TNFα expression decreased, accompanied by an increased IL-6 expression. Thus, the application of MSC appears to be a promising therapeutic approach to induce LG regeneration in patients suffering from severe DED/ADDE.
Collapse
Affiliation(s)
- Jana Dietrich
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl-von-Ossietzky University, 26121, Oldenburg, Germany.
| | - Lolita Ott
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Mathias Roth
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Joana Witt
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Gerd Geerling
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Sonja Mertsch
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl-von-Ossietzky University, 26121, Oldenburg, Germany
| | - Stefan Schrader
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl-von-Ossietzky University, 26121, Oldenburg, Germany
| |
Collapse
|
16
|
Saeedi P, Halabian R, Imani Fooladi AA. A revealing review of mesenchymal stem cells therapy, clinical perspectives and Modification strategies. Stem Cell Investig 2019; 6:34. [PMID: 31620481 DOI: 10.21037/sci.2019.08.11] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022]
Abstract
Multipotent mesenchymal stem cells (MSCs) have been considerably inspected as effective tool for cell-based therapy of inflammatory, immune-mediated, and degenerative diseases, attributed to their immunomodulatory, immunosuppressive, and regenerative potentials. In the present review, we focus on recent research findings of the clinical applications and therapeutic potential of this cell type, MSCs' mechanisms of therapy, strategies to improve their therapeutic potentials such as manipulations and preconditioning, and potential/unexpected risks which should be considered as a prerequisite step before clinical use. The potential risks would probably include undesirable immune responses, tumor formation and the transmission of incidental agents. Then, we also review some of the milestones in the field, briefly discuss challenges and highlight the new guideline suggested for future directions and perspectives.
Collapse
Affiliation(s)
- Pardis Saeedi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Sharifi S, Zununi Vahed S, Ahmadian E, Maleki Dizaj S, Abedi A, Hosseiniyan Khatibi SM, Samiei M. Stem Cell Therapy: Curcumin Does the Trick. Phytother Res 2019; 33:2927-2937. [DOI: 10.1002/ptr.6482] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/21/2019] [Accepted: 08/04/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Simin Sharifi
- Dental and Periodontal Research CenterTabriz University of Medical Sciences Tabriz Iran
| | | | - Elham Ahmadian
- Kidney Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research CenterTabriz University of Medical Sciences Tabriz Iran
| | - Atefeh Abedi
- Faculty of DentistryTabriz University of Medical Sciences Tabriz Iran
| | | | - Mohammad Samiei
- Faculty of DentistryTabriz University of Medical Sciences Tabriz Iran
- Stem Cell Research CenterTabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
18
|
Rahimi S, Roushandeh AM, Ebrahimi A, Samadani AA, Kuwahara Y, Roudkenar MH. CRISPR/Cas9-mediated knockout of Lcn2 effectively enhanced CDDP-induced apoptosis and reduced cell migration capacity of PC3 cells. Life Sci 2019; 231:116586. [DOI: 10.1016/j.lfs.2019.116586] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/02/2019] [Accepted: 06/17/2019] [Indexed: 01/21/2023]
|
19
|
Dietrich J, Roth M, König S, Geerling G, Mertsch S, Schrader S. Analysis of lacrimal gland derived mesenchymal stem cell secretome and its impact on epithelial cell survival. Stem Cell Res 2019; 38:101477. [PMID: 31181482 DOI: 10.1016/j.scr.2019.101477] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/10/2019] [Accepted: 05/31/2019] [Indexed: 01/03/2023] Open
Abstract
In situ regeneration of lacrimal gland (LG) tissue would be a promising approach to curatively treat dry eye disease (DED). Mesenchymal stem cells (MSC) exhibit therapeutic effects in a variety of pathological conditions and our group recently reported that their number increases in regenerating mouse LG. Since the therapeutic effects are suggested to arise from secreted trophic factors, the application of MSC-secreted proteins seems to be a promising approach to induce/enhance LG regeneration. Therefore, this study aims to optimize the isolation of murine LG-MSC and analyze their secretome to investigate its potential for LG epithelial cell survival in vitro. For optimization, LG-MSC were isolated by an explant technique or cell sorting and their secretome was investigated under normal and inflammatory conditions. Results showed that the secretome of MSC had beneficial effects on the viability of ethanol-damaged LG epithelial cells. Additional, Lipocalin-2, prosaposin, ras GTPase-activating protein-binding protein 1 (Rac1) and signal transducer and activator of transcription 1 (STAT1), proteins that were up-regulated under inflammatory conditions, further improved the cell survival of ethanol-damaged LG epithelial cells. Interestingly, recovery of cell viability was highest, when the cells were incubated with STAT1. Summarizing, this study identified promising proteins for further studies on LG regeneration.
Collapse
Affiliation(s)
- Jana Dietrich
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius Hospital, University of Oldenburg, 26121 Oldenburg, Germany.
| | - Mathias Roth
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, University of Duesseldorf, 40225 Duesseldorf, Germany
| | - Simone König
- Core Unit Proteomics, Interdisciplinary Center for Clinical Research, University of Muenster, 48149 Muenster, Germany
| | - Gerd Geerling
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, University of Duesseldorf, 40225 Duesseldorf, Germany
| | - Sonja Mertsch
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius Hospital, University of Oldenburg, 26121 Oldenburg, Germany
| | - Stefan Schrader
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius Hospital, University of Oldenburg, 26121 Oldenburg, Germany
| |
Collapse
|
20
|
Wang-Yang L, You-Liang Z, Tiao L, Peng Z, Wu-Ji X, Xiao-Long L, Xin-Yu Q, Hui X. Pretreatment with Lithospermic Acid Attenuates Oxidative Stress- induced Apoptosis in Bone Marrow-derived Mesenchymal Stem Cells via Anti-oxidation and Activation of PI3K/Akt Pathway. DIGITAL CHINESE MEDICINE 2019. [DOI: 10.1016/j.dcmed.2019.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
21
|
Mesenchymal stem cell-based therapy for autoimmune diseases: emerging roles of extracellular vesicles. Mol Biol Rep 2019; 46:1533-1549. [PMID: 30623280 DOI: 10.1007/s11033-019-04588-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/03/2019] [Indexed: 02/07/2023]
Abstract
In autoimmune disease body's own immune system knows healthy cells as undesired and foreign cells. Over 80 types of autoimmune diseases have been recognized. Currently, at clinical practice, treatment strategies for autoimmune disorders are based on relieving symptoms and preventing difficulties. In other words, there is no effective and useful therapy up to now. It has been well-known that mesenchymal stem cells (MSCs) possess immunomodulatory effects. This strongly suggests that MSCs might be as a novel modality for treatment of autoimmune diseases. Supporting this notion a few preclinical and clinical studies indicate that MSCs ameliorate autoimmune disorders. Interestingly, it has been found that the beneficial effects of MSCs in autoimmune disorders are not relying only on direct cell-to-cell communication but on their capability to produce a broad range of paracrine factors including growth factors, cytokines and extracellular vehicles (EVs). EVs are multi-signal messengers that play a serious role in intercellular signaling through carrying cargo such as mRNA, miRNA, and proteins. Numerous studies have shown that MSC-derived EVs are able to mimic the effects of the cell of origin on immune cells. In this review, we discuss the current studies dealing with MSC-based therapies in autoimmune diseases and provide a vision and highlight in order to introduce MSC-derived EVs as an alternative and emerging modality for autoimmune disorders.
Collapse
|
22
|
Karabulutoglu M, Finnon R, Imaoka T, Friedl AA, Badie C. Influence of diet and metabolism on hematopoietic stem cells and leukemia development following ionizing radiation exposure. Int J Radiat Biol 2018; 95:452-479. [PMID: 29932783 DOI: 10.1080/09553002.2018.1490042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE The review aims to discuss the prominence of dietary and metabolic regulators in maintaining hematopoietic stem cell (HSC) function, long-term self-renewal, and differentiation. RESULTS Most adult stem cells are preserved in a quiescent, nonmotile state in vivo which acts as a "protective state" for stem cells to reduce endogenous stress provoked by DNA replication and cellular respiration as well as exogenous environmental stress. The dynamic balance between quiescence, self-renewal and differentiation is critical for supporting a functional blood system throughout life of an organism. Stress-conditions, for example ionizing radiation exposure can trigger the blood forming HSCs to proliferate and migrate through extramedullary tissues to expand the number of HSCs and increase hematopoiesis. In addition, a wealth of investigation validated that deregulation of this balance plays a critical pathogenic role in various different hematopoietic diseases including the leukemia development. CONCLUSION The review summarizes the current knowledge on how alterations in dietary and metabolic factors could alter the risk of leukemia development following ionizing radiation exposure by inhibiting or even reversing the leukemic progression. Understanding the influence of diet, metabolism, and epigenetics on radiation-induced leukemogenesis may lead to the development of practical interventions to reduce the risk in exposed populations.
Collapse
Affiliation(s)
- Melis Karabulutoglu
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK.,b CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Oxford , UK
| | - Rosemary Finnon
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| | - Tatsuhiko Imaoka
- c Department of Radiation Effects Research, National Institute of Radiological Sciences , National Institutes for Quantum and Radiological Science and Technology , Chiba , Japan
| | - Anna A Friedl
- d Department of Radiation Oncology , University Hospital, LMU Munich , Munich , Germany
| | - Christophe Badie
- a Cancer Mechanisms and Biomarkers group, Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards , Public Health England , Didcot , UK
| |
Collapse
|
23
|
Bashiri H, Amiri F, Hosseini A, Hamidi M, Mohammadi Roushandeh A, Kuwahara Y, Jalili MA, Habibi Roudkenar M. Dual Preconditioning: A Novel Strategy to Withstand Mesenchymal Stem Cells against Harsh Microenvironments. Adv Pharm Bull 2018; 8:465-470. [PMID: 30276143 PMCID: PMC6156477 DOI: 10.15171/apb.2018.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/21/2018] [Accepted: 07/19/2018] [Indexed: 02/06/2023] Open
Abstract
Purpose: Poor survival rate of mesenchymal stem cells (MSCs) following their transplantation is one of the major challenges in their therapeutic application. Therefore, it is necessary to augment the viability of the MSCs in order to improve their therapeutic efficacy. Several strategies have been used to overcome this problem. Preconditioning of MSCs with oxidative stresses has gained a lot of attention. Therefore, in the present study, we investigated the effects of simultaneous preconditioning of MSCs with hydrogen peroxide and serum deprivation stresses on their survival and resistance to stressful conditions. Methods: MSCs were isolated from human umbilical cord blood. To perform simultaneous preconditioning, the cells were cultured in DMEM medium containing 1, 2.5 and 5 percent FBS and different concentrations of H2O2 (5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 80 and 100 µM) for 24 hrs. Then, the cells were cultured in recovery culture medium. Finally, one group of the cells was exposed to a lethal concentration of H2O2 (300µM), and the other cells were cultivated in FBS free DMEM medium as the lethal situation. In addition, the percentage of apoptotic cells was analyzed using Caspase 3 assay kit. Results: Simultaneous preconditioning of the MSCs with 15µM H2O2 plus serum deprivation, 2.5% FBS, significantly increased the resistance of the cells to the toxicity induced following their cultivation in FBS free DMEM medium. It exerted the protective effect on the cells after treating with the lethal dose of H2O2 as well. Conclusion: Simultaneous preconditioning of MSCs with oxidative and serum deprivation stresses enhances their survival against harsh conditions, which might increase the viability and stability of the MSCs following their transplantation.
Collapse
Affiliation(s)
- Hamed Bashiri
- Department of Medical Laboratory Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Islamic Republic of Iran
| | - Fatemeh Amiri
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Islamic Republic of Iran
| | - Ali Hosseini
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Islamic Republic of Iran
| | - Masoud Hamidi
- Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Amaneh Mohammadi Roushandeh
- Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Yoshikazu Kuwahara
- Division of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Mohammad Ali Jalili
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Islamic Republic of Iran
| | - Mehryar Habibi Roudkenar
- Cardiovascular Disease Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
24
|
Moon GJ, Cho YH, Kim DH, Sung JH, Son JP, Kim S, Cha JM, Bang OY. Serum-mediated Activation of Bone Marrow-derived Mesenchymal Stem Cells in Ischemic Stroke Patients: A Novel Preconditioning Method. Cell Transplant 2018; 27:485-500. [PMID: 29774769 PMCID: PMC6038038 DOI: 10.1177/0963689718755404] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stroke induces complex and dynamic, local and systemic changes including inflammatory
reactions, immune responses, and repair and recovery processes. Mesenchymal stem cells
(MSCs) have been shown to enhance neurological recovery after stroke. We hypothesized that
serum factors play a critical role in the activation of bone marrow (BM) MSCs after stroke
such as by increasing proliferation, paracrine effects, and rejuvenation. Human MSCs
(hMSCs) were grown in fetal bovine serum (FBS), normal healthy control serum (NS), or
stroke patient serum (SS). MSCs cultured in growth medium with 10% SS or NS exhibited
higher proliferation indices than those cultured with FBS (P < 0.01).
FBS-, NS-, and SS-hMSCs showed differences in the expression of trophic factors; vascular
endothelial growth factor, glial cell–derived neurotrophic factor, and fibroblast growth
factor were densely expressed in samples cultured with SS (P < 0.01).
In addition, SS-MSCs revealed different cell cycle– or aging-associated messenger RNA
expression in a later passage, and β-galactosidase staining showed the senescence of MSCs
observed during culture expansion was lower in MSCs cultured with SS than those cultured
with NS or FBS (P < 0.01). Several proteins related to the activity of
receptors, growth factors, and cytokines were more prevalent in the serum of stroke
patients than in that of normal subjects. Neurogenesis and angiogenesis were markedly
increased in rats that had received SS-MSCs (P < 0.05), and these rats
showed significant behavioral improvements (P < 0.01). Our results
indicate that stroke induces a process of recovery via the activation of MSCs. Culture
methods for MSCs using SS obtained during the acute phase of a stroke could constitute a
novel MSC activation method that is feasible and efficient for the neurorestoration of
stroke.
Collapse
Affiliation(s)
- Gyeong Joon Moon
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,2 Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea.,3 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Buk-gu, Daegu, South Korea
| | - Yeon Hee Cho
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Dong Hee Kim
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| | - Ji Hee Sung
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Jeong Pyo Son
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| | - Sooyoon Kim
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Jae Min Cha
- 6 Medical Device Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Oh Young Bang
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,7 Department of Neurology, Samsung Medical Center, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| |
Collapse
|
25
|
Abbasi-Malati Z, Roushandeh AM, Kuwahara Y, Roudkenar MH. Mesenchymal Stem Cells on Horizon: A New Arsenal of Therapeutic Agents. Stem Cell Rev Rep 2018; 14:484-499. [DOI: 10.1007/s12015-018-9817-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
26
|
Parmar T, Parmar VM, Perusek L, Georges A, Takahashi M, Crabb JW, Maeda A. Lipocalin 2 Plays an Important Role in Regulating Inflammation in Retinal Degeneration. THE JOURNAL OF IMMUNOLOGY 2018; 200:3128-3141. [PMID: 29602770 DOI: 10.4049/jimmunol.1701573] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/05/2018] [Indexed: 12/17/2022]
Abstract
It has become increasingly important to understand how retinal inflammation is regulated because inflammation plays a role in retinal degenerative diseases. Lipocalin 2 (LCN2), an acute stress response protein with multiple innate immune functions, is increased in ATP-binding cassette subfamily A member 4 (Abca4) -/- retinol dehydrogenase 8 (Rdh8) -/- double-knockout mice, an animal model for Stargardt disease and age-related macular degeneration (AMD). To examine roles of LCN2 in retinal inflammation and degeneration, Lcn2-/-Abca4-/-Rdh8-/- triple-knockout mice were generated. Exacerbated inflammation following light exposure was observed in Lcn2-/-Abca4-/-Rdh8-/- mice as compared with Abca4-/-Rdh8-/- mice, with upregulation of proinflammatory genes and microglial activation. RNA array analyses revealed an increase in immune response molecules such as Ccl8, Ccl2, and Cxcl10 To further probe a possible regulatory role for LCN2 in retinal inflammation, we examined the in vitro effects of LCN2 on NF-κB signaling in human retinal pigmented epithelial (RPE) cells differentiated from induced pluripotent stem cells derived from healthy donors. We found that LCN2 induced expression of antioxidant enzymes heme oxygenase 1 and superoxide dismutase 2 in these RPE cells and could inhibit the cytotoxic effects of H2O2 and LPS. ELISA revealed increased LCN2 levels in plasma of patients with Stargardt disease, retinitis pigmentosa, and age-related macular degeneration as compared with healthy controls. Finally, overexpression of LCN2 in RPE cells displayed protection from cell death. Overall these results suggest that LCN2 is involved in prosurvival responses during cell stress and plays an important role in regulating inflammation during retinal degeneration.
Collapse
Affiliation(s)
- Tanu Parmar
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106
| | - Vipul M Parmar
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106
| | - Lindsay Perusek
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106
| | - Anouk Georges
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo 650-0047, Japan
| | - John W Crabb
- Cole Eye Institute, Cleveland Clinic, OH 44195; and
| | - Akiko Maeda
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106; .,Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo 650-0047, Japan.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
27
|
Sadeghi F, Etebari M, Habibi Roudkenar M, Jahanian-Najafabadi A. Lipocalin2 Protects Human Embryonic Kidney Cells against Cisplatin-Induced Genotoxicity. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:147-154. [PMID: 29755547 PMCID: PMC5937086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Cisplatin is one of the most useful chemotherapeutics which performs its cytotoxic effect via accumulation of platinum resulting in oxidative stress, and destruction of cell DNA. This could probably cause secondary cancers in healthy tissues. Lipocalin2 (Lcn2) is a protein which its expression is increased in oxidative stresses. Therefore, the present study was performed to evaluate the protective effects of Lcn2 up-regulation on cisplatin genotoxicity. In order to up-regulate Lcn2 expression, HEK293 cells were transfected with pcDNA3.1-Lcn2 vector. Afterwards, stable cells consistently expressing Lcn2 were selected via screening with G418 antibiotic. Next, overexpression of Lcn2 was evaluated by RT-PCR and ELISA, comparing to the control non-transfected cells. Then, in order to evaluate the cytoprotective effects of Lcn2 overexpression, transfected and non-transfected cells were subjected to cisplatin treatment followed by MTT and alkaline Comet assays. RT-PCR and ELISA assays confirmed up-regulation of Lcn2 by the stable cells. MTT assay of the Lcn2 over-expressing cells showed higher IC50 values comparing to the non-transfected cells. Furthermore, the Comet assay confirmed Lcn2 protective effects on the cisplatin (1 µg/mL) induced genotoxicity. In the present study, for the first time, we showed the protective effect of Lcn2 on cisplatin induced genotoxicity. Therefore, one of the probable mechanisms of Lcn2 cytoprotctive effects under oxidative stress conditions could be due to the prevention of genotoxicity. However, further evaluations in this regard must be considered.
Collapse
Affiliation(s)
- Fatemeh Sadeghi
- Student Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran.
| | - Mahmoud Etebari
- Department of Toxicology and Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran.
| | - Mehryar Habibi Roudkenar
- Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran.
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran.,Corresponding author: E-mail:
| |
Collapse
|
28
|
Hossain MI, Horie M, Yoshioka N, Kurose M, Yamamura K, Takebayashi H. Motoneuron degeneration in the trigeminal motor nucleus innervating the masseter muscle in Dystonia musculorum mice. Neurochem Int 2017; 119:159-170. [PMID: 29061384 DOI: 10.1016/j.neuint.2017.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/26/2017] [Accepted: 10/17/2017] [Indexed: 12/11/2022]
Abstract
Dystonia musculorum (dt) mice, which have a mutation in the Dystonin (Dst) gene, are used as animal models to investigate the human disease known as hereditary sensory and autonomic neuropathy type VI. Massive neuronal cell death is observed, mainly in the peripheral nervous system (PNS) of dt mice. We and others have recently reported a histopathological feature of these mice that neurofilament (NF) accumulates in various areas of the central nervous system (CNS), including motor pathways. Although dt mice show motor disorder and growth retardation, the causes for these are still unknown. Here we performed histopathological analyses on motor units of the trigeminal motor nucleus (Mo5 nucleus), because they are a good system to understand neuronal responses in the mutant CNS, and abnormalities in this system may lead to problems in mastication, with subsequent growth retardation. We report that motoneurons with NF accumulation in the Mo5 nuclei of DstGt homozygous mice express the stress-induced genes CHOP, ATF3, and lipocalin 2 (Lcn2). We also show a reduced number of Mo5 motoneurons and a reduced size of Mo5 nuclei in DstGt homozygous mice, possibly due to apoptosis, given the presence of cleaved caspase 3-positive Mo5 motoneurons. In the mandibular (V3) branches of the trigeminal nerve, which contains axons of Mo5 motoneurons and trigeminal sensory neurons, there was infiltration of Iba1-positive macrophages. Finally, we report atrophy of the masseter muscles in DstGt homozygous mice, which showed abnormal nuclear localization of myofibrils and increased expression of atrogin-1 mRNA, a muscle atrophy-related gene and weaker masseter muscle strength with uncontrolled muscle activity by electromyography (EMG). Taken together, our findings strongly suggest that mastication in dt mice is affected due to abnormalities of Mo5 motoneurons and masseter muscles, leading to growth retardation at the post-weaning stages.
Collapse
Affiliation(s)
- M Ibrahim Hossain
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Masao Horie
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; Transdisciplinary Research Program, Niigata University, Niigata 951-8510, Japan
| | - Masayuki Kurose
- Division of Oral Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Kensuke Yamamura
- Division of Oral Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan.
| |
Collapse
|
29
|
Roushandeh AM, Bahadori M, Roudkenar MH. Mesenchymal Stem Cell-based Therapy as a New Horizon for Kidney Injuries. Arch Med Res 2017. [PMID: 28625316 DOI: 10.1016/j.arcmed.2017.03.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Today, the prevalence of kidney diseases is increasing around the world, but there has still been no effective medical treatment. The therapeutic choices are confined to supportive cares and preventive strategies. Currently, mesenchymal stem cells (MSCs)-based cell therapy was proposed for the treatment of kidney injuries. However, after the transplantation of MSCs, they are exposed to masses of cytotoxic factors involving an inflammatory cytokine storm, a nutritionally-poor hypoxic environment and oxidative stresses that finally lead to minimize the efficacy of MSCs based cell therapy. Therefore, several innovative strategies were developed in order to potentiate MSCs to withstand the unfavorable microenvironments of the injured kidney tissues and improve their therapeutic potentials. This review aims to introduce MSCs as a new modality in the treatment of renal failure. Here, we discuss the clinical trials of MSCs-based therapy in kidney diseases as well as the in vivo studies dealing with MSCs application in kidney injuries mainly from the proliferation, differentiation, migration and survival points of view. The obstacles and challenges of this new modality in kidney injuries are also discussed.
Collapse
Affiliation(s)
| | - Marzie Bahadori
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mehryar Habibi Roudkenar
- Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences Rasht, Iran.
| |
Collapse
|
30
|
Effects of Antioxidant Supplements on the Survival and Differentiation of Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5032102. [PMID: 28770021 PMCID: PMC5523230 DOI: 10.1155/2017/5032102] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022]
Abstract
Although physiological levels of reactive oxygen species (ROS) are required to maintain the self-renewal capacity of stem cells, elevated ROS levels can induce chromosomal aberrations, mitochondrial DNA damage, and defective stem cell differentiation. Over the past decade, several studies have shown that antioxidants can not only mitigate oxidative stress and improve stem cell survival but also affect the potency and differentiation of these cells. Further beneficial effects of antioxidants include increasing genomic stability, improving the adhesion of stem cells to culture media, and enabling researchers to manipulate stem cell proliferation by using different doses of antioxidants. These findings can have several clinical implications, such as improving neurogenesis in patients with stroke and neurodegenerative diseases, as well as improving the regeneration of infarcted myocardial tissue and the banking of spermatogonial stem cells. This article reviews the cellular and molecular effects of antioxidant supplementation to cultured or transplanted stem cells and draws up recommendations for further research in this area.
Collapse
|
31
|
Ectopic Expression of Innate Immune Protein, Lipocalin-2, in Lactococcus lactis Protects Against Gut and Environmental Stressors. Inflamm Bowel Dis 2017; 23:1120-1132. [PMID: 28445245 PMCID: PMC5469687 DOI: 10.1097/mib.0000000000001134] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Lipocalin-2 (Lcn2) is a multifunctional innate immune protein that exhibits antimicrobial activity by the sequestration of bacterial siderophores, regulates iron homeostasis, and augments cellular tolerance to oxidative stress. Studies in the murine model of colitis have demonstrated that Lcn2 deficiency exacerbates colitogenesis; however, the therapeutic potential of Lcn2 supplementation has yet to be elucidated. In light of its potential mucoprotective functions, we, herein, investigated whether expression of Lcn2 in the probiotic bacterium can be exploited to alleviate experimental colitis. METHODS Murine Lcn2 was cloned into the pT1NX plasmid and transformed into Lactococcus lactis to generate L. lactis-expressing Lcn2 (Lactis-Lcn2) or the empty plasmid (Lactis-Con). Lactis-Lcn2 was characterized by immunoblot and enzyme-linked immunosorbent assay and tested for its antimicrobial efficacy on Escherichia coli. The capacity of Lactis-Lcn2 and Lactis-Con to withstand adverse conditions was tested using in vitro viability assays. Dextran sodium sulfate colitis model was used to investigate the colonization ability and therapeutic potential of Lactis-Lcn2 and Lactis-Con. RESULTS Lcn2 derived from Lactis-Lcn2 inhibited the growth of E. coli and reduced the bioactivity of enterobactin (E. coli-derived siderophore) in vitro. Lactis-Lcn2 displayed enhanced tolerance to adverse pH, high concentration of bile acids, and oxidative stress in vitro and survived better in the inflamed gut than Lactis-Con. Consistent with these features, Lactis-Lcn2 displayed better mucoprotection against intestinal inflammation than Lactis-Con when administered into mice with dextran sulfate sodium-induced acute colitis. CONCLUSIONS Our findings suggest that Lcn2 expression can be exploited to enhance the survivability of probiotic bacteria during inflammation, which could further improve its efficacy to treat experimental colitis.
Collapse
|
32
|
Roudkenar MH, Halabian R, Tehrani HA, Amiri F, Jahanian-Najafabadi A, Roushandeh AM, Abbasi-Malati Z, Kuwahara Y. Lipocalin 2 enhances mesenchymal stem cell-based cell therapy in acute kidney injury rat model. Cytotechnology 2017; 70:103-117. [PMID: 28573544 DOI: 10.1007/s10616-017-0107-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/09/2017] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury (AKI) is one of the most common health-threatening diseases in the world. There is still no effective medical treatment for AKI. Recently, Mesenchymal stem cell (MSC)-based therapy has been proposed for treatment of AKI. However, the microenvironment of damaged kidney tissue is not favorable for survival of MSCs which would be used for therapeutic intervention. In this study, we genetically manipulated MSCs to up-regulate lipocalin-2 (Lcn2) and investigated whether the engineered MSCs (MSC-Lcn2) could improve cisplatin-induced AKI in a rat model. Our results revealed that up-regulation of Lcn2 in MSCs efficiently enhanced renal function. MSC Lcn2 up-regulates expression of HGF, IGF, FGF and VEGF growth factors. In addition, they reduced molecular biomarkers of kidney injury such as KIM-1 and Cystatin C, while increased the markers of proximal tubular epithelium such as AQP-1 and CK18 following cisplatin-induced AKI. Overall, here we over-expressed Lcn2, a well-known cytoprotective factor against acute ischemic renal injury, in MSCs. This not only potentiated beneficial roles of MSCs for cell therapy purposes but also suggested a new modality for treatment of AKI.
Collapse
Affiliation(s)
- Mehryar Habibi Roudkenar
- Department of Medical Biotechnology, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran. .,Neuroscience Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Raheleh Halabian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Amiri
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | | | - Zahra Abbasi-Malati
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Yoshikazu Kuwahara
- Department of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsushima, Aoba-ku, Sendai, 981-8558, Miyagi, Japan
| |
Collapse
|
33
|
Tsai TL, Li WJ. Identification of Bone Marrow-Derived Soluble Factors Regulating Human Mesenchymal Stem Cells for Bone Regeneration. Stem Cell Reports 2017; 8:387-400. [PMID: 28162996 PMCID: PMC5312426 DOI: 10.1016/j.stemcr.2017.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/15/2022] Open
Abstract
Maintaining properties of human bone marrow-derived mesenchymal stem cells (BMSCs) in culture for regenerative applications remains a great challenge. An emerging approach of constructing a culture environment mimicking the bone marrow niche to regulate BMSC activities has been developed. In this study, we have demonstrated a systematic approach to identify soluble factors of interest extracted from human bone marrow and used them in BMSC culture for tissue regeneration. We have found that lipocalin-2 and prolactin are key factors in bone marrow, involved in regulating BMSC activities. Treating the cell with lipocalin-2 and prolactin delays cellular senescence of BMSCs and primes the cell for osteogenesis and chondrogenesis. We have also demonstrated that BMSCs pretreated with lipocalin-2 and prolactin can enhance the repair of calvarial defects in mice. Together, our study provides research evidence of using a viable approach to prime BMSC properties in vitro for improving cell-based tissue regeneration in vivo. Soluble factors extracted from bone marrow maintain BMSC properties in culture LCN2 and PRL of bone marrow delay cellular senescence of BMSCs LCN2 and PRL prime cultured BMSCs to enhance bone regeneration in mice
Collapse
Affiliation(s)
- Tsung-Lin Tsai
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
34
|
Li W, Liu Y, Wang B, Luo Y, Hu N, Chen D, Zhang X, Xiong Y. Protective effect of berberine against oxidative stress-induced apoptosis in rat bone marrow-derived mesenchymal stem cells. Exp Ther Med 2016; 12:4041-4048. [PMID: 28101183 DOI: 10.3892/etm.2016.3866] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 09/06/2016] [Indexed: 12/13/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) have the potential to be used for the treatment of delayed union, nonunion or persistent bone defects in MSC-based cell therapy. However, implantation of BMSCs into the fracture site is confronted with apoptosis on account of harsh conditions and oxidative stress. In the present study, the anti-apoptotic effects of berberine (BBR) on BMSCs subjected to hydrogen peroxide (H2O2) are investigated, and the potential underlying mechanisms are explored. Oxidative injury was induced by exposure to H2O2, and cell viability was assessed using a cell counting kit-8 assay. The apoptosis of BMSCs was measured by Hoechst 33258 and Annexin V-fluorescein isothiocyanate/propidium iodide assay. Reactive oxygen species staining and superoxide dismutase (SOD) assay were applied to assess the anti-oxidative effect of BBR. Finally, western blot was performed to measure the expression levels of phosphorylated (p)-Akt, B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax) and cleaved caspase-3. In the present study, it was identified that BBR remarkably attenuated H2O2-induced apoptotic cell death via quenching ROS production and increasing SOD activity. Further studies indicated that BBR can reduce apoptosis by upregulating the expression level of p-Akt and Bcl-2, and downregulating the expression levels of Bax and cleaved caspase-3. Taken together, the results of the present study demonstrate that pretreatment with BBR could alleviate H2O2-induced apoptosis in rat BMSCs in vitro.
Collapse
Affiliation(s)
- Wangyang Li
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, P.R. China
| | - Yamei Liu
- Department of Diagnostics of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Bin Wang
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, P.R. China
| | - Yiwen Luo
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, P.R. China
| | - Nianhong Hu
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, P.R. China
| | - Dongfeng Chen
- Department of Anatomy, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Xunchao Zhang
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, P.R. China
| | - Yunpu Xiong
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, P.R. China
| |
Collapse
|
35
|
Miyamoto T, Kashima H, Yamada Y, Kobara H, Asaka R, Ando H, Higuchi S, Ida K, Mvunta DH, Shiozawa T. Lipocalin 2 Enhances Migration and Resistance against Cisplatin in Endometrial Carcinoma Cells. PLoS One 2016; 11:e0155220. [PMID: 27168162 PMCID: PMC4864227 DOI: 10.1371/journal.pone.0155220] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 04/26/2016] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Lipocalin 2 (LCN2) is a secretory protein that is involved in various physiological processes including iron transport. We previously identified LCN2 as an up-regulated gene in endometrial carcinoma, and found that the overexpression of LCN2 and its receptor, SLC22A17, was associated with a poor prognosis. However, the functions and mechanism of action of LCN2 currently remain unclear. METHODS The LCN2-overexpressing endometrial carcinoma cell lines, HHUA and RL95-2, and LCN2-low-expressing one, HEC1B, were used. The effects of LCN2 on cell migration, cell viability, and apoptosis under various stresses, including ultraviolet (UV) irradiation and cisplatin treatment, were examined using the scratch wound healing assay, WST-1 assay, and Apostrand assay, respectively. RESULTS LCN2-silencing using shRNA method significantly reduced the migration ability of cells (p<0.05). Cytotoxic stresses significantly decreased the viability of LCN2-silenced cells more than that of control cells. In contrast, LCN2 overexpression was significantly increased cisplatin resistance. These effects were canceled by the addition of the iron chelator, deferoxamine. After UV irradiation, the expression of phosphorylated Akt (pAkt) was decreased in LCN2-silenced cells, and the PI3K inhibitor canceled the difference induced in UV sensitivity by LCN2. The cisplatin-induced expression of pAkt was not affected by LCN2; however, the expression of p53 and p21 was increased by LCN2-silencing. CONCLUSIONS These results indicated that LCN2 was involved in the migration and survival of endometrial carcinoma cells under various stresses in an iron-dependent manner. The survival function of LCN2 may be exerted through the PI3K pathway and suppression of the p53-p21 pathway. These functions of LCN2 may increase the malignant potential of endometrial carcinoma cells.
Collapse
Affiliation(s)
- Tsutomu Miyamoto
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
- * E-mail:
| | - Hiroyasu Kashima
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Yasushi Yamada
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Hisanori Kobara
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Ryoichi Asaka
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Hirofumi Ando
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Shotaro Higuchi
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Koichi Ida
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - David Hamisi Mvunta
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| | - Tanri Shiozawa
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390–8621, Japan
| |
Collapse
|
36
|
Zhaleh F, Amiri F, Mohammadzadeh-Vardin M, Bahadori M, Harati MD, Roudkenar MH, Saki S. Nuclear factor erythroid-2 related factor 2 overexpressed mesenchymal stem cells transplantation, improves renal function, decreases injuries markers and increases repair markers in glycerol-induced Acute kidney injury rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:323-9. [PMID: 27114803 PMCID: PMC4834123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVES Recently cell therapy is a promising therapeutic modality for many types of disease including acute kidney injury (AKI). Due to the unique biological properties, mesenchymal stem cells (MSCs) are attractive cells in this regard. This study aims to transplant MSCs equipped with nuclear factor E2-related factor 2 (Nrf2) in rat experimental models of acute kidney and evaluate regeneration potential of injured kidney especially expression of injury and repaired biomarkers. MATERIALS AND METHODS Nrf2 was overexpressed in bone marrow-derived MSCs by pcDNA.3.1 plasmid. AKI was induced using glycerol in rat models. The regenerative potential of Nrf2-overexpressed MSCs was evaluated in AKI-Induced animal models using biochemical and histological methods after transplantation. Expression of repaired genes, AQP1 and CK-18, as well as injury markers, Kim-1 and Cystatin C, was also assayed in engrafted kidney sections. RESULTS Our results revealed that transplantation of Nrf2-overexpressed MSCs into AKI-induced rats decreased blood urea nitrogen and creatinine and ameliorated kidney regeneration throughout 14 days. Upregulation of repaired markers and downregulation of injury markers were considerable 14 days after transplantation. CONCLUSIONS Overexpression of Nrf2 in MSCs suggests a new strategy to increase efficiency of MSC-based cell therapy in AKI.
Collapse
Affiliation(s)
- Fateme Zhaleh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Fatemeh Amiri
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohammad Mohammadzadeh-Vardin
- Department of Anatomical Sciences and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Marzie Bahadori
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | | | - Mehryar Habibi Roudkenar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Sasan Saki
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Islamic Azad University, Arak Branch, Arak, Iran,Corresponding author: Sasan Saki. Department of Medical Laboratory Sciences, Faculty of Medical sciences, Islamic Azad University, Arak Branch, Arak, Iran. Tel: +086-33412508;
| |
Collapse
|
37
|
Yamada Y, Miyamoto T, Kashima H, Kobara H, Asaka R, Ando H, Higuchi S, Ida K, Shiozawa T. Lipocalin 2 attenuates iron-related oxidative stress and prolongs the survival of ovarian clear cell carcinoma cells by up-regulating the CD44 variant. Free Radic Res 2016; 50:414-25. [PMID: 26729415 DOI: 10.3109/10715762.2015.1134795] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Ovarian clear cell carcinoma (CCC) arises from ovarian endometriosis. Intra-cystic fluid contains abundant amounts of free iron, which causes persistent oxidative stress, a factor that has been suggested to induce malignant transformation. However, the mechanisms linking oxidative stress and carcinogenesis in CCC currently remain unclear. Lipocalin 2 (LCN2), a multifunctional secretory protein, functions as an iron transporter as well as an antioxidant. Therefore, we herein examined the roles of LCN2 in the regulation of intracellular iron concentrations, oxidative stress, DNA damage, and antioxidative functions using LCN2-overexpressing (ES2), and LCN2-silenced (RMG-1) CCC cell lines. The results of calcein staining indicated that the up-regulated expression of LCN2 correlated with increases in intracellular iron concentrations. However, a DCFH-DA assay and 8OHdG staining revealed that LCN2 reduced intracellular levels of reactive oxygen species and DNA damage. Furthermore, the expression of LCN2 suppressed hydrogen peroxide-induced apoptosis and prolonged cell survival, suggesting an antioxidative role for LCN2. The expression of mRNAs and proteins for various oxidative stress-catalyzing enzymes, such as heme oxygenase (HO), superoxide dismutase (SOD), and glutathione peroxidase, was not affected by LCN2, whereas the intracellular concentration of the potent antioxidant, glutathione (GSH), was increased by LCN2. Furthermore, the expression of xCT, a cystine transporter protein, and CD44 variant 8-10 (CD44v), a stem cell marker, was up-regulated by LCN2. Although LCN2 increased intracellular iron concentrations, LCN2-induced GSH may catalyze and override oxidative stress via CD44v and xCT, and subsequently enhance the survival of CCC cells in oxidative stress-rich endometriosis.
Collapse
Affiliation(s)
- Yasushi Yamada
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Tsutomu Miyamoto
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hiroyasu Kashima
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hisanori Kobara
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Ryoichi Asaka
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Hirofumi Ando
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Shotaro Higuchi
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Koichi Ida
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| | - Tanri Shiozawa
- a Department of Obstetrics and Gynecology , Shinshu University School of Medicine , Matsumoto , Japan
| |
Collapse
|
38
|
Roles of NGAL and MMP-9 in the tumor microenvironment and sensitivity to targeted therapy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:438-448. [PMID: 26278055 DOI: 10.1016/j.bbamcr.2015.08.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/08/2015] [Accepted: 08/10/2015] [Indexed: 12/15/2022]
Abstract
Various, diverse molecules contribute to the tumor microenvironment and influence invasion and metastasis. In this review, the roles of neutrophil gelatinase-associated lipocalin (NGAL) and matrix metalloproteinase-9 (MMP-9) in the tumor microenvironment and sensitivity to therapy will be discussed. The lipocalin family of proteins has many important functions. For example when NGAL forms a complex with MMP-9 it increases its stability which is important in cancer metastasis. Small hydrophobic molecules are bound by NGAL which can alter their entry into and efflux from cells. Iron transport and storage are also influenced by NGAL activity. Regulation of iron levels is important for survival in the tumor microenvironment as well as metastasis. Innate immunity is also regulated by NGAL as it can have bacteriostatic properties. NGAL and MMP-9 expression may also affect the sensitivity of cancer cells to chemotherapy as well as targeted therapy. Thus NGAL and MMP-9 play important roles in key processes involved in metastasis as well as response to therapy. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
|
39
|
Targeting delivery of lipocalin 2-engineered mesenchymal stem cells to colon cancer in order to inhibit liver metastasis in nude mice. Tumour Biol 2015; 36:6011-8. [PMID: 25740061 DOI: 10.1007/s13277-015-3277-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/18/2015] [Indexed: 01/05/2023] Open
Abstract
One of the major obstacles in cancer therapy is the lack of anticancer agent specificity to tumor tissues. The strategy of cell-based therapy is a promising therapeutic option for cancer treatment. The specific tumor-oriented migration of mesenchymal stem cells (MSCs) makes them a useful vehicle to deliver anticancer agents. In this study, we genetically manipulated bone marrow-derived mesenchymal stem cells with their lipocalin 2 (Lcn2) in order to inhibit liver metastasis of colon cancer in nude mice. Lcn2 was successfully overexpressed in transfected MSCs. The PCR results of SRY gene confirmed the presence of MSCs in cancer liver tissue. This study showed that Lcn2-engineered MSCs (MSC-Lcn2) not only inhibited liver metastasis of colon cancer but also downregulated the expression of vascular endothelial growth factor (VEGF) in the liver. Overall, MSCs by innate tropism toward cancer cells can deliver the therapeutic agent, Lcn2, and inhibit cancer metastasis. Hence, it could be a new modality for efficient targeted delivery of anticancer agent to liver metastasis.
Collapse
|
40
|
Amiri F, Jahanian-Najafabadi A, Roudkenar MH. In vitro augmentation of mesenchymal stem cells viability in stressful microenvironments : In vitro augmentation of mesenchymal stem cells viability. Cell Stress Chaperones 2015; 20:237-51. [PMID: 25527070 PMCID: PMC4326383 DOI: 10.1007/s12192-014-0560-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/02/2014] [Accepted: 12/07/2014] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are under intensive investigation for use in cell-based therapies because their differentiation abilities, immunomodulatory effects, and homing properties offer potential for significantly augmenting regenerative capacity of many tissues. Nevertheless, major impediments to their therapeutic application, such as low proliferation and survival rates remain as obstacles to broad clinical use of MSCs. Another major challenge to evolution of MSC-based therapies is functional degradation of these cells as a result of their exposure to oxidative stressors during isolation. Indeed, oxidative stress-mediated MSC depletion occurs due to inflammatory processes associated with chemotherapy, radiotherapy, and expression of pro-apoptotic factors, and the microenvironment of damaged tissue in patients receiving MSC therapy is typically therapeutic not favorable to their survival. For this reason, any strategies that enhance the viability and proliferative capacity of MSCs associated with their therapeutic use are of great value. Here, recent strategies used by various researchers to improve MSC allograft function are reviewed, with particular focus on in vitro conditioning of MSCs in preparation for clinical application. Preconditioning, genetic manipulation, and optimization of MSC culture conditions are some examples of the methodologies described in the present article, along with novel strategies such as treatment of MSCs with secretome and MSC-derived microvesicles. This topic material is likely to find value as a guide for both research and clinical use of MSC allografts and for improvement of the value that use of these cells brings to health care.
Collapse
Affiliation(s)
- Fatemeh Amiri
- />Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ali Jahanian-Najafabadi
- />Department of Pharmaceutical Biotechnology, School of Pharmacy, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | - Mehryar Habibi Roudkenar
- />Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
41
|
Diverse functional roles of lipocalin-2 in the central nervous system. Neurosci Biobehav Rev 2015; 49:135-56. [DOI: 10.1016/j.neubiorev.2014.12.006] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 11/28/2014] [Accepted: 12/04/2014] [Indexed: 12/16/2022]
|
42
|
Cremers NAJ, Lundvig DMS, van Dalen SCM, Schelbergen RF, van Lent PLEM, Szarek WA, Regan RF, Carels CE, Wagener FADTG. Curcumin-induced heme oxygenase-1 expression prevents H2O2-induced cell death in wild type and heme oxygenase-2 knockout adipose-derived mesenchymal stem cells. Int J Mol Sci 2014; 15:17974-99. [PMID: 25299695 PMCID: PMC4227200 DOI: 10.3390/ijms151017974] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/11/2014] [Accepted: 09/23/2014] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cell (MSC) administration is a promising adjuvant therapy to treat tissue injury. However, MSC survival after administration is often hampered by oxidative stress at the site of injury. Heme oxygenase (HO) generates the cytoprotective effector molecules biliverdin/bilirubin, carbon monoxide (CO) and iron/ferritin by breaking down heme. Since HO-activity mediates anti-apoptotic, anti-inflammatory, and anti-oxidative effects, we hypothesized that modulation of the HO-system affects MSC survival. Adipose-derived MSCs (ASCs) from wild type (WT) and HO-2 knockout (KO) mice were isolated and characterized with respect to ASC marker expression. In order to analyze potential modulatory effects of the HO-system on ASC survival, WT and HO-2 KO ASCs were pre-treated with HO-activity modulators, or downstream effector molecules biliverdin, bilirubin, and CO before co-exposure of ASCs to a toxic dose of H2O2. Surprisingly, sensitivity to H2O2-mediated cell death was similar in WT and HO-2 KO ASCs. However, pre-induction of HO-1 expression using curcumin increased ASC survival after H2O2 exposure in both WT and HO-2 KO ASCs. Simultaneous inhibition of HO-activity resulted in loss of curcumin-mediated protection. Co-treatment with glutathione precursor N-Acetylcysteine promoted ASC survival. However, co-incubation with HO-effector molecules bilirubin and biliverdin did not rescue from H2O2-mediated cell death, whereas co-exposure to CO-releasing molecules-2 (CORM-2) significantly increased cell survival, independently from HO-2 expression. Summarizing, our results show that curcumin protects via an HO-1 dependent mechanism against H2O2-mediated apoptosis, and likely through the generation of CO. HO-1 pre-induction or administration of CORMs may thus form an attractive strategy to improve MSC therapy.
Collapse
Affiliation(s)
- Niels A J Cremers
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Ditte M S Lundvig
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Stephanie C M van Dalen
- Department of Rheumatology, Experimental Rheumatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Rik F Schelbergen
- Department of Rheumatology, Experimental Rheumatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Peter L E M van Lent
- Department of Rheumatology, Experimental Rheumatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Walter A Szarek
- Department of Chemistry, Queen's University, Kingston, ON K7L 3N6, Canada.
| | - Raymond F Regan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Carine E Carels
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Frank A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
43
|
Bahmani B, Roudkenar MH, Halabian R, Jahanian-Najafabadi A, Amiri F, Jalili MA. Lipocalin 2 decreases senescence of bone marrow-derived mesenchymal stem cells under sub-lethal doses of oxidative stress. Cell Stress Chaperones 2014; 19:685-93. [PMID: 24452457 PMCID: PMC4147076 DOI: 10.1007/s12192-014-0496-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/03/2014] [Accepted: 01/08/2014] [Indexed: 01/02/2023] Open
Abstract
The regenerative potential of mesenchymal stem cells (MSCs) is impaired by cellular senescence, a multi factorial process that has various functions. However, pathways and molecules involved in senescence have not been fully identified. Lipocalin 2 (Lcn2) has been the subject of intensive research, due to its contribution to many physiological and pathophysiological conditions. The implication of Lcn2 has been reported in many conditions where senescence also occurs. In the present study, we evaluated the role of Lcn2 in the occurrence of senescence in human bone marrow-derived mesenchymal stem cells (hB-MSCs) under oxidative conditions. When hB-MSCs were genetically engineered to over-express Lcn2 (MSC-Lcn2) and exposed to H2O2, the proliferation rate of the cells increased. However, the number of colonies and the number of cells that made up each colony in both MSC-V and MSC-Lcn2 cells decreased compared to those cultivated under normal conditions. Our results revealed that over-expression of recombinant Lcn2 in hB-MSCs decreases senescence induced by H2O2 treatment. Senescent cells were observed in aged hB-MSCs; however, no alteration in the expression level of Lcn2 was detected compared to earlier passages. Finally, a higher amount of Lcn2 protein was detected in the plasma of the elderly than in young people. Our findings suggest that Lcn2 might restore the health and regeneration potential of MSCs by decreasing senescence.
Collapse
Affiliation(s)
- Bahareh Bahmani
- />Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, P.O. Box 14665–1157, Tehran, Iran
| | - Mehryar Habibi Roudkenar
- />Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, P.O. Box 14665–1157, Tehran, Iran
| | - Raheleh Halabian
- />Applied Microbiology Research Center, Medical Science of Baqiyatallah University, Tehran, Iran
| | - Ali Jahanian-Najafabadi
- />Department of Pharmaceutical Biotechnology, and Bioinformatics Research Center, School of Pharmacy, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | - Fatemeh Amiri
- />Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, P.O. Box 14665–1157, Tehran, Iran
| | - Mohammad Ali Jalili
- />Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, P.O. Box 14665–1157, Tehran, Iran
| |
Collapse
|
44
|
Halabian R, Roudkenar MH, Jahanian-Najafabadi A, Hosseini KM, Tehrani HA. Co-culture of bone marrow-derived mesenchymal stem cells overexpressing lipocalin 2 with HK-2 and HEK293 cells protects the kidney cells against cisplatin-induced injury. Cell Biol Int 2014; 39:152-63. [PMID: 25049146 DOI: 10.1002/cbin.10344] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 06/25/2014] [Indexed: 12/16/2022]
Abstract
Conditioned medium of mesenchymal stem cells (MSCs) is now being used for its cytoprotective effects, especially when the cells are equipped with cytoprotective factors to strengthen them against unfavorable microenvironments. Overexpression of Lcn2 in MSCs mimics in vivo kidney injury. Hence, unraveling how Lcn2-engineered MSCs affect kidney cells has been investigated. Cisplatin treated HK-2 or HEK293 kidney cells were co-cultivated with Lcn2 overexpressing MSCs in upper and lower chambers of transwell plates. Proliferation, apoptosis, and expression of growth factors and cytokines were assessed in the kidney cells. Co-cultivation with the MSCs-Lcn2 not only inhibited cisplatin-induced cytotoxicity in the HK-2 and HEK293 cells, but increased proliferation rate, prevented cisplatin-induced apoptosis, and increased expression of growth factors and the amount of antioxidants in the kidney cells. Thus Lcn2-engineered MSCs can ameliorate and repair injured kidney cells in vitro, which strongly suggests there are beneficial effects of the MSCs-Lcn2 in cell therapy of kidney injury.
Collapse
Affiliation(s)
- Raheleh Halabian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | |
Collapse
|