1
|
Kibritoglu E, Yuksel H. Numerical analysis of coil designs to expedite fracture healing using dielectrophoresis with S method. Comput Biol Med 2025; 192:110213. [PMID: 40279972 DOI: 10.1016/j.compbiomed.2025.110213] [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: 10/21/2024] [Revised: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Classical methods for speeding up fracture healing usually rely on direct electrical stimulation and electromagnetic fields to boost the levels of growth factors at the fracture site. However, these techniques often concentrate on bone cells themselves rather than addressing the critical blood flow dynamics necessary for effective healing. This study introduces a mathematical model designed to explore the potential of dielectrophoretic forces (DEPFs) in improving blood flow at the fracture site. By adjusting blood flow, the model seeks to enhance the delivery of vital nutrients, hormones, and growth factors, including endothelial cells (ECs), vascular endothelial growth factor (VEGF) and oxygen, which are essential for accelerating the fracture healing process. METHOD The proposed approach includes a new technique, termed the S method, which assesses the non-uniformity of DEPFs by algebraically analyzing the electric field lines associated with positive and negative dielectrophoresis. We developed analytical equations to simulate various coil configurations, focusing on long bone fractures where blood flow is vertically oriented. The DEPF Factor (χDEPF) was used to measure the ratio of blood flow velocity in the presence of DEPFs compared to the absence of DEPFs, thus indicating the effectiveness of DEPF in enhancing blood flow. RESULTS The simulation results revealed that DEPF reaches its peak efficacy at the gamma dispersion band, with the most significant enhancement occurring at a frequency of 15 MHz. Specifically, the average values of χDEPF were 1.8, 3.2, and 7.9 for the catenary, lintearia, and valeria coils, respectively. Our computational model, which incorporated VEGF, ECs, and oxygen tension, demonstrated that the catenary coil slightly improved healing rates in impaired fractures, the lintearia coil normalized healing times between impaired and normal fractures, and the valeria coil not only accelerated healing in impaired fractures but also enhanced healing in normal fractures. CONCLUSIONS This paper's findings suggest that the valeria coil exhibits the best DEPF functionality, making it the optimal configuration for future experimental studies aimed at evaluating the efficacy of DEPF in promoting fracture healing. The ability of DEPFs to significantly enhance blood flow could represent a substantial advancement in the treatment of both normal and impaired fractures.
Collapse
Affiliation(s)
- Erman Kibritoglu
- Department of Electrical and Electronics Engineering, Bogazici University, Bebek, İstanbul, 34342, Turkey
| | - Heba Yuksel
- Department of Electrical and Electronics Engineering, Bogazici University, Bebek, İstanbul, 34342, Turkey.
| |
Collapse
|
2
|
Bu F, Yuan X, Cui X, Guo R. Bibliometric Analysis and Visualized Study of Research on Mesenchymal Stem Cells in Ischemic Stroke. Stem Cell Rev Rep 2025:10.1007/s12015-025-10878-9. [PMID: 40257541 DOI: 10.1007/s12015-025-10878-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND One of the major global causes of death and disability is ischemic stroke (IS). Mesenchymal stem cells (MSCs) emerge as a cell-based therapy for numerous diseases. Recently, research on the role of MSCs in ischemic stroke has developed rapidly worldwide. Bibliometric analysis of MSCs for IS has not yet been published, though. AIM Through bibliometric analysis, the aim of this study was to assess the current state of research on MSCs in the field of ischemic stroke research worldwide and to identify important results, major research areas, and emerging trends. METHODS Publications related to MSCs in ischemic stroke from January 1, 2002, to December 31, 2022, were obtained from the Web of Science Core Collection (WoSCC). We used HistCite, VOSViewer, CiteSpace, and Bibliometrix for bibliometric analysis and visualization. We employed the Total Global Citation Score (TGCS) to assess the impact of publications. RESULTS The bibliometric analysis included a total of 2,048 publications. The 1,386 papers used in this study were authored by 200 individuals across 200 organizations in 72 countries, published in 202 journals. Cesar V Borlongan published the most documents among high-productivity authors. Michael Chopp was the author with the highest average number of citations per paper, with an average paper citation time of 118.54. We found that research of MSCs in ischemic stroke developed rapidly starting in 2008. Neurosciences were the most productive journals, and Chinese researchers have produced the most research papers in this subject. The most cited article is "Systemic administration of exosomes released from mesenchymal stromal cells promotes functional recovery and neurovascular plasticity after stroke in rats". CONCLUSION This study uses both numbers and descriptions to thoroughly review the research on MSCs related to IS. This information provides valuable experience for researchers to carry out MSCs' work on IS.
Collapse
Affiliation(s)
- Fanwei Bu
- Xinxiang First People's Hospital, Xinxiang, China
| | | | - Xiaocan Cui
- Xinxiang First People's Hospital, Xinxiang, China
| | - Ruyue Guo
- Henan University of Chinese Medicine, Zhengzhou, China.
| |
Collapse
|
3
|
Sikorová M, Klener P, Tonarová P, Kalbáčová MH. Interactions between leukemia and feeders in co-cultivation under hypoxia. BMC Cancer 2025; 25:678. [PMID: 40229651 PMCID: PMC11995666 DOI: 10.1186/s12885-025-13988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Leukemia is driven by complex interactions within the inherently hypoxic bone marrow microenvironment, impacting both disease progression and therapeutic resistance. Co-cultivation of leukemic cells with feeder cells has emerged as a valuable tool to mimic the bone marrow niche. This study explores the interplay between human commercial SD-1 and patient-derived UPF26K leukemic cell lines with feeders - human fibroblasts (NHDF) and mesenchymal stem cells (hMSCs) under normoxic and hypoxic conditions. RESULTS Co-cultivation with feeders significantly enhances proliferation and glycolytic activity in the SD-1 cells, improving their viability, while this interaction inhibits the growth and glucose metabolism of the feeders, particularly NHDF. In contrast, UPF26K cells show reduced proliferation when co-cultivated with the feeders while this interaction stimulates NHDF and hMSCs proliferation and glycolysis but reduce their mitochondrial metabolism with hypoxia amplifying these effects. CONCLUSIONS Cells that switch to glycolysis during co-cultivation, particularly under hypoxia, benefit most from these low oxygen conditions. Due to this leukemic cells' response heterogeneity, targeting microenvironmental interactions and oxygen levels is crucial for personalized leukemia therapy. Advancing co-cultivation models, particularly through innovations like spheroids, can further enhance in vitro studies of primary leukemic cells and support the testing of novel therapies.
Collapse
Affiliation(s)
- Miriama Sikorová
- Institute of Pathological Physiology, 1st Faculty of Faculty of Medicine, Charles University, U nemocnice 5, Prague, 128 53, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, 1st Faculty of Faculty of Medicine, Charles University, U nemocnice 5, Prague, 128 53, Czech Republic
| | - Pavla Tonarová
- Institute of Pathological Physiology, 1st Faculty of Faculty of Medicine, Charles University, U nemocnice 5, Prague, 128 53, Czech Republic
| | - Marie Hubálek Kalbáčová
- Institute of Pathological Physiology, 1st Faculty of Faculty of Medicine, Charles University, U nemocnice 5, Prague, 128 53, Czech Republic.
| |
Collapse
|
4
|
Zhou H, Xiang W, Zhou G, Rodrigues-Lima F, Guidez F, Wang L. Metabolic dysregulation in myelodysplastic neoplasm: impact on pathogenesis and potential therapeutic targets. Med Oncol 2024; 42:23. [PMID: 39644425 DOI: 10.1007/s12032-024-02575-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024]
Abstract
Despite significant advancements in the research of the pathogenesis mechanisms of Myelodysplastic Neoplasm (MDS) in recent years, there are still many gaps to fill. The advancement of metabolomics studies has led to a research booming in clarifying the impact of metabolic abnormalities during the pathogenesis of MDS. The present review primarily focuses on the dysregulated metabolic pathways, exploring the influences on the pathogenesis of MDS and their roles during the course of the disease. Furthermore, we discuss the potential of relevant metabolic pathways as therapeutic targets, along with the latest metabolic-related treatment drugs and approaches.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wenqiong Xiang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Guangyu Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Fernando Rodrigues-Lima
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle Et Adaptative, 75013, Paris, France
| | - Fabien Guidez
- Université Paris Cité, Institut de Recherche Saint Louis INSERM UMR_S1131, 75010, Paris, France
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
5
|
Artamonov MY, Sokov EL. Intraosseous Delivery of Mesenchymal Stem Cells for the Treatment of Bone and Hematological Diseases. Curr Issues Mol Biol 2024; 46:12672-12693. [PMID: 39590346 PMCID: PMC11592824 DOI: 10.3390/cimb46110752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Mesenchymal stem cells are used most in regenerative medicine due to their capacities in differentiation and immune modulation. The intraosseous injection of MSC into the bone has been recommended because of expected outcomes for retention, bioavailability, and enhanced therapeutic efficacy, particularly in conditions involving the bone, such as osteoporosis and osteonecrosis. A review of the intraosseous delivery of mesenchymal stem cells in comparison with intravenous and intra-arterial delivery methods will be subjected to critical examination. This delivery mode fares better regarding paracrine signaling and immunomodulation attributes, which are the cornerstone of tissue regeneration and inflammation reduction. The local complications and technical challenges still apply with this method. This study was more focused on further research soon to be conducted to further elucidate long-term safety and efficacy of intraosseous mesenchymal stem cell therapy. Though much has been achieved with very impressive progress in this field, it is worth noting that more studies need to be put into place so that this technique can be established as a routine approach, especially with further research in biomaterials, gene therapy, and personalized medicine.
Collapse
Affiliation(s)
| | - Evgeniy L. Sokov
- Department of Algology and Rehabilitation, Peoples’ Friendship University, Moscow 117198, Russia;
| |
Collapse
|
6
|
Raabe J, Wittig I, Laurette P, Stathopoulou K, Brand T, Schulze T, Klampe B, Orthey E, Cabrera-Orefice A, Meisterknecht J, Thiemann E, Laufer SD, Shibamiya A, Reinsch M, Fuchs S, Kaiser J, Yang J, Zehr S, Wrona KM, Lorenz K, Lukowski R, Hansen A, Gilsbach R, Brandes RP, Ulmer BM, Eschenhagen T, Cuello F. Physioxia rewires mitochondrial complex composition to protect stem cell viability. Redox Biol 2024; 77:103352. [PMID: 39341035 PMCID: PMC11466565 DOI: 10.1016/j.redox.2024.103352] [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: 05/27/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are an invaluable tool to study molecular mechanisms on a human background. Culturing stem cells at an oxygen level different from their microenvironmental niche impacts their viability. To understand this mechanistically, dermal skin fibroblasts of 52 probands were reprogrammed into hiPSCs, followed by either hyperoxic (20 % O2) or physioxic (5 % O2) culture and proteomic profiling. Analysis of chromosomal stability by Giemsa-banding revealed that physioxic -cultured hiPSC clones exhibited less pathological karyotypes than hyperoxic (e.g. 6 % vs. 32 % mosaicism), higher pluripotency as evidenced by higher Stage-Specific Embryonic Antigen 3 positivity, higher glucose consumption and lactate production. Global proteomic analysis demonstrated lower abundance of several subunits of NADH:ubiquinone oxidoreductase (complex I) and an underrepresentation of pathways linked to oxidative phosphorylation and cellular senescence. Accordingly, release of the pro-senescent factor IGFBP3 and β-galactosidase staining were lower in physioxic hiPSCs. RNA- and ATAC-seq profiling revealed a distinct hypoxic transcription factor-binding footprint, amongst others higher expression of the HIF1α-regulated target NDUFA4L2 along with increased chromatin accessibility of the NDUFA4L2 gene locus. While mitochondrial DNA content did not differ between groups, physioxic hiPSCs revealed lower polarized mitochondrial membrane potential, altered mitochondrial network appearance and reduced basal respiration and electron transfer capacity. Blue-native polyacrylamide gel electrophoresis coupled to mass spectrometry of the mitochondrial complexes detected higher abundance of NDUFA4L2 and ATP5IF1 and loss of incorporation into complex IV or V, respectively. Taken together, physioxic culture of hiPSCs improved chromosomal stability, which was associated with downregulation of oxidative phosphorylation and senescence and extensive re-wiring of mitochondrial complex composition.
Collapse
Affiliation(s)
- Janice Raabe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ilka Wittig
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany
| | - Patrick Laurette
- Institute of Experimental Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Konstantina Stathopoulou
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Theresa Brand
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Thomas Schulze
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Birgit Klampe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ellen Orthey
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Alfredo Cabrera-Orefice
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Jana Meisterknecht
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Ellen Thiemann
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Sandra D Laufer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Aya Shibamiya
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Marina Reinsch
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Sigrid Fuchs
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jennifer Kaiser
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jiaqi Yang
- Institute of Pharmacy, Experimental Pharmacology, University Tübingen, 72076 Tübingen, Germany
| | - Simonida Zehr
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Kinga M Wrona
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Robert Lukowski
- Institute of Pharmacy, Experimental Pharmacology, University Tübingen, 72076 Tübingen, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Ralf P Brandes
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Bärbel M Ulmer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
7
|
Mendiratta M, Mendiratta M, Ganguly S, Rai S, Gupta R, Kumar L, Bakhshi S, Dadhwal V, Pushpam D, Malik PS, Pramanik R, Aggarwal M, Gupta AK, Dhawan R, Seth T, Mahapatra M, Nayak B, Singh TD, Kumar S, Mir RA, Kaur G, GuruRao H, Singh M, Prasad CP, Prakash H, Mohanty S, Sahoo RK. Concurrent hypoxia and apoptosis imparts immune programming potential in mesenchymal stem cells: Lesson from acute graft-versus-host-disease model. Stem Cell Res Ther 2024; 15:381. [PMID: 39468660 PMCID: PMC11520827 DOI: 10.1186/s13287-024-03947-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have emerged as promising candidates for immune modulation in various diseases that are associated with dysregulated immune responses like Graft-versus-Host-Disease (GVHD). MSCs are pleiotropic and the fate of MSCs following administration is a major determinant of their therapeutic efficacy. METHODS Human MSCs were derived from bone marrow (BM) and Wharton's Jelly (WJ) and preconditioned through exposure to hypoxia and induction of apoptosis, either sequentially or simultaneously. The immune programming potential of preconditioned MSCs was evaluated by assessing their effects on T cell proliferation, induction of Tregs, programming of effector T-cell towards Th2 phenotype, macrophage polarization in the direct co-culture of MSCs and aGVHD patients-derived PBMNCs. Additionally, efferocytosis of MSCs and relative change in the expression of immunomodulatory soluble factors were examined. RESULTS Our study demonstrated that hypoxia preconditioned apoptotic MSCs (BM-MSCs, WJ-MSCs) bear more immune programming ability in a cellular model of acute Graft-versus-Host-Disease (aGVHD). Our findings revealed that WJ-MSCsHYP+APO were superior to BM-MSCsHYP+APO for immune regulation. These induced the differentiation of CD4+T-cell into Tregs, enhanced Th2 effector responses, and simultaneously mitigated Th1- and Th17 responses. Additionally, this approach led to the polarization of M1 macrophages toward an M2 phenotype. CONCLUSION Our study highlights the potential of WJ-MSCs conditioned with hypoxia and apoptosis concurrently, as a promising therapeutic strategy for aGVHD. It underscores the importance of considering MSC apoptosis in optimizing MSCs-based cellular therapy protocols for enhanced therapeutic efficacy in aGvHD.
Collapse
Affiliation(s)
- Mohini Mendiratta
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Meenakshi Mendiratta
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Shuvadeep Ganguly
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sandeep Rai
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Lalit Kumar
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vatsla Dadhwal
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Deepam Pushpam
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Prabhat Singh Malik
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Raja Pramanik
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Mukul Aggarwal
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Aditya Kumar Gupta
- Department of Pediatric Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rishi Dhawan
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Tulika Seth
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Manoranjan Mahapatra
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Baibaswata Nayak
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Thoudam Debraj Singh
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sachin Kumar
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Riyaz Ahmed Mir
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Gurvinder Kaur
- Laboratory Oncology Unit, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Hariprasad GuruRao
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Mayank Singh
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Hridayesh Prakash
- Amity Centre for Translational Research, Amity University, Sector - 125, Noida, 201313, India.
| | - Sujata Mohanty
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
8
|
Burzi IS, Parchi PD, Barachini S, Pardini E, Sardo Infirri G, Montali M, Petrini I. Hypoxia Promotes the Stemness of Mesangiogenic Progenitor Cells and Prevents Osteogenic but not Angiogenic Differentiation. Stem Cell Rev Rep 2024; 20:1830-1842. [PMID: 38914791 PMCID: PMC11457687 DOI: 10.1007/s12015-024-10749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
The stem cell niche in the bone marrow is a hypoxic environment, where the low oxygen tension preserves the pluripotency of stem cells. We have identified mesangiogenic progenitor cells (MPC) exhibiting angiogenic and mesenchymal differentiation capabilities in vitro. The effect of hypoxia on MPC has not been previously explored. In this study, MPCs were isolated from volunteers' bone marrow and cultured under both normoxic and hypoxic conditions (3% O2). MPCs maintained their characteristic morphology and surface marker expression (CD18 + CD31 + CD90-CD73-) under hypoxia. However, hypoxic conditions led to reduced MPC proliferation in primary cultures and hindered their differentiation into mesenchymal stem cells (MSCs) upon exposure to differentiative medium. First passage MSCs derived from MPC appeared unaffected by hypoxia, exhibiting no discernible differences in proliferative potential or cell cycle. However, hypoxia impeded the subsequent osteogenic differentiation of MSCs, as evidenced by decreased hydroxyapatite deposition. Conversely, hypoxia did not impact the angiogenic differentiation potential of MPCs, as demonstrated by spheroid-based assays revealing comparable angiogenic sprouting and tube-like formation capabilities under both hypoxic and normoxic conditions. These findings indicate that hypoxia preserves the stemness phenotype of MPCs, inhibits their differentiation into MSCs, and hampers their osteogenic maturation while leaving their angiogenic potential unaffected. Our study sheds light on the intricate effects of hypoxia on bone marrow-derived MPCs and their differentiation pathways.
Collapse
Affiliation(s)
- Irene Sofia Burzi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Paolo Domenico Parchi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56125, Pisa, Italy
| | - Eleonora Pardini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Gisella Sardo Infirri
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Marina Montali
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56125, Pisa, Italy
| | - Iacopo Petrini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy.
| |
Collapse
|
9
|
Gandolfi S, Sanouj A, Chaput B, Coste A, Sallerin B, Varin A. The role of adipose tissue-derived stromal cells, macrophages and bioscaffolds in cutaneous wound repair. Biol Direct 2024; 19:85. [PMID: 39343924 PMCID: PMC11439310 DOI: 10.1186/s13062-024-00534-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
Skin healing is a complex and dynamic physiological process that follows mechanical alteration of the skin barrier. Under normal conditions, this complex process can be divided into at least three continuous and overlapping phases: an inflammatory reaction, a proliferative phase that leads to tissue reconstruction and a phase of tissue remodeling. Macrophages critically contribute to the physiological cascade for tissue repair. In fact, as the inflammatory phase progresses, macrophage gene expression gradually shifts from pro-inflammatory M1-like to pro-resolutive M2-like characteristics, which is critical for entry into the repair phase. A dysregulation in this macrophage' shift phenotype leads to the persistence of the inflammatory phase. Mesenchymal stromal cells and specifically the MSC-derived from adipose tissue (ADSCs) are more and more use to treat inflammatory diseases and several studies have demonstrated that ADSCs promote the wound healing thanks to their neoangiogenic, immunomodulant and regenerative properties. In several studies, ADSCs and macrophages have been injected directly into the wound bed, but the delivery of exogenous cells directly to the wound raise the problem of cell engraftment and preservation of pro-resolutive phenotype and viability of the cells. Complementary approaches have therefore been explored, such as the use of biomaterials enriched with therapeutic cell to improve cell survival and function. This review will present a background of the current scaffold models, using adipose derived stromal-cells and macrophage as therapeutic cells for wound healing, through a discussion on the potential impact for future applications in skin regeneration. According to the PRISMA statement, we resumed data from investigations reporting the use ADSCs and bioscaffolds and data from macrophages behavior with functional biomaterials in wound healing models. In the era of tissue engineering, functional biomaterials, that can maintain cell delivery and cellular viability, have had a profound impact on the development of dressings for the treatment of chronic wounds. Promising results have been showed in pre-clinical reports using ADSCs- and macrophages-based scaffolds to accelerate and to improve the quality of the cutaneous healing.
Collapse
Affiliation(s)
- S Gandolfi
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France.
- Department of Plastic and Reconstructive Surgery, Toulouse University Hospital, 1 Av. Pr.Jean Poulhès, 31400, Toulouse, France.
| | - A Sanouj
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France
| | - B Chaput
- Department of Plastic and Reconstructive Surgery, Toulouse University Hospital, 1 Av. Pr.Jean Poulhès, 31400, Toulouse, France
| | - A Coste
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France
| | - B Sallerin
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France
- Department of Pharmacology, Toulouse University Hospital, 1 Av Pr.Jean Poulhès, 31400, Toulouse, France
| | - A Varin
- FLAMES Team, Restore Institute, Inserm, Toulouse III Paul Sabatier University, 4Bis Av. H. Curien, 31100, Toulouse, France
| |
Collapse
|
10
|
Zhang H, Li L, Sun X, Hou B, Luo C. Research and development of microenvironment's influence on stem cells from the apical papilla - construction of novel research microdevices: tooth-on-a-chip. Biomed Microdevices 2024; 26:33. [PMID: 39023652 DOI: 10.1007/s10544-024-00715-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2024] [Indexed: 07/20/2024]
Abstract
Stem cells are crucial in tissue engineering, and their microenvironment greatly influences their behavior. Among the various dental stem cell types, stem cells from the apical papilla (SCAPs) have shown great potential for regenerating the pulp-dentin complex. Microenvironmental cues that affect SCAPs include physical and biochemical factors. To research optimal pulp-dentin complex regeneration, researchers have developed several models of controlled biomimetic microenvironments, ranging from in vivo animal models to in vitro models, including two-dimensional cultures and three-dimensional devices. Among these models, the most powerful tool is a microfluidic microdevice, a tooth-on-a-chip with high spatial resolution of microstructures and precise microenvironment control. In this review, we start with the SCAP microenvironment in the regeneration of pulp-dentin complexes and discuss research models and studies related to the biological process.
Collapse
Affiliation(s)
- Hexuan Zhang
- Center for Microscope Enhanced Dentistry, School of Stomatology, Capital Medical University, Beijing, China
- Department of Endodontics and Operative Dentistry, School of Stomatology, Capital Medical University, Beijing, China
| | - Lingjun Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Xiaoqiang Sun
- Department of Endodontics and Operative Dentistry, School of Stomatology, Capital Medical University, Beijing, China.
| | - Benxiang Hou
- Center for Microscope Enhanced Dentistry, School of Stomatology, Capital Medical University, Beijing, China.
| | - Chunxiong Luo
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.
| |
Collapse
|
11
|
Jin L, Zhou S, Zhao S, Long J, Huang Z, Zhou J, Zhang Y. Early short-term hypoxia promotes epidermal cell migration by activating the CCL2-ERK1/2 pathway and epithelial-mesenchymal transition during wound healing. BURNS & TRAUMA 2024; 12:tkae017. [PMID: 38887221 PMCID: PMC11182653 DOI: 10.1093/burnst/tkae017] [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: 09/08/2023] [Revised: 01/05/2024] [Indexed: 06/20/2024]
Abstract
Background Due to vasculature injury and increased oxygen consumption, the early wound microenvironment is typically in a hypoxic state. We observed enhanced cell migration ability under early short-term hypoxia. CCL2 belongs to the CC chemokine family and was found to be increased in early hypoxic wounds and enriched in the extracellular signal-regulated kinase (ERK)1/2 pathway in our previous study. However, the underlying mechanism through which the CCL2-ERK1/2 pathway regulates wound healing under early short-term hypoxia remains unclear. Activation of epithelial-mesenchymal transition (EMT) is a key process in cancer cell metastasis, during which epithelial cells acquire the characteristics of mesenchymal cells and enhance cell motility and migration ability. However, the relationship between epithelial cell migration and EMT under early short-term hypoxia has yet to be explored. Methods HaCaT cells were cultured to verify the effect of early short-term hypoxia on migration through cell scratch assays. Lentiviruses with silenced or overexpressed CCL2 were used to explore the relationship between CCL2 and migration under short-term hypoxia. An acute full-thickness cutaneous wound rat model was established with the application of an ERK inhibitor to reveal the hidden role of the ERK1/2 pathway in the early stage of wound healing. The EMT process was verified in all the above experiments through western blotting. Results In our study, we found that short-term hypoxia promoted cell migration. Mechanistically, hypoxia promoted cell migration through mediating CCL2. Overexpression of CCL2 via lentivirus promoted cell migration, while silencing CCL2 via lentivirus inhibited cell migration and the production of related downstream proteins. In addition, we found that CCL2 was enriched in the ERK1/2 pathway, and the application of an ERK inhibitor in vivo and in vitro verified the upstream and downstream relationships between the CCL2 pathway and ERK1/2. Western blot results both in vivo and in vitro demonstrated that early short-term hypoxia promotes epidermal cell migration by activating the CCL2-ERK1/2 pathway and EMT during wound healing. Conclusions Our work demonstrated that hypoxia in the early stage serves as a stimulus for triggering wound healing through activating the CCL2-ERK1/2 pathway and EMT, which promote epidermal cell migration and accelerate wound closure. These findings provide additional detailed insights into the mechanism of wound healing and new targets for clinical treatment.
Collapse
Affiliation(s)
- Linbo Jin
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Shiqi Zhou
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Shihan Zhao
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Junhui Long
- Department of Dermatology, Southwest Hospital Jiangbei Area (The 958th hospital of Chinese People’s Liberation Army), Chongqing, China
| | - Zhidan Huang
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Junli Zhou
- Department of Burn and Plastic Surgery, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Yiming Zhang
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| |
Collapse
|
12
|
Shi Y, Wang S, Wang K, Yang R, Liu D, Liao H, Qi Y, Qiu K, Hu Y, Wen H, Xu K. Relieving Macrophage Dysfunction by Inhibiting SREBP2 Activity: A Hypoxic Mesenchymal Stem Cells-Derived Exosomes Loaded Multifunctional Hydrogel for Accelerated Diabetic Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309276. [PMID: 38247194 DOI: 10.1002/smll.202309276] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/18/2023] [Indexed: 01/23/2024]
Abstract
Macrophage dysfunction is one of the primary factors leading to the delayed healing of diabetic wounds. Hypoxic bone marrow mesenchymal stem cells-derived exosomes (hyBMSC-Exos) have been shown to play an active role in regulating cellular function through the carried microRNAs. However, the administration of hyBMSC-Exos alone in diabetic wounds usually brings little effect, because the exosomes are inherently unstable and have a short retention time at the wounds. In this study, a multifunctional hydrogel based on gallic acid (GA) conjugated chitosan (Chi-GA) and partially oxidized hyaluronic acid (OHA) is prepared for sustained release of hyBMSC-Exos. The hydrogel not only exhibits needs-satisfying physicochemical properties, but also displays outstanding biological performances such as low hemolysis rate, strong antibacterial capacity, great antioxidant ability, and excellent biocompatibility. It has the ability to boost the stability of hyBMSC-Exos, leading to a continuous and gradual release of the exosomes at wound locations, ultimately enhancing the exosomes' uptake efficiency by target cells. Most importantly, hyBMSC-Exos loaded hydrogel shows an excellent ability to promote diabetic wound healing by regulating macrophage polarization toward M2 phenotype. This may be because exosomal miR-4645-5p and antioxidant property of the hydrogel synergistically inhibit SREBP2 activity in macrophages. This study presents a productive approach for managing diabetic wounds.
Collapse
Affiliation(s)
- Yan Shi
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Shang Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Kai Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, P. R. China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, 510650, P. R. China
| | - Dewu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Huaiwei Liao
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yuhan Qi
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Keqing Qiu
- Dermatological Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yanghong Hu
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330006, P. R. China
| | - Huicai Wen
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Kui Xu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, P. R. China
| |
Collapse
|
13
|
Das S, Valoor R, Ratnayake P, Basu B. Low-Concentration Gelatin Methacryloyl Hydrogel with Tunable 3D Extrusion Printability and Cytocompatibility: Exploring Quantitative Process Science and Biophysical Properties. ACS APPLIED BIO MATERIALS 2024; 7:2809-2835. [PMID: 38602318 DOI: 10.1021/acsabm.3c01194] [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] [Indexed: 04/12/2024]
Abstract
Three-dimensional (3D) bioprinting of hydrogels with a wide spectrum of compositions has been widely investigated. Despite such efforts, a comprehensive understanding of the correlation among the process science, buildability, and biophysical properties of the hydrogels for a targeted clinical application has not been developed in the scientific community. In particular, the quantitative analysis across the entire developmental path for 3D extrusion bioprinting of such scaffolds is not widely reported. In the present work, we addressed this gap by using widely investigated biomaterials, such as gelatin methacryloyl (GelMA), as a model system. Using extensive experiments and quantitative analysis, we analyzed how the individual components of methacrylated carboxymethyl cellulose (mCMC), needle-shaped nanohydroxyapatite (nHAp), and poly(ethylene glycol)diacrylate (PEGDA) with GelMA as baseline matrix of the multifunctional bioink can influence the biophysical properties, printability, and cellular functionality. The complex interplay among the biomaterial ink formulations, viscoelastic properties, and printability toward the large structure buildability (structurally stable cube scaffolds with 15 mm edge) has been explored. Intriguingly, the incorporation of PEGDA into the GelMA/mCMC matrix offered improved compressive modulus (∼40-fold), reduced swelling ratio (∼2-fold), and degradation rates (∼30-fold) compared to pristine GelMA. The correlation among microstructural pore architecture, biophysical properties, and cytocompatibility is also established for the biomaterial inks. These photopolymerizable bio(material)inks served as the platform for the growth and development of bone and cartilage matrix when human mesenchymal stem cells (hMSCs) are either seeded on two-dimensional (2D) substrates or encapsulated on 3D scaffolds. Taken together, this present study unequivocally establishes a significant step forward in the development of a broad spectrum of shape-fidelity compliant bioink for the 3D bioprinting of multifunctional scaffolds and emphasizes the need for invoking more quantitative analysis in establishing process-microstructure-property correlation.
Collapse
Affiliation(s)
- Soumitra Das
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Remya Valoor
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Praneeth Ratnayake
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
14
|
Wang X, Li F, Wu S, Xing W, Fu J, Wang R, He Y. Research progress on optimization of in vitro isolation, cultivation and preservation methods of dental pulp stem cells for clinical application. Front Bioeng Biotechnol 2024; 12:1305614. [PMID: 38633667 PMCID: PMC11021638 DOI: 10.3389/fbioe.2024.1305614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Due to high proliferative capacity, multipotent differentiation, immunomodulatory abilities, and lack of ethical concerns, dental pulp stem cells (DPSCs) are promising candidates for clinical application. Currently, clinical research on DPSCs is in its early stages. The reason for the failure to obtain clinically effective results may be problems with the production process of DPSCs. Due to the different preparation methods and reagent formulations of DPSCs, cell characteristics may be affected and lead to inconsistent experimental results. Preparation of clinical-grade DPSCs is far from ready. To achieve clinical application, it is essential to transit the manufacturing of stem cells from laboratory grade to clinical grade. This review compares and analyzes experimental data on optimizing the preparation methods of DPSCs from extraction to resuscitation, including research articles, invention patents and clinical trials. The advantages and disadvantages of various methods and potential clinical applications are discussed, and factors that could improve the quality of DPSCs for clinical application are proposed. The aim is to summarize the current manufacture of DPSCs in the establishment of a standardized, reliable, safe, and economic method for future preparation of clinical-grade cell products.
Collapse
Affiliation(s)
- Xinxin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College of the Ministry of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Fenyao Li
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College of the Ministry of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Shuting Wu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College of the Ministry of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Wenbo Xing
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College of the Ministry of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jiao Fu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College of the Ministry of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Ruoxuan Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College of the Ministry of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College of the Ministry of Medicine, Wuhan University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Neuhaus F, Lieber S, Shinkevich V, Steitz AM, Raifer H, Roth K, Finkernagel F, Worzfeld T, Burchert A, Keber C, Nist A, Stiewe T, Reinartz S, Beutgen VM, Graumann J, Pauck K, Garn H, Gaida M, Müller R, Huber M. Reciprocal crosstalk between Th17 and mesothelial cells promotes metastasis-associated adhesion of ovarian cancer cells. Clin Transl Med 2024; 14:e1604. [PMID: 38566518 PMCID: PMC10988119 DOI: 10.1002/ctm2.1604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND IL-17A and TNF synergistically promote inflammation and tumorigenesis. Their interplay and impact on ovarian carcinoma (OC) progression are, however, poorly understood. We addressed this question focusing on mesothelial cells, whose interaction with tumor cells is known to play a pivotal role in transcoelomic metastasis formation. METHODS Flow-cytometry and immunohistochemistry experiments were employed to identify cellular sources of IL-17A and TNF. Changes in transcriptomes and secretomes were determined by bulk and single cell RNA sequencing as well as affinity proteomics. Functional consequences were investigated by microscopic analyses and tumor cell adhesion assays. Potential clinical implications were assessed by immunohistochemistry and survival analyses. RESULTS We identified Th17 cells as the main population of IL-17A- and TNF producers in ascites and detected their accumulation in early omental metastases. Both IL-17A and its receptor subunit IL-17RC were associated with short survival of OC patients, pointing to a role in clinical progression. IL-17A and TNF synergistically induced the reprogramming of mesothelial cells towards a pro-inflammatory mesenchymal phenotype, concomitantly with a loss of tight junctions and an impairment of mesothelial monolayer integrity, thereby promoting cancer cell adhesion. IL-17A and TNF synergistically induced the Th17-promoting cytokines IL-6 and IL-1β as well as the Th17-attracting chemokine CCL20 in mesothelial cells, indicating a reciprocal crosstalk that potentiates the tumor-promoting role of Th17 cells in OC. CONCLUSIONS Our findings reveal a novel function for Th17 cells in the OC microenvironment, which entails the IL-17A/TNF-mediated induction of mesothelial-mesenchymal transition, disruption of mesothelial layer integrity and consequently promotion of OC cell adhesion. These effects are potentiated by a positive feedback loop between mesothelial and Th17 cells. Together with the observed clinical associations and accumulation of Th17 cells in omental micrometastases, our observations point to a potential role in early metastases formation and thus to new therapeutic options.
Collapse
Affiliation(s)
- Felix Neuhaus
- Institute of Systems ImmunologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Sonja Lieber
- Institute of Systems ImmunologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | | | - Anna Mary Steitz
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Hartmann Raifer
- Institute of Systems ImmunologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
- FACS Core FacilityCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Kathrin Roth
- Cell Imaging Core Facility, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Florian Finkernagel
- Bioinformatics Core Facility, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Thomas Worzfeld
- Institute of PharmacologyPhilipps UniversityMarburgGermany
- Department of PharmacologyMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Andreas Burchert
- Department of HematologyOncology and ImmunologyUniversity Hospital Giessen and MarburgMarburgGermany
| | - Corinna Keber
- Comprehensive Biomaterial Bank Marburg (CBBMR) and Institute of PathologyPhilipps UniversityMarburgGermany
| | - Andrea Nist
- Genomics Core FacilityInstitute of Molecular OncologyMember of the German Center for Lung Research (DZL)Philipps UniversityMarburgGermany
| | - Thorsten Stiewe
- Genomics Core FacilityInstitute of Molecular OncologyMember of the German Center for Lung Research (DZL)Philipps UniversityMarburgGermany
| | - Silke Reinartz
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Vanessa M. Beutgen
- Institute of Translational Proteomics and Translational Proteomics Core FacilityBiochemical Pharmacological CentrePhilipps UniversityMarburgGermany
| | - Johannes Graumann
- Institute of Translational Proteomics and Translational Proteomics Core FacilityBiochemical Pharmacological CentrePhilipps UniversityMarburgGermany
| | - Kim Pauck
- Translational Inflammation Research Division and Core Facility for Single Cell MultiomicsPhilipps UniversityMarburgGermany
| | - Holger Garn
- Translational Inflammation Research Division and Core Facility for Single Cell MultiomicsPhilipps UniversityMarburgGermany
| | - Matthias Gaida
- Institute of PathologyUniversity Medical Center Mainz, Johannes Gutenberg UniversityMainzGermany
- TRON, Translational Oncology at the University Medical CenterJohannes Gutenberg UniversityMainzGermany
- Research Center for ImmunotherapyUniversity Medical Center Mainz, Johannes Gutenberg UniversityMainzGermany
| | - Rolf Müller
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Magdalena Huber
- Institute of Systems ImmunologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| |
Collapse
|
16
|
Rasouli M, Fattahi R, Nuoroozi G, Zarei-Behjani Z, Yaghoobi M, Hajmohammadi Z, Hosseinzadeh S. The role of oxygen tension in cell fate and regenerative medicine: implications of hypoxia/hyperoxia and free radicals. Cell Tissue Bank 2024; 25:195-215. [PMID: 37365484 DOI: 10.1007/s10561-023-10099-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
Oxygen pressure plays an integral role in regulating various aspects of cellular biology. Cell metabolism, proliferation, morphology, senescence, metastasis, and angiogenesis are some instances that are affected by different tensions of oxygen. Hyperoxia or high oxygen concentration, enforces the production of reactive oxygen species (ROS) that disturbs physiological homeostasis, and consequently, in the absence of antioxidants, cells and tissues are directed to an undesired fate. On the other side, hypoxia or low oxygen concentration, impacts cell metabolism and fate strongly through inducing changes in the expression level of specific genes. Thus, understanding the precise mechanism and the extent of the implication of oxygen tension and ROS in biological events is crucial to maintaining the desired cell and tissue function for application in regenerative medicine strategies. Herein, a comprehensive literature review has been performed to find out the impacts of oxygen tensions on the various behaviors of cells or tissues.
Collapse
Affiliation(s)
- Mehdi Rasouli
- Student Research Committee, Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Fattahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Ghader Nuoroozi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Zeinab Zarei-Behjani
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maliheh Yaghoobi
- Engineering Department, Faculty of Chemical Engineering, Zanjan University, Zanjan, Iran
| | - Zeinab Hajmohammadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran.
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Yasan GT, Gunel-Ozcan A. Hypoxia and Hypoxia Mimetic Agents As Potential Priming Approaches to Empower Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2024; 19:33-54. [PMID: 36642875 DOI: 10.2174/1574888x18666230113143234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 01/17/2023]
Abstract
Mesenchymal stem cells (MSC) exhibit self-renewal capacity and multilineage differentiation potential, making them attractive for research and clinical application. The properties of MSC can vary depending on specific micro-environmental factors. MSC resides in specific niches with low oxygen concentrations, where oxygen functions as a metabolic substrate and a signaling molecule. Conventional physical incubators or chemically hypoxia mimetic agents are applied in cultures to mimic the original low oxygen tension settings where MSC originated. This review aims to focus on the current knowledge of the effects of various physical hypoxic conditions and widely used hypoxia-mimetic agents-PHD inhibitors on mesenchymal stem cells at a cellular and molecular level, including proliferation, stemness, differentiation, viability, apoptosis, senescence, migration, immunomodulation behaviors, as well as epigenetic changes.
Collapse
Affiliation(s)
| | - Aysen Gunel-Ozcan
- Department of Stem Cell Sciences, Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| |
Collapse
|
18
|
Fleischhammer TM, Dienemann S, Ulber N, Pepelanova I, Lavrentieva A. Detection of Hypoxia in 2D and 3D Cell Culture Systems Using Genetically Encoded Fluorescent Hypoxia Sensors. Methods Mol Biol 2024; 2755:31-48. [PMID: 38319567 DOI: 10.1007/978-1-0716-3633-6_2] [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] [Indexed: 02/07/2024]
Abstract
In vivo oxygen availability varies widely between cellular microenvironments, depending on the tissue of origin and its cellular niche. It has long been known that too high or too low oxygen concentrations can act as a biological stressor. Thus, the precise control of oxygen availability should be a consideration for cell culture optimization, especially in the field of three-dimensional (3D) cell culture. In this chapter, we describe a system for visualizing oxygen limitations at a cellular level using human adipose tissue-derived mesenchymal stem cells (hAD-MSCs) that were genetically modified to express a fluorescent hypoxia sensor. This sensor can detect the activation of hypoxia-induced factors (HIF) transcription factors that lead to the expression of the oxygen-independent fluorescent protein, UnaG, at low oxygen concentrations. The response of these hypoxia reporter cells can be evaluated in two-dimensional (2D) and 3D cultivation platforms during exposure to hypoxia (1% O2) and normoxia (21% O2) using fluorescence microscopy and flow cytometry. We show that hypoxia reporter MSCs exhibit a hypoxia-induced fluorescence signal in both 2D and 3D cultivation platforms with fast decay kinetics after reoxygenation, rendering it a valuable tool for studying the cellular microenvironment and regenerative potential of hAD-MSCs in an in vivo-like setting.
Collapse
Affiliation(s)
| | - Sandra Dienemann
- Institute of Technical Chemistry, Leibniz University of Hanover, Hanover, Germany
| | - Nico Ulber
- Institute of Technical Chemistry, Leibniz University of Hanover, Hanover, Germany
| | - Iliyana Pepelanova
- Institute of Technical Chemistry, Leibniz University of Hanover, Hanover, Germany
| | - Antonina Lavrentieva
- Institute of Technical Chemistry, Leibniz University of Hanover, Hanover, Germany.
| |
Collapse
|
19
|
Rovere M, Reverberi D, Arnaldi P, Palamà MEF, Gentili C. Spheroid size influences cellular senescence and angiogenic potential of mesenchymal stromal cell-derived soluble factors and extracellular vesicles. Front Bioeng Biotechnol 2023; 11:1297644. [PMID: 38162179 PMCID: PMC10756914 DOI: 10.3389/fbioe.2023.1297644] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction: The secretome of mesenchymal stromal cells (MSCs) serves as an innovative tool employed in the regenerative medicine approach. In this particular context, three-dimensional (3D) culture systems are widely utilized to better replicate in vivo conditions and facilitate prolonged cell maintenance during culture. The use of spheroids enables the preservation of the classical phenotypical characteristics of MSCs. However, the distinct microenvironment within the spheroid may impact the secretome, thereby enhancing the angiogenic properties of adult MSCs that typically possess a reduced angiogenic potential compared to MSCs derived from perinatal tissues due to the hypoxia created in the internal region of the spheroid. Methods: In this study, large spheroids (2,600 cells, ∼300 μm diameter) and small spheroids (1,000 cells, ∼200 μm diameter) were used to examine the role of spheroid diameter in the generation of nutrients and oxygen gradients, cellular senescence, and the angiogenic potential of secreted factors and extracellular vesicles (EVs). Results: In this study, we demonstrate that large spheroids showed increased senescence and a secretome enriched in pro-angiogenic factors, as well as pro-inflammatory and anti-angiogenic cytokines, while small spheroids exhibited decreased senescence and a secretome enriched in pro-angiogenic molecules. We also demonstrated that 3D culture led to a higher secretion of EVs with classical phenotypic characteristics. Soluble factors and EVs from small spheroids exhibited higher angiogenic potential in a human umbilical vein endothelial cell (HUVEC) angiogenic assay. Discussion: These findings highlighted the necessity of choosing the appropriate culture system for obtaining soluble factors and EVs for specific therapeutic applications.
Collapse
Affiliation(s)
- Matteo Rovere
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | | - Pietro Arnaldi
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | | - Chiara Gentili
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
20
|
Kumlin M, Ungerstedt J, Cai H, Leonard E, Felländer-Tsai L, Qian H. The functional and molecular impact of triamcinolone acetonide on primary human bone marrow mesenchymal stem cells. Sci Rep 2023; 13:21787. [PMID: 38066109 PMCID: PMC10709330 DOI: 10.1038/s41598-023-48448-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Traumatic or degenerative joint pain is abundant in the population. Symptom relief by intra- and periarticular glucocorticoid administration is frequently used, however may have potentially devastating effects, changing the normal healing process of the joint. Mesenchymal stem cells (MSCs) are important for wound-healing processes due to their multipotency in regenerating osteoblasts, chondrocytes and adipocytes but also have immunomodulatory properties. The aim of this study was to investigate the impact of triamcinolone acetonide (TA) a common glucocorticoid administrated intra- and periarticularly, on human bone marrow derived MSC viability, functionality, multi-lineage differentiation and transcriptomic output. We found that TA treatment induced apoptosis and promoted adipogenesis while impairing chondrogenesis of MSCs. RNA sequencing indicated that TA modulated the inflammatory response of MSCs, which may have an impact on the immunologic environment where the inflammatory phase is a physiological part of the natural healing process. These data indicate that triamcinolone acetonide should be used with consideration bearing the patient's outcome in mind, with the intention to optimize joint recovery and homeostasis.
Collapse
Affiliation(s)
- Maritha Kumlin
- Division of Orthopaedics and Biotechnology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, K54, 141 86, Stockholm, Sweden.
| | - Johanna Ungerstedt
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute, Stockholm, Sweden
- ME Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Huan Cai
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute, Stockholm, Sweden
| | - Elory Leonard
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute, Stockholm, Sweden
| | - Li Felländer-Tsai
- Division of Orthopaedics and Biotechnology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, K54, 141 86, Stockholm, Sweden
| | - Hong Qian
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
21
|
Mahjoor M, Fakouri A, Farokhi S, Nazari H, Afkhami H, Heidari F. Regenerative potential of mesenchymal stromal cells in wound healing: unveiling the influence of normoxic and hypoxic environments. Front Cell Dev Biol 2023; 11:1245872. [PMID: 37900276 PMCID: PMC10603205 DOI: 10.3389/fcell.2023.1245872] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/11/2023] [Indexed: 10/31/2023] Open
Abstract
The innate and adaptive immune systems rely on the skin for various purposes, serving as the primary defense against harmful environmental elements. However, skin lesions may lead to undesirable consequences such as scarring, accelerated skin aging, functional impairment, and psychological effects over time. The rising popularity of mesenchymal stromal cells (MSCs) for skin wound treatment is due to their potential as a promising therapeutic option. MSCs offer advantages in terms of differentiation capacity, accessibility, low immunogenicity, and their central role in natural wound-healing processes. To accelerate the healing process, MSCs promote cell migration, angiogenesis, epithelialization, and granulation tissue development. Oxygen plays a critical role in the formation and expansion of mammalian cells. The term "normoxia" refers to the usual oxygen levels, defined at 20.21 percent oxygen (160 mm of mercury), while "hypoxia" denotes oxygen levels of 2.91 percent or less. Notably, the ambient O2 content (20%) in the lab significantly differs from the 2%-9% O2 concentration in their natural habitat. Oxygen regulation of hypoxia-inducible factor-1 (HIF-1) mediated expression of multiple genes plays a crucial role in sustaining stem cell destiny concerning proliferation and differentiation. This study aims to elucidate the impact of normoxia and hypoxia on MSC biology and draw comparisons between the two. The findings suggest that expanding MSC-based regenerative treatments in a hypoxic environment can enhance their growth kinetics, genetic stability, and expression of chemokine receptors, ultimately increasing their effectiveness.
Collapse
Affiliation(s)
- Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arshia Fakouri
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hojjatollah Nazari
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Fatemeh Heidari
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
22
|
Soh S, Han S, Ka HI, Mun SH, Kim W, Oh G, Yang Y. Adiponectin affects the migration ability of bone marrow-derived mesenchymal stem cells via the regulation of hypoxia inducible factor 1α. Cell Commun Signal 2023; 21:158. [PMID: 37370133 PMCID: PMC10294307 DOI: 10.1186/s12964-023-01143-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/22/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Bone marrow (BM) is progressively filled with adipocytes during aging process. Thus, BM adipocytes-derived adiponectin (APN) affects the function of bone marrow-derived mesenchymal stem cells (BMSCs). However, little is known about the effect of APN on migration ability of BMSCs cultured under hypoxic conditions, which is similar to the BM microenvironment. RESULTS We found that the population and migration ability of BMSCs from APN KO mice was higher than that of WT mice due to increased stability of hypoxia inducible factor 1α (HIF1α). Stem cell factor (SCF)-activated STAT3 stimulated the induction of HIF1α which further stimulated SCF production, indicating that the SCF/STAT3/HIF1α positive loop was highly activated in the absence of APN. It implies that APN negatively regulated this positive loop by stimulating HIF1α degradation via the inactivation of GSK3β. Furthermore, APN KO BMSCs were highly migratory toward EL-4 lymphoma, and the interaction between CD44 in BMSCs and hyaluronic acid (HA) from EL-4 enhanced the migration of BMSCs. On the other hand, the migrated BMSCs recruited CD8+ T cells into the EL-4 tumor tissue, resulting in the retardation of tumor growth. Additionally, gradually increased APN in BM on the aging process affects migration and related functions of BMSCs, thus aged APN KO mice showed more significant suppression of EL-4 growth than young APN KO mice due to higher migration and recruitment of CD8+ T cells. CONCLUSION APN deficiency enhances CD44-mediated migration ability of BMSCs in the hypoxic conditions by the SCF/STAT3/HIF1α positive loop and influences the migration ability of BMSCs for a longer time depending on the aging process. Video Abstract.
Collapse
Affiliation(s)
- Sujung Soh
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Sora Han
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Hye In Ka
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Se Hwan Mun
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Woojung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Gaeun Oh
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Young Yang
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
23
|
Ong HT, Prêle CM, Dilley RJ. Using RNA-seq to identify suitable housekeeping genes for hypoxia studies in human adipose-derived stem cells. BMC Mol Cell Biol 2023; 24:16. [PMID: 37062833 PMCID: PMC10108514 DOI: 10.1186/s12860-023-00475-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/15/2023] [Indexed: 04/18/2023] Open
Abstract
BACKGROUND Hypoxic culture conditions have been used to study the impact of oxygen deprivation has on gene expression in a number of disease models. However, hypoxia response elements present in the promoter regions of some commonly used housekeeping genes, such as GAPDH and PGK1, can confound the relative gene expression analysis. Thus, there is ongoing debate as to which housekeeping gene is appropriate for studies investigating hypoxia-induced cell responses. Specifically, there is still contradicting information for which housekeeping genes are stable in hypoxia cultures of mesenchymal stem cells. In this study, candidate housekeeping genes curated from the literature were matched to RNAseq data of normoxic and hypoxic human adipose-derived stem cell cultures to determine if gene expression was modulated by hypoxia or not. Expression levels of selected candidates were used to calculate coefficient of variation. Then, accounting for the mean coefficient of variation, and normalised log twofold change, genes were ranked and shortlisted, before validating with qRT-PCR. Housekeeping gene suitability were then determined using GeNorm, NormFinder, BestKeeper, comparative[Formula: see text], RefFinder, and the Livak method. RESULTS Gene expression levels of 78 candidate genes identified in the literature were analysed in the RNAseq dataset generated from hADSC cultured under Nx and Hx conditions. From the dataset, 15 candidates with coefficient of variation ≤ 0.15 were identified, where differential expression analysis results further shortlisted 8 genes with least variation in expression levels. The top 4 housekeeping gene candidates, ALAS1, RRP1, GUSB, and POLR2B, were chosen for qRT-PCR validation. Additionally, 18S, a ribosomal RNA commonly used as housekeeping gene but not detected in the RNAseq method, was added to the list of housekeeping gene candidates to validate. From qRT-PCR results, 18S and RRP1 were determined to be stably expressed in cells cultured under hypoxic conditions. CONCLUSIONS We have demonstrated that 18S and RRP1 are suitable housekeeping genes for use in hypoxia studies with human adipose-derived stem cell and should be used in combination. Additionally, these data shown that the commonly used GAPDH and PGK1 are not suitable housekeeping genes for investigations into the effect of hypoxia in human adipose-derived stem cell.
Collapse
Affiliation(s)
- Huan Ting Ong
- Ear Science Institute Australia, Nedlands, Western Australia, Australia.
- Ear Sciences Centre, The University of Western Australia, Nedlands, Western Australia, Australia.
| | - Cecilia M Prêle
- Ear Science Institute Australia, Nedlands, Western Australia, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- Institute for Respiratory Health, The University of Western Australia, Nedlands, Western Australia, Australia
- Discipline of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, Western Australia, Australia
| | - Rodney J Dilley
- Ear Science Institute Australia, Nedlands, Western Australia, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
24
|
Theodoridis K, Aggelidou E, Manthou ME, Kritis A. Hypoxia Promotes Cartilage Regeneration in Cell-Seeded 3D-Printed Bioscaffolds Cultured with a Bespoke 3D Culture Device. Int J Mol Sci 2023; 24:ijms24076040. [PMID: 37047021 PMCID: PMC10094683 DOI: 10.3390/ijms24076040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
In this study, we investigated the effect of oxygen tension on the expansion of ADMSCs and on their differentiation toward their chondrocytic phenotype, regenerating a lab-based cartilaginous tissue with superior characteristics. Controversial results with reference to MSCs that were cultured under different hypoxic levels, mainly in 2D culturing settings combined with or without other biochemical stimulus factors, prompted our team to study the role of hypoxia on MSCs chondrogenic differentiation within an absolute 3D environment. Specifically, we used 3D-printed honeycomb-like PCL matrices seeded with ADMSCs in the presence or absence of TGF and cultured with a prototype 3D cell culture device, which was previously shown to favor nutrient/oxygen supply, cell adhesion, and infiltration within scaffolds. These conditions resulted in high-quality hyaline cartilage that was distributed uniformly within scaffolds. The presence of the TGF medium was necessary to successfully produce cartilaginous tissues with superior molecular and increased biomechanical properties. Despite hypoxia's beneficial effect, it was overall not enough to fully differentiate ADMSCs or even promote cell expansion within 3D scaffolds alone.
Collapse
Affiliation(s)
- Konstantinos Theodoridis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- CGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
| | - Eleni Aggelidou
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- CGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- Basic and Translational Research Unit (BTRU) of Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
| | - Maria-Eleni Manthou
- Laboratory of Histology, Embryology and Anthropology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
| | - Aristeidis Kritis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- CGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- Basic and Translational Research Unit (BTRU) of Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
| |
Collapse
|
25
|
Cahill T, da Silveira WA, Renaud L, Wang H, Williamson T, Chung D, Chan S, Overton I, Hardiman G. Investigating the effects of chronic low-dose radiation exposure in the liver of a hypothermic zebrafish model. Sci Rep 2023; 13:918. [PMID: 36650199 PMCID: PMC9845366 DOI: 10.1038/s41598-022-26976-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/22/2022] [Indexed: 01/18/2023] Open
Abstract
Mankind's quest for a manned mission to Mars is placing increased emphasis on the development of innovative radio-protective countermeasures for long-term space travel. Hibernation confers radio-protective effects in hibernating animals, and this has led to the investigation of synthetic torpor to mitigate the deleterious effects of chronic low-dose-rate radiation exposure. Here we describe an induced torpor model we developed using the zebrafish. We explored the effects of radiation exposure on this model with a focus on the liver. Transcriptomic and behavioural analyses were performed. Radiation exposure resulted in transcriptomic perturbations in lipid metabolism and absorption, wound healing, immune response, and fibrogenic pathways. Induced torpor reduced metabolism and increased pro-survival, anti-apoptotic, and DNA repair pathways. Coupled with radiation exposure, induced torpor led to a stress response but also revealed maintenance of DNA repair mechanisms, pro-survival and anti-apoptotic signals. To further characterise our model of induced torpor, the zebrafish model was compared with hepatic transcriptomic data from hibernating grizzly bears (Ursus arctos horribilis) and active controls revealing conserved responses in gene expression associated with anti-apoptotic processes, DNA damage repair, cell survival, proliferation, and antioxidant response. Similarly, the radiation group was compared with space-flown mice revealing shared changes in lipid metabolism.
Collapse
Affiliation(s)
- Thomas Cahill
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK
| | - Willian Abraham da Silveira
- School of Health, Science and Wellbeing, Department of Biological Sciences, Science Centre, Staffordshire University, Leek Road, Stoke-On-Trent, ST4 2DF, UK
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France
| | - Ludivine Renaud
- Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Hao Wang
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK
| | - Tucker Williamson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Sherine Chan
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
- JLABS at the Children's National Research and Innovation Campus, Washington, DC, 20012, USA
| | - Ian Overton
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Gary Hardiman
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK.
- Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
26
|
Murphy RJ, Gunasingh G, Haass NK, Simpson MJ. Growth and adaptation mechanisms of tumour spheroids with time-dependent oxygen availability. PLoS Comput Biol 2023; 19:e1010833. [PMID: 36634128 PMCID: PMC9876349 DOI: 10.1371/journal.pcbi.1010833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/25/2023] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Tumours are subject to external environmental variability. However, in vitro tumour spheroid experiments, used to understand cancer progression and develop cancer therapies, have been routinely performed for the past fifty years in constant external environments. Furthermore, spheroids are typically grown in ambient atmospheric oxygen (normoxia), whereas most in vivo tumours exist in hypoxic environments. Therefore, there are clear discrepancies between in vitro and in vivo conditions. We explore these discrepancies by combining tools from experimental biology, mathematical modelling, and statistical uncertainty quantification. Focusing on oxygen variability to develop our framework, we reveal key biological mechanisms governing tumour spheroid growth. Growing spheroids in time-dependent conditions, we identify and quantify novel biological adaptation mechanisms, including unexpected necrotic core removal, and transient reversal of the tumour spheroid growth phases.
Collapse
Affiliation(s)
- Ryan J. Murphy
- Mathematical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- * E-mail:
| | - Gency Gunasingh
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Nikolas K. Haass
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Matthew J. Simpson
- Mathematical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
27
|
Ren X, Gao X, Cheng Y, Xie L, Tong L, Li W, Chu PK, Wang H. Maintenance of multipotency of bone marrow mesenchymal stem cells on poly(ε-caprolactone) nanoneedle arrays through the enhancement of cell-cell interaction. Front Bioeng Biotechnol 2023; 10:1076345. [PMID: 36698633 PMCID: PMC9870049 DOI: 10.3389/fbioe.2022.1076345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
Mesenchymal stem cells (MSCs), with high self-renewal ability and multipotency, are commonly used as the seed cells for tissue engineering. However, the reduction and loss of multipotential ability after necessary expansion in vitro set up a heavy obstacle to the clinical application of MSCs. Here in this study, we exploit the autologous crystallization ability of biocompatible poly (ε-caprolactone) (PCL) to obtain uniformly distributed nanoneedle arrays. By controlling the molecular weight of PCL, nanoneedle with a width of 2 μm and height of 50 nm, 80 nm, and 100 nm can be successfully fabricated. After surface chemical modification with polydopamine (PDA), the water contact angle of the fabricated PCL nanoneedle arrays are reduced from 84° to almost 60° with no significant change of the nanostructure. All the fabricated substrates are cultured with bone marrow MSCs (BMMSCs), and the adhesion, spreading, proliferation ability and multipotency of cells on different substrates are investigated. Compared with the BMMSCs cultured on pure PCL nanoneedle arrays, the decoration of PDA can improve the adhesion and spreading of cells and further change them from aggregated distribution to laminar distribution. Nevertheless, the laminar distribution of cultured cells leads to a weak cell-cell interaction, and hence the multipotency of BMMSCs cultured on the PCL-PDA substrates is decimated. On the contrary, the pure PCL nanoneedle arrays can be used to maintain the multipotency of BMMSCs via clustered growth, and the PCL1 nanoneedle array with a height of 50 nm is more promising than the other 2 with regard to the highest proliferation rate and best multipotential differentiation ability of cultured cells. Interestingly, there is a positive correlation between the strength of cell-cell interaction and the multipotency of stem cells in vitro. In conclusion, we have successfully maintained the multipotency of BMMSCs by using the PCL nanoneedle arrays, especially the PCL1 nanoneedle array with a height of 50 nm, as the substrates for in vitro extension, and further revealed the importance of cell-cell interaction on the multipotency of MSCs. The study provides a theoretical basis for the behavioral regulation of MSCs, and is instructive to the design of tissue engineering scaffolds.
Collapse
Affiliation(s)
- Xiaoxue Ren
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaoting Gao
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China,University of Chinese Academy of Sciences, Beijing, China
| | - Yicheng Cheng
- Department of Stomatology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China,*Correspondence: Yicheng Cheng, ; Wei Li, ; Huaiyu Wang,
| | - Lingxia Xie
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Liping Tong
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China,*Correspondence: Yicheng Cheng, ; Wei Li, ; Huaiyu Wang,
| | - Paul K. Chu
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Huaiyu Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China,*Correspondence: Yicheng Cheng, ; Wei Li, ; Huaiyu Wang,
| |
Collapse
|
28
|
Sivakumar S, Lieber S, Librizzi D, Keber C, Sommerfeld L, Finkernagel F, Roth K, Reinartz S, Bartsch JW, Graumann J, Müller‐Brüsselbach S, Müller R. Basal cell adhesion molecule promotes metastasis-associated processes in ovarian cancer. Clin Transl Med 2023; 13:e1176. [PMID: 36647260 PMCID: PMC9842900 DOI: 10.1002/ctm2.1176] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Basal cell adhesion molecule (BCAM) is a laminin α5 (LAMA5) binding membrane-bound protein with a putative role in cancer. Besides full-length BCAM1, an isoform lacking most of the cytoplasmic domain (BCAM2), and a soluble form (sBCAM) of unknown function are known. In ovarian carcinoma (OC), all BCAM forms are abundant and associated with poor survival, yet BCAM's contribution to peritoneal metastatic spread remains enigmatic. METHODS Biochemical, omics-based and real-time cell assays were employed to identify the source of sBCAM and metastasis-related functions of different BCAM forms. OC cells, explanted omentum and a mouse model of peritoneal colonisation were used in loss- and gain-of-function experiments. RESULTS We identified ADAM10 as a major BCAM sheddase produced by OC cells and identified proteolytic cleavage sites proximal to the transmembrane domain. Recombinant soluble BCAM inhibited single-cell adhesion and migration identically to membrane-bound isoforms, confirming its biological activity in OC. Intriguingly, this seemingly anti-tumorigenic potential of BCAM contrasts with a novel pro-metastatic function discovered in the present study. Thus, all queried BCAM forms decreased the compactness of tumour cell spheroids by inhibiting LAMA5 - integrin β1 interactions, promoted spheroid dispersion in a three-dimensional collagen matrix, induced clearance of mesothelial cells at spheroid attachment sites in vitro and enhanced invasion of spheroids into omental tissue both ex vivo and in vivo. CONCLUSIONS Membrane-bound BCAM as well as sBCAM shed by ADAM10 act as decoys rather than signalling receptors to modulate metastasis-related functions. While BCAM appears to have tumour-suppressive effects on single cells, it promotes the dispersion of OC cell spheroids by regulating LAMA5-integrin-β1-dependent compaction and thereby facilitating invasion of metastatic target sites. As peritoneal dissemination is majorly mediated by spheroids, these findings offer an explanation for the association of BCAM with a poor clinical outcome of OC, suggesting novel therapeutic options.
Collapse
Affiliation(s)
- Suresh Sivakumar
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Sonja Lieber
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Damiano Librizzi
- Small Animal Imaging Core FacilityCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Corinna Keber
- Institute for PathologyPhilipps UniversityMarburgGermany
| | - Leah Sommerfeld
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Florian Finkernagel
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
- Bioinformatics Core FacilityCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Katrin Roth
- Cell Imaging Core FacilityCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Silke Reinartz
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | | | - Johannes Graumann
- Biomolecular Mass SpectrometryMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Institute for Translational ProteomicsPhilipps UniversityMarburgGermany
| | - Sabine Müller‐Brüsselbach
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Rolf Müller
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| |
Collapse
|
29
|
da Graça Cabreira M, Wang X, Critsinelis A, Setegne M, Lotfi P, Wan YW, Barrios G, Mei Z, Gee AP, Buja LM, Perin E. Environmental oxygen affects ex vivo growth and proliferation of mesenchymal progenitors by modulating mitogen-activated protein kinase and mammalian target of rapamycin signaling. Cytotherapy 2022; 24:1201-1210. [PMID: 36109320 DOI: 10.1016/j.jcyt.2022.06.005] [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: 12/16/2021] [Revised: 04/16/2022] [Accepted: 06/13/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AIMS Stem and progenitor cells of hematopoietic and mesenchymal lineages reside in the bone marrow under low oxygen (O2) saturation. O2 levels used in ex vivo expansion of multipotent mesenchymal stromal cells (MSCs) affect proliferation, metabolism and differentiation. METHODS Using cell-based assays and transcriptome and proteome data, the authors compared MSC cultures simultaneously grown under a conventional 19.95% O2 atmosphere or at 5% O2. RESULTS In 5% O2, MSCs showed better proliferation and higher self-renewal ability, most probably sustained by enhanced signaling activity of mitogen-activated protein kinase and mammalian target of rapamycin pathways. Non-oxidative glycolysis-based energy metabolism supported growth and proliferation in 5% O2 cultures, whereas MSCs grown under 19.95% O2 also utilized oxidative phosphorylation. Cytoprotection mechanisms used by cells under 5% O2 differed from 19.95% O2 suggesting differences in the triggers of cell stress between these two O2 conditions. CONCLUSIONS Based on the potential benefits for the growth and metabolism of MSCs, the authors propose the use of 5% O2 for MSC culture.
Collapse
Affiliation(s)
| | - Xiaohong Wang
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Mekedlawit Setegne
- Chemistry-Biology Interface Predoctoral Training Program, Stanford University, Stanford, California, USA
| | - Parisa Lotfi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Ying-Wooi Wan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Gabriela Barrios
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, Texas, USA
| | - Zhuyong Mei
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas, USA
| | - Adrian P Gee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas, USA
| | - Louis Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, USA
| | - Emerson Perin
- Center for Clinical Research, Texas Heart Institute, Houston, Texas, USA
| |
Collapse
|
30
|
Zhang J, Zhang W, Sun T, Wang J, Li Y, Liu J, Li Z. The Influence of Intervertebral Disc Microenvironment on the Biological Behavior of Engrafted Mesenchymal Stem Cells. Stem Cells Int 2022; 2022:8671482. [PMID: 36387746 PMCID: PMC9663214 DOI: 10.1155/2022/8671482] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 12/01/2024] Open
Abstract
Intervertebral disc degeneration is the main cause of low back pain. Traditional treatment methods cannot repair degenerated intervertebral disc tissue. The emergence of stem cell therapy makes it possible to regenerate and repair degenerated intervertebral disc tissue. At present, mesenchymal stem cells are the most studied, and different types of mesenchymal stem cells have their own characteristics. However, due to the harsh and complex internal microenvironment of the intervertebral disc, it will affect the biological behaviors of the implanted mesenchymal stem cells, such as viability, proliferation, migration, and chondrogenic differentiation, thereby affecting the therapeutic effect. This review is aimed at summarizing the influence of each intervertebral disc microenvironmental factor on the biological behavior of mesenchymal stem cells, so as to provide new ideas for using tissue engineering technology to assist stem cells to overcome the influence of the microenvironment in the future.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011 Liaoning, China
| | - Wentao Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011 Liaoning, China
| | - Tianze Sun
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011 Liaoning, China
| | - Jinzuo Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011 Liaoning, China
| | - Ying Li
- Stem Cell Clinical Research Centers, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021 Liaoning, China
| | - Jing Liu
- Stem Cell Clinical Research Centers, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021 Liaoning, China
| | - Zhonghai Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, 116011 Liaoning, China
- Stem Cell Clinical Research Centers, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021 Liaoning, China
| |
Collapse
|
31
|
Pachane BC, Nunes ACC, Cataldi TR, Micocci KC, Moreira BC, Labate CA, Selistre-de-Araujo HS, Altei WF. Small Extracellular Vesicles from Hypoxic Triple-Negative Breast Cancer Cells Induce Oxygen-Dependent Cell Invasion. Int J Mol Sci 2022; 23:ijms232012646. [PMID: 36293503 PMCID: PMC9604480 DOI: 10.3390/ijms232012646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 12/04/2022] Open
Abstract
Hypoxia, a condition of low oxygenation frequently found in triple-negative breast tumors (TNBC), promotes extracellular vesicle (EV) secretion and favors cell invasion, a complex process in which cell morphology is altered, dynamic focal adhesion spots are created, and ECM is remodeled. Here, we investigated the invasive properties triggered by TNBC-derived hypoxic small EV (SEVh) in vitro in cells cultured under hypoxic (1% O2) and normoxic (20% O2) conditions, using phenotypical and proteomic approaches. SEVh characterization demonstrated increased protein abundance and diversity over normoxic SEV (SEVn), with enrichment in pro-invasive pathways. In normoxic cells, SEVh promotes invasive behavior through pro-migratory morphology, invadopodia development, ECM degradation, and matrix metalloprotease (MMP) secretion. The proteome profiling of 20% O2-cultured cells exposed to SEVh determined enrichment in metabolic processes and cell cycles, modulating cell health to escape apoptotic pathways. In hypoxia, SEVh was responsible for proteolytic and catabolic pathway inducement, interfering with integrin availability and gelatinase expression. Overall, our results demonstrate the importance of hypoxic signaling via SEV in tumors for the early establishment of metastasis.
Collapse
Affiliation(s)
- Bianca Cruz Pachane
- Biochemistry and Molecular Biology Laboratory, Department of Physiological Sciences, Universidade Federal de São Carlos—UFSCar, São Carlos 13565-905, SP, Brazil
| | - Ana Carolina Caetano Nunes
- Biochemistry and Molecular Biology Laboratory, Department of Physiological Sciences, Universidade Federal de São Carlos—UFSCar, São Carlos 13565-905, SP, Brazil
| | - Thais Regiani Cataldi
- Max Feffer Plant Genetics Laboratory, Department of Genetics, University of São Paulo—ESALQ, Piracicaba 13418-900, SP, Brazil
| | - Kelli Cristina Micocci
- Center for the Study of Social Insects, São Paulo State University “Julio de Mesquita Filho”, Rio Claro 14884-900, SP, Brazil
| | - Bianca Caruso Moreira
- Biochemistry and Molecular Biology Laboratory, Department of Physiological Sciences, Universidade Federal de São Carlos—UFSCar, São Carlos 13565-905, SP, Brazil
| | - Carlos Alberto Labate
- Max Feffer Plant Genetics Laboratory, Department of Genetics, University of São Paulo—ESALQ, Piracicaba 13418-900, SP, Brazil
| | - Heloisa Sobreiro Selistre-de-Araujo
- Biochemistry and Molecular Biology Laboratory, Department of Physiological Sciences, Universidade Federal de São Carlos—UFSCar, São Carlos 13565-905, SP, Brazil
| | - Wanessa Fernanda Altei
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil
- Radiation Oncology Department, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil
- Correspondence:
| |
Collapse
|
32
|
Qin Q, Liu Y, Yang Z, Aimaijiang M, Ma R, Yang Y, Zhang Y, Zhou Y. Hypoxia-Inducible Factors Signaling in Osteogenesis and Skeletal Repair. Int J Mol Sci 2022; 23:ijms231911201. [PMID: 36232501 PMCID: PMC9569554 DOI: 10.3390/ijms231911201] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sufficient oxygen is required to maintain normal cellular and physiological function, such as a creature’s development, breeding, and homeostasis. Lately, some researchers have reported that both pathological hypoxia and environmental hypoxia might affect bone health. Adaptation to hypoxia is a pivotal cellular event in normal cell development and differentiation and in pathological settings such as ischemia. As central mediators of homeostasis, hypoxia-inducible transcription factors (HIFs) can allow cells to survive in a low-oxygen environment and are essential for the regulation of osteogenesis and skeletal repair. From this perspective, we summarized the role of HIF-1 and HIF-2 in signaling pathways implicated in bone development and skeletal repair and outlined the molecular mechanism of regulation of downstream growth factors and protein molecules such as VEGF, EPO, and so on. All of these present an opportunity for developing therapies for bone regeneration.
Collapse
|
33
|
Tian Y, Fang J, Zeng F, Chen Y, Pei Y, Gu F, Ding C, Niu G, Gu B. The role of hypoxic mesenchymal stem cells in tumor immunity. Int Immunopharmacol 2022; 112:109172. [PMID: 36087506 DOI: 10.1016/j.intimp.2022.109172] [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: 07/08/2022] [Revised: 08/06/2022] [Accepted: 08/14/2022] [Indexed: 11/09/2022]
Abstract
The emerging evidence has shown that mesenchymal stem cells (MSCs) not only exert a significant role in the occurrence and development of tumors, but also have immunosuppressive potential in tumor immunity. Hypoxia is a sign of solid tumors, but how functions of hypoxic MSCs alter in the tumor microenvironment (TME) remains less well and comprehensively described. Herein, we mostly describe and investigate recent advances in our comprehension of the emerging effects of different tissue derived MSCs in hypoxia condition on tumor progression and development, as well as bidirectional influence between hypoxic MSCs and immune cells of the TME. Furthermore, we also discuss the potential drug-resistant and therapeutic role of hypoxic MSCs. It can be envisaged that novel and profound insights into the functionality of hypoxic MSCs and the underlying mechanisms in tumor and tumor immunity will promote the meaningful and promising treatment strategies against tumor.
Collapse
Affiliation(s)
- Yiqing Tian
- Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Jian Fang
- The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230022, Anhui, PR China
| | - Fanpeng Zeng
- Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Yongqiang Chen
- Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Yunfeng Pei
- Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Feng Gu
- Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Chen Ding
- Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China.
| | - Guoping Niu
- Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China.
| | - Bing Gu
- Laboratory Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510000, PR China.
| |
Collapse
|
34
|
The secretome obtained under hypoxic preconditioning from human adipose-derived stem cells exerts promoted anti-apoptotic potentials through upregulated autophagic process. Mol Biol Rep 2022; 49:8859-8870. [PMID: 35941418 DOI: 10.1007/s11033-022-07736-z] [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: 07/21/2021] [Revised: 06/14/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Hypoxic preconditioning (HP) is a stem cell preconditioning modality designed to augment the therapeutic effects of mesenchymal stem cells (MSCs). Although autophagy is expected to play a role in HP, very little is known regarding the relationship between HP and autophagy. METHODS AND RESULTS The adipose-derived stem cell (ASC)-secretome obtained under normoxia (NCM) and ASC-secretome obtained under HP (HCM) were obtained by culturing ASCs for 24 h under normoxic (21% partial pressure of O2) and hypoxic (1% partial pressure of O2) conditions, respectively. Subsequently, to determine the in vivo effects of HCM, each secretome was injected into 70% partially hepatectomized mice, and liver specimens were obtained. HCM significantly reduced the apoptosis of thioacetamide-treated AML12 hepatocytes and promoted the autophagic processes of the cells (P < 0.05). Autophagy blockage by either bafilomycin A1 or ATG5 siRNA significantly abrogated the anti-apoptotic effect of HCM (P < 0.05), demonstrating that HCM exerts its anti-apoptotic effect by promoting autophagy. The effect of HCM - reduction of cell apoptosis and promotion of autophagic process - was also demonstrated in a mouse model. CONCLUSIONS HP appears to induce ASCs to release a secretome with enhanced anti-apoptotic effects by promoting the autophagic process of ASCs.
Collapse
|
35
|
Alsobaie S, Alsobaie T, Mantalaris S. Rho-Associated Protein Kinase Inhibitor and Hypoxia Synergistically Enhance the Self-Renewal, Survival Rate, and Proliferation of Human Stem Cells. STEM CELLS AND CLONING: ADVANCES AND APPLICATIONS 2022; 15:43-52. [PMID: 35812359 PMCID: PMC9259205 DOI: 10.2147/sccaa.s365776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/22/2022] [Indexed: 01/16/2023]
Abstract
Introduction High-efficacy single-cell cloning of human-induced pluripotent cells (IPSCs) remains a major challenge. The development of a culture method that supports single-cell passaging while maintaining reproducibility, homogeneity, scalability, and cell expansion to clinically relevant numbers is necessary for clinical application. Methods To address this issue, we combined the use of the rho-associated protein kinase (ROCK) inhibitor Y-27632 and hypoxic conditions in culture to produce a novel, efficient single-cell culture method for human IPSCs and embryonic stem cells. Results Through immunocytochemistry, alkaline phosphatase assays, and flow cytometry, we demonstrated that our method enabled high single-cell proliferation while maintaining self-renewal and pluripotency abilities. Discussion We showed the beneficial effect of the interaction between hypoxia and ROCK inhibition in regulating cell proliferation, pluripotency, and single-cell survival of pluripotent cells.
Collapse
Affiliation(s)
- Sarah Alsobaie
- Department of Clinical Laboratory Science, King Saud University, Riyadh, 11451, Saudi Arabia
- Correspondence: Sarah Alsobaie, Department of Clinical Laboratory Science, King Saud University, Prince Turki Alawal Street, Riyadh, 11451, Saudi Arabia, Tel +966 507191011, Fax +966 114677580, Email
| | - Tamador Alsobaie
- Biological Systems Engineering Laboratory, Department of Chemical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Sakis Mantalaris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
| |
Collapse
|
36
|
Taurino G, Deshmukh R, Villar VH, Chiu M, Shaw R, Hedley A, Shokry E, Sumpton D, Dander E, D'Amico G, Bussolati O, Tardito S. Mesenchymal stromal cells cultured in physiological conditions sustain citrate secretion with glutamate anaplerosis. Mol Metab 2022; 63:101532. [PMID: 35752287 PMCID: PMC9254159 DOI: 10.1016/j.molmet.2022.101532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/31/2022] [Accepted: 06/17/2022] [Indexed: 11/19/2022] Open
Abstract
Bone marrow mesenchymal stromal cells (MSCs) have immunomodulatory and regenerative potential. However, culture conditions govern their metabolic processes and therapeutic efficacy. Here we show that culturing donor-derived MSCs in Plasmax™, a physiological medium with the concentrations of nutrients found in human plasma, supports their proliferation and stemness, and prevents the nutritional stress induced by the conventional medium DMEM. The quantification of the exchange rates of metabolites between cells and medium, untargeted metabolomics, stable isotope tracing and transcriptomic analysis, performed at physiologically relevant oxygen concentrations (1%O2), reveal that MSCs rely on high rate of glucose to lactate conversion, coupled with parallel anaplerotic fluxes from glutamine and glutamate to support citrate synthesis and secretion. These distinctive traits of MSCs shape the metabolic microenvironment of bone marrow niche and can influence nutrient cross-talks under physiological and pathological conditions.
Collapse
Affiliation(s)
- Giuseppe Taurino
- Laboratory of General Pathology, Dept. of Medicine and Surgery, University of Parma, 43125, Parma, Italy; MRH - Microbiome Research Hub, Parco Area delle Scienze 11/A, University of Parma, 43124 Parma, Italy
| | - Ruhi Deshmukh
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Victor H Villar
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Martina Chiu
- Laboratory of General Pathology, Dept. of Medicine and Surgery, University of Parma, 43125, Parma, Italy
| | - Robin Shaw
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Ann Hedley
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Engy Shokry
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - David Sumpton
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Erica Dander
- Centro Ricerca Tettamanti, Pediatric Dept., University of Milano-Bicocca, Fondazione MBBM, Monza, 20900, Italy
| | - Giovanna D'Amico
- Centro Ricerca Tettamanti, Pediatric Dept., University of Milano-Bicocca, Fondazione MBBM, Monza, 20900, Italy
| | - Ovidio Bussolati
- Laboratory of General Pathology, Dept. of Medicine and Surgery, University of Parma, 43125, Parma, Italy; MRH - Microbiome Research Hub, Parco Area delle Scienze 11/A, University of Parma, 43124 Parma, Italy.
| | - Saverio Tardito
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
37
|
Effects of Hypoxia on Proliferation and Differentiation in Belgian Blue and Hanwoo Muscle Satellite Cells for the Development of Cultured Meat. Biomolecules 2022; 12:biom12060838. [PMID: 35740963 PMCID: PMC9221279 DOI: 10.3390/biom12060838] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 02/01/2023] Open
Abstract
Among future food problems, the demand for meat is expected to increase rapidly, but the production efficiency of meat, which is a protein source, is very low compared to other foods. To address this problem, research on the development and production of cultured meat as an alternative meat source using muscle stem cells in vitro has recently been undertaken. Many studies have been conducted on myosatellite cells for medical purposes, but studies on alternative meat production are rare. In vitro cell culture mimics the in vivo environment for cell growth. The satellite cell niche is closer to hypoxic (2% O2) than normoxic (20% O2) conditions. The aim of this study was to investigate the efficient oxygen conditions of myosatellite cell cultures for the production of cultured meat. The bovine satellite cell counts and mRNA (Pax7, Myf5 and HIF1α) levels were higher in hypoxia than normoxia (p < 0.05). Through Hoechst-positive nuclei counts, and expression of Pax7, MyoD and myosin protein by immunofluorescence, it was confirmed that muscle cells performed normal proliferation and differentiation. Myoblast fusion was higher under hypoxic conditions (p < 0.05), and the myotube diameters were also thicker (p < 0.05). In the myotube, the number of cells was high in hypoxia, and the expression of the total protein amounts, differentiation marker mRNA (myogenin, myosin and TOM20), and protein markers (myosin and TOM20) was also high. The study results demonstrated that the proliferation and differentiation of bovine myosatellite cells were promoted more highly under hypoxic conditions than under normoxic conditions. Therefore, hypoxic cultures that promote the proliferation and differentiation of bovine myosatellite cells may be an important factor in the development of cultured meat.
Collapse
|
38
|
Silvestro S, Diomede F, Chiricosta L, Zingale VD, Marconi GD, Pizzicannella J, Valeri A, Avanzini MA, Calcaterra V, Pelizzo G, Mazzon E. The Role of Hypoxia in Improving the Therapeutic Potential of Mesenchymal Stromal Cells. A Comparative Study From Healthy Lung and Congenital Pulmonary Airway Malformations in Infants. Front Bioeng Biotechnol 2022; 10:868486. [PMID: 35774062 PMCID: PMC9237219 DOI: 10.3389/fbioe.2022.868486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) play an important role in the field of regenerative medicine thanks to their immunomodulatory properties and their ability to secrete paracrine factors. The use of MSCs has also been tested in children with congenital lung diseases inducing fibrosis and a decrease in lung function. Congenital malformations of the pulmonary airways (CPAM) are the most frequently encountered lung lesion that results from defects in early development of airways. Despite the beneficial properties of MSCs, interventions aimed at improving the outcome of cell therapy are needed. Hypoxia may be an approach aimed to ameliorate the therapeutic potential of MSCs. In this regard, we evaluated the transcriptomic profile of MSCs collected from pediatric patients with CPAM, analyzing similarities and differences between healthy tissue (MSCs-lung) and cystic tissue (MSCs-CPAM) both in normoxia and in cells preconditioned with hypoxia (0.2%) for 24 h. Study results showed that hypoxia induces cell cycle activation, increasing in such a way the cell proliferation ability, and enhancing cell anaerobic metabolism in both MSCs-lung and MSCs-CPAM-lung. Additionally, hypoxia downregulated several pro-apoptotic genes preserving MSCs from apoptosis and, at the same time, improving their viability in both comparisons. Finally, data obtained indicates that hypoxia leads to a greater expression of genes involved in the regulation of the cytoskeleton in MSCs-lung than MSCs-CPAM.
Collapse
Affiliation(s)
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | | | | | - Guya Diletta Marconi
- Department of Medical, Oral and Biotechnological Sciences, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | | | - Andrea Valeri
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy
| | - Maria Antonietta Avanzini
- Cell Factory, Pediatric Hematology Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Valeria Calcaterra
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy
- Pediatric Department, Children’s Hospital “Vittore Buzzi”, Milano, Italy
| | - Gloria Pelizzo
- Pediatric Surgery Department, Children’s Hospital “Vittore Buzzi”, Milano, Italy
- Department of Biomedical and Clinical Sciences-L. Sacco, University of Milan, Milan, Italy
| | | |
Collapse
|
39
|
Ngai HW, Kim DH, Hammad M, Gutova M, Aboody K, Cox CD. Stem Cell-based therapies for COVID-19-related acute respiratory distress syndrome. J Cell Mol Med 2022; 26:2483-2504. [PMID: 35426198 PMCID: PMC9077311 DOI: 10.1111/jcmm.17265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
As the number of confirmed cases and resulting death toll of the COVID-19 pandemic continue to increase around the globe - especially with the emergence of new mutations of the SARS-CoV-2 virus in addition to the known alpha, beta, gamma, delta and omicron variants - tremendous efforts continue to be dedicated to the development of interventive therapeutics to mitigate infective symptoms or post-viral sequelae in individuals for which vaccines are not accessible, viable or effective in the prevention of illness. Many of these investigations aim to target the associated acute respiratory distress syndrome, or ARDS, which induces damage to lung epithelia and other physiologic systems and is associated with progression in severe cases. Recently, stem cell-based therapies have demonstrated preliminary efficacy against ARDS based on a number of preclinical and preliminary human safety studies, and based on promising outcomes are now being evaluated in phase II clinical trials for ARDS. A number of candidate stem cell therapies have been found to exhibit low immunogenicity, coupled with inherent tropism to injury sites. In recent studies, these have demonstrated the ability to modulate suppression of pro-inflammatory cytokine signals such as those characterizing COVID-19-associated ARDS. Present translational studies are aiming to optimize the safety, efficacy and delivery to fully validate stem cell-based strategies targeting COVID-19 associated ARDS for viable clinical application.
Collapse
Affiliation(s)
- Hoi Wa Ngai
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Dae Hong Kim
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Mohamed Hammad
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Karen Aboody
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | | |
Collapse
|
40
|
van der Sluis N, Scheers EC, Krenning G, van der Lei B, Oonk MH, van Dongen JA. Autologous lipoaspirate as a new treatment of vulvar lichen sclerosus: A review on literature. Exp Dermatol 2022; 31:689-699. [PMID: 35276020 PMCID: PMC9314062 DOI: 10.1111/exd.14561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/03/2022] [Accepted: 03/05/2022] [Indexed: 11/29/2022]
Abstract
Lichen sclerosus (LS) is a chronic inflammatory dermatosis that mostly affects the genital and anal skin areas. Symptoms may vary from pruritis and pain to sexual dysfunction; however, LS can also be asymptomatic. LS occurs at all ages and in both sexes. Approximately 5% of all women affected by vulvar LS will develop vulvar squamous cell carcinoma. Topical treatment is safe but less effective resulting in chronic course in most patients, who suffer from persistent itching and pain. In severe cases of therapy-resistant LS, there is no adequate treatment. Fat grafting is a novel regenerative therapy to reduce dermal fibrosis. The therapeutic effect of adipose tissue grafts for LS is already investigated in various pioneering studies. This review provides an overview of these studies and the putative mechanisms-of-action of fat grafting to treat LS.
Collapse
Affiliation(s)
- Nanouk van der Sluis
- Department of Plastic SurgeryUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Department of Plastic‐, Reconstructive‐ and Hand SurgeryMedisch Spectrum TwenteEnschedeThe Netherlands
| | - Esther C.A.H. Scheers
- Department of Obstetrics and GynecologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Guido Krenning
- Department of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenGroningenThe Netherlands
| | - Berend van der Lei
- Department of Plastic SurgeryUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Maaike H.M. Oonk
- Department of Obstetrics and GynecologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Joris A. van Dongen
- Department of Plastic‐, Reconstructive‐ and Hand Surgery, Utrecht University Medical CenterUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
41
|
Impact of Microenvironmental Changes during Degeneration on Intervertebral Disc Progenitor Cells: A Comparison with Mesenchymal Stem Cells. Bioengineering (Basel) 2022; 9:bioengineering9040148. [PMID: 35447707 PMCID: PMC9025850 DOI: 10.3390/bioengineering9040148] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/22/2022] Open
Abstract
Intervertebral disc (IVD) degeneration occurs with natural ageing and is linked to low back pain, a common disease. As an avascular tissue, the microenvironment inside the IVD is harsh. During degeneration, the condition becomes even more compromised, presenting a significant challenge to the survival and function of the resident cells, as well as to any regeneration attempts using cell implantation. Mesenchymal stem cells (MSCs) have been proposed as a candidate stem cell tool for IVD regeneration. Recently, endogenous IVD progenitor cells have been identified inside the IVD, highlighting their potential for self-repair. IVD progenitor cells have properties similar to MSCs, with minor differences in potency and surface marker expression. Currently, it is unclear how IVD progenitor cells react to microenvironmental factors and in what ways they possibly behave differently to MSCs. Here, we first summarized the microenvironmental factors presented in the IVD and their changes during degeneration. Then, we analyzed the available studies on the responses of IVD progenitor cells and MSCs to these factors, and made comparisons between these two types of cells, when possible, in an attempt to achieve a clear understanding of the characteristics of IVD progenitor cells when compared to MSCs; as well as, to provide possible clues to cell fate after implantation, which may facilitate future manipulation and design of IVD regeneration studies.
Collapse
|
42
|
Marchus CR, Knudson JA, Morrison AE, Strawn IK, Hartman AJ, Shrestha D, Pancheri NM, Glasgow I, Schiele NR. Low-cost, open-source cell culture chamber for regulating physiologic oxygen levels. HARDWAREX 2022; 11:e00253. [PMID: 35509920 PMCID: PMC9058583 DOI: 10.1016/j.ohx.2021.e00253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
The physiological oxygen levels for several mammalian cell types in vivo are considered to be hypoxic (low oxygen tension), but the vast majority of in vitro mammalian cell culture is conducted at atmospheric oxygen levels of around 21%. In order to understand the impact of low oxygen environments on cells, oxygen levels need to be regulated during in vitro culture. Two common methods for simulating a hypoxic environment are through the regulation of gas composition or chemical induction. Chemically mimicking hypoxia can have adverse effects such as reducing cell viability, making oxygen regulation in cell culture chambers crucial for long-term culture. However, oxygen-regulating cell culture incubators and commercial hypoxia chambers may not always be a viable option due to cost and limited customization. Other low-cost chambers have been developed, but they tend to lack control systems or are fairly small scale. Thus, the objective of this project was to design and develop a low-cost, open-source, controllable, and reproducible hypoxia chamber that can fit inside a standard cell culture incubator. This design allows for the control of O2 between 1 and 21%, while maintaining CO2 levels at 5%, as well as monitoring of temperature, pressure, and relative humidity. Testing showed our hypoxia chamber was able to maintain CO2 levels at 5% and hypoxic O2 levels at 1% and 5% for long-term cell culture. This simple and easy-to-manufacture design uses off the shelf components, and the total material cost was $832.47 (USD).
Collapse
Affiliation(s)
- Colin R.N. Marchus
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Jacob A. Knudson
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Alexandra E. Morrison
- University of Idaho, Department of Electrical and Computer Engineering, Moscow, ID, United States
| | - Isabell K. Strawn
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Andrew J. Hartman
- University of Idaho, Department of Electrical and Computer Engineering, Moscow, ID, United States
| | - Dev Shrestha
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Nicholas M. Pancheri
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Ian Glasgow
- University of Idaho, Department of Mechanical Engineering, Moscow, ID, United States
| | - Nathan R. Schiele
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| |
Collapse
|
43
|
Edifying the Focal Factors Influencing Mesenchymal Stem Cells by the Microenvironment of Intervertebral Disc Degeneration in Low Back Pain. Pain Res Manag 2022; 2022:6235400. [PMID: 35386857 PMCID: PMC8977320 DOI: 10.1155/2022/6235400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/26/2022] [Accepted: 03/07/2022] [Indexed: 02/07/2023]
Abstract
Intervertebral disc degeneration (IVDD) is one of the main triggers of low back pain, which is most often associated with patient morbidity and high medical costs. IVDD triggers a wide range of pathologies and clinical syndromes like paresthesia, weakness of extremities, and intermittent/chronic back pain. Mesenchymal stem cells (MSCs) have demonstrated to possess immunomodulatory functions as well as the capability of differentiating into chondrocytes under appropriate microenvironment conditions, which makes them potentially epitome for intervertebral disc (IVD) regeneration. The IVD microenvironment is composed by niche of cells, and their chemical and physical milieus have been exhibited to have robust influence on MSC behavior as well as differentiation. Nevertheless, the contribution of MSCs to the IVD milieu conditions in healthy as well as degeneration situations is still a matter of debate. It is still not clear which factors, if any, are essential for effective and efficient MSC survival, proliferation, and differentiation. IVD microenvironment clues such as nucleopulpocytes, potential of hydrogen (pH), osmotic changes, glucose, hypoxia, apoptosis, pyroptosis, and hydrogels are capable of influencing the MSCs aimed for the treatment of IVDD. Therefore, clinical usage of MSCs ought to take into consideration these microenvironment clues during treatment. Alteration in these factors could function as prognostic indicators during the treatment of patients with IVDD using MSCs. Thus, standardized valves for these microenvironment clues are warranted.
Collapse
|
44
|
Insight in Hypoxia-Mimetic Agents as Potential Tools for Mesenchymal Stem Cell Priming in Regenerative Medicine. Stem Cells Int 2022; 2022:8775591. [PMID: 35378955 PMCID: PMC8976669 DOI: 10.1155/2022/8775591] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-mimetic agents are new potential tools in MSC priming instead of hypoxia incubators or chambers. Several pharmaceutical/chemical hypoxia-mimetic agents can be used to induce hypoxia in the tissues: deferoxamine (DFO), dimethyloxaloylglycine (DMOG), 2,4-dinitrophenol (DNP), cobalt chloride (CoCl2), and isoflurane (ISO). Hypoxia-mimetic agents can increase cell proliferation, preserve or enhance differentiation potential, increase migration potential, and induce neovascularization in a concentration- and stem cell source-dependent manner. Moreover, hypoxia-mimetic agents may increase HIF-1α, changing the metabolism and enhancing glycolysis like hypoxia. So, there is clear evidence that treatment with hypoxia-mimetic agents is beneficial in regenerative medicine, preserving stem cell capacities. These agents are not studied so wildly as hypoxia but, considering the low cost and ease of use, are believed to find application as pretreatment of many diseases such as ischemic heart disease and myocardial fibrosis and promote cardiac and cartilage regeneration. The knowledge of MSC priming is critical in evaluating safety procedures and use in clinics. In this review, similarities and differences between hypoxia and hypoxia-mimetic agents in terms of their therapeutic efficiency are considered in detail. The advantages, challenges, and future perspectives in MSC priming with hypoxia mimetic agents are also discussed.
Collapse
|
45
|
Franco RAG, McKenna E, Robey PG, Shajib MS, Crawford RW, Doran MR, Futrega K. Inhibition of BMP signaling with LDN 193189 can influence bone marrow stromal cell fate but does not prevent hypertrophy during chondrogenesis. Stem Cell Reports 2022; 17:616-632. [PMID: 35180395 PMCID: PMC9039850 DOI: 10.1016/j.stemcr.2022.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/22/2023] Open
Abstract
Bone morphogenetic protein (BMP) cascades are upregulated during bone marrow-derived stromal cell (BMSC) chondrogenesis, contributing to hypertrophy and preventing effective BMSC-mediated cartilage repair. Previous work demonstrated that a proprietary BMP inhibitor prevented BMSC hypertrophy, yielding stable cartilage tissue. Because of the significant therapeutic potential of a molecule capable of hypertrophy blockade, we evaluated the capacity of a commercially available BMP type I receptor inhibitor with similar properties, LDN 193189, to prevent BMSC hypertrophy. Using 14-day microtissue chondrogenic induction cultures we found that LDN 193189 permitted BMSC chondrogenesis but did not prevent hypertrophy. LDN 193189 was sufficiently potent to counter mineralization and adipogenesis in response to exogenous BMP-2 in osteogenic induction cultures. LDN 193189 did not modify BMSC behavior in adipogenic induction cultures. Although LDN 193189 is effective in countering BMP signaling in a manner that influences BMSC fate, this blockade is insufficient to prevent hypertrophy.
Collapse
Affiliation(s)
- Rose Ann G Franco
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia
| | - Eamonn McKenna
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Md Shaffiulah Shajib
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Ross W Crawford
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia
| | - Michael R Doran
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia; Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Mater Research Institute - University of Queensland, Brisbane, Australia.
| | - Kathryn Futrega
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Australia; Translational Research Institute, Brisbane, Australia; Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA.
| |
Collapse
|
46
|
Egger D, Lavrentieva A, Kugelmeier P, Kasper C. Physiologic isolation and expansion of human mesenchymal stem/stromal cells for manufacturing of cell-based therapy products. Eng Life Sci 2022; 22:361-372. [PMID: 35382547 PMCID: PMC8961040 DOI: 10.1002/elsc.202100097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 01/04/2023] Open
Abstract
The utilization of mesenchymal stem/stromal cells raises new hopes in treatment of diseases and pathological conditions, while at the same time bringing immense challenges for researchers, manufacturers and physicians. It is essential to consider all steps along the in vitro fabrication of cell-based products in order to reach efficient and reproducible treatment outcomes. Here, the optimal protocols for isolation, cultivation and differentiation of mesenchymal stem cells are required. In this review we discuss these aspects and their influence on the final cell-based product quality. We demonstrate that physiological in vitro cell cultivation conditions play a crucial role in therapeutic functionalities of cultivated cells. We show that three-dimensional cell culture, dynamic culture conditions and physiologically relevant in vitro oxygen concentrations during isolation and expansion make a decisive contribution towards the improvement of cell-based products in regenerative medicine.
Collapse
Affiliation(s)
- Dominik Egger
- Department of BiotechnologyUniversity of Natural Resources and Life ScienceViennaAustria
| | | | | | - Cornelia Kasper
- Department of BiotechnologyUniversity of Natural Resources and Life ScienceViennaAustria
| |
Collapse
|
47
|
Herger N, Bermudez-Lekerika P, Farshad M, Albers CE, Distler O, Gantenbein B, Dudli S. Should Degenerated Intervertebral Discs of Patients with Modic Type 1 Changes Be Treated with Mesenchymal Stem Cells? Int J Mol Sci 2022; 23:ijms23052721. [PMID: 35269863 PMCID: PMC8910866 DOI: 10.3390/ijms23052721] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/16/2022] Open
Abstract
Low back pain (LBP) has been among the leading causes of disability for the past 30 years. This highlights the need for improvement in LBP management. Many clinical trials focus on developing treatments against degenerative disc disease (DDD). The multifactorial etiology of DDD and associated risk factors lead to a heterogeneous patient population. It comes as no surprise that the outcomes of clinical trials on intradiscal mesenchymal stem cell (MSC) injections for patients with DDD are inconsistent. Intradiscal MSC injections have demonstrated substantial pain relief and significant disability-related improvements, yet they have failed to regenerate the intervertebral disc (IVD). Increasing evidence suggests that the positive outcomes in clinical trials might be attributed to the immunomodulatory potential of MSCs rather than to their regenerative properties. Therefore, patient stratification for inflammatory DDD phenotypes may (i) better serve the mechanisms of action of MSCs and (ii) increase the treatment effect. Modic type 1 changes—pathologic inflammatory, fibrotic changes in the vertebral bone marrow—are frequently observed adjacent to degenerated IVDs in chronic LBP patients and represent a clinically distinct subpopulation of patients with DDD. This review discusses whether degenerated IVDs of patients with Modic type 1 changes should be treated with an intradiscal MSC injection.
Collapse
Affiliation(s)
- Nick Herger
- Center of Experimental Rheumatology, University Hospital Zurich and Balgrist University Hospital, University of Zurich, CH-8008 Zurich, Switzerland; (N.H.); (O.D.)
| | - Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, CH-3008 Bern, Switzerland; (P.B.-L.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, CH-3010 Bern, Switzerland;
| | - Mazda Farshad
- Department of Orthopaedics, Balgrist University Hospital, CH-8008 Zurich, Switzerland;
| | - Christoph E. Albers
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, CH-3010 Bern, Switzerland;
| | - Oliver Distler
- Center of Experimental Rheumatology, University Hospital Zurich and Balgrist University Hospital, University of Zurich, CH-8008 Zurich, Switzerland; (N.H.); (O.D.)
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, CH-3008 Bern, Switzerland; (P.B.-L.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, CH-3010 Bern, Switzerland;
| | - Stefan Dudli
- Center of Experimental Rheumatology, University Hospital Zurich and Balgrist University Hospital, University of Zurich, CH-8008 Zurich, Switzerland; (N.H.); (O.D.)
- Correspondence: ; Tel.: +41-4451-07511
| |
Collapse
|
48
|
Chang X, Zhou F, Bu L, Wang N, Deng J, Wang S. Semaphorin 3A attenuates the hypoxia suppression of osteogenesis in periodontal ligament stem cells. J Periodontal Res 2022; 57:425-433. [PMID: 35037251 DOI: 10.1111/jre.12973] [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: 07/29/2021] [Revised: 12/25/2021] [Accepted: 01/06/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE AND BACKGROUND The occurrence and development of periodontitis are closely related to hypoxia of the periodontal microenvironment. Periodontal ligament stem cells (PDLSCs) are considered to have potential to regenerate periodontal tissues. Semaphorin 3A (Sema3A) plays an essential role in promoting osteogenesis. However, the effect of Sema3A on osteogenesis of PDLSCs under hypoxia remains unclear. The aim of this study was to investigate the effect of Sema3A on osteogenesis of PDLSCs under hypoxia. METHODS Isolated PDLSCs were identified using flow cytometry. Adipogenic differentiation potential was identified by oil red O staining. Osteogenesis was measured using Alizarin Red S staining and ALP staining. Intracellular hypoxia was induced using cobalt chloride (CoCl2 ). The expression level of hypoxia-inducible factor-1α (HIF-1α) was detected via ELISA. Expression of osteogenic markers and Sema3A was analyzed using western blot and real-time PCR. RESULTS The proliferation and osteogenesis of PDLSCs were markedly inhibited with increased concentrations of CoCl2 . Under the treatment with a low concentration of CoCl2 , expression of related osteogenic markers and Sema3A decreased in a time-dependent manner. ARS and ALP staining results also showed that osteogenic calcification decreased under hypoxia. Apigenin, an inhibitor of HIF-1α, effectively up-regulated expression of Sema3A and osteogenic markers with CoCl2 treatment. Moreover, exogenous Sema3A significantly increased the expression of osteogenesis-related markers and mineralization of PDLSCs according to ALP and ARS staining with CoCl2 treatment. CONCLUSIONS Hypoxia markedly inhibited osteogenesis of PDLSCs. Sema3A explicitly attenuated the hypoxia suppression of osteogenesis in PDLSCs.
Collapse
Affiliation(s)
- Xiaochi Chang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
| | - Fengyi Zhou
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Lingxue Bu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Nan Wang
- School of Stomatology of Qingdao University, Qingdao, China.,Department of Stomatology, Hospital of the PLA Navy, Qingdao, China
| | - Jing Deng
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
| | - Shuai Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
| |
Collapse
|
49
|
Loopmans S, Stockmans I, Carmeliet G, Stegen S. Isolation and in vitro characterization of murine young-adult long bone skeletal progenitors. Front Endocrinol (Lausanne) 2022; 13:930358. [PMID: 35979436 PMCID: PMC9376626 DOI: 10.3389/fendo.2022.930358] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Skeletal stem and progenitor cells (SSPCs) constitute a reservoir of bone-forming cells necessary for bone development, modeling and remodeling, as well as for fracture healing. Recent advances in tools to identify and isolate SSPCs have revealed that cells with multipotent properties are present not only in neonatal bone, but also in adult bone marrow and periosteum. The long bone metaphysis and endosteum have been proposed as an additional SSPC niche, although in vitro approaches to study their cellular and molecular characteristics are still limited. Here, we describe a comprehensive procedure to isolate and culture SSPCs derived from the metaphysis and endosteum of young-adult mice. Based on flow cytometry analysis of known SSPC markers, we found the presence of putative multipotent SSPCs, similar to neonatal bone tissue. In vitro, metaphyseal/endosteal SSPCs possess self-renewing capacity, and their multipotency is underscored by the ability to differentiate into the osteogenic and adipogenic lineage, while chondrogenic potential is limited. Expansion of metaphyseal/endosteal SSPCs under low oxygen conditions increases their proliferation capacity, while progenitor properties are maintained, likely reflecting their hypoxic niche in vivo. Collectively, we propose a validated isolation and culture protocol to study metaphyseal/endosteal SSPC biology in vitro.
Collapse
|
50
|
Choudhery MS. Strategies to improve regenerative potential of mesenchymal stem cells. World J Stem Cells 2021; 13:1845-1862. [PMID: 35069986 PMCID: PMC8727227 DOI: 10.4252/wjsc.v13.i12.1845] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/31/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
In the last few decades, stem cell-based therapies have gained attention worldwide for various diseases and disorders. Adult stem cells, particularly mesenchymal stem cells (MSCs), are preferred due to their significant regenerative potential in cellular therapies and are currently involved in hundreds of clinical trials. Although MSCs have high self-renewal as well as differentiation potential, such abilities are compromised with "advanced age" and "disease status" of the donor. Similarly, cell-based therapies require high cell number for clinical applications that often require in vitro expansion of cells. It is pertinent to note that aged individuals are the main segment of population for stem cell-based therapies, however; autologous use of stem cells for such patients (aged and diseased) does not seem to give optimal results due to their compromised potential. In vitro expansion to obtain large numbers of cells also negatively affects the regenerative potential of MSCs. It is therefore essential to improve the regenerative potential of stem cells compromised due to "in vitro expansion", "donor age" and "donor disease status" for their successful autologous use. The current review has been organized to address the age and disease depleted function of resident adult stem cells, and the strategies to improve their potential. To combat the problem of decline in the regenerative potential of cells, this review focuses on the strategies that manipulate the cell environment such as hypoxia, heat shock, caloric restriction and preconditioning with different factors.
Collapse
Affiliation(s)
- Mahmood S Choudhery
- Department of Biomedical Sciences, King Edward Medical University, Lahore 54000, Punjab, Pakistan
- Department of Genetics and Molecular Biology, University of Health Sciences, Lahore 54600, Punjab, Pakistan.
| |
Collapse
|