1
|
Yun YG, Yeo D, Tran TTT, Shin SJ, Shin JS, Lim NK, Lee JH, Kim HW. The characterization of the biological effect of hypoxia-mimetic condition on angiogenic potential in mesenchymal stem cells derived from different origins. Cells Dev 2025:204039. [PMID: 40541762 DOI: 10.1016/j.cdev.2025.204039] [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: 12/11/2024] [Revised: 05/29/2025] [Accepted: 06/16/2025] [Indexed: 06/22/2025]
Abstract
Under physiological conditions, adult stem cells are located in a special microenvironment, known as the niche, which is characterized by low oxygen levels. Hypoxia is a crucial factor regulating stem cell behavior, and in vitro normoxic culture often leads to the loss of pluripotency, resulting in reduced therapeutic potential of cells. In this study, we aimed to investigate the effects of cobalt chloride (CoCl2), a hypoxia-mimetic agent, on mesenchymal stem cells (MSCs) derived from different sources by inducing hypoxia-inducible factor-1α. Cell proliferation, migration, survival, and metabolic activity were assessed. CoCl2 treatment decreased the proliferation and migration and enhanced the survival of MSCs. Furthermore, a metabolic shift from oxidative phosphorylation to glycolysis was observed in cells. Overall, our findings provide important insights into the mechanisms by which hypoxic conditions differentially influence stem cells, facilitating the improvement of their therapeutic potential.
Collapse
Affiliation(s)
- Yeo Gyun Yun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Donghyeon Yeo
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Trang Thanh Thien Tran
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jin Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Ji-Sun Shin
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea; Department of Pediatric Dentistry, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
| | - Nam Kyu Lim
- Department of Plastic and Reconstructive Surgery, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea.
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
2
|
Rybkowska P, Kawalec M, Dymkowska D, Radoszkiewicz K, Zabłocka B, Zabłocki K, Sarnowska A. Activity and function of auxiliary fluxes of glucose metabolism in response to physiological normoxia (5 % O 2) during long-term Adipose-Derived Stem/Stromal cell culture. Eur J Cell Biol 2025; 104:151486. [PMID: 40187000 DOI: 10.1016/j.ejcb.2025.151486] [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] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Energy metabolism homeostasis emerges as a dominant element influencing mesenchymal stem/stromal cells' trajectory of development. The predominant glycolysis activity is a primary driver of cell proliferation and maintenance of the high-energetic state. Here, we examined the functions of two crucial auxiliary pathways: the phosphate-pentose pathway (PPP) and fructose-2,6-biphosphate pathway (FBP) to evaluate their impact on the therapeutic potential of Adipose-Derived Stem/Stromal cells (ASCs) during prolonged culture in various oxygen conditions: 5 % O2 - physiological normoxia or 21 % O2 - atmospheric oxygen. Our findings demonstrate that ASCs cultured in 5 % O2 increased the rate of proliferation, migration, and expression of stemness factors, which is prominent during the initial and middle passages. Additionally, ASCs cultured in a 5 % O2 exhibited heightened protection mechanisms against free radicals, increased LDH gene expression, and elevated extracellular acidification rate (ECAR). By estimating the HIF-1α level, we concluded that 5 % oxygen conditions were insufficient to induce a profound hypoxic state in ASCs. However, at the protein level, both the PPP and FBP pathways appeared to be more active in young (2-passage) cells, regardless of oxygen conditions, and their activity diminished over time. Additionally, the chemical suppression of G6PDH by Polydatin and inhibition of PFKFB3 by PFK-158 in ASCs (passage-2) revealed dose- and time-dependent effect on decreasing migratory capabilities of cells. Nevertheless, our work underscores the adaptable nature of ASC metabolism to prevailing external conditions, with the aging of the culture contributing to the decline in glycolysis-associated auxiliary pathways.
Collapse
Affiliation(s)
- Paulina Rybkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland.
| | - Maria Kawalec
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland
| | - Dorota Dymkowska
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3 Street, Warsaw 02-093, Poland
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland
| | - Barbara Zabłocka
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland
| | - Krzysztof Zabłocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3 Street, Warsaw 02-093, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, Warsaw 02-106, Poland.
| |
Collapse
|
3
|
Wang Y, Li Q, Li H, Yang X, Fang H, Bi R, Zhu S. Heterogeneous Characteristics of the CD90 + Progenitors in the Fibrocartilage of Different Joints. Cartilage 2025; 16:190-201. [PMID: 37750508 PMCID: PMC12066840 DOI: 10.1177/19476035231200359] [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: 06/12/2023] [Revised: 07/24/2023] [Accepted: 08/25/2023] [Indexed: 09/27/2023] Open
Abstract
ObjectiveThis study aimed to isolate and compare the mesenchymal stem cell characteristics of CD90+ cells from different fibrocartilage tissues in the temporomandibular joint (TMJ), the knee joint, and the intervertebral joint to further understand the similarities and differences of these 4 fibrocartilage tissues.MethodsCD90+ cells were isolated from TMJ disc, condylar cartilage, meniscus, and intervertebral disc by using magnetic-activated cell sorting. Cellular assays including 4.5-ethynyl-2'-deoxyuridine labeling, multilineage differentiation, colony formation, and cell migration were conducted to compare their mesenchymal stem cell characteristics. Immunofluorescent staining was performed for observing the expression of actively proliferating CD90+ cells within the tissues. H&E staining and Safranine O staining were used to compare the histological features.ResultsThe CD90+ cells derived from these 4 fibrocartilage tissues exhibited comparable cell proliferation abilities. However, the cells from the TMJ disc displayed limited multilineage differentiation potential, colony formation, and cell migration abilities in comparison with the cells from the other fibrocartilage tissues. In vivo, there was relatively more abundant expression of CD90+ cells in the TMJ disc during the early postnatal stage. The limited EDU+ cell numbers signified a low proliferation capacity of CD90+ cells in the TMJ disc. In addition, we observed a significant decrease in cell density and a restriction in the synthesis of extracellular proteoglycans in the TMJ disc.ConclusionOur study highlights the spatial heterogeneity of CD90+ cells in the fibrocartilages of different joint tissues, which may contribute to the limited cartilage repair capacity in the TMJ disc.
Collapse
Affiliation(s)
- Yiru Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianli Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haohan Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xianni Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Han Fang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ruiye Bi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Songsong Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Orthognathic and Temporomandibular Joint Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Zhou X, Lv Z, Chen Z, Xu Y, Lin C, Liu L, Chen H, Niu B, Cui W, Zhang Y. Manipulation of Oxygen Tension in Damaged Regions via Hypoxia-Induced IPN Hydrogel Microspheres for Intervertebral Disc Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417570. [PMID: 40231808 PMCID: PMC12165120 DOI: 10.1002/advs.202417570] [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] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/28/2025] [Indexed: 04/16/2025]
Abstract
Disruption of low oxygen tension homeostasis during intervertebral disc degeneration inhibits endogenous stem cell viability and function, posing a challenge for endogenous regeneration. Here, to achieve sustained hypoxia manipulation, constructed hypoxia-inducible interpenetrating polymer network (IPN) hydrogel microspheres (HIMS) are constructed by microfluidics to integrate the hypoxic system with a stabilizing network. The IPN is synthesized through a two-step polymerization process, consisting of rapid photo-crosslinked gelatin methacrylate anhydride (GM) polymer I and slow enzyme-crosslinked vanillin-grafted gelatin (GV) polymer II. The enzymatic reaction between GV and laccase is able to create a hypoxic microenvironment to modulate oxygen tension in situ within the injured region. HIMS can reduce microenvironmental oxygen tension by 1/3 and maintain a hypoxic microenvironment for up to 5 days, thereby activating the PI3K/AKT/HIF-1α signaling pathway in endogenous stem cells to promote differentiation into nucleus pulposus-like cells. Additionally, NSC-Exos are loaded onto HIMS to trigger endogenous progenitor/stem cell recruitment and migration. Both in vitro and in vivo assays demonstrate that NSC-Exos@HIMS facilitates stem cell recruitment, targets differentiation, and stimulates extracellular matrix synthesis. Overall, the microspheres established herein provide a novel strategy for manipulating oxygen tension and enhancing endogenous tissue regeneration in injured regions during intervertebral disc degeneration.
Collapse
Affiliation(s)
- Xingdie Zhou
- Department of Spine SurgeryRenji HospitalShanghai Jiao Tong University School of Medicine160 Pujian RoadShanghai200127P. R. China
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- School of Materials Science and EngineeringShanghai UniversityNanchen Road 333Shanghai200444P. R. China
| | - Zhendong Lv
- Department of Spine SurgeryRenji HospitalShanghai Jiao Tong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| | - Zehao Chen
- Department of OrthopaedicsLaboratory of Key Technology and Materials in Minimally Invasive Spine SurgeryCenter for Spinal Minimally Invasive ResearchHongqiao International Institute of MedicineTongren HospitalShanghai Jiao Tong University School of MedicineShanghai200336China
| | - Yiming Xu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Chao Lin
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Department of OrthopaedicsLaboratory of Key Technology and Materials in Minimally Invasive Spine SurgeryCenter for Spinal Minimally Invasive ResearchHongqiao International Institute of MedicineTongren HospitalShanghai Jiao Tong University School of MedicineShanghai200336China
| | - Li Liu
- School of Materials Science and EngineeringShanghai UniversityNanchen Road 333Shanghai200444P. R. China
| | - Hao Chen
- Department of Spine SurgeryRenji HospitalShanghai Jiao Tong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| | - Bing Niu
- School of Life SciencesShanghai UniversityNanchen Road 333Shanghai200444P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yuhui Zhang
- Department of Spine SurgeryRenji HospitalShanghai Jiao Tong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| |
Collapse
|
5
|
Li W, Chen M, Zhang L. Muscle Stem Cell Microenvironment and Functions in Muscle Regeneration. Biomolecules 2025; 15:765. [PMID: 40563407 DOI: 10.3390/biom15060765] [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: 04/02/2025] [Revised: 05/22/2025] [Accepted: 05/22/2025] [Indexed: 06/28/2025] Open
Abstract
Muscle stem cells (MuSCs) are the key to muscle regeneration. The activation and maintenance of MuSCs require the precise regulation of their microenvironments. Myofibers and other cells including endothelial cells, fibroblasts, and immune cell populations constitute the cell components of the MuSC niche. The communication between these cell populations and MuSCs play an essential role in muscle repair. Furthermore, the physical and chemical stimulations around MuSCs also affect the cell behaviors of MuSCs. Extracellular matrix (ECM) and the factors stored in it generate a repair-promoting niche for efficient muscle regeneration. Understanding the mechanism of muscle stem cell regulation is the basis of clinically optimizing muscle repair. In this review, we discuss recent findings about the microenvironments of MuSCs and their functions in muscle regeneration, which would shed light on new targets and strategies for muscle injury treatment.
Collapse
Affiliation(s)
- Wenjing Li
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| | - Minyou Chen
- College of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
6
|
Li M, Wang X, He W, Zhou H. Drug-tolerant persister cells in acute myeloid leukemia: pressing challenge and promising new strategies for treatment. Front Med (Lausanne) 2025; 12:1586552. [PMID: 40443513 PMCID: PMC12120853 DOI: 10.3389/fmed.2025.1586552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Acute myeloid leukemia (AML) exhibits a pronounced ability to develop drug resistance and undergo disease relapse. Recent research has noticed that resistance to treatments could substantially be attributed to drug-tolerant persister (DTP) cells, which are capable of surviving under therapeutic pressures. These are transient, reversibly dormant cells with the capability to act as a reservoir for disease relapse. DTP cells utilize diverse adaptive strategies to optimize the ecological niche, undergo metabolic reprogramming, and interact with microenvironment. The persister state of AML is established through transient cellular reprogramming, thus allowing cells to survive the initial phase of drug therapy and develop drug resistance. Our review explores the identification and phenotypic characteristics of AML DTP cells, as well as their clinical relevance. We summarize the mechanisms underlying the persistence of AML DTP cells and the molecular attributes that define the DTP state. We further address the current challenges and future prospects of DTP-targeting approaches. Understanding these features may provide critical insights into novel therapeutic strategies aimed at targeting AML DTP cells, especially in the new era of immunotherapy against AML.
Collapse
Affiliation(s)
- Meng Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoli Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjuan He
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Perfetto M, Ishfaq M, Mohideen A, Rondelli CM, Gillis S, Tejero J, Stratman AN, Riggins R, Yien YY. FAM210B regulates iron homeostasis and sex-specific responses in stress erythropoiesis. Exp Hematol 2025:104797. [PMID: 40355042 DOI: 10.1016/j.exphem.2025.104797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/14/2025]
Abstract
Iron is required for redox homeostasis but poses toxicity risks due to its redox activity. Erythropoiesis hence requires tight regulation of iron utilization for hemoglobin synthesis. The requirement for iron in erythropoiesis has necessitated the evolution of mechanisms to handle the iron required for hemoglobinization. FAM210B was identified as a regulator of mitochondrial iron import and heme synthesis in erythroid cell culture and zebrafish models. Here, we demonstrate that although FAM210B is required for erythroid differentiation and heme synthesis under standard cell culture conditions, holotransferrin supplementation was sufficient to chemically complement the iron-deficient phenotype. To investigate the role of FAM210B in erythropoiesis, we used knockout mice. Although Fam210b-/- mice were viable and did not exhibit overt erythropoietic defects in the bone marrow, the male mice exhibited an increase in serum transferrin, suggesting sex-specific alterations in systemic iron sensing. On phlebotomy-induced stress erythropoiesis, Fam210b-/- mice exhibited differences in serum transferrin levels, and more starkly, had markedly smaller spleens, indicating defects in stress response. Fam210b-/- males had defects in neutrophil and monocyte numbers, as well as decreased erythroid progenitor numbers during erythropoietic stress. Together, our findings show that Fam210b plays a key role in the splenic response to erythropoietic stress. Our findings reveal a critical role for FAM210B in mediating splenic stress erythropoiesis and suggest it may act as a sex-specific regulator, potentially linked to androgen signaling.
Collapse
Affiliation(s)
- Mark Perfetto
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA; Department of Medicine, Division of Classical Hematology, University of Pittsburgh, Pittsburgh, PA
| | - Muhammad Ishfaq
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA; Department of Medicine, Division of Classical Hematology, University of Pittsburgh, Pittsburgh, PA
| | - Aiden Mohideen
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA; Department of Medicine, Division of Classical Hematology, University of Pittsburgh, Pittsburgh, PA
| | | | - Samantha Gillis
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Jesus Tejero
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Amber N Stratman
- Department of Cell Biology and Physiology, Washington University in St Louis School of Medicine, St Louis, M
| | - Rebecca Riggins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC
| | - Yvette Y Yien
- Pittsburgh Heart Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA; Department of Medicine, Division of Classical Hematology, University of Pittsburgh, Pittsburgh, PA.
| |
Collapse
|
8
|
Filippi L, Innocenti F, Pascarella F, Scaramuzzo RT, Morganti R, Bagnoli P, Cammalleri M, Dal Monte M, Calvani M, Pini A. β 3-Adrenoceptor Agonism to Mimic the Biological Effects of Intrauterine Hypoxia: Taking Great Strides Toward a Pharmacological Artificial Placenta. Med Res Rev 2025; 45:842-866. [PMID: 39604126 PMCID: PMC11976384 DOI: 10.1002/med.22092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/24/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
At different stages of life, from embryonic to postnatal, varying oxygen concentrations modulate cellular gene expression by enhancing or repressing hypoxia-inducible transcription factors. During embryonic/fetal life, these genes encode proteins involved in adapting to a low-oxygen environment, including the induction of specific enzymes related to glycolytic metabolism, erythropoiesis, angiogenesis, and vasculogenesis. However, oxygen concentrations fluctuate during intrauterine life, enabling the induction of tissue-specific differentiation processes. Fetal well-being is thus closely linked to the physiological benefits of a dynamically hypoxic environment. Premature birth entails the precocious exposure of the immature fetus to a more oxygen-rich environment compared to the womb. As a result, preterm newborns face a condition of relative hyperoxia, which alters the postnatal development of organs and contributes to prematurity-related diseases. However, until recently, the molecular mechanism by which high oxygen tension alters normal fetal differentiation remained unclear. In this review, we discuss the research trajectory followed by our research group, which suggests that early exposure to a relatively hyperoxic environment may impair preterm neonates due to reduced expression of the β3-adrenoceptor. Additionally, we explore how these impairments could be prevented through the pharmacological stimulation of the remaining β3-adrenoceptors. Recent preclinical studies demonstrate that pharmacological stimulation of the β3-adrenoceptor can decouple exposure to hyperoxia from its harmful effects, offering a glimpse of the possibility to recreating the conditions typical of intrauterine life, even after premature birth.
Collapse
Affiliation(s)
- Luca Filippi
- Neonatology UnitAzienda Ospedaliero‐Universitaria PisanaPisaItaly
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | | | | | | | - Riccardo Morganti
- Section of StatisticsAzienda Ospedaliero‐Universitaria PisanaPisaItaly
| | - Paola Bagnoli
- Department of Biology, Unit of General PhysiologyUniversity of PisaPisaItaly
| | - Maurizio Cammalleri
- Department of Biology, Unit of General PhysiologyUniversity of PisaPisaItaly
| | - Massimo Dal Monte
- Department of Biology, Unit of General PhysiologyUniversity of PisaPisaItaly
| | - Maura Calvani
- Department of Pediatric Hematology‐OncologyMeyer Children's Hospital IRCCSFlorenceItaly
| | - Alessandro Pini
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| |
Collapse
|
9
|
Liu J, Chen Z, Deng L, Yao C, Zhou Z, Zhou C, Bin Y, Liu M, Wang L, Wang L, Wang Z. Metal-phenolic networks specifically eliminate hypoxic tumors by instigating oxidative and proteotoxic stresses. Bioact Mater 2025; 47:361-377. [PMID: 40026824 PMCID: PMC11870026 DOI: 10.1016/j.bioactmat.2025.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/04/2025] [Accepted: 01/18/2025] [Indexed: 03/05/2025] Open
Abstract
Hypoxia, a prevalent characteristic of solid tumors, substantially impairs the efficacy of cancer treatments. However, there are no feasible clinical approaches for treating hypoxic tumors. Here, we develop metal-phenolic networks (CuGI) utilizing the natural glycolysis inhibitor (epigallocatechin gallate) and the essential metal element in the human body (copper ions), specifically targeting and annihilating hypoxic cancer cells. CuGI redirects the metabolic pathway of hypoxic cancer cells from anaerobic glycolysis to oxidative phosphorylation, thereby enhancing reactive oxygen species production and promoting oligomerization of lipoylated proteins in the tricarboxylic acid cycle. Through targeted induction of oxidative and proteotoxic stresses, CuGI induces apoptosis and cuproptosis specifically in cancer cells under hypoxic conditions while sparing normal cells. Moreover, cancer cell membrane-coated CuGI (CuGI@CM) exhibits enhanced tumor penetration effect and demonstrates commendable biocompatibility, effectively suppressing colorectal tumor growth. Importantly, CuGI@CM, when combined with vascular disruptors or radiotherapy which aggravate tumor hypoxia, synergistically potentiates therapeutic efficacy. Thus, CuGI represents a specific and potent nanotherapeutic capable of selectively eliminating hypoxic tumors, offering promise in combination therapies to address tumor hypoxia.
Collapse
Affiliation(s)
- Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zuoyu Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lixue Deng
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chundong Yao
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhixin Zhou
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cheng Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yawen Bin
- Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Miaodeng Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liping Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Clinical Laboratory and Active Health Smart Equipment, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
10
|
Liu X, Cui Y, Gong J, Yu X, Cui Y, Xuan Y. SETD5 facilitates stemness and represses ferroptosis via m6A-mediating PKM2 stabilization in non-small cell lung cancer. Oncogene 2025:10.1038/s41388-025-03426-9. [PMID: 40307507 DOI: 10.1038/s41388-025-03426-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
SETD5, an atypical member of the histone lysine methyltransferase family known for its association with cancer stemness, is a significant predictor of unfavorable survival outcomes in non-small cell lung cancer (NSCLC). However, the function of SETD5 in NSCLC stemness remains unclear, and whether it is an active H3K36me3 is controversial. Consequently, further investigation is required to clarify the pivotal role of SETD5 in NSCLC stemness and its related mechanism. Thus, this study employed the NSCLC tissue microarray and bioinformatics tools to analyze SETD5 expression and determine its effect on stemness and investigated the role of SETD5 in the metastasis of NSCLC using in vitro and in vivo analyses. The findings indicated high SETD5 expression in embryonic and NSCLC tissues, which was related to the pathological tumor stage, lymph node metastasis, and clinical stage, indicating that SETD5 could be used as a biomarker and prognostic factor in NSCLC. In addition, we found that SETD5 can promote glycolysis, thereby inhibiting ferroptosis and promoting the stemness of NSCLC, causing tumor metastasis and adverse prognosis in patients. In terms of mechanism, SETD5 as H3K36me3 facilitates the m6A modification of METTL14 and the recruitment of YTHDF1 and mediates PKM2 nuclear translocation and phosphorylation of p-PKM2 Tyr105, regulating GPX4 mediated ferroptosis resistance and SOX9 mediated stemness in NSCLC. The findings emphasize that SETD5 may serve as a promising indicator of stemness in NSCLC, which can help develop therapeutic targets for NSCLC and prognostic evaluation. This study provides evidence that SETD5 as H3K36me3 facilitates the m6A modification of METTL14 and the recruitment of YTHDF1 and mediates the nuclear translocation of PKM2, regulating GPX4 mediated ferroptosis resistance and SOX9 mediated stemness, causing tumor metastasis and adverse prognosis in patients.
Collapse
Affiliation(s)
- Xingzhe Liu
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | - Yuzhen Cui
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Jie Gong
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | - Xinhui Yu
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yan Cui
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yanhua Xuan
- Department of Pathology, Yanbian University College of Medicine, Yanji, China.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China.
- Institute of Regenerative Medicine, Yanbian University College of Medicine, Yanji, China.
| |
Collapse
|
11
|
Yuan G, Wang J, Qiu S, Zhu Y, Cheng Q, Li L, Sha J, Yang X, Yuan Y. Improving in vitro induction efficiency of human primordial germ cell-like cells using N2B27 or NAC-based medium. J Biomed Res 2025; 39:1-14. [PMID: 40204653 DOI: 10.7555/jbr.38.20240433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Primordial germ cells (PGCs), the precursors of oocytes or spermatozoa, are highly pluripotent. In recent years, the in vitro induction of human primordial germ cell-like cells (hPGCLCs) has advanced significantly. However, the stability and efficacy of obtaining hPGCLCs in vitro still require further improvement. In the current study, we identified a novel induction system by using Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 (DMEM/F-12) as the basal medium supplemented with B27 and N2 (referred to as N2B27) in combination with four cytokines: bone morphogenetic protein 4 (BMP4), stem cell factor (SCF), epidermal growth factor (EGF), and leukemia inhibitory factor (LIF). The hPGCLCs induced under these conditions closely resemble PGCs from 4 to 5-week-old embryos at the transcriptome level. Compared with traditional GK15 (GMEM supplemented with 15% Knockout™ SR)-based induction conditions, the N2B27 system significantly increased the speed and efficacy of hPGCLC induction. RNA sequencing analysis revealed that this improvement resulted from an increased cell capacity to cope with hypoxic stress and avoid apoptosis. The N2B27 medium promoted an increase in mitochondrial activity, enabling cells to better cope with hypoxic stress while also reducing the production of reactive oxygen species. Moreover, by gradient concentration experiments, we demonstrated that addition of the common antioxidant N-acetyl-L-cysteine at an optimized concentration further enhanced the efficiency of PGCLC induction under GK15 conditions. Thus, our study established an optimized induction system that enhances the efficiency of hPGCLC differentiation by improving cellular resilience to hypoxic stress and apoptosis.
Collapse
Affiliation(s)
- Gege Yuan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiachen Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Shuangshuang Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yunfei Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qing Cheng
- Women's Hospital of Nanjing Medical University, Women and Children's Healthcare Hospital, Nanjing, Jiangsu 211100, China
| | - Laihua Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyu Yang
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029 China
| | - Yan Yuan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
12
|
Divvela SSK, Gallorini M, Gellisch M, Patel GD, Saso L, Brand-Saberi B. Navigating redox imbalance: the role of oxidative stress in embryonic development and long-term health outcomes. Front Cell Dev Biol 2025; 13:1521336. [PMID: 40206404 PMCID: PMC11979171 DOI: 10.3389/fcell.2025.1521336] [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: 11/01/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
Embryonic development is a complex process of concurrent events comprising cell proliferation, differentiation, morphogenesis, migration, and tissue remodeling. To cope with the demands arising from these developmental processes, cells increase their nutrient uptake, which subsequently increases their metabolic activity. Mitochondria play a key role in the maintenance of metabolism and production of reactive oxygen species (ROS) as a natural byproduct. Regulation of ROS by antioxidants is critical and tightly regulated during embryonic development, as dysregulation results in oxidative stress that damages essential cellular components such as DNA, proteins, and lipids, which are crucial for cellular maintenance and in extension development. However, during development, exposure to certain exogenous factors or damage to cellular components can result in an imbalance between ROS production and its neutralization by antioxidants, leading to detrimental effects on the developmental process. In this review article, we highlight the crucial role of redox homeostasis in normal development and how disruptions in redox balance may result in developmental defects.
Collapse
Affiliation(s)
| | - Marialucia Gallorini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Morris Gellisch
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Gaurav Deepak Patel
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
13
|
Kaufman D, Winkler S, Heuer C, Shibli A, Snezhko A, Livshits GI, Bahnemann J, Ben-Yoav H. Automated electrochemical oxygen sensing using a 3D-printed microfluidic lab-on-a-chip system. LAB ON A CHIP 2025; 25:1404-1415. [PMID: 39763425 DOI: 10.1039/d4lc00962b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
Dissolved oxygen is crucial for metabolism, growth, and other complex physiological and pathological processes; however, standard physiological models (such as organ-on-chip systems) often use ambient oxygen levels, which do not reflect the lower levels that are typically found in vivo. Additionally, the local generation of reactive oxygen species (ROS; a key factor in physiological systems) is often overlooked in biology-mimicking models. Here, we present a microfluidic system that integrates electrochemical dissolved oxygen sensors with lab-on-a-chip technology to monitor the physiological oxygen concentrations and generate hydrogen peroxide (H2O2; a specific ROS). This microfluidic lab-on-a-chip system was fabricated using high-resolution 3D printing technology in a one-step process. It incorporates a micromixer, an on-chip bubble-trap, an electrochemical cell with fabricated gold or platinum black-coated working electrodes as well as an Ag/AgCl reference electrode, and a commercial optical oxygen sensor for validation. This device enables an automated variation of the oxygen levels as well as sensitive electrochemical oxygen monitoring (limit of detection = 11.9 ± 0.3 μM), with a statistically significant correlation with the optical sensor. The proposed system can serve as a tool to characterize and evaluate custom-made electrodes. Indeed, we envision that in the future it will be used to regulate dissolved oxygen levels and oxygen species in real time in organ-on-chip systems.
Collapse
Affiliation(s)
- Daniel Kaufman
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| | - Steffen Winkler
- Institute of Physics, University of Augsburg, Universitätsstraße 1, 86159 Augsburg, Germany.
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, 8615, Augsburg, Germany
| | - Christopher Heuer
- Institute of Physics, University of Augsburg, Universitätsstraße 1, 86159 Augsburg, Germany.
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, 8615, Augsburg, Germany
| | - Ahed Shibli
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| | - Alexander Snezhko
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| | - Gideon I Livshits
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| | - Janina Bahnemann
- Institute of Physics, University of Augsburg, Universitätsstraße 1, 86159 Augsburg, Germany.
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, 8615, Augsburg, Germany
| | - Hadar Ben-Yoav
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel.
| |
Collapse
|
14
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [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: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
15
|
Fan MH, Zhang XZ, Jiang YL, Pi JK, Zhang JY, Zhang YQ, Xing F, Xie HQ. Exosomes from hypoxic urine-derived stem cells facilitate healing of diabetic wound by targeting SERPINE1 through miR-486-5p. Biomaterials 2025; 314:122893. [PMID: 39418849 DOI: 10.1016/j.biomaterials.2024.122893] [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: 05/24/2024] [Revised: 09/02/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024]
Abstract
Vascular pathologies and injuries are important factors for the delayed wound healing in diabetes. Previous studies have demonstrated that hypoxic environments could induce formation of new blood vessels by regulating intercellular communication and cellular behaviors. In this study, we have enhanced the angiogenic potential of exosomes by subjecting urine-derived stem cells (USCs) to hypoxic preconditioning. To prolong the retention of exosomes at the wound site, we have also engineered a novel dECM hydrogel termed SISMA, which was modified from porcine small intestinal submucosa (SIS). For its rapid and controllable gelation kinetics, excellent biocompatibility, and exosome release capability, the SISMA hydrogel has proven to be a reliable delivery vehicle for exosomes. The hypoxia-induced exosomes-loaded hydrogel has promoted endothelial cell proliferation, migration, and tube formation. More importantly, as evidenced by significant in vivo vascular regeneration in the early stages post-injury, it has facilitated tissue repair. This may because miR-486-5p in H-exo inhibit SERPINE1 activity in endothelial cell. Additionally, miRNA sequencing analysis suggested that the underlying mechanism for enhanced angiogenesis may be associated with the activation of classical HIF-1α signaling pathway. In summary, our study has presented a novel non-invasive, cell-free therapeutic approach for accelerating diabetes wound healing and development of a practical and efficient exosomes delivery platform.
Collapse
Affiliation(s)
- Ming-Hui Fan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Xiu-Zhen Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Yan-Lin Jiang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Jin-Kui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Ji-Ye Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Yue-Qi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Fei Xing
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, 610212, PR China.
| |
Collapse
|
16
|
Leelasangsai W, Thummachot K, Thammasarnsophon P, Srion A, Suwanprateeb J, Patntirapong S. Cobalt-Incorporated Hydroxyapatite Conditioned Media Promotes In Vitro Scratch Wound Healing and Mesenchymal Stem Cell Migration. J Funct Biomater 2025; 16:72. [PMID: 40137351 PMCID: PMC11942815 DOI: 10.3390/jfb16030072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 03/27/2025] Open
Abstract
Cell migration of mesenchymal stem cells (MSCs) is critical for bone healing and remodeling. Cobalt is a well-known hypoxia mimic, which can enhance MSC migration. Therefore, the objective of this study was to investigate the migratory response of MSCs to a developed cobalt-incorporated hydroxyapatite (HACo) material. HACo was fabricated by a simple ion exchange procedure at concentrations ranging from 40 to 8000 μM into disc shape. HACo discs were incubated in the media and conditioned media (CM; HACoCM) were collected for MSC culture. HACM served as a control. MSCs were cultured until reaching 90% confluence before the wound was generated by scraping. Time-lapse imaging of wound migration was monitored, recorded, and assessed. Statistical analysis was performed by one-way ANOVA followed by a Dunnett's test. The wound area gradually declined from 0 to 40 h for all samples. HACoCM at 40 µM (HACo40CM) promoted wound closure at the early period of wound healing. Both HACo40CM and HACo8000CM enhanced the distance and velocity of individual cell migration. However, only HACo40CM affected cell persistence and direction at the early period of cell migration. Exposure to HACoCM accelerated the speed of MSC migration, which is necessary for wound healing. The migratory ability of individual cells could help the rate of wound healing. Therefore, HACo materials may serve as potential biomaterials for enhanced bone healing.
Collapse
Affiliation(s)
- Weerapat Leelasangsai
- Department of Implantology, Faculty of Dentistry, Thammasat University, Pathum Thani 12120, Thailand;
| | - Krongrat Thummachot
- Faculty of Dentistry, Thammasat University, Pathum Thani 12120, Thailand; (K.T.); (P.T.)
| | | | - Autcharaporn Srion
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center (MTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (A.S.); (J.S.)
| | - Jintamai Suwanprateeb
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center (MTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (A.S.); (J.S.)
- Thammasat University Center of Excellence in Computational Mechanics and Medical Engineering, Thammasat University, Pathum Thani 12120, Thailand
| | - Somying Patntirapong
- Faculty of Dentistry, Thammasat University, Pathum Thani 12120, Thailand; (K.T.); (P.T.)
- Thammasat University Research Unit in Dental and Bone Substitute Biomaterials, Faculty of Dentistry, Thammasat University, Pathum Thani 12120, Thailand
| |
Collapse
|
17
|
Daumova L, Manakov D, Petrak J, Sovilj D, Behounek M, Andera L, Vit O, Souckova O, Havranek O, Dolnikova A, Renesova N, Tuskova L, Winkowska L, Bettazova N, Kupcova K, Kalbacova MH, Sikorova M, Trneny M, Klener P. Long-term adaptation of lymphoma cell lines to hypoxia is mediated by diverse molecular mechanisms that are targetable with specific inhibitors. Cell Death Discov 2025; 11:65. [PMID: 39966387 PMCID: PMC11836139 DOI: 10.1038/s41420-025-02341-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
A large body of evidence suggests that hypoxia drives aggressive molecular features of malignant cells irrespective of cancer type. Non-Hodgkin lymphomas (NHL) are the most common hematologic malignancies characterized by frequent involvement of diverse hypoxic microenvironments. We studied the impact of long-term deep hypoxia (1% O2) on the biology of lymphoma cells. Only 2 out of 6 tested cell lines (Ramos, and HBL2) survived ≥ 4 weeks under hypoxia. The hypoxia-adapted (HA)b Ramos and HBL2 cells had a decreased proliferation rate accompanied by significant suppression of both oxidative phosphorylation and glycolytic pathways. Transcriptome and proteome analyses revealed marked downregulation of genes and proteins of the mitochondrial respiration complexes I and IV, and mitochondrial ribosomal proteins. Despite the observed suppression of glycolysis, the proteome analysis of both HA cell lines showed upregulation of several proteins involved in the regulation of glucose utilization including the active catalytic component of prolyl-4-hydroxylase P4HA1, an important druggable oncogene. HA cell lines demonstrated increased transcription of key regulators of auto-/mitophagy, e.g., neuritin, BCL2 interacting protein 3 (BNIP3), BNIP3-like protein, and BNIP3 pseudogene. Adaptation to hypoxia was further associated with deregulation of apoptosis, namely upregulation of BCL2L1/BCL-XL, overexpression of BCL2L11/BIM, increased binding of BIM to BCL-XL, and significantly increased sensitivity of both HA cell lines to A1155463, a BCL-XL inhibitor. Finally, in both HA cell lines AKT kinase was hyperphosphorylated and the cells showed increased sensitivity to copanlisib, a pan-PI3K inhibitor. In conclusion, our data report on several shared mechanisms of lymphoma cell adaptation to long-term hypoxia including: 1. Upregulation of proteins responsible for glucose utilization, 2. Degradation of mitochondrial proteins for potential mitochondrial recycling (by mitophagy), and 3. Increased dependence on BCL-XL and PI3K-AKT signaling for survival. In translation, inhibition of glycolysis, BCL-XL, or PI3K-AKT cascade may result in targeted elimination of HA lymphoma cells.
Collapse
Affiliation(s)
- Lenka Daumova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dmitry Manakov
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Petrak
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Dana Sovilj
- Institute of Biotechnology, Czech Academy of Sciences / BIOCEV, Vestec, Czech Republic
| | - Matej Behounek
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Ladislav Andera
- Institute of Biotechnology, Czech Academy of Sciences / BIOCEV, Vestec, Czech Republic
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Vit
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Olga Souckova
- OMICS Mass Spectrometry Core Facility, Biology Departments, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Ondrej Havranek
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alex Dolnikova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nicol Renesova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Liliana Tuskova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lucie Winkowska
- CLIP, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Nardjas Bettazova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kristyna Kupcova
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marie Hubalek Kalbacova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Miriama Sikorova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Trneny
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
18
|
Di Giovannantonio M, Hartley F, Elshenawy B, Barberis A, Hudson D, Shafique HS, Allott VES, Harris DA, Lord SR, Haider S, Harris AL, Buffa FM, Harris BHL. Defining hypoxia in cancer: A landmark evaluation of hypoxia gene expression signatures. CELL GENOMICS 2025; 5:100764. [PMID: 39892389 PMCID: PMC11872601 DOI: 10.1016/j.xgen.2025.100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/04/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Tumor hypoxia drives metabolic shifts, cancer progression, and therapeutic resistance. Challenges in quantifying hypoxia have hindered the exploitation of this potential "Achilles' heel." While gene expression signatures have shown promise as surrogate measures of hypoxia, signature usage is heterogeneous and debated. Here, we present a systematic pan-cancer evaluation of 70 hypoxia signatures and 14 summary scores in 104 cell lines and 5,407 tumor samples using 472 million length-matched random gene signatures. Signature and score choice strongly influenced the prediction of hypoxia in vitro and in vivo. In cell lines, the Tardon signature was highly accurate in both bulk and single-cell data (94% accuracy, interquartile mean). In tumors, the Buffa and Ragnum signatures demonstrated superior performance, with Buffa/mean and Ragnum/interquartile mean emerging as the most promising for prospective clinical trials. This work delivers recommendations for experimental hypoxia detection and patient stratification for hypoxia-targeting therapies, alongside a generalizable framework for signature evaluation.
Collapse
Affiliation(s)
- Matteo Di Giovannantonio
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Fiona Hartley
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Badran Elshenawy
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Alessandro Barberis
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Dan Hudson
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK; The Rosalind Franklin Institute, Didcot, UK
| | | | | | | | - Simon R Lord
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Syed Haider
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Adrian L Harris
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK
| | - Francesca M Buffa
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK; CompBio Lab, Department of Computing Sciences, Bocconi University, Milan, Italy; AI and Systems Biology Lab, IFOM - Istituto Fondazione di Oncologia Molecolare ETS, Milan, Italy.
| | - Benjamin H L Harris
- Computational Biology and Integrative Genomics Lab, Department of Oncology, University of Oxford, Oxford, UK; St. Catherine's College, University of Oxford, Oxford, UK; Cutrale Perioperative and Ageing Group, Imperial College London, London, UK.
| |
Collapse
|
19
|
Song Y, Liang F, Tian W, Rayhill E, Ye L, Tian X. Optimizing therapeutic outcomes: preconditioning strategies for MSC-derived extracellular vesicles. Front Pharmacol 2025; 16:1509418. [PMID: 39995418 PMCID: PMC11847897 DOI: 10.3389/fphar.2025.1509418] [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: 10/10/2024] [Accepted: 01/23/2025] [Indexed: 02/26/2025] Open
Abstract
Mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (MSC-EVs) are increasingly recognized for their therapeutic potential in regenerative medicine, driven by their capabilities in immunomodulation and tissue repair. However, MSCs present risks such as immunogenic responses, malignant transformation, and the potential to transmit infectious pathogens due to their intrinsic proliferative and differentiative abilities. In contrast, MSC-EVs, particularly exosomes (MSC-exosomes, 30-150 nm in diameter), offer a safer therapeutic profile. These acellular vesicles mitigate risks associated with immune rejection and tumorigenesis and are inherently incapable of forming ectopic tissues, thereby enhancing their clinical safety and applicability. This review highlights the therapeutic promise of MSC-exosomes especially focusing on the modulation of miRNA (one of bioactive molecules in MSC-EVs) profiles through various preconditioning strategies such as exposure to hypoxia, chemotherapeutic agents, inflammatory cytokines, and physical stimuli. Such conditioning is shown to optimize their therapeutic potential. Key miRNAs including miR-21, miR-146, miR-125a, miR-126, and miR-181a are particularly noted for their roles in facilitating tissue repair and modulating inflammatory responses. These functionalities position MSC-exosomes as a valuable tool in personalized medicine, particularly in the case of exosome-based interventions. Despite the potential of MSC-EVs, this review also acknowledged the limitations of traditional MSC therapies and advocates for a strategic pivot towards exosome-based modalities to enhance therapeutic outcomes. By discussing recent advances in detail and identifying remaining pitfalls, this review aims to guide future directions in improving the efficacy of MSC-exosome-based therapeutics. Additionally, miRNA variability in MSC-EVs presents challenges due to the diverse roles of miRNAs play in regulating gene expression and cell behavior. The miRNA content of MSC-EVs can be influenced by preconditioning strategies and differences in isolation and purification methods, which may alter the expression profiles of specific miRNAs, contributing to differences in their therapeutic effects.
Collapse
Affiliation(s)
- Yuqi Song
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Fengrui Liang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Weikun Tian
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Erin Rayhill
- Biology Department, Hamilton College, Clinton, NY, United States
| | - Liping Ye
- Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Xinghan Tian
- Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
20
|
Li W, Lu M, Shang J, Zhou J, Lin L, Liu Y, Zhao D, Zhu X. Hypoxic mesenchymal stem cell-derived exosomal circDennd2a regulates granulosa cell glycolysis by interacting with LDHA. Stem Cell Res Ther 2024; 15:484. [PMID: 39695793 DOI: 10.1186/s13287-024-04098-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is an ovarian dysfunction disorder that significantly impacts female fertility. Ovarian granulosa cells (GCs) are crucial somatic components supporting oocyte development that rely on glycolysis for energy production, which is essential for follicular growth. Hypoxia-induced exosomal circRNAs regulate glycolysis, but their biological functions and molecular mechanisms in POI are largely unexplored. The present comprehensive investigation revealed a substantial reduction in ovarian glycolysis levels in POI rats. Notably, hypoxia-induced exosomes originating from mesenchymal stem cells (HM-Exs) exhibit a remarkable capacity to enhance ovarian glycolysis, mitigate GCs apoptosis, reinstate disrupted estrous cycles, modulate sex hormone levels, and curtail the presence of atretic follicles. These restorative actions collectively contribute to fostering fertility revival in POI-afflicted rats. METHODS Cyclophosphamide was administered for 2 weeks to induce POI rat model, and POI rats were randomly divided into three groups and treated with PBS, NM-Exs and HM-Exs, respectively. Ovarian function and fertility were assessed at the end of the study and ovarian tissues were collected for analysis of energy metabolites. The relationship between circDennd2a and POI was explored in vitro by qRT-PCR, Western blotting, CCK-8 assay, EdU staining, TUNEL staining, extracellular acidification rate (ECAR) measurements, and ATP, lactate and pyruvate level assays. RESULTS Our findings revealed depletion of circDennd2a in serum samples and GCs from individuals suffering from POI. The introduction of HM-Exs-derived circDennd2a (HM-Exs-circDennd2a) effectively counteracted GCs apoptosis by enhancing glycolytic processes and driving cellular proliferation. CircDennd2a interacted with lactate dehydrogenase A (LDHA), which served as a catalyst to increase LDHA enzymatic activity and facilitate the conversion of NADH to NAD+. This biochemical cascade worked synergistically to sustain glycolytic function within GCs. CONCLUSION This study revealed that HM-Exs-circDennd2a promoted LDHA activity and enhanced GCs glycolytic capacity, both of which support its use as a potential clinical diagnostic and therapeutic target for POI.
Collapse
Affiliation(s)
- Wenxin Li
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, Jiangsu Province, 212001, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Minjun Lu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, Jiangsu Province, 212001, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Junyu Shang
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, Jiangsu Province, 212001, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jiamin Zhou
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, Jiangsu Province, 212001, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Li Lin
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, Jiangsu Province, 212001, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yueqin Liu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, Jiangsu Province, 212001, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Dan Zhao
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, Jiangsu Province, 212001, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaolan Zhu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, Jiangsu Province, 212001, China.
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China.
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
21
|
Zhang J, Ren N, Chen S, Liu K, Xiong L, Zheng X. Itga11 promotes osteogenic differentiation, inhibits angiogenesis and proliferation of mesenchymal stem cells under hypoxia. Tissue Cell 2024; 91:102616. [PMID: 39566247 DOI: 10.1016/j.tice.2024.102616] [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: 03/15/2024] [Revised: 10/26/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE This study aimed to explore the role and mechanism of hypoxic environment in rat bone mesenchymal stem cells (rBMSCs) proliferation, osteogenic differentiation and angiogenesis. METHODS Cell proliferation, angiogenesis and osteogenic differentiation were assessed using the CCK-8 assay, tube formation assay and alizarin red staining, respectively. Transcriptomic databases for rBMSCs under hypoxic (1 % O2) and normoxic (18 % O2) conditions were constructed to identify differentially expressed genes (DEGs), which were then subjected to gene function annotation and KEGG pathway analysis. To modulate the expression of Itga11, siRNA targeting Itga11 (si-Itga11) and a negative control (si-con), as well as pcDNA-Itga11 and an empty control plasmid (pcDNA), were employed to induce silencing or overexpression of Itga11. The protein levels were evaluated using Western blot analysis. RESULTS Hypoxia stimulated the proliferation and angiogenesis of rBMSCs but suppressed their osteogenic differentiation. Differential expression analysis identified 541 upregulated and 277 downregulated genes in the hypoxic group compared to the normoxic group. KEGG pathway enrichment analysis suggested that the hypoxic response in rBMSCs is closely associated with the Pi3k /Akt signaling pathway. Itga11 was significantly downregulated in rBMSCs under hypoxic conditions. Overexpression of Itga11 in rBMSCs inhibited their proliferation and angiogenesis and enhanced osteogenic differentiation, while its knockdown had the opposite effect. Itga11 was found to activate the Pi3k /Akt signaling pathway in rBMSCs. CONCLUSION Itga11 facilitates osteogenic differentiation and suppresses angiogenesis and proliferation of MSCs under hypoxia by activating the Pi3k /Akt signaling pathway.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Pharmacy, Hunan Vocational College of Science and Technology, Changsha, Hunan 410004, PR China.
| | - Na Ren
- Department of Pharmacy, Hunan Vocational College of Science and Technology, Changsha, Hunan 410004, PR China
| | - Shujuan Chen
- Department of Pharmacy, Hunan Vocational College of Science and Technology, Changsha, Hunan 410004, PR China
| | - Kun Liu
- School of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, PR China
| | - Lei Xiong
- School of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, PR China
| | - Xing Zheng
- Department of Pharmacy, Hunan Vocational College of Science and Technology, Changsha, Hunan 410004, PR China
| |
Collapse
|
22
|
Pawlik MT, Rinneberg G, Koch A, Meyringer H, Loew TH, Kjellberg A. Is there a rationale for hyperbaric oxygen therapy in the patients with Post COVID syndrome? : A critical review. Eur Arch Psychiatry Clin Neurosci 2024; 274:1797-1817. [PMID: 39545965 PMCID: PMC11579208 DOI: 10.1007/s00406-024-01911-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/16/2024] [Indexed: 11/17/2024]
Abstract
The SARS-CoV-2 pandemic has resulted in 762 million infections worldwide from 2020 to date, of which approximately ten percent are suffering from the effects after infection in 2019 (COVID-19) [1, 40]. In Germany, it is now assumed that at least one million people suffer from post-COVID condition with long-term consequences. These have been previously reported in diseases like Myalgic Encephalomyelitis (ME) and Chronic Fatigue Syndrome (CFS). Symptoms show a changing variability and recent surveys in the COVID context indicate that 10-30 % of outpatients, 50 to 70% of hospitalised patients suffer from sequelae. Recent data suggest that only 13% of all ill people were completely free of symptoms after recovery [3, 9]. Current hypotheses consider chronic inflammation, mitochondrial dysfunction, latent viral persistence, autoimmunity, changes of the human microbiome or multilocular sequelae in various organ system after infection. Hyperbaric oxygen therapy (HBOT) is applied since 1957 for heart surgery, scuba dive accidents, CO intoxication, air embolisms and infections with anaerobic pathogens. Under hyperbaric pressure, oxygen is physically dissolved in the blood in higher concentrations and reaches levels four times higher than under normobaric oxygen application. Moreover, the alternation of hyperoxia and normoxia induces a variety of processes at the cellular level, which improves oxygen supply in areas of locoregional hypoxia. Numerous target gene effects on new vessel formation, anti-inflammatory and anti-oedematous effects have been demonstrated [74]. The provision of intermittently high, local oxygen concentrations increases repair and regeneration processes and normalises the predominance of hyperinflammation. At present time only one prospective, randomized and placebo-controlled study exists with positive effects on global cognitive function, attention and executive function, psychiatric symptoms and pain interference. In conclusion, up to this date HBO is the only scientifically proven treatment in a prospective randomized controlled trial to be effective for cognitive improvement, regeneration of brain network and improvement of cardiac function. HBOT may have not only theoretical but also potential impact on targets of current pathophysiology of Post COVID condition, which warrants further scientific studies in patients.
Collapse
Affiliation(s)
- M T Pawlik
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany.
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel c/o German Naval Medical Institute, Kronshagen, Germany.
| | - G Rinneberg
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany
| | - A Koch
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel c/o German Naval Medical Institute, Kronshagen, Germany
| | - H Meyringer
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany
| | - T H Loew
- Department of Psychosomatic Medicine, University Hospital Regensburg, Regensburg, Germany
| | - A Kjellberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- Perioperative Medicine and Intensive Care, Medical Unit Intensive Care and Thoracic surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
23
|
Mi L, Zhang H. Myriad factors and pathways influencing tumor radiotherapy resistance. Open Life Sci 2024; 19:20220992. [PMID: 39655194 PMCID: PMC11627069 DOI: 10.1515/biol-2022-0992] [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: 05/07/2024] [Revised: 09/09/2024] [Accepted: 09/30/2024] [Indexed: 12/12/2024] Open
Abstract
Radiotherapy is a cornerstone in the treatment of various tumors, yet radioresistance often leads to treatment failure and tumor recurrence. Several factors contribute to this resistance, including hypoxia, DNA repair mechanisms, and cancer stem cells. This review explores the diverse elements that drive tumor radiotherapy resistance. Historically, resistance has been attributed to cellular repair and tumor repopulation, but recent research has expanded this understanding. The tumor microenvironment - characterized by hypoxia, immune evasion, and stromal interactions - further complicates treatment. Additionally, molecular mechanisms such as aberrant signaling pathways, epigenetic modifications, and non-B-DNA structures play significant roles in mediating resistance. This review synthesizes current knowledge, highlighting the interplay of these factors and their clinical implications. Understanding these mechanisms is crucial for developing strategies to overcome resistance and improve therapeutic outcomes in cancer patients.
Collapse
Affiliation(s)
- Lanjuan Mi
- School of Life and Health Sciences, Huzhou College, Hu Zhou, China
| | - Hongquan Zhang
- The First Affiliated Hospital of Huzhou University, Hu Zhou, China
| |
Collapse
|
24
|
Jiang N, Ying G, Yin Y, Guo J, Lozada J, Valdivia Padilla A, Gómez A, Gomes de Melo BA, Lugo Mestre F, Gansevoort M, Palumbo M, Calá N, Garciamendez-Mijares CE, Kim GA, Takayama S, Gerhard-Herman MD, Zhang YS. A closed-loop modular multiorgan-on-chips platform for self-sustaining and tightly controlled oxygenation. Proc Natl Acad Sci U S A 2024; 121:e2413684121. [PMID: 39541351 PMCID: PMC11588096 DOI: 10.1073/pnas.2413684121] [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] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
To mimic physiological microenvironments in organ-on-a-chip systems, physiologically relevant parameters are required to precisely access drug metabolism. Oxygen level is a critical microenvironmental parameter to maintain cellular or tissue functions and modulate their behaviors. Current organ-on-a-chip setups are oftentimes subjected to the ambient incubator oxygen level at 21%, which is higher than most if not all physiological oxygen concentrations. Additionally, the physiological oxygen level in each tissue is different ranging from 0.5 to 13%. Here, a closed-loop modular multiorgan-on-chips platform is developed to enable not only real-time monitoring of the oxygen levels but, more importantly, tight control of them in the range of 4 to 20% across each connected microtissue-on-a-chip in the circulatory culture medium. This platform, which consists of microfluidic oxygen scavenger(s), an oxygen generator, a monitoring/controller system, and bioreactor(s), allows for independent, precise upregulation and downregulation of dissolved oxygen in the perfused culture medium to meet the physiological oxygen level in each modular microtissue compartment, as needed. Furthermore, drug studies using the platform demonstrate that the oxygen level affects drug metabolism in the parallelly connected liver, kidney, and arterial vessel microtissues without organ-organ interactions factored in. Overall, this platform can promote the performances of organ-on-a-chip devices in drug screening by providing more physiologically relevant and independently adjustable oxygen microenvironments for desired organ types on a single- or a multiorgan-on-chip(s) configuration.
Collapse
Affiliation(s)
- Nan Jiang
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
- Department of Physics, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Guoliang Ying
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Yixia Yin
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Jie Guo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Jorge Lozada
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Alejandra Valdivia Padilla
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Ameyalli Gómez
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Bruna Alice Gomes de Melo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Francisco Lugo Mestre
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Merel Gansevoort
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Marcello Palumbo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Noemi Calá
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Carlos Ezio Garciamendez-Mijares
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Ge-Ah Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA30318
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA30318
| | - Marie Denis Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Yu Shrike Zhang
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
- Harvard Stem Cell Institute, Harvard University,Cambridge, MA02138
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA02142
| |
Collapse
|
25
|
Cherubini A, Rusconi F, Piras R, Wächtershäuser KN, Dossena M, Barilani M, Mei C, Hof L, Sordi V, Pampaloni F, Dolo V, Piemonti L, Lazzari L. Exploring human pancreatic organoid modelling through single-cell RNA sequencing analysis. Commun Biol 2024; 7:1527. [PMID: 39558019 PMCID: PMC11574267 DOI: 10.1038/s42003-024-07193-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/31/2024] [Indexed: 11/20/2024] Open
Abstract
Human organoids have been proposed to be powerful tools mimicking the physiopathological processes of the organs of origin. Recently, human pancreatic organoids (hPOs) have gained increasing attention due to potential theragnostic and regenerative medicine applications. However, the cellular components of hPOs have not been defined precisely. In this work, we finely characterized these structures, focusing first on morphology and identity-defining molecular features under long-term culture conditions. Next, we focused our attention on hPOs cell type composition using single-cell RNA sequencing founding a complex heterogeneity in ductal components, ranging from progenitor components to terminally differentiated ducts. Furthermore, an extensive comparison of human pancreatic organoids with previously reported transcriptomics signature of human and mouse pancreatic ductal populations, confirmed the functional pancreatic duct subpopulation heterogeneity. Finally, we showed that pancreatic organoid cells follow a precise developmental trajectory and utilize diverse signalling mechanisms, including EGF and SPP1, to facilitate cell-cell communication and maturation. Together our results offer an in-depth description of human pancreatic organoids providing a strong foundation for future in vitro diagnostic and translational studies of pancreatic health and disease.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Precision Medicine Lab - Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Francesco Rusconi
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberta Piras
- Department of Radiation Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kaja Nicole Wächtershäuser
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Marta Dossena
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mario Barilani
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia Mei
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lotta Hof
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Valeria Sordi
- IRCCS Ospedale San Raffaele, San Raffaele Diabetes Research Institute, Milan, Italy
| | - Francesco Pampaloni
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Lorenzo Piemonti
- IRCCS Ospedale San Raffaele, San Raffaele Diabetes Research Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenza Lazzari
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
26
|
Lee W, Lin SL, Chiang CS, Chen JY, Chieng WW, Huang SR, Chang TY, Linju Yen B, Hung MC, Chang KC, Lee HT, Jeng LB, Shyu WC. Role of HIF-1α-Activated IL-22/IL-22R1/Bmi1 Signaling Modulates the Self-Renewal of Cardiac Stem Cells in Acute Myocardial Ischemia. Stem Cell Rev Rep 2024; 20:2194-2214. [PMID: 39264501 PMCID: PMC11554697 DOI: 10.1007/s12015-024-10774-8] [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: 08/06/2024] [Indexed: 09/13/2024]
Abstract
Impaired tissue regeneration negatively impacts on left ventricular (LV) function and remodeling after acute myocardial infarction (AMI). Little is known about the intrinsic regulatory machinery of ischemia-induced endogenous cardiac stem cells (eCSCs) self-renewing divisions after AMI. The interleukin 22 (IL-22)/IL-22 receptor 1 (IL-22R1) pathway has emerged as an important regulator of several cellular processes, including the self-renewal and proliferation of stem cells. However, whether the hypoxic environment could trigger the self-renewal of eCSCs via IL-22/IL-22R1 activation remains unknown. In this study, the upregulation of IL-22R1 occurred due to activation of hypoxia-inducible factor-1α (HIF-1α) under hypoxic and ischemic conditions. Systemic IL-22 administration not only attenuated cardiac remodeling, inflammatory responses, but also promoted eCSC-mediated cardiac repair after AMI. Unbiased RNA microarray analysis showed that the downstream mediator Bmi1 regulated the activation of CSCs. Therefore, the HIF-1α-induced IL-22/IL-22R1/Bmi1 cascade can modulate the proliferation and activation of eCSCs in vitro and in vivo. Collectively, investigating the HIF-1α-activated IL-22/IL-22R1/Bmi1 signaling pathway might offer a new therapeutic strategy for AMI via eCSC-induced cardiac repair.
Collapse
Affiliation(s)
- Wei Lee
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
| | - Syuan-Ling Lin
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Chih-Sheng Chiang
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University (CMU), Taichung, 404, Taiwan
- Neuroscience and Brain Disease Center and New Drug Development Center, CMU, Taichung, 404, Taiwan
| | - Jui-Yu Chen
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Wee-Wei Chieng
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Shu-Rou Huang
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Ting-Yu Chang
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan, 350, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences and Research Centers for Cancer Biology and Molecular Medicine, CMU, Taichung, 404, Taiwan
| | - Kuan-Cheng Chang
- Division of Cardiovascular Medicine, Department of Medicine, CMUH, Taichung, 404, Taiwan
- School of Medicine, CMU, Taichung, 404, Taiwan
| | - Hsu-Tung Lee
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, 404, Taiwan
| | - Long-Bin Jeng
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
- Organ Transplantation Center, CMUH, Taichung, 404, Taiwan
| | - Woei-Cherng Shyu
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University (CMU), Taichung, 404, Taiwan.
- Neuroscience and Brain Disease Center and New Drug Development Center, CMU, Taichung, 404, Taiwan.
- Department of Neurology, CMUH, Taichung, 404, Taiwan.
- Department of Occupational Therapy, Asia University, No. 2, Yude Rd., North Dist, Taichung City, 404332, Taiwan.
| |
Collapse
|
27
|
Cooper PO, Yang J, Wang HH, Broman MM, Jayasundara SM, Sahoo SS, Yan B, Awdalkreem GD, Cresswell GM, Wang L, Goossens E, Lanman NA, Doerge RW, Zheng F, Cheng L, Alqahtani S, Crist SA, Braun RE, Kazemian M, Jerde TJ, Ratliff TL. Inflammation impacts androgen receptor signaling in basal prostate stem cells through interleukin 1 receptor antagonist. Commun Biol 2024; 7:1390. [PMID: 39455902 PMCID: PMC11511867 DOI: 10.1038/s42003-024-07071-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: 12/04/2023] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Chronic prostate inflammation in patients with benign prostate hyperplasia (BPH) correlates with the severity of symptoms. How inflammation contributes to prostate enlargement and/or BPH symptoms and the underlying mechanisms remain unclear. In this study, we utilize a unique transgenic mouse model that mimics chronic non-bacterial prostatitis in men and investigate the impact of inflammation on androgen receptor (AR) in basal prostate stem cells (bPSC) and their differentiation in vivo. We find that inflammation significantly enhances AR levels and activity in bPSC. More importantly, we identify interleukin 1 receptor antagonist (IL-1RA) as a crucial regulator of AR in bPSC during inflammation. IL-1RA is one of the top molecules upregulated by inflammation, and inhibiting IL-1RA reverses the enhanced AR activity in organoids derived from inflamed bPSC. Additionally, IL-1RA appears to activate AR by counteracting IL-1α's inhibitory effect. Furthermore, using a lineage tracing model, we observe that inflammation induces bPSC proliferation and differentiation into luminal cells even under castrate conditions, indicating that AR activation driven by inflammation is sufficient to promote bPSC proliferation and differentiation. Taken together, our study uncovers mechanisms through which inflammation modulates AR signaling in bPSC and induces bPSC luminal differentiation that may contribute to prostate hyperplasia.
Collapse
Affiliation(s)
- Paula O Cooper
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Jiang Yang
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Cancer Research, West Lafayette, IN, USA.
| | - Hsing-Hui Wang
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Immune Monitoring and Genomics Facility, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Meaghan M Broman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | | | | | - Bingyu Yan
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Gada D Awdalkreem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | - Gregory M Cresswell
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Flow Cytometry Core Facility, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Liang Wang
- Department of Pharmacology and Toxicology, Department of Urology, Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Emery Goossens
- Department of Statistics, Purdue University, West Lafayette, IN, USA
| | - Nadia A Lanman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | - Rebecca W Doerge
- Department of Statistics, Purdue University, West Lafayette, IN, USA
- Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Faye Zheng
- Department of Statistics, Purdue University, West Lafayette, IN, USA
- Sorcero, Inc., Washington, DC, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Department of Surgery (Urology), Brown University Warren Alpert Medical School, the Legorreta Cancer Center at Brown University, and Brown University Health, Providence, RI, USA
| | - Saeed Alqahtani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | - Scott A Crist
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Carver College of Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
| | | | - Majid Kazemian
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Travis J Jerde
- Department of Pharmacology and Toxicology, Department of Urology, Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Timothy L Ratliff
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Cancer Research, West Lafayette, IN, USA.
| |
Collapse
|
28
|
Zhang H, Jin C, Hua J, Chen Z, Gao W, Xu W, Zhou L, Shan L. Roles of Microenvironment on Mesenchymal Stem Cells Therapy for Osteoarthritis. J Inflamm Res 2024; 17:7069-7079. [PMID: 39377043 PMCID: PMC11457791 DOI: 10.2147/jir.s475617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/21/2024] [Indexed: 10/09/2024] Open
Abstract
Osteoarthritis (OA) induced microenvironmental alterations are a common and unavoidable phenomenon that greatly exacerbate the pathologic process of OA. Imbalances in the synthesis and degradation of cartilage extracellular matrix (ECM) have been reported to be associated with an adverse microenvironment. Stem cell therapy is a promising treatment for OA, and mesenchymal stem cells (MSCs) are the main cell sources for this therapy. With multispectral differentiation and immunomodulation, MSCs can effectively regulate the microenvironment of articular cartilage, ameliorate inflammation, promote regeneration of damaged cartilage, and ultimately alleviate OA symptoms. However, the efficacy of MSCs in the treatment of OA is greatly influenced by articular cavity microenvironments. This article reviews the five microenvironments of OA articular cavity, including inflammatory microenvironment, senescence microenvironment, hypoxic microenvironment, high glucose microenvironment and high lipid environment, focus on the positive and negative effects of OA microenvironments on the fate of MSCs. In this regard, we emphasize the mechanisms of the current use of MSCs in OA treatment, as well as its limitations and challenges.
Collapse
Affiliation(s)
- Haiyan Zhang
- The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Chaoying Jin
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiaqing Hua
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Wenxin Gao
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Wenting Xu
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Letian Shan
- The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
29
|
Huang X, Lin Z, Zheng ZM, Shi JL, Lu KY, Wang JR, Li MQ, Shao J. A Hypoxia-Decidual Macrophage Regulatory Axis in Normal Pregnancy and Spontaneous Miscarriage. Int J Mol Sci 2024; 25:9710. [PMID: 39273657 PMCID: PMC11395248 DOI: 10.3390/ijms25179710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
The significance of hypoxia at the maternal-fetal interface is proven to be self-explanatory in the context of pregnancy. During the first trimester, low oxygen conditions play a crucial role in processes such as angiogenesis, trophoblast invasion and differentiation, and immune regulation. Recently, there has been increasing research on decidual macrophages, which contribute to the maintenance of immune tolerance, placental and fetal vascular development, and spiral artery remodeling, to investigate the effects of hypoxia on their biological behaviors. On these grounds, this review describes the dynamic changes in oxygen levels at the maternal-fetal interface throughout gestation, summarizing current knowledge on how the hypoxic environment sustains a successful pregnancy by regulating retention, differentiation and efferocytosis of decidual macrophages. Additionally, we explore the relationship between spontaneous miscarriages and an abnormal hypoxia-macrophage axis, shedding light on the underlying mechanisms. However, further studies are essential to elucidate these pathways in greater detail and to develop targeted interventions that could improve pregnancy outcomes.
Collapse
Affiliation(s)
- Xu Huang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
| | - Zhi Lin
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200010, China
| | - Zi-Meng Zheng
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jia-Lu Shi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200010, China
| | - Ke-Yu Lu
- Xing Lin College, Nantong University, Nantong 226236, China
| | - Jia-Rui Wang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200010, China
| | - Ming-Qing Li
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jun Shao
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200010, China
| |
Collapse
|
30
|
Meng H, Huan Y, Zhang K, Yi X, Meng X, Kang E, Wu S, Deng W, Wang Y. Quiescent Adult Neural Stem Cells: Developmental Origin and Regulatory Mechanisms. Neurosci Bull 2024; 40:1353-1363. [PMID: 38656419 PMCID: PMC11365920 DOI: 10.1007/s12264-024-01206-1] [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: 09/26/2023] [Accepted: 01/02/2024] [Indexed: 04/26/2024] Open
Abstract
The existence of neural stem cells (NSCs) in the adult mammalian nervous system, although small in number and restricted to the sub-ventricular zone of the lateral ventricles, the dentate gyrus of the hippocampus, and the olfactory epithelium, is a gift of evolution for the adaptive brain function which requires persistent plastic changes of these regions. It is known that most adult NSCs are latent, showing long cell cycles. In the past decade, the concept of quiescent NSCs (qNSCs) has been widely accepted by researchers in the field, and great progress has been made in the biology of qNSCs. Although the spontaneous neuronal regeneration derived from adult NSCs is not significant, understanding how the behaviors of qNSCs are regulated sheds light on stimulating endogenous NSC-based neuronal regeneration. In this review, we mainly focus on the recent progress of the developmental origin and regulatory mechanisms that maintain qNSCs under normal conditions, and that mobilize qNSCs under pathological conditions, hoping to give some insights for future study.
Collapse
Affiliation(s)
- Han Meng
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu Huan
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, 110016, China
| | - Kun Zhang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xuyang Yi
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xinyu Meng
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- School of Life Science and Research Center for Natural Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yan'an, 716000, China
| | - Enming Kang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Wenbing Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 510631, China.
| | - Yazhou Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
31
|
Chettouh-Hammas N, Grillon C. Physiological skin oxygen levels: An important criterion for skin cell functionality and therapeutic approaches. Free Radic Biol Med 2024; 222:259-274. [PMID: 38908804 DOI: 10.1016/j.freeradbiomed.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The skin is made up of different layers with various gradients, which maintain a complex microenvironment, particularly in terms of oxygen levels. However, all types of skin cells are cultured in conventional incubators that do not reproduce physiological oxygen levels. Instead, they are cultured at atmospheric oxygen levels, a condition that is far removed from physiology and may lead to the generation of free radicals known to induce skin ageing. This review aims to summarize the current literature on the effect of physiological oxygen levels on skin cells, highlight the shortcomings of current in vitro models, and demonstrate the importance of respecting skin oxygen levels. We begin by clarifying the terminology used about oxygen levels and describe the specific distribution of oxygen in the skin. We review and discuss how skin cells adapt their oxygen consumption and metabolism to oxygen levels environment, as well as the changes that are induced, particularly, their redox state, life cycle and functions. We examine the effects of oxygen on both simple culture models and more complex reconstructed skin models. Finally, we present the implications of oxygen modulation for a more therapeutic approach.
Collapse
Affiliation(s)
- Nadira Chettouh-Hammas
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071, Orléans, Cedex 2, France.
| | - Catherine Grillon
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071, Orléans, Cedex 2, France.
| |
Collapse
|
32
|
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
|
33
|
Li Y, Xie J, Du X, Chen Y, Wang C, Liu T, Yi Z, Wang Y, Zhao M, Li X, Shi S. Oridonin, a small molecule inhibitor of cancer stem cell with potent cytotoxicity and differentiation potential. Eur J Pharmacol 2024; 975:176656. [PMID: 38754536 DOI: 10.1016/j.ejphar.2024.176656] [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: 02/08/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/18/2024]
Abstract
Cancer stem cells (CSCs) drive malignant tumor progression, recurrence, and metastasis with unique characteristics, including self-renewal and resistance to conventional treatments. Conventional differentiation inducers, although promising, have limited cytotoxicity and may inadvertently enhance CSC stemness. To address these challenges, ongoing efforts are dedicated to developing strategies that can effectively combine both cytotoxicity and differentiation-inducing effects. In this study, we introduce oridonin (Ori), a small molecule with dual differentiation-inducing and cytotoxicity properties capable of eliminating tumor CSCs. We isolated CSCs in B16F10 cells using the Hoechst side population method and assessed the differentiation effect of Ori. Ori's differentiation-inducing effect was further evaluated using human acute promyelocytic leukemia. The cytotoxic potential of Ori against MCF-7 and B16F10 cell lines was assessed through various methods. In vivo anti-tumor and anti-CSC efficacy of Ori was investigated using mouse melanoma and CSCs melanoma models. Safety evaluation included zebrafish embryotoxicity and mouse acute toxicity experiments. As a result, Ori effectively dismantles tumorspheres, inhibits proliferation, and reduces the expression of CSC-specific markers. It induces significant differentiation, especially in the case of NB4. Additionally, Ori upregulates TP53 expression, mitigates the hypoxic tumor microenvironment, suppresses stemness, and inhibits PD-L1 expression, prompting a robust anti-cancer immune response. Ori demonstrates pronounced cytotoxicity, inducing notable pro-apoptotic effects on B16F10 and MCF-7 cells, with specific triggering of mitochondrial apoptosis. Importantly, Ori maintains a commendable biosafety record. The dual-action prowess of Ori not only induces the differentiation of CSCs but also dispatches differentiated and residual tumor cells, effectively thwarting the relentless march of tumor progression.
Collapse
Affiliation(s)
- Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinjin Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuan Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tiantian Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhiwen Yi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
34
|
Bhattacharya R, Brown JS, Gatenby RA, Ibrahim-Hashim A. A gene for all seasons: The evolutionary consequences of HIF-1 in carcinogenesis, tumor growth and metastasis. Semin Cancer Biol 2024; 102-103:17-24. [PMID: 38969311 DOI: 10.1016/j.semcancer.2024.06.003] [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: 02/06/2024] [Revised: 04/23/2024] [Accepted: 06/06/2024] [Indexed: 07/07/2024]
Abstract
Oxygen played a pivotal role in the evolution of multicellularity during the Cambrian Explosion. Not surprisingly, responses to fluctuating oxygen concentrations are integral to the evolution of cancer-a disease characterized by the breakdown of multicellularity. Poorly organized tumor vasculature results in chaotic patterns of blood flow characterized by large spatial and temporal variations in intra-tumoral oxygen concentrations. Hypoxia-inducible growth factor (HIF-1) plays a pivotal role in enabling cells to adapt, metabolize, and proliferate in low oxygen conditions. HIF-1 is often constitutively activated in cancers, underscoring its importance in cancer progression. Here, we argue that the phenotypic changes mediated by HIF-1, in addition to adapting the cancer cells to their local environment, also "pre-adapt" them for proliferation at distant, metastatic sites. HIF-1-mediated adaptations include a metabolic shift towards anaerobic respiration or glycolysis, activation of cell survival mechanisms like phenotypic plasticity and epigenetic reprogramming, and formation of tumor vasculature through angiogenesis. Hypoxia induced epigenetic reprogramming can trigger epithelial to mesenchymal transition in cancer cells-the first step in the metastatic cascade. Highly glycolytic cells facilitate local invasion by acidifying the tumor microenvironment. New blood vessels, formed due to angiogenesis, provide cancer cells a conduit to the circulatory system. Moreover, survival mechanisms acquired by cancer cells in the primary site allow them to remodel tissue at the metastatic site generating tumor promoting microenvironment. Thus, hypoxia in the primary tumor promoted adaptations conducive to all stages of the metastatic cascade from the initial escape entry into a blood vessel, intravascular survival, extravasation into distant tissues, and establishment of secondary tumors.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Department of Cancer Biology, University of South Florida, United States; Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, United States
| | - Joel S Brown
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, United States; Department of Evolutionary Biology, University of Illinois, at Chicago, United States
| | - Robert A Gatenby
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, United States; Department of Radiology, H. Lee Moffitt Cancer Center, United States.
| | - Arig Ibrahim-Hashim
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, United States.
| |
Collapse
|
35
|
Zhao X, Luo T, Qiu Y, Yang Z, Wang D, Wang Z, Zeng J, Bi Z. Mechanisms of traditional Chinese medicine overcoming of radiotherapy resistance in breast cancer. Front Oncol 2024; 14:1388750. [PMID: 38993643 PMCID: PMC11237312 DOI: 10.3389/fonc.2024.1388750] [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: 02/20/2024] [Accepted: 05/30/2024] [Indexed: 07/13/2024] Open
Abstract
Breast cancer stands as the most prevalent malignancy among women, with radiotherapy serving as a primary treatment modality. Despite radiotherapy, a subset of breast cancer patients experiences local recurrence, attributed to the intrinsic resistance of tumors to radiation. Therefore, there is a compelling need to explore novel approaches that can enhance cytotoxic effects through alternative mechanisms. Traditional Chinese Medicine (TCM) and its active constituents exhibit diverse pharmacological actions, including anti-tumor effects, offering extensive possibilities to identify effective components capable of overcoming radiotherapy resistance. This review delineates the mechanisms underlying radiotherapy resistance in breast cancer, along with potential candidate Chinese herbal medicines that may sensitize breast cancer cells to radiotherapy. The exploration of such herbal interventions holds promise for improving therapeutic outcomes in the context of breast cancer radiotherapy resistance.
Collapse
Affiliation(s)
- Xiaohui Zhao
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Oncology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-Sen University, Shanwei, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ting Luo
- Department of Oncology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-Sen University, Shanwei, China
| | - Yuting Qiu
- Department of Oncology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-Sen University, Shanwei, China
| | - Zhiwei Yang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Danni Wang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zairui Wang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiale Zeng
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuofei Bi
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
36
|
Chen W, Wang P, Liu C, Han Y, Zhao F. Male Germ Cell Specification in Plants. Int J Mol Sci 2024; 25:6643. [PMID: 38928348 PMCID: PMC11204311 DOI: 10.3390/ijms25126643] [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: 05/01/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Germ cells (GCs) serve as indispensable carriers in both animals and plants, ensuring genetic continuity across generations. While it is generally acknowledged that the timing of germline segregation differs significantly between animals and plants, ongoing debates persist as new evidence continues to emerge. In this review, we delve into studies focusing on male germ cell specifications in plants, and we summarize the core gene regulatory circuits in germ cell specification, which show remarkable parallels to those governing meristem homeostasis. The similarity in germline establishment between animals and plants is also discussed.
Collapse
Affiliation(s)
- Wenqian Chen
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi’an 710129, China; (W.C.); (P.W.); (C.L.); (Y.H.)
| | - Pan Wang
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi’an 710129, China; (W.C.); (P.W.); (C.L.); (Y.H.)
| | - Chan Liu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi’an 710129, China; (W.C.); (P.W.); (C.L.); (Y.H.)
| | - Yuting Han
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi’an 710129, China; (W.C.); (P.W.); (C.L.); (Y.H.)
| | - Feng Zhao
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi’an 710129, China; (W.C.); (P.W.); (C.L.); (Y.H.)
- Collaborative Innovation Center of Northwestern Polytechnical University, Shanghai 201108, China
| |
Collapse
|
37
|
Tian Y, Cheng Z, Ge D, Xu Z, Wang H, Li X, Tian H, Liu F, Luo D, Wang Y. ROS are required for the germinative cell proliferation and metacestode larval growth of Echinococcus multilocularis. Front Microbiol 2024; 15:1410504. [PMID: 38912347 PMCID: PMC11190091 DOI: 10.3389/fmicb.2024.1410504] [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: 04/01/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024] Open
Abstract
The potentially lethal zoonotic disease alveolar echinococcosis (AE) is caused by the metacestode larval stages of the tapeworm Echinococcus multilocularis. Metacestode growth and proliferation occurs within the inner organs of mammalian hosts, which is associated with complex molecular parasite-host interactions. The host has developed various ways to resist a parasitic infection, and the production of reactive oxygen species (ROS) is one of the most important strategies. Here, we found that scavenging of ROS reduced metacestode larval growth and germinative cell proliferation in in vivo models. Furthermore, using in vitro-cultured metacestode vesicles, we found that increased ROS levels enhanced metacestode growth and germinative cell proliferation, which was achieved by positively activating the ROS-EmERK-EmHIF1α axis. These results indicate that, beside its capacity to damage the parasite, ROS also play critical roles in metacestode growth and germinative cell proliferation. This study suggests that the effects of ROS on parasite may be bidirectional during AE infection, reflecting the parasite's adaptation to the oxidative stress microenvironment.
Collapse
Affiliation(s)
- Ye Tian
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhe Cheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Defeng Ge
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhijian Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Huijuan Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiazhen Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Huimin Tian
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Fan Liu
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Damin Luo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yanhai Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
38
|
Olson SL, Akbar RJ, Gorniak A, Fuhr LI, Borahay MA. Hypoxia in uterine fibroids: role in pathobiology and therapeutic opportunities. OXYGEN (BASEL, SWITZERLAND) 2024; 4:236-252. [PMID: 38957794 PMCID: PMC11218552 DOI: 10.3390/oxygen4020013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Uterine fibroids are the most common tumors in females affecting up to 70% of women world-wide, yet targeted therapeutic options are limited. Oxidative stress has recently surfaced as a key driver of fibroid pathogenesis and provides insights into hypoxia-induced cell transformation, extracellular matrix pathophysiology, hypoxic cell signaling cascades, and uterine biology. Hypoxia drives fibroid tumorigenesis through (1) promoting myometrial stem cell proliferation, (2) causing DNA damage propelling transformation of stem cells to tumor initiating cells, and (3) driving excess extracellular matrix (ECM) production. Common fibroid-associated DNA mutations include MED12 mutations, HMGA2 overexpression, and Fumarate hydratase loss of function. Evidence suggests an interaction between hypoxia signaling and these mutations. Fibroid development and growth are promoted by hypoxia-triggered cell signaling via various pathways including HIF-1, TGFβ, and Wnt/β-catenin. Fibroid-associated hypoxia persists due to antioxidant imbalance, ECM accumulation, and growth beyond adequate vascular supply. Current clinically available fibroid treatments do not take advantage of hypoxia-targeting therapies. Growing pre-clinical and clinical studies identify ROS inhibitors, anti-HIF-1 agents, Wnt/β-catenin inhibition, and TGFβ cascade inhibitors as agents that may reduce fibroid development and growth through targeting hypoxia.
Collapse
Affiliation(s)
- Sydney L. Olson
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | | | - Adrianna Gorniak
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Laura I. Fuhr
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Mostafa A. Borahay
- Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
39
|
Eweida A, Sandberg E, Ritthaler O, Fleckenstein J, Abo-Madyan Y, Giordano FA, Schulte M, Kneser U, Harhaus L. Hypoxia as a stimulus for tissue formation: The concept of organogenesis in microsurgically vascularized tissue engineering constructs. J Craniomaxillofac Surg 2024; 52:707-714. [PMID: 38582676 DOI: 10.1016/j.jcms.2024.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/12/2024] [Indexed: 04/08/2024] Open
Abstract
Axial vascularization of tissue constructs is essential to maintain an adequate blood supply for a stable regeneration of a clinically relevant tissue size. The versatility of the arterio-venous loop (AVL) has been previously shown in various small and large animal models as well as in clinical reports for bone regeneration. We have previously demonstrated the capability of the AVL to induce axial vascularization and to support the nourishment of tissue constructs in small animal models after applying high doses of ionizing radiation comparable to those applied for adjuvant radiotherapy after head and neck cancer. We hypothesize that this robust ability to induce regeneration after irradiation could be related to a state of hypoxia inside the constructs that triggers the HIF1 (hypoxia induced factor 1) - SDF1 (stromal derived factor 1) axis leading to chemotaxis of progenitor cells and induction of tissue regeneration and vascularization. We analyzed the expression of HIF1 and SDF1 via immunofluorescence in axially vascularized bone tissue engineering constructs in Lewis rats 2 and 5 weeks after local irradiation with 9Gy or 15Gy. We also analyzed the expression of various genes for osteogenic differentiation (collagen 1, RUNX, alkaline phosphatase and osteonectin) via real time PCR analysis. The expression of HIF1 and SDF1 was enhanced two weeks after irradiation with 15Gy in comparison to non-irradiated constructs. The expression of osteogenic markers was enhanced at the 5-weeks time point with significant results regarding collagen, alkaline phosphatase and osteonectin. These results indicate that the hypoxia within the AVL constructs together with an enhanced SDF1 expression probably play a role in promoting tissue differentiation. The process of tissue generation triggered by hypoxia in the vicinity of a definite vascular axis with enhanced tissue differentiation over time resembles hereby the well-known concept of organogenesis in fetal life.
Collapse
Affiliation(s)
- Ahmad Eweida
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany; Department of Head, Neck and Endocrine Surgery, Faculty of Medicine, University of Alexandria, Alkhartoum Square, 5372066, Alexandria, Egypt.
| | - Elli Sandberg
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany
| | - Oliver Ritthaler
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany
| | - Jens Fleckenstein
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Yasser Abo-Madyan
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Frank Anton Giordano
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Matthias Schulte
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany
| | - Ulrich Kneser
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany
| | - Leila Harhaus
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Str. 13, 67071, Ludwigshafen, Germany
| |
Collapse
|
40
|
Chen C, Song C, Liu B, Wang Y, Jia J, Pang K, Wang Y, Wang P. Activation of BMP4/SMAD pathway by HIF-1α in hypoxic environment promotes osteogenic differentiation of BMSCs and leads to ectopic bone formation. Tissue Cell 2024; 88:102376. [PMID: 38608407 DOI: 10.1016/j.tice.2024.102376] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 03/15/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVE Heterotopic ossification (HO), also known as ossifying myositis, is a condition that produces abnormal bone and cartilage tissue in the soft tissues. Hypoxia inducible factor lα (HIF-lα) regulates the expression of various genes, which is closely related to the promotion of bone formation, and Drosophila mothers against decapentaplegic protein (SMAD) mediates the signal transduction in the Bone morphogenetic protein (BMP) signaling pathway, which affects the function of osteoblasts and osteoclasts, and thus plays a key role in the regulation of bone remodeling. We aimed to investigate the mechanism by which HIF-1α induces osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in a hypoxic environment. METHODS A cellular hypoxia model was constructed to verify the expression of HIF-1α, while alizarin red staining was performed to observe the osteogenic differentiation ability of bone marrow mesenchymal stem cells (BMSCs). Alizarin red staining was used to analyze the late mineralization ability of the cells. Western blot analysis was performed to analyze the expression levels of osteogenesis-related factors OCN, OPN proteins as well as the pathway proteins BMP4, p-Smad1/5/8, and Smad1. We also constructed a rat model of ectopic bone formation, observed ectopic ossification by X-ray, and verified the success of the rat model by ELISA of HIF-1α. HE staining was used to observe the matrix and trabecular structure of bone, and Masson staining was used to observe the collagen and trabecular structure of bone. Immunohistochemistry analyzed the expression of OCN and OPN in ectopic bone tissues, and WB analyzed the expression of pathway proteins BMP4, p-Smad1/5/8 and Smad1 in ectopic bone tissues to verify the signaling pathway of ectopic bone formation. RESULTS Our results indicate that hypoxic environment upregulates HIF-1a expression and activates BMP4/SMAD signaling pathway. This led to an increase in ALP content and enhanced expression of the osteogenesis-related factors OCN and OPN, resulting in enhanced osteogenic differentiation of BMSCs. The results of our in vivo experiments showed that rats inoculated with BMSCs overexpressing HIF-1α showed bony structures in tendon tissues, enhanced expression of the bone signaling pathways BMP4 and p-Smad1/5/8, and enhanced expression levels of the osteogenic-related factors OCN and OPN, resulting in the formation of ectopic bone. CONCLUSIONS These data further suggest a novel mechanistic view that hypoxic bone marrow BMSCs activate the BMP4/SMAD pathway by up-regulating the expression level of HIF-1α, thereby promoting the secretion of osteogenic factors leading to ectopic bone formation.
Collapse
Affiliation(s)
- Cong Chen
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Chunhao Song
- Department of Medical Imaging, Weihai Wendeng District People Hospital, Weihai 264200, China
| | - Bo Liu
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Yitao Wang
- Department of Laboratory, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Jun Jia
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Kai Pang
- Department of Operations Management, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Yuanhao Wang
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China
| | - Peng Wang
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai 264200, China.
| |
Collapse
|
41
|
Huynh GT, Tunny SS, Frith JE, Meagher L, Corrie SR. Organosilica Nanosensors for Monitoring Spatiotemporal Changes in Oxygen Levels in Bacterial Cultures. ACS Sens 2024; 9:2383-2394. [PMID: 38687178 DOI: 10.1021/acssensors.3c02747] [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: 05/02/2024]
Abstract
Oxygen plays a central role in aerobic metabolism, and while many approaches have been developed to measure oxygen concentration in biological environments over time, monitoring spatiotemporal changes in dissolved oxygen levels remains challenging. To address this, we developed a ratiometric core-shell organosilica nanosensor for continuous, real-time optical monitoring of oxygen levels in biological environments. The nanosensors demonstrate good steady state characteristics (KpSV = 0.40 L/mg, R2 = 0.95) and respond reversibly to changes in oxygen concentration in buffered solutions and report similar oxygen level changes in response to bacterial cell growth (Escherichia coli) in comparison to a commercial bulk optode-based sensing film. We further demonstrated that the oxygen nanosensors could be distributed within a growing culture of E. coli and used to record oxygen levels over time and in different locations within a static culture, opening the possibility of spatiotemporal monitoring in complex biological systems.
Collapse
Affiliation(s)
- Gabriel T Huynh
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO) Manufacturing, Clayton, VIC 3168, Australia
| | - Salma S Tunny
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Simon R Corrie
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
42
|
Granath-Panelo M, Kajimura S. Mitochondrial heterogeneity and adaptations to cellular needs. Nat Cell Biol 2024; 26:674-686. [PMID: 38755301 DOI: 10.1038/s41556-024-01410-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/21/2024] [Indexed: 05/18/2024]
Abstract
Although it is well described that mitochondria are at the epicentre of the energy demands of a cell, it is becoming important to consider how each cell tailors its mitochondrial composition and functions to suit its particular needs beyond ATP production. Here we provide insight into mitochondrial heterogeneity throughout development as well as in tissues with specific energy demands and discuss how mitochondrial malleability contributes to cell fate determination and tissue remodelling.
Collapse
Affiliation(s)
- Melia Granath-Panelo
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA.
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Shingo Kajimura
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
43
|
Shi Y, Yang X, Min J, Kong W, Hu X, Zhang J, Chen L. Advancements in culture technology of adipose-derived stromal/stem cells: implications for diabetes and its complications. Front Endocrinol (Lausanne) 2024; 15:1343255. [PMID: 38681772 PMCID: PMC11045945 DOI: 10.3389/fendo.2024.1343255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/29/2024] [Indexed: 05/01/2024] Open
Abstract
Stem cell-based therapies exhibit considerable promise in the treatment of diabetes and its complications. Extensive research has been dedicated to elucidate the characteristics and potential applications of adipose-derived stromal/stem cells (ASCs). Three-dimensional (3D) culture, characterized by rapid advancements, holds promise for efficacious treatment of diabetes and its complications. Notably, 3D cultured ASCs manifest enhanced cellular properties and functions compared to traditional monolayer-culture. In this review, the factors influencing the biological functions of ASCs during culture are summarized. Additionally, the effects of 3D cultured techniques on cellular properties compared to two-dimensional culture is described. Furthermore, the therapeutic potential of 3D cultured ASCs in diabetes and its complications are discussed to provide insights for future research.
Collapse
Affiliation(s)
- Yinze Shi
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Xueyang Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Jie Min
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Xiang Hu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Jiaoyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| |
Collapse
|
44
|
Li L, Liu Y, Zhi N, Ji Y, Xu J, Mao G, Wang Y, Ma J, Wang Y. Hypoxic preconditioning accelerates the healing of ischemic intestinal injury by activating HIF-1α/PPARα pathway-mediated fatty acid oxidation. Cell Death Discov 2024; 10:164. [PMID: 38575595 PMCID: PMC10994932 DOI: 10.1038/s41420-024-01937-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024] Open
Abstract
Hypoxic preconditioning (HPC) has been shown to improve organ tolerance to subsequent severe hypoxia or ischemia. However, its impact on intestinal ischemic injury has not been well studied. In this study, we evaluated the effects of HPC on intestinal ischemia in rats. Intestinal rehabilitation, levels of fatty acid oxidation (FAO) by-products, intestinal stem cells (ISCs), levels of hypoxia-inducible factor 1 subunit α (HIF-1α) and its downstream genes such as peroxisome proliferator-activated receptor α (PPARα), and carnitine palmitoyltransferase 1a (CPT1A) were assessed at distinct time intervals following intestinal ischemia with or without the interference of HIF-1α. Our data showed that HPC facilitates the restoration of the intestinal structure and enhances the FAO, by boosting intestinal stem cells. Additionally, HIF-1α, PPARα, and CPT1A mRNA and their protein levels were generally up-regulated in the small intestine of HPC rats as compared to the control group. Our vitro experiment also shows low-oxygen induces highly levels of HIF-1α and its downstream genes, with a concurrent increase in FAO products in IEC-6 cells. Furthermore, the above phenomenon could be reversed by silencing HIF-1α. In conclusion, we hypothesize that HPC can stimulate the activation of intestinal stem cells via HIF-1α/PPARα pathway-mediated FAO, thereby accelerating the healing process post ischemic intestinal injury.
Collapse
Affiliation(s)
- Linxia Li
- Department of Aerospace Medicine, Air Force Medical University, 710032, Xi'an, China
| | - Yanqi Liu
- Department of Aerospace Medicine, Air Force Medical University, 710032, Xi'an, China
| | - Na Zhi
- Department of Aerospace Medicine, Air Force Medical University, 710032, Xi'an, China
| | - Yaoxuan Ji
- Department of Aerospace Medicine, Air Force Medical University, 710032, Xi'an, China
| | - Jialing Xu
- Department of Aerospace Medicine, Air Force Medical University, 710032, Xi'an, China
| | - Guoyun Mao
- Department of Aerospace Medicine, Air Force Medical University, 710032, Xi'an, China
| | - Yazhou Wang
- Department of Neurobiology and Institute of Neurosciences, Air Force Medical University, 710032, Xi'an, China
| | - Jin Ma
- Department of Aerospace Medicine, Air Force Medical University, 710032, Xi'an, China.
| | - Yunying Wang
- Department of Aerospace Medicine, Air Force Medical University, 710032, Xi'an, China.
| |
Collapse
|
45
|
Zhu Y, Liu T, Deng X, Sheng D, Chen J, Kuang Y, Dai Z, Chen H. Ultrasound-mediated intra-/extracellular dual intervening effect combined with all-trans retinoic acid for cancer stemness inhibition. NANO TODAY 2024; 55:102207. [DOI: 10.1016/j.nantod.2024.102207] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
46
|
Su Y, Yu Z, Yang Y, Wong K, Li X. Distribution-Agnostic Deep Learning Enables Accurate Single-Cell Data Recovery and Transcriptional Regulation Interpretation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307280. [PMID: 38380499 PMCID: PMC11040354 DOI: 10.1002/advs.202307280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/16/2024] [Indexed: 02/22/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) is a robust method for studying gene expression at the single-cell level, but accurately quantifying genetic material is often hindered by limited mRNA capture, resulting in many missing expression values. Existing imputation methods rely on strict data assumptions, limiting their broader application, and lack reliable supervision, leading to biased signal recovery. To address these challenges, authors developed Bis, a distribution-agnostic deep learning model for accurately recovering missing sing-cell gene expression from multiple platforms. Bis is an optimal transport-based autoencoder model that can capture the intricate distribution of scRNA-seq data while addressing the characteristic sparsity by regularizing the cellular embedding space. Additionally, they propose a module using bulk RNA-seq data to guide reconstruction and ensure expression consistency. Experimental results show Bis outperforms other models across simulated and real datasets, showcasing superiority in various downstream analyses including batch effect removal, clustering, differential expression analysis, and trajectory inference. Moreover, Bis successfully restores gene expression levels in rare cell subsets in a tumor-matched peripheral blood dataset, revealing developmental characteristics of cytokine-induced natural killer cells within a head and neck squamous cell carcinoma microenvironment.
Collapse
Affiliation(s)
- Yanchi Su
- School of Artificial IntelligenceJilin UniversityChangchun130012China
| | - Zhuohan Yu
- School of Artificial IntelligenceJilin UniversityChangchun130012China
| | - Yuning Yang
- Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONM5S 3E1Canada
| | - Ka‐Chun Wong
- Department of Computer ScienceCity University of Hong KongHong Kong SAR999077China
| | - Xiangtao Li
- School of Artificial IntelligenceJilin UniversityChangchun130012China
| |
Collapse
|
47
|
Zeng J, Geng X, Zhao Z, Zhou W. Tipping the balance: The dynamics of stem cell maintenance and stress responses in plant meristems. CURRENT OPINION IN PLANT BIOLOGY 2024; 78:102510. [PMID: 38266375 DOI: 10.1016/j.pbi.2024.102510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/24/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024]
Abstract
Plant meristems contain pools of dividing stem cells that produce new organs for plant growth and development. Environmental factors, including biotic and abiotic stresses and nutrient availability, affect meristem activity and thus the architecture of roots and shoots; understanding how meristems react to changing environmental conditions will shed light on how plants optimize nutrient acquisition and acclimate to different environmental conditions. This review highlights recent exciting advances in this field, mainly in Arabidopsis. We discuss the signaling pathways, genetic regulators, and molecular mechanisms involved in the response of plant meristems to environmental and nutrient cues, and compare the similarities and differences of stress responses between the shoot and root apical meristems.
Collapse
Affiliation(s)
- Jian Zeng
- Department of Stem Cell Biology, Centre for Organismal Studies, Heidelberg University, Heidelberg 69120, Germany
| | - Xin Geng
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Zhong Zhao
- CAS Center for Excellence in Molecular Plant Sciences, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China.
| | - Wenkun Zhou
- State Key Laboratory of Plant Environmental Resilience, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
48
|
Ruszkowska-Ciastek B, Kwiatkowska K, Marques-da-Silva D, Lagoa R. Cancer Stem Cells from Definition to Detection and Targeted Drugs. Int J Mol Sci 2024; 25:3903. [PMID: 38612718 PMCID: PMC11011379 DOI: 10.3390/ijms25073903] [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: 02/26/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Cancers remain the second leading cause of mortality in the world. Preclinical and clinical studies point an important role of cancer/leukaemia stem cells (CSCs/LSCs) in the colonisation at secondary organ sites upon metastatic spreading, although the precise mechanisms for specific actions are still not fully understood. Reviewing the present knowledge on the crucial role of CSCs/LSCs, their plasticity, and population heterogeneity in treatment failures in cancer patients is timely. Standard chemotherapy, which acts mainly on rapidly dividing cells, is unable to adequately affect CSCs with a low proliferation rate. One of the proposed mechanisms of CSC resistance to anticancer agents is the fact that these cells can easily shift between different phases of the cell cycle in response to typical cell stimuli induced by anticancer drugs. In this work, we reviewed the recent studies on CSC/LSC alterations associated with disease recurrence, and we systematised the functional assays, markers, and novel methods for CSCs screening. This review emphasises CSCs' involvement in cancer progression and metastasis, as well as CSC/LSC targeting by synthetic and natural compounds aiming at their elimination or modulation of stemness properties.
Collapse
Affiliation(s)
- Barbara Ruszkowska-Ciastek
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 85-094 Bydgoszcz, Poland
| | - Katarzyna Kwiatkowska
- Department of Laboratory Diagnostics, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland;
| | - Dorinda Marques-da-Silva
- Laboratory of Separation and Reaction Engineering-Laboratory of Catalysis and Materials (LSRE-LCM), Polytechnic Institute of Leiria, 2411-901 Leiria, Portugal; (D.M.-d.-S.); (R.L.)
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Ricardo Lagoa
- Laboratory of Separation and Reaction Engineering-Laboratory of Catalysis and Materials (LSRE-LCM), Polytechnic Institute of Leiria, 2411-901 Leiria, Portugal; (D.M.-d.-S.); (R.L.)
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| |
Collapse
|
49
|
Scodellaro C, Pina RR, Ferreira FC, Sanjuan-Alberte P, Fernandes TG. Unlocking the Potential of Stem Cell Microenvironments In Vitro. Bioengineering (Basel) 2024; 11:289. [PMID: 38534563 DOI: 10.3390/bioengineering11030289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/07/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
The field of regenerative medicine has recently witnessed groundbreaking advancements that hold immense promise for treating a wide range of diseases and injuries. At the forefront of this revolutionary progress are stem cells. Stem cells typically reside in specialized environments in vivo, known as microenvironments or niches, which play critical roles in regulating stem cell behavior and determining their fate. Therefore, understanding the complex microenvironments that surround stem cells is crucial for advancing treatment options in regenerative medicine and tissue engineering applications. Several research articles have made significant contributions to this field by exploring the interactions between stem cells and their surrounding niches, investigating the influence of biomechanical and biochemical cues, and developing innovative strategies for tissue regeneration. This review highlights the key findings and contributions of these studies, shedding light on the diverse applications that may arise from the understanding of stem cell microenvironments, thus harnessing the power of these microenvironments to transform the landscape of medicine and offer new avenues for regenerative therapies.
Collapse
Affiliation(s)
- Chiara Scodellaro
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Raquel R Pina
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Paola Sanjuan-Alberte
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
50
|
Herger N, Heggli I, Mengis T, Devan J, Arpesella L, Brunner F, Distler O, Dudli S. Impacts of priming on distinct immunosuppressive mechanisms of mesenchymal stromal cells under translationally relevant conditions. Stem Cell Res Ther 2024; 15:65. [PMID: 38443999 PMCID: PMC10916130 DOI: 10.1186/s13287-024-03677-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND The multimodal properties of mesenchymal stromal cells (MSCs), particularly their ability to modulate immune responses is of high interest in translational research. Pro-inflammatory, hypoxic, and 3D culture priming are promising and often used strategies to improve the immunosuppressive potency of MSCs, but the underlying mechanisms are not well understood. Therefore, the aims of this study were (i) to compare the effects of pro-inflammatory, hypoxic, and 3D culture priming on the in vitro immunosuppressive potential of MSCs, (ii) to assess if immunosuppressive priming effects are temporally preserved under standard and translationally relevant culture conditions, and (iii) to investigate if the three priming strategies engage the same immunosuppressive mechanisms. METHODS Functional in vitro T cell suppressive potency measurements were conducted to assess the impact of pro-inflammatory, hypoxic, and 3D culture priming on the immunosuppressive potential of human bone marrow-derived MSCs. Primed MSCs were either cultured under standard cell culture conditions or translationally relevant culture conditions, and their transcriptomic adaptations were monitored over time. Next-generation sequencing was performed to assess if different priming strategies activate distinct immunosuppressive mechanisms. RESULTS (i) Pro-inflammatory, hypoxic, and 3D culture priming induced profound transcriptomic changes in MSCs resulting in a significantly enhanced T cell suppressive potential of pro-inflammatory and 3D culture primed MSCs. (ii) Priming effects rapidly faded under standard cell culture conditions but were partially preserved under translationally relevant conditions. Interestingly, continuous 3D culture priming of MSCs maintained the immunosuppressive potency of MSCs. (iii) Next-generation sequencing revealed that priming strategy-specific differentially expressed genes are involved in the T cell suppressive capacity of MSCs, indicating that different priming strategies engage distinct immunosuppressive mechanisms. CONCLUSION Priming can be a useful approach to improve the immunosuppressive potency of MSCs. However, future studies involving primed MSCs should carefully consider the significant impact of translationally relevant conditions on the preservation of priming effects. Continuous 3D culture could act as a functionalized formulation, supporting the administration of MSC spheroids for a sustainably improved immunosuppressive potency.
Collapse
Affiliation(s)
- Nick Herger
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland.
| | - Irina Heggli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Tamara Mengis
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Jan Devan
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Leonardo Arpesella
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Florian Brunner
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stefan Dudli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Balgrist Campus, Zurich, Switzerland
| |
Collapse
|