1
|
Feng Y, Qiu H, Chen D. Regulation of Stem Cell Function by NAD . Physiology (Bethesda) 2025; 40:0. [PMID: 39907078 PMCID: PMC12167693 DOI: 10.1152/physiol.00052.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+), a coenzyme in cellular metabolism, has never ceased to capture the fascination of scientists since its discovery in 1906. The expansion of NAD+'s function from cellular metabolism to DNA repair, gene regulation, cell signaling, and aging reflects the central role of cellular metabolism in orchestrating the diverse cellular pathways. In the past decade, NAD+ has emerged as a key regulator of stem cells, opening the door to potential approaches for regenerative medicine. Here we reflect on how the field of NAD+ regulation of stem cells has evolved since a decade ago, when sirtuins, NAD+-dependent enzymes, were shown to be critical regulators of stem cells. We review the recent development on how NAD+ is regulated in stem cells to influence fate decision. We discuss the difference in NAD+ regulation of normal and cancer stem cells. Finally, we consider the consequences of NAD+ regulation of stem cells for health and diseases.
Collapse
Affiliation(s)
- Yufan Feng
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Huixian Qiu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
2
|
Zhou H, Liu Y, Tian GG, Wu J. Nicotinamide mononucleotide promotes female germline stem cell proliferation by activating the H4K16ac-Hmgb1-Fyn-PLD signaling pathway through epigenetic remodeling. Cell Biosci 2025; 15:48. [PMID: 40247362 PMCID: PMC12004683 DOI: 10.1186/s13578-025-01387-w] [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: 02/04/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Nicotinamide mononucleotide (NMN), an endogenous nucleotide essential for various physiological processes, has an unclear role and regulatory mechanisms in female germline stem cell (FGSC) development. RESULTS We demonstrate that NMN significantly enhances FGSC viability and proliferation. Quantitative acetylation proteomics revealed that NMN markedly increases the acetylation of histone H4 at lysine 16 (H4K16ac). Subsequent chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq) identified high mobility group box 1 (Hmgb1) as a downstream target of H4K16ac, a finding further validated by ChIP-qPCR. Knockdown of Hmgb1 reduced FGSC proliferation by disrupting cell cycle progression, inducing apoptosis, and decreasing chromatin accessibility. High-throughput chromosome conformation capture (Hi-C) analysis showed that Hmgb1 knockdown induced A/B compartment switching, increased the number of topologically associating domains (TADs), and decreased chromatin loop formation in FGSCs. Notably, the chromatin loop at the promoter region of Fyn proto-oncogene (Fyn) disappeared following Hmgb1 knockdown. ChIP-qPCR and dual-luciferase reporter assays further confirmed the interaction between Hmgb1 and the Fyn promoter. Importantly, Fyn overexpression reversed the inhibitory effects of Hmgb1 knockdown on FGSC proliferation. Proteomic analysis suggested this rescue was mediated through the phospholipase D (PLD) signaling pathway, as Fyn overexpression selectively enhanced the phosphorylation of PLD1 at threonine 147 without affecting serine 561. Furthermore, treatment with 5-fluoro-2-indolyldechlorohaloamide, a PLD inhibitor, nullified the pro-proliferative effects of Fyn overexpression. CONCLUSIONS Our findings reveal that NMN promotes FGSC proliferation by activating the H4K16ac-Hmgb1-Fyn-PLD signaling pathway through epigenetic remodeling. These results deepen our understanding of FGSC proliferation and highlight potential therapeutic avenues for advancing FGSC applications in reproductive medicine.
Collapse
Affiliation(s)
- Hong Zhou
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yujie Liu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Geng G Tian
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Ji Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
3
|
Gao J, Meng X, Yang X, Xie C, Tian C, Gong J, Zhang J, Dai S, Gao T. The protection of nicotinamide riboside against diabetes mellitus-induced bone loss via OXPHOS. Bone 2025; 193:117411. [PMID: 39884488 DOI: 10.1016/j.bone.2025.117411] [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: 09/28/2024] [Revised: 01/21/2025] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
Diabetes mellitus is a global disease that results in various complications, including diabetic osteoporosis. Prior studies have indicated a correlation between low levels of nicotinamide adenine dinucleotide (NAD+) and diabetes-related complications. Nicotinamide riboside (NR), a widely utilized precursor vitamin of NAD+, has been demonstrated to enhance age-related osteoporosis through the Sirt1/FOXO/β-catenin pathway in osteoblast progenitors. However, the impact of NR on bone health in diabetes mellitus remains unclear. In this study, we assessed the potential effects of NR on bone in diabetic mice. NR was administered to high-fat diet (HFD)/streptozotocin (STZ)-induced type 2 diabetic mice (T2DM), and various parameters, including metabolic indicators, bone quality, bone metabolic markers, and RNA sequences, were measured. Our findings confirmed that HFD/STZ-induced T2DM impaired bone microstructures, resulting in bone loss. NR effectively ameliorated insulin resistance, improved bone microarchitecture, and bone quality, reduced bone resorption, enhanced the Forkhead box O (FOXO) signaling pathway, mitigated the nuclear factor kappa B (NF-kB) signaling pathway, and ameliorated the disorder of the oxidative phosphorylation process (OXPHOS) in diabetic mice. In conclusion, NR demonstrated the capacity to alleviate T2DM-induced bone loss through the modulation of OXPHOS in type 2 diabetic mice. Our results underscore the potential of NR as a therapeutic target for addressing T2DM-related bone metabolism and associated diseases. Further cell-based studies under diabetic conditions, such as in vitro cultures of key cell types (e.g., osteoblasts and osteoclasts), are necessary to validate these findings.
Collapse
Affiliation(s)
- Jie Gao
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao 266000, China; School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Xiangyuan Meng
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Xingxiang Yang
- School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Chenqi Xie
- School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Chunyan Tian
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jianbao Gong
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao 266000, China
| | - Junwei Zhang
- Shandong Wendeng Osteopathic Hospital, Weihai 264400, China
| | - Shiyou Dai
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao 266000, China.
| | - Tianlin Gao
- School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
4
|
Liu L, Zhang J, Cui R, Wang N, Zhang Y, Liu L, Zhang X, Liu Q. SIRT1 and exercise-induced bone metabolism: a regulatory nexus. Front Cell Dev Biol 2025; 13:1522821. [PMID: 40206398 PMCID: PMC11979185 DOI: 10.3389/fcell.2025.1522821] [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: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025] Open
Abstract
Regular exercise positively influences bone health, enhances bone density and strength, and reduces the risk of osteoporosis. Silent information regulator of transcription 1 (SIRT1) is a deacetylase that plays a pivotal role in the regulation of various biological processes. In this review, we explore the role of SIRT1 in modulating bone metabolism in response to exercise. SIRT1 regulates crucial cellular processes, including inflammation, aging, autophagy, and oxidative stress, in bone cells such as bone marrow mesenchymal stem cells, osteoblasts, and osteoclasts, in response to exercise-induced stimuli. Notably, exercise influences bone metabolism by modulating muscle metabolism and neurotransmitters, with SIRT1 acting as a key mediator. A comprehensive understanding of SIRT1's regulatory mechanisms will facilitate a deeper exploration of the principles underlying exercise-induced improvements in bone metabolism, ultimately providing novel insights into the treatment of bone metabolic disorders.
Collapse
Affiliation(s)
- Lijie Liu
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Jiale Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Runhong Cui
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Na Wang
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Yun Zhang
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Lifei Liu
- Department of Rehabilitation, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Xinan Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Qingfeng Liu
- Department of General Surgery, Jinqiu Hospital of Liaoning Province, Shenyang, China
| |
Collapse
|
5
|
Chen X, Huang X, Zhang X, Chen Z. Metabolism-epigenetic interaction-based bone and dental regeneration: From impacts and mechanisms to treatment potential. Bone 2025; 192:117382. [PMID: 39730093 DOI: 10.1016/j.bone.2024.117382] [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: 09/05/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Metabolic pathways exhibit fluctuating activities during bone and dental loss and defects, suggesting a regulated metabolic plasticity. Skeletal remodeling is an energy-demanding process related to altered metabolic activities. These metabolic changes are frequently associated with epigenetic modifications, including variations in the expression or activity of enzymes modified through epigenetic mechanisms, which directly or indirectly impact cellular metabolism. Metabolic reprogramming driven by bone and dental conditions alters the epigenetic landscape by modulating the activities of DNA and histone modification enzymes at the metabolite level. Epigenetic mechanisms modulate the expression of metabolic genes, consequently influencing the metabolome. The interplay between epigenetics and metabolomics is crucial in maintaining bone and dental homeostasis by preserving cell proliferation and pluripotency. This review, therefore, aims to examine the effects of metabolic reprogramming in bone and dental-related cells on the regulation of epigenetic modifications, particularly acetylation, methylation, and lactylation. We also discuss the effects of chromatin-modifying enzymes on metabolism and the potential therapeutic benefits of dietary compounds as epigenetic modulators. In this review, we highlight the inconsistencies in current research findings and suggest potential approaches to translate fundamental insights into clinical treatments for bone and tooth diseases.
Collapse
Affiliation(s)
- Xinyi Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Centre for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Centre of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Xiaoyuan Huang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Centre for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Centre of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Xiatong Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Centre for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Centre of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhuo Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Centre for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Centre of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
6
|
Huang YS, Gao JW, Ao RF, Liu XY, Wu DZ, Huang JL, Tu C, Zhuang JS, Zhu SY, Zhong ZM. Accumulation of advanced oxidation protein products aggravates bone-fat imbalance during skeletal aging. J Orthop Translat 2025; 51:24-36. [PMID: 39902100 PMCID: PMC11788738 DOI: 10.1016/j.jot.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/30/2024] [Accepted: 12/24/2024] [Indexed: 02/05/2025] Open
Abstract
Background Skeletal aging is characterized by a decrease in bone mass and an increase in marrowfat content. Advanced oxidation protein products (AOPPs) accumulate easily with aging and disrupt redox homeostasis. We examined whether AOPPs accumulation contributes to the bone-fat imbalance during skeletal aging. Methods Both young and aged mice were employed to assess the changes of AOPPs levels and its contribution to bone-fat imbalance during skeletal aging. Primary bone marrow mesenchymal stromal cells (MSCs) were used to examine the potential role of AOPPs in age-related switch between osteogenic and adipogenic differentiation. Aged mice were also gavaged by non-selective antioxidant N-acetyl-L-cysteine (NAC), followed by close monitoring of the changes in AOPPs levels and bone-fat metabolism. Furthermore, young mice were chronically exposed to AOPPs and then evaluated for the changes of bone mass and marrow adiposity. Results The levels of AOPPs in serum and bone marrow were markedly higher in aged mice than that in young mice. Age-related accumulation of AOPPs was accompanied by reduced bone formation, increased marrow adiposity and deterioration of bone microstructure. Reduced AOPPs accumulation by antioxidant NAC leaded to improvement of the bone-fat imbalance in aged mice. Similarly, the bone-fat imbalance was induced by chronic AOPPs loading in young mice. Compared with MSCs from young mice, MSCs from aged mice tended to differentiate into adipocytes rather than osteoblasts and displayed cellular senescence. Exposure of primary MSCs to AOPPs resulted in the switch from osteogenic to adipogenic lineage and cellular senescence. AOPPs challenge also increased intracellular ROS generation by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondria. The antioxidant NAC, after scavenging ROS, ameliorated the AOPPs-induced lineage switch and senescence in MSCs by inhibiting the PI3K/AKT/mTOR pathway. Conclusion Our findings revealed the involvement of AOPPs in age-related switch between osteogenic and adipogenic differentiation, and illuminated a novel potential mechanism underlying bone-fat imbalance during skeletal aging. The translational potential of this article Reducing AOPPs accumulation and its cascading effects on MSCs might be an attractive strategy for delaying skeletal aging.
Collapse
Affiliation(s)
- Yu-Sheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia-Wen Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rui-Feng Ao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin-Yu Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Di-Zheng Wu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun-Long Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Shen Zhuang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Si-Yuan Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Tian RC, Zhang RY, Ma CF. Rejuvenation of Bone Marrow Mesenchymal Stem Cells: Mechanisms and Their Application in Senile Osteoporosis Treatment. Biomolecules 2025; 15:276. [PMID: 40001580 PMCID: PMC11853522 DOI: 10.3390/biom15020276] [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: 12/16/2024] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Bone marrow mesenchymal stromal cells (BM-MSCs) are multipotent cells present in bone marrow; they play a crucial role in the process of bone formation. Cellular senescence is defined as a stable state of cell cycle arrest that impairs the functioning of cells. Research has shown that aging triggers a state of senescence in BM-MSCs, leading to a reduced capacity for osteogenic differentiation and the accumulation of senescent cells, which can accelerate the onset of various diseases. Therefore, it is essential to explore mechanisms and strategies for the rejuvenation of senescent BM-MSCs. Senile osteoporosis (SOP) is a metabolic bone disease characterized by reduced bone formation. The senescence of BM-MSCs is considered one of the most important factors in the occurrence and development of SOP. Therefore, the rejuvenation of BM-MSCs for the treatment of SOP represents a promising strategy. This work provides a summary of the functional alterations observed in senescent BM-MSCs and a systematic review of the mechanisms that facilitate the rejuvenation of senescent BM-MSCs. Additionally, we analyze the progress in and the limitations associated with the application of rejuvenated senescent BM-MSCs to treat SOP, with the aim of providing new insights for the prevention and treatment of SOP.
Collapse
Affiliation(s)
- Rui-Chuan Tian
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing 100142, China;
- Graduate School, China Medical University, Shenyang 110002, China
| | - Ru-Ya Zhang
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, China;
| | - Chu-Fan Ma
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing 100142, China;
- Graduate School, China Medical University, Shenyang 110002, China
| |
Collapse
|
8
|
Yu C, Sun R, Yang W, Gu T, Ying X, Ye L, Zheng Y, Fan S, Zeng X, Yao S. Exercise ameliorates osteopenia in mice via intestinal microbial-mediated bile acid metabolism pathway. Theranostics 2025; 15:1741-1759. [PMID: 39897551 PMCID: PMC11780523 DOI: 10.7150/thno.104186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/09/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Physical exercise is essential for skeletal integrity and bone health. The gut microbiome, as a pivotal modulator of overall physiologic states, is closely associated with skeletal homeostasis and bone metabolism. However, the potential role of intestinal microbiota in the exercise-mediated bone gain remains unclear. Methods: We conducted microbiota depletion and fecal microbiota transplantation (FMT) in ovariectomy (OVX) mice and aged mice to investigate whether the transfer of gut ecological traits could confer the exercise-induced bone protective effects. The study analyzed the gut microbiota and metabolic profiles via 16S rRNA gene sequencing and LC-MS untargeted metabolomics to identify key microbial communities and metabolites responsible for bone protection. Transcriptome sequencing and RNA interference were employed to explore the molecular mechanisms. Results: We found that gut microbiota depletion hindered the osteogenic benefits of exercise, and FMT from exercised osteoporotic mice effectively mitigated osteopenia. Comprehensive profiling of the microbiome and metabolome revealed that the exercise-matched FMT reshaped intestinal microecology and metabolic landscape. Notably, alterations in bile acid metabolism, specifically the enrichment of taurine and ursodeoxycholic acid, mediated the protective effects on bone mass. Mechanistically, FMT from exercised mice activated the apelin signaling pathway and restored the bone-fat balance in recipient MSCs. Conclusion: Our study underscored the important role of the microbiota-metabolic axis in the exercise-mediated bone gain, heralding a potential breakthrough in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Rongtai Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Wentao Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Tianyuan Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Xiaozhang Ying
- Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang 310016, China
| | - Lin Ye
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Yang Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
- Research Institute of Orthopedics, The Affiliated Jiangnan Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Xiangjun Zeng
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Shasha Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| |
Collapse
|
9
|
Reeves J, Tournier P, Becquart P, Carton R, Tang Y, Vigilante A, Fang D, Habib SJ. Rejuvenating aged osteoprogenitors for bone repair. eLife 2024; 13:RP104068. [PMID: 39692737 DOI: 10.7554/elife.104068] [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] [Indexed: 12/19/2024] Open
Abstract
Aging is marked by a decline in tissue regeneration, posing significant challenges to an increasingly older population. Here, we investigate age-related impairments in calvarial bone healing and introduce a novel two-part rejuvenation strategy to restore youthful repair. We demonstrate that aging negatively impacts the calvarial bone structure and its osteogenic tissues, diminishing osteoprogenitor number and function and severely impairing bone formation. Notably, increasing osteogenic cell numbers locally fails to rescue repair in aged mice, identifying the presence of intrinsic cellular deficits. Our strategy combines Wnt-mediated osteoprogenitor expansion with intermittent fasting, which leads to a striking restoration of youthful levels of bone healing. We find that intermittent fasting improves osteoprogenitor function, benefits that can be recapitulated by modulating NAD+-dependent pathways or the gut microbiota, underscoring the multifaceted nature of this intervention. Mechanistically, we identify mitochondrial dysfunction as a key component in age-related decline in osteoprogenitor function and show that both cyclical nutrient deprivation and Nicotinamide mononucleotide rejuvenate mitochondrial health, enhancing osteogenesis. These findings offer a promising therapeutic avenue for restoring youthful bone repair in aged individuals, with potential implications for rejuvenating other tissues.
Collapse
Affiliation(s)
- Joshua Reeves
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Pierre Tournier
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Pierre Becquart
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Robert Carton
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Yin Tang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Alessandra Vigilante
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Dong Fang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Shukry J Habib
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
10
|
Yu F, Zhang G, Weng J, Jia G, Fang C, Xu H, Xiong A, Qin H, Qi T, Yang Q, Yuan G, Zeng H, Zhu Y. Repair of Osteoporotic Bone Defects in Rats via the Sirtuin 1-Wnt/β-catenin Signaling Pathway by Novel Icariin/Porous Magnesium Alloy Scaffolds. Biomater Res 2024; 28:0090. [PMID: 39655164 PMCID: PMC11625907 DOI: 10.34133/bmr.0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/08/2024] [Accepted: 09/24/2024] [Indexed: 12/12/2024] Open
Abstract
The slow rate of bone regeneration and repair in osteoporotic defects is one of the difficulties of clinical work. To prepare a novel icariin (ICA)/porous magnesium alloy scaffold and to investigate its effectiveness and possible mechanism in repairing osteoporotic bone defects, bilateral ovariectomy was performed on Sprague-Dawley rats. Then, a cylindrical bone defect was created in the model, and a novel ICA/porous magnesium alloy scaffold was prepared and implanted into the defect. Eight or 12 weeks after repairing, specimens and micro-computed tomography (CT) data were collected. Microscopic observation was fulfilled through hematoxylin and eosin, Goldner, Masson, periodic acid-Schiff, and Sirius red staining. The expression of proteins was detected by immunohistochemical staining. The novel ICA/porous magnesium alloy scaffold was noncytotoxic and biologically safe. After it was implanted into the defect for 8 or 12 weeks, the surface color and smoothness, depth, and area of the defect were better than those in the control group. Besides, there was sufficient osteoid tissue, more mineralized bones, more collagen fibers, and more polysaccharide components in the defect repaired with the ICA/porous magnesium alloy scaffold. These conditions are closer to those of real bones. Moreover, the repair effect improved with the repair time. Compared with those in the control group, the expression levels of Sirtuin 1(SIRT1), Wnt5a, β-catenin, glycogen synthase kinase 3β, alkaline phosphatase, runt-related transcription factor 2, bone morphogenetic protein-2, and osteocalcin proteins were elevated in bone tissue after the scaffold was implanted into the defect for 8 weeks (all P < 0.05). The novel ICA/porous magnesium alloy scaffold promotes the repair of osteoporotic bone defects in rats, a process that may be achieved through activation of the SIRT1-Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Fei Yu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials,
Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China
| | - Geng Zhang
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials,
Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China
| | - Jian Weng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials,
Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China
| | - Gaozhi Jia
- School of Intelligent Manufacturing and Equipment,
Shenzhen Institute of Information Technology, Shenzhen 518172, China
| | - Chongzhou Fang
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Huihui Xu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials,
Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China
| | - Ao Xiong
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials,
Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China
| | - Haotian Qin
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials,
Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China
| | - Tiantian Qi
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials,
Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China
| | - Qi Yang
- Department of Medical Ultrasound,
Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Guangyin Yuan
- Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hui Zeng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- Department of Orthopedics,
Shenzhen Second People’s Hospital, Shenzhen 518035, China
| | - Yuanchao Zhu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen, Guangdong, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials,
Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China
| |
Collapse
|
11
|
Zhou K, Du L, Ding R, Xu L, Shi S, Wang S, Wang Z, Zhang G, He G, Zhao Z, Tang BZ. Photocatalytic therapy via photoinduced redox imbalance in biological system. Nat Commun 2024; 15:10551. [PMID: 39632877 PMCID: PMC11618361 DOI: 10.1038/s41467-024-55060-w] [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: 03/19/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024] Open
Abstract
Redox balance is essential for sustaining normal physiological metabolic activities of life. In this study, we present a photocatalytic system to perturb the balance of NADH/NAD+ in oxygen-free conditions, achieving photocatalytic therapy to cure anaerobic bacterial infected periodontitis. Under light irradiation, the catalyst TBSMSPy+ can bind bacterial DNA and initiate the generation of radical species through a multi-step electron transfer process. It catalyzes the conversion from NADH to NAD+ (the turnover frequency up to 60.7 min-1), inhibits ATP synthesis, disrupts the energy supply required for DNA replication, and successfully accomplishes photocatalytic sterilization in an oxygen-free environment. The catalyst participates in the redox reaction, interfering with the balance of NADH/NAD+ contents under irradiation, so we termed this action as photoinduced redox imbalance. Additionally, animal experiments in male rats also validate that the TBSMSPy+ could effectively catalyze the NADH oxidation, suppress metabolism and stimulate osteogenesis. Our research substantiates the concept of photoinduced redox imbalance and the application of photocatalytic therapy, further advocating the development of such catalyst based on photoinduced redox imbalance strategy for oxygen-free phototherapy.
Collapse
Grants
- 52003228 National Natural Science Foundation of China (National Science Foundation of China)
- 52273197 National Natural Science Foundation of China (National Science Foundation of China)
- National Key Research and Development Program of China (2023YFB3810001), Shenzhen Key Laboratory of Functional Aggregate Materials (ZDSYS 20211021111400001), Science, Technology and Innovation Commission of Shenzhen Municipality (JCYJ 2021324134613038, KQTD 20210811090142053, JCYJ20220818103007014, GJHZ 20210705141810031), the Innovation and Technology Commission (ITC-CNERC14SC01), the Open Fund of Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates (2021-kllma-08), Guangzhou 510640, China (South China University of Technology). Guangzhou Science and Technology Planning Project (202201010439). Guangdong Basic and Applied Basic Research Foundation (2023A1515110346, 2021A1515110826). Guangzhou Science and Technology Planning Project (202201010439).
Collapse
Affiliation(s)
- Kun Zhou
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Lili Du
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Rui Ding
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Letian Xu
- Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou, China
| | - Shuai Shi
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Siyuan Wang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China
| | - Zaiyu Wang
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Guoqing Zhang
- University of Science and Technology of China, Hefei, Anhui, China
| | - Gang He
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| | - Zheng Zhao
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China.
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China.
| | - Ben Zhong Tang
- Clinical Translational Research Center of Aggregation-Induced Emission, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China.
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong, China.
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China.
- AIE Institute, Guangzhou Development District, Huangpu, Guangdong, China.
| |
Collapse
|
12
|
Chikhaoui A, Zayoud K, Kraoua I, Bouchoucha S, Tebourbi A, Turki I, Yacoub-Youssef H. Supplementation with nicotinamide limits accelerated aging in affected individuals with cockayne syndrome and restores antioxidant defenses. Aging (Albany NY) 2024; 16:13271-13287. [PMID: 39611850 PMCID: PMC11719109 DOI: 10.18632/aging.206160] [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: 03/11/2024] [Accepted: 10/21/2024] [Indexed: 11/30/2024]
Abstract
Cockayne syndrome (CS) is a segmental progeroid syndrome characterized by defects in the DNA excision repair pathway, predisposing to neurodegenerative manifestations. It is a rare genetic disorder and an interesting model for studying premature aging. Oxidative stress and autophagy play an important role in the aging process. The study of these two processes in a model of accelerated aging and the means to counteract them would lead to the identification of relevant biomarkers with therapeutic value for healthy aging. Here we investigated the gene expression profiles of several oxidative stress-related transcripts derived from CS-affected individuals and healthy elderly donors. We also explored the effect of nicotinamide supplementation on several genes related to inflammation and autophagy. Gene expression analysis revealed alterations in two main pathways. This involves the activation of arachidonic acid metabolism and the repression of the NRF2 pathway in affected individuals with CS. The supplementation with nicotinamide adjusted these abnormalities by enhancing autophagy and decreasing inflammation. Furthermore, CSA/CSB-dependent depletion of the mitochondrial DNA polymerase-γ catalytic subunit (POLG1) was restored following nicotinamide supplementation in CS-affected individuals' fibroblasts. This study reveals the link between oxidative stress and accelerated aging in affected individuals with CS and highlights new biomarkers of cellular senescence. However, further analyses are needed to confirm these results, which could not be carried out, mainly due to the unavailability of crucial samples of this rare disease.
Collapse
Affiliation(s)
- Asma Chikhaoui
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| | - Kouloud Zayoud
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| | - Ichraf Kraoua
- Department of Neuropediatrics, National Institute of Neurology Mongi Ben Hamida, Tunis 2092, Tunisia
| | - Sami Bouchoucha
- Orthopedics Department, Béchir Hamza Children’s Hospital, Tunis 2092, Tunisia
| | - Anis Tebourbi
- Orthopedic and Trauma Surgery Department, Mongi Slim Hospital, La Marsa 2046, Tunisia
| | - Ilhem Turki
- Department of Neuropediatrics, National Institute of Neurology Mongi Ben Hamida, Tunis 2092, Tunisia
| | - Houda Yacoub-Youssef
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| |
Collapse
|
13
|
Li C, Zheng C, Pu Y, Zhou H, Li Y, Wang W, Chen X, Zhang C, Chen Y. Vericiguat enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction through microRNA-1180-3p/ETS1 pathway. Cell Signal 2024; 125:111512. [PMID: 39551415 DOI: 10.1016/j.cellsig.2024.111512] [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: 09/18/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
Reversing cardiac fibrosis contributes to the restoration of cardiac function in acute myocardial infarction (MI). Exosomes-derived mesenchymal stem cells (MSCs) have been established as potential biomarkers of cardiovascular diseases. While vericiguat has demonstrated promising outcomes in MI via reverse hypertrophy and fibrosis, previous studies about vericiguat pretreatment with MSCs is limited. We aim at exploring whether exosomes derived from vericiguat pretreatment MSCs could augment cardioprotective function and the underlying mechanisms. In our study, exosomes isolated from MSCs (MSC-Exo) and pretreated with vericiguat (MSCVER-Exo) were administered to cardiac fibroblasts (CFs) in vitro and male infarcted Sprague-Dawley rat hearts in vivo. In vivo, MSCVER-Exo could significantly improve cardiac function and attenuate cardiac fibrosis and decrease the expression of α-smooth muscle actin (α-SMA), Ι and III collagen (Col Ι and Col III) compared to MSC-Exo treatment. In vitro, MSCVER-Exo could also restrain proliferation, migration, and the profibrotic genes expression in CFs. miR-1180-3p was enrich in MSCVER-Exo. Besides, miR-1180-3p could be delivered to CFs via Exo and alleviated TGF-β1-induced fibrosis through inhibiting ETS1 signaling. The elucidation of this mechanism suggested that exosomes derived from vericiguat pretreatment MSCs could improve cardioprotective effects through promoting CFs function. MiR-1180-3p targeting ETS1 played an important role in antifibrosis.
Collapse
Affiliation(s)
- Chunyu Li
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chongming Zheng
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yanan Pu
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215000, China
| | - Haoyang Zhou
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215000, China
| | - Ying Li
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215000, China
| | - Weiwei Wang
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xufeng Chen
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Cheng Zhang
- Long Jiang Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Yan Chen
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215000, China; Department of Emergency Management, School of Health Policy & Management, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
14
|
Shi Y, Peng J, Liu M, Qi X, Li S, Li Q, Jiang Q, Zheng L, Xu J, Zhao Y, Zhang Y. Nicotinamide mononucleotide enhances fracture healing by promoting skeletal stem cell proliferation. Theranostics 2024; 14:5999-6015. [PMID: 39346542 PMCID: PMC11426247 DOI: 10.7150/thno.98149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/04/2024] [Indexed: 10/01/2024] Open
Abstract
The process of skeletal regeneration initiated by stem cells following injury, especially in fractures, is significantly impaired by aging and adverse factors. Nicotinamide mononucleotide (NMN), a critical endogenous precursor of nicotinamide adenine dinucleotide (NAD), has garnered extensive attention for its multifaceted regulatory functions in living organisms and its wide-ranging therapeutic potential. However, whether NMN contributes to trauma-induced skeletal regeneration remains unclear. Methods: The transverse femoral shaft fracture model was employed to evaluate the potential advantages of NMN administration for overall repair during the initial fracture stages in male mice through micro-CT analysis, histochemistry, and biomechanical testing. The pro-proliferative function of NMN on skeletal stem cells (SSCs) was investigated through flow cytometry, qRT-PCR, NAD content measurement, and cell proliferation assay. Results: In this study, we observed that the administration of NMN during the initial phase of fracture in mice led to a larger callus and corresponding improvement in micro-CT parameters. NMN enhances the cartilaginous component of the callus by elevating the NAD content, consequently accelerating subsequent endochondral ossification and the fracture healing process. Subsequent analyses elucidated that NMN was beneficial in promoting the expansion of diverse stem cells in vivo and in vitro potentially via modulation of the Notch signaling pathway. Moreover, the depletion of macrophages profoundly obstructs the proliferation of SSCs. Conclusion: Our discoveries provide a potential strategy for enhancing fracture healing through stimulation of callus SSC proliferation at an early stage, shedding light on the translational value of NMN as an enhancer for skeletal regeneration and highlighting the pivotal role of macrophage-stem cell interactions in governing the regenerative influence of NMN on stem cells.
Collapse
Affiliation(s)
- Yitian Shi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Jiayin Peng
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, PR China
| | - Mengfan Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Xiling Qi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Siyu Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Qiangqiang Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, PR China
| | - Liming Zheng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Yun Zhao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, PR China
| | - Yifeng Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
- Shanghai Clinical Research and Trial Center, Shanghai 200000, PR China
| |
Collapse
|
15
|
Peng K, Yao YX, Lu X, Wang WJ, Zhang YH, Zhao H, Wang H, Xu DX, Tan ZX. Mitochondrial dysfunction-associated alveolar epithelial senescence is involved in CdCl 2-induced COPD-like lung injury. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135103. [PMID: 38972203 DOI: 10.1016/j.jhazmat.2024.135103] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
An earlier study found that respiratory cadmium chloride (CdCl2) exposure caused COPD-like lung injury. This study aimed to explore whether mitochondrial dysfunction-mediated alveolar epithelial senescence is involved in CdCl2-induced COPD-like lung injury. Adult C57BL/6 mice were exposed to CdCl2 (10 mg/L) aerosol for six months. Beta-galactosidase-positive cells, p21 and p16 were increased in CdCl2-exposed mouse lungs. The in vitro experiments showed that γ-H2AX was elevated in CdCl2-exposed alveolar epithelial cells. The cGAS-STING pathway was activated in CdCl2-exposed alveolar epithelial cells and mouse lungs. Cxcl1, Cxcl9, Il-10, Il-1β and Mmp2, several senescence-associated secretory phenotypes (SASP), were upregulated in CdCl2-exposed alveolar epithelial cells. Mechanistically, CdCl2 exposure caused SIRT3 reduction and mitochondrial dysfunction in mouse lungs and alveolar epithelial cells. The in vitro experiment found that Sirt3 overexpression attenuated CdCl2-induced alveolar epithelial senescence and SASP. The in vivo experiments showed that Sirt3 gene knockout exacerbated CdCl2-induced alveolar epithelial senescence, alveolar structure damage, airway inflammation and pulmonary function decline. NMN, an NAD+ precursor, attenuated CdCl2-induced alveolar epithelial senescence and SASP in mouse lungs. Moreover, NMN supplementation prevented CdCl2-induced COPD-like alveolar structure damage, epithelial-mesenchymal transition and pulmonary function decline. These results suggest that mitochondrial dysfunction-associated alveolar epithelial senescence is involved in CdCl2-induced COPD-like lung injury.
Collapse
Affiliation(s)
- Kun Peng
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ya-Xin Yao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xue Lu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Wen-Jing Wang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi-Hao Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China.
| | - Zhu-Xia Tan
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
16
|
Liao G, Xie Y, Peng H, Li T, Zou X, Yue F, Guo J, Rong L. Advancements in NMN biotherapy and research updates in the field of digestive system diseases. J Transl Med 2024; 22:805. [PMID: 39215316 PMCID: PMC11363601 DOI: 10.1186/s12967-024-05614-9] [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: 05/02/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Nicotinamide mononucleotide (NMN), a crucial intermediate in NAD + synthesis, can rapidly transform into NAD + within the body after ingestion. NMN plays a pivotal role in several important biological processes, including energy metabolism, cellular aging, circadian rhythm regulation, DNA repair, chromatin remodeling, immunity, and inflammation. NMN has emerged as a key focus of research in the fields of biomedicine, health care, and food science. Recent years have witnessed extensive preclinical studies on NMN, offering valuable insights into the pathogenesis of age- and aging-related diseases. Given the sustained global research interest in NMN and the substantial market expectations for the future, here, we comprehensively review the milestones in research on NMN biotherapy over the past 10 years. Additionally, we highlight the current research on NMN in the field of digestive system diseases, identifying existing problems and challenges in the field of NMN research. The overarching aim of this review is to provide references and insights for the further exploration of NMN within the spectrum of digestive system diseases.
Collapse
Affiliation(s)
- Guanyi Liao
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Yuchen Xie
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Hong Peng
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Tianke Li
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Xinsen Zou
- Department of Intensive Unit Care, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Faguo Yue
- Sleep and Psychology Center, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Jinjun Guo
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China.
| | - Li Rong
- Department of Gastroenterology, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China.
| |
Collapse
|
17
|
Chen Y, Xiao H, Liu Z, Teng F, Yang A, Geng B, Sheng X, Xia Y. Sirt1: An Increasingly Interesting Molecule with a Potential Role in Bone Metabolism and Osteoporosis. Biomolecules 2024; 14:970. [PMID: 39199358 PMCID: PMC11352324 DOI: 10.3390/biom14080970] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Osteoporosis (OP) is a common metabolic bone disease characterized by low bone mass, decreased bone mineral density, and degradation of bone tissue microarchitecture. However, our understanding of the mechanisms of bone remodeling and factors affecting bone mass remains incomplete. Sirtuin1 (SIRT1) is a nicotinamide adenine dinucleotide-dependent deacetylase that regulates a variety of cellular metabolisms, including inflammation, tumorigenesis, and bone metabolism. Recent studies have emphasized the important role of SIRT1 in bone homeostasis. This article reviews the role of SIRT1 in bone metabolism and OP and also discusses therapeutic strategies and future research directions for targeting SIRT1.
Collapse
Affiliation(s)
- Yi Chen
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Hefang Xiao
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Zirui Liu
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Fei Teng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Ao Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Xiaoyun Sheng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Y.C.); (H.X.); (Z.L.); (F.T.); (A.Y.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
18
|
Zheng C, Li Y, Wu X, Gao L, Chen X. Advances in the Synthesis and Physiological Metabolic Regulation of Nicotinamide Mononucleotide. Nutrients 2024; 16:2354. [PMID: 39064797 PMCID: PMC11279976 DOI: 10.3390/nu16142354] [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: 06/25/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Nicotinamide mononucleotide (NMN), the direct precursor of nicotinamide adenine dinucleotide (NAD+), is involved in the regulation of many physiological and metabolic reactions in the body. NMN can indirectly affect cellular metabolic pathways, DNA repair, and senescence, while also being essential for maintaining tissues and dynamic metabolic equilibria, promoting healthy aging. Therefore, NMN has found many applications in the food, pharmaceutical, and cosmetics industries. At present, NMN synthesis strategies mainly include chemical synthesis and biosynthesis. Despite its potential benefits, the commercial production of NMN by organic chemistry approaches faces environmental and safety problems. With the rapid development of synthetic biology, it has become possible to construct microbial cell factories to produce NMN in a cost-effective way. In this review, we summarize the chemical and biosynthetic strategies of NMN, offering an overview of the recent research progress on host selection, chassis cell optimization, mining of key enzymes, metabolic engineering, and adaptive fermentation strategies. In addition, we also review the advances in the role of NMN in aging, metabolic diseases, and neural function. This review provides comprehensive technical guidance for the efficient biosynthesis of NMN as well as a theoretical basis for its application in the fields of food, medicine, and cosmetics.
Collapse
Affiliation(s)
- Chuxiong Zheng
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China;
| | - Yumeng Li
- National Technology Innovation Center for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China; (Y.L.); (X.W.)
| | - Xin Wu
- National Technology Innovation Center for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China; (Y.L.); (X.W.)
| | - Le Gao
- National Technology Innovation Center for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China; (Y.L.); (X.W.)
| | - Xiaoyi Chen
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China;
| |
Collapse
|
19
|
Chandrasekara CMN, Gemikonakli G, Mach J, Sang R, Anwer AG, Agha A, Goldys EM, Hilmer SN, Campbell JM. Ageing and Polypharmacy in Mesenchymal Stromal Cells: Metabolic Impact Assessed by Hyperspectral Imaging of Autofluorescence. Int J Mol Sci 2024; 25:5830. [PMID: 38892017 PMCID: PMC11171960 DOI: 10.3390/ijms25115830] [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: 04/22/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
The impact of age on mesenchymal stromal cell (MSC) characteristics has been well researched. However, increased age is concomitant with increased prevalence of polypharmacy. This adjustable factor may have further implications for the functionality of MSCs and the effectiveness of autologous MSC procedures. We applied hyperspectral microscopy of cell autofluorescence-a non-invasive imaging technique used to characterise cytometabolic heterogeneity-to identify changes in the autofluorescence signals of MSCs from (1) young mice, (2) old mice, (3) young mice randomised to receive polypharmacy (9-10 weeks of oral therapeutic doses of simvastatin, metoprolol, oxycodone, oxybutynin and citalopram), and (4) old mice randomised to receive polypharmacy. Principal Component Analysis and Logistic Regression Analysis were used to assess alterations in spectral and associated metabolic characteristics. Modelling demonstrated that cells from young mice receiving polypharmacy had less NAD(P)H and increased porphyrin relative to cells from old control mice, allowing for effective separation of the two groups (AUC of ROC curve > 0.94). Similarly, cells from old polypharmacy mice were accurately separated from those from young controls due to lower levels of NAD(P)H (p < 0.001) and higher porphyrin (p < 0.001), allowing for an extremely accurate logistic regression (AUC of ROC curve = 0.99). This polypharmacy regimen may have a more profound impact on MSCs than ageing, and can simultaneously reduce optical redox ratio (ORR) and increase porphyrin levels. This has implications for the use of autologous MSCs for older patients with chronic disease.
Collapse
Affiliation(s)
- Chandrasekara M. N. Chandrasekara
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (C.M.N.C.); (R.S.); (A.G.A.); (A.A.); (E.M.G.)
| | - Gizem Gemikonakli
- Laboratory of Ageing and Pharmacology, Kolling Institute, Northern Sydney Local Health District and Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; (G.G.); (J.M.); (S.N.H.)
| | - John Mach
- Laboratory of Ageing and Pharmacology, Kolling Institute, Northern Sydney Local Health District and Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; (G.G.); (J.M.); (S.N.H.)
| | - Rui Sang
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (C.M.N.C.); (R.S.); (A.G.A.); (A.A.); (E.M.G.)
| | - Ayad G. Anwer
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (C.M.N.C.); (R.S.); (A.G.A.); (A.A.); (E.M.G.)
| | - Adnan Agha
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (C.M.N.C.); (R.S.); (A.G.A.); (A.A.); (E.M.G.)
| | - Ewa M. Goldys
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (C.M.N.C.); (R.S.); (A.G.A.); (A.A.); (E.M.G.)
| | - Sarah N. Hilmer
- Laboratory of Ageing and Pharmacology, Kolling Institute, Northern Sydney Local Health District and Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; (G.G.); (J.M.); (S.N.H.)
| | - Jared M. Campbell
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (C.M.N.C.); (R.S.); (A.G.A.); (A.A.); (E.M.G.)
| |
Collapse
|
20
|
Yen BL, Wang LT, Wang HH, Hung CP, Hsu PJ, Chang CC, Liao CY, Sytwu HK, Yen ML. Excess glucose alone depress young mesenchymal stromal/stem cell osteogenesis and mitochondria activity within hours/days via NAD +/SIRT1 axis. J Biomed Sci 2024; 31:49. [PMID: 38735943 PMCID: PMC11089752 DOI: 10.1186/s12929-024-01039-0] [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: 10/13/2023] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND The impact of global overconsumption of simple sugars on bone health, which peaks in adolescence/early adulthood and correlates with osteoporosis (OP) and fracture risk decades, is unclear. Mesenchymal stromal/stem cells (MSCs) are the progenitors of osteoblasts/bone-forming cells, and known to decrease their osteogenic differentiation capacity with age. Alarmingly, while there is correlative evidence that adolescents consuming greatest amounts of simple sugars have the lowest bone mass, there is no mechanistic understanding on the causality of this correlation. METHODS Bioinformatics analyses for energetics pathways involved during MSC differentiation using human cell information was performed. In vitro dissection of normal versus high glucose (HG) conditions on osteo-/adipo-lineage commitment and mitochondrial function was assessed using multi-sources of non-senescent human and murine MSCs; for in vivo validation, young mice was fed normal or HG-added water with subsequent analyses of bone marrow CD45- MSCs. RESULTS Bioinformatics analyses revealed mitochondrial and glucose-related metabolic pathways as integral to MSC osteo-/adipo-lineage commitment. Functionally, in vitro HG alone without differentiation induction decreased both MSC mitochondrial activity and osteogenesis while enhancing adipogenesis by 8 h' time due to depletion of nicotinamide adenine dinucleotide (NAD+), a vital mitochondrial co-enzyme and co-factor to Sirtuin (SIRT) 1, a longevity gene also involved in osteogenesis. In vivo, HG intake in young mice depleted MSC NAD+, with oral NAD+ precursor supplementation rapidly reversing both mitochondrial decline and osteo-/adipo-commitment in a SIRT1-dependent fashion within 1 ~ 5 days. CONCLUSIONS We found a surprisingly rapid impact of excessive glucose, a single dietary factor, on MSC SIRT1 function and osteogenesis in youthful settings, and the crucial role of NAD+-a single molecule-on both MSC mitochondrial function and lineage commitment. These findings have strong implications on future global OP and disability risks in light of current worldwide overconsumption of simple sugars.
Collapse
Affiliation(s)
- B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), No.35, Keyan Road, Zhunan, 35053, Taiwan.
| | - Li-Tzu Wang
- Department of Obstetrics & Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, No.1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 250, Wuxing Street, Taipei, 11042, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, No.250, Wuxing Street, Taipei, 11042, Taiwan
| | - Hsiu-Huang Wang
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), No.35, Keyan Road, Zhunan, 35053, Taiwan
| | - Chin-Pao Hung
- Department of Obstetrics & Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, No.1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan
| | - Pei-Ju Hsu
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), No.35, Keyan Road, Zhunan, 35053, Taiwan
| | - Chia-Chi Chang
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), No.35, Keyan Road, Zhunan, 35053, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), No.161, Section 6, Minquan East Road, Taipei, 11490, Taiwan
| | - Chien-Yu Liao
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), No.35, Keyan Road, Zhunan, 35053, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases & Vaccinology, NHRI, No.35, Keyan Road, Zhunan, 35053, Taiwan
- Graduate Institute of Microbiology & Immunology, NDMC, No.161, Section 6, Minquan East Road, Taipei, 11490, Taiwan
| | - Men-Luh Yen
- Department of Obstetrics & Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, No.1, Section 1, Jen-Ai Road, Taipei, 10051, Taiwan.
| |
Collapse
|
21
|
Wen F, Xu A, Wei W, Yang S, Xi Z, Ge Y, Wu S, Ju Z. Nicotinamide Mononucleotide Supplementation Alleviates Doxorubicin-Induced Multi-Organ Fibrosis. Int J Mol Sci 2024; 25:5303. [PMID: 38791345 PMCID: PMC11120852 DOI: 10.3390/ijms25105303] [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: 04/15/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic agent known for its multi-organ toxicity, especially in the heart, which limits its clinical application. The toxic side effects of DOX, including DNA damage, oxidative stress, mitochondrial dysfunction and cell apoptosis, are intricately linked to the involvement of nicotinamide adenine dinucleotide (NAD+). To assess the effectiveness of the NAD+ precursor nicotinamide mononucleotide (NMN) in counteracting the multi-organ toxicity of DOX, a mouse model was established through DOX administration, which led to significant reductions in NAD+ in tissues with evident injury, including the heart, liver and lungs. NMN treatment alleviated both multi-organ fibrosis and mortality in mice. Mechanistically, tissue fibrosis, macrophage infiltration and DOX-related cellular damage, which are potentially implicated in the development of multi-organ fibrosis, could be attenuated by NAD+ restoration. Our findings provide compelling evidence for the benefits of NMN supplementation in mitigating the adverse effects of chemotherapeutic drugs on multiple organs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu Wu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (F.W.); (Y.G.)
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (F.W.); (Y.G.)
| |
Collapse
|
22
|
Li J, Yan S, Yang X, Ren X, Qu H, Song J. Nicotinamide mononucleotide based hyaluronic acid methacryloyl hybrid hydrogel regulating stem cells fate for bone regeneration via SIRT1/RUNX2 signaling. Int J Biol Macromol 2024; 261:129905. [PMID: 38311136 DOI: 10.1016/j.ijbiomac.2024.129905] [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: 06/25/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
Efficient bone reconstruction, especially of the critical size after bone damage, remains a challenge in the clinic. Bone marrow mesenchymal stem cell (BMSC) osteogenic differentiation is considered as a promising strategy for bone repair. Nicotinamide adenine dinucleotide (NAD+) regulating BMSC fate and cellular function enhance osteogenesis, but is hardly delivered and lack of targeting. Herein, a novel and biocompatible scaffold was fabricated to locally deliver a precursor of NAD+, nicotinamide mononucleotide (NMN) to the bone defect site, and its bone repair capability and healing mechanism were clarified. NMN-based hyaluronic acid methacryloyl hybrid hydrogel scaffold (denoted as NMN/HAMA) was prepared via photopolymerization. In vitro RT-qPCR analysis, western blotting, Elisa and alizarin red S staining assays demonstrated that the NMN/HAMA hybrid hydrogel regulated BMSCs cellular function in favour of osteogenic differentiation and mineralization by upregulating the mRNA and proteins expression of the osteogenic genes type I pro-collagen (Col-1), bone morphogenic protein 4 (BMP4), and runt-related transcription factor 2 (RUNX2) via the SIRT1 pathway. Implantation of such hybrid hydrogels significantly enhanced bone regeneration in rodent critical calvarial defect models. Furthermore, restoration of the bone defect with NMN administration was inhibited in Prx1 Cre+; SIRT1flox/flox mice, confirming that the NMN/HAMA hybrid hydrogel scaffold promoted bone regeneration via the SIRT1-RUNX2 pathway. These results imply that NMN-based scaffold may be a promising and economic strategy for the treatment of bone defects.
Collapse
Affiliation(s)
- Jing Li
- Center of Digestive Endoscopy, Guangdong Second Provincial General Hospital, Guangzhou 510310, China; Department of Anatomy, School of Medicine College, Jinan University, Guangzhou 510632, China.
| | - Shuyu Yan
- Center of Digestive Endoscopy, Guangdong Second Provincial General Hospital, Guangzhou 510310, China; Department of Anatomy, School of Medicine College, Jinan University, Guangzhou 510632, China
| | - Xiaoqiao Yang
- Center of Digestive Endoscopy, Guangdong Second Provincial General Hospital, Guangzhou 510310, China
| | - Ximing Ren
- Center of Digestive Endoscopy, Guangdong Second Provincial General Hospital, Guangzhou 510310, China
| | - Hongying Qu
- Center of Digestive Endoscopy, Guangdong Second Provincial General Hospital, Guangzhou 510310, China; Medical Department for Digestive Diseases, the Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510310, China.
| | - Jie Song
- Center of Digestive Endoscopy, Guangdong Second Provincial General Hospital, Guangzhou 510310, China; Medical Department for Digestive Diseases, the Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510310, China.
| |
Collapse
|
23
|
He X, Chen M, Zhang X, Cheng X, Chen Y, Shen H, Yang H, Shi Q, Niu J. Apigenin alleviates osteoporosis by orchestrating SIRT1/HIF1α signaling in mesenchymal stem cells. FUNDAMENTAL RESEARCH 2024. [DOI: 10.1016/j.fmre.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
24
|
Chiang CH, Lin YH, Kao YC, Weng SC, Chen CM, Liou YM. Mechanistic study of the Aldo-keto reductase family 1 member A1 in regulating mesenchymal stem cell fate decision toward adipogenesis and osteogenesis. Life Sci 2024; 336:122336. [PMID: 38092142 DOI: 10.1016/j.lfs.2023.122336] [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: 05/05/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023]
Abstract
AIMS Akr1A1 is a glycolytic enzyme catalyzing the reduction of aldehyde to alcohol. This study aims to delineate the role of Akr1A1 in regulating the adipo-osteogenic lineage differentiation of mesenchymal stem cells (MSCs). MAIN METHODS MSCs derived from human bone marrow and Wharton Jelly together with gain- and loss-of-function analysis as well as supplementation with the S-Nitrosoglutathione reductase (GSNOR) inhibitor N6022 were used to study the function of Akr1A1 in controlling MSC lineage differentiation into osteoblasts and adipocytes. KEY FINDINGS Akr1A1 expression, PKM2 activity, and lactate production were found to be decreased in osteoblast-committed MSCs, but PGC-1α increased to induce mitochondrial oxidative phosphorylation. Increased Akr1A1 inhibited the SIRT1-dependent pathway for decreasing the expressions of PGC-1α and TAZ but increasing PPAR γ in adipocyte-committed MSCs, hence promoting glycolysis in adipogenesis. In contrast, Akr1A1 expression, PKM2 activity and lactate production were all increased in adipocyte-differentiated cells with decreased PGC-1α for switching energy utilization to glycolytic metabolism. Reduced Akr1A1 expression in osteoblast-committed cells relieves its inhibition of SIRT1-mediated activation of PGC-1α and TAZ for facilitating osteogenesis and mitochondrial metabolism. SIGNIFICANCE Several metabolism-involved regulators including Akr1A1, SIRT1, PPARγ, PGC-1α and TAZ were differentially expressed in osteoblast- and adipocyte-committed MSCs. More importantly, Akr1A1 was identified as a new key regulator for controlling the MSC lineage commitment in favor of adipogenesis but detrimental to osteogenesis. Such information should be useful to develop perspective new therapeutic agents to reverse the adipo-osteogenic differentiation of BMSCs, in a way to increase in osteogenesis but decrease in adipogenesis.
Collapse
Affiliation(s)
- Chen Hao Chiang
- Department of Orthopaedics, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Yi-Hui Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Yu-Cuieh Kao
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Shuo-Chun Weng
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; Institute of Clinical Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Center for Geriatrics and Gerontology, Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Ying-Ming Liou
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
25
|
Cai M, Chen Y, Lin Y, Hu Z, Li L, Huang H, Lin J. SIRT1 Asn346 sugar chain promoting collagen deacetylation protective effect on osteoblasts under stress. Biochem Biophys Res Commun 2023; 682:148-155. [PMID: 37806254 DOI: 10.1016/j.bbrc.2023.09.075] [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: 09/04/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023]
Abstract
Silencing type information regulator homolog 1 (SIRT1) is a class of nicotinamide adenine dinucleotide (NAD+) dependent deacetylases, which is the convergence point of important physiological processes in vivo, namely, osteoblast aging, energy metabolism, and bone remodeling. To verify whether the O-acetylglucosamine (O-GlcNAc) modification of SIRT1 in the nucleus of osteoblasts enhances its deacetylase activity under stress and protects osteoblasts through the RANK/RANKL signaling pathway by collagen deacetylation. The R language and online data research identified SIRT1 as being involved in bone metabolism. Enrichment analysis showed that SIRT1 is involved in osteoblast transcription, apoptosis, and deacetylation pathways. Interactive Immuno-blotting and immunofluorescence experiments revealed that SIRT1 and O-glycosylation catalytic enzyme (OGT) were localized in the nucleus. Mass Spectrometry analysis showed that O-glycosylation occurred on the asparagine at the 346th position of SIRT1, and N346th was located in the central domain of SIRT1. Furthermore, the protein structure analysis of PyMol also proved that the OGT binding region was in the central domain of SIRT1. Under physiological conditions, both wtSIRT1 and SIRT1N346R can inhibit RANKL-mediated transcriptional activation. The RT-PCR detection results showed that wtSIRT1 reduced RANKL transcription under the conditions of apoptotic agent treatment. The finding that SIRT1 can regulate the physiological process of bone remodeling through the RANK/RANKL signaling pathway in osteoblasts under stress. The O-glycosylation and deacetylation activity of SIRT1 significantly increased, regulating the balance between osteoblast survival and apoptosis by deacetylation of key proteins such as RANKL.
Collapse
Affiliation(s)
- Min Cai
- Department of Geriatric Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Yaoqi Chen
- Department of Geriatric Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China; Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Yiting Lin
- Department of Geriatric Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China; Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhangjie Hu
- Department of Geriatric Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China; Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Lizhi Li
- Department of Geriatric Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China; Department of Pediatric Surgery, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Huping Huang
- Department of Gastroenterology, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China.
| | - Jianli Lin
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
26
|
Zhao N, Zhu X, Xie L, Guan X, Tang L, Jiang G, Pang T. The Combination of Citicoline and Nicotinamide Mononucleotide Induces Neurite Outgrowth and Mitigates Vascular Cognitive Impairment via SIRT1/CREB Pathway. Cell Mol Neurobiol 2023; 43:4261-4277. [PMID: 37812361 PMCID: PMC11407720 DOI: 10.1007/s10571-023-01416-7] [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: 06/09/2023] [Accepted: 09/17/2023] [Indexed: 10/10/2023]
Abstract
Vascular dementia (VD) is characterized with vascular cognitive impairment (VCI), which currently has few effective therapies in clinic. Neuronal damage and white matter injury are involved in the pathogenesis of VCI. Citicoline has been demonstrated to exhibit neuroprotection and neurorepair to improve cognition in cerebrovascular diseases. Nicotinamide adenine dinucleotide (NAD+)-dependent sirtuin (SIRT) signaling pathway constitutes a strong intrinsic defense system against various stresses including neuroinflammation in VCI. Our hypothesis is that the combined use of citicoline and the precursor of NAD+, nicotinamide mononucleotide (NMN), could enhance action on cognitive function in VCI. We investigated the synergistic effect of these two drugs in the rat model of VCI by bilateral common carotid artery occlusion (BCCAO). Citicoline significantly enhanced neurite outgrowth in Neuro-2a cells, and the combination of citicoline and NMN remarkably induced neurite outgrowth in Neuro-2a cells and primary cortical neuronal cells with an optimal proportion of 4:1. In the rat model of BCCAO, when two drugs in combination of 160 mg/kg citicoline and 40 mg/kg NMN, this combination administrated at 7 days post-BCCAO significantly improved the cognitive impairment in BCCAO rats compared with vehicle group by the analysis of the Morris water maze and the novel object recognition test. This combination also decreased microglial activation and neuroinflammation, and protected white matter integrity indicated by the increased myelin basic protein (MBP) expression through activation of SIRT1/TORC1/CREB signaling pathway. Our results suggest that the combination of citicoline and NMN has a synergistic effect for the treatment of VD associated with VCI.
Collapse
Affiliation(s)
- Ning Zhao
- State Key Laboratory of Natural Medicines, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Xiaofeng Zhu
- State Key Laboratory of Natural Medicines, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Luyang Xie
- State Key Laboratory of Natural Medicines, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Xin Guan
- State Key Laboratory of Natural Medicines, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Leilei Tang
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, 728 Yucai North Road, Hangzhou, 311200, People's Republic of China
| | - Guojun Jiang
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, 728 Yucai North Road, Hangzhou, 311200, People's Republic of China.
| | - Tao Pang
- State Key Laboratory of Natural Medicines, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
27
|
Jiang X, Li W, Ge L, Lu M. Mesenchymal Stem Cell Senescence during Aging:From Mechanisms to Rejuvenation Strategies. Aging Dis 2023; 14:1651-1676. [PMID: 37196126 PMCID: PMC10529739 DOI: 10.14336/ad.2023.0208] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/08/2023] [Indexed: 05/19/2023] Open
Abstract
In cell transplantation therapy, mesenchymal stem cells(MSCs)are ideal seed cells due to their easy acquisition and cultivation, strong regenerative capacity, multi-directional differentiation abilities, and immunomodulatory effects. Autologous MSCs are better applicable compared with allogeneic MSCs in clinical practice. The elderly are the main population for cell transplantation therapy, but as donor aging, MSCs in the tissue show aging-related changes. When the number of generations of in vitro expansion is increased, MSCs will also exhibit replicative senescence. The quantity and quality of MSCs decline during aging, which limits the efficacy of autologous MSCs transplantation therapy. In this review, we examine the changes in MSC senescence as a result of aging, discuss the progress of research on mechanisms and signalling pathways of MSC senescence, and discuss possible rejuvenation strategies of aged MSCs to combat senescence and enhance the health and therapeutic potential of MSCs.
Collapse
Affiliation(s)
- Xinchen Jiang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
- Hunan provincical key laboratory of Neurorestoratology, the Second Affiliated Hospital, Hunan Normal University, Changsha, China.
| | - Wenshui Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
- Hunan provincical key laboratory of Neurorestoratology, the Second Affiliated Hospital, Hunan Normal University, Changsha, China.
| | - Lite Ge
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
- Hunan provincical key laboratory of Neurorestoratology, the Second Affiliated Hospital, Hunan Normal University, Changsha, China.
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China, Changsha
| | - Ming Lu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
- Hunan provincical key laboratory of Neurorestoratology, the Second Affiliated Hospital, Hunan Normal University, Changsha, China.
| |
Collapse
|
28
|
Chinnapaka S, Malekzadeh H, Tirmizi Z, Arellano JA, Ejaz A. Nicotinamide Riboside Improves Stemness of Human Adipose-Derived Stem Cells and Inhibits Terminal Adipocyte Differentiation. Pharmaceuticals (Basel) 2023; 16:1134. [PMID: 37631051 PMCID: PMC10458272 DOI: 10.3390/ph16081134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Adipose tissue plays a crucial role in maintaining metabolic homeostasis by serving as a storage site for excess fat and protecting other organs from the detrimental effects of lipotoxicity. However, the aging process is accompanied by a redistribution of fat, characterized by a decrease in insulin-sensitive subcutaneous adipose depot and an increase in insulin-resistant visceral adipose depot. This age-related alteration in adipose tissue distribution has implications for metabolic health. Adipose-derived stem cells (ASCs) play a vital role in the regeneration of adipose tissue. However, aging negatively impacts the stemness and regenerative potential of ASCs. The accumulation of oxidative stress and mitochondrial dysfunction-associated cellular damage contributes to the decline in stemness observed in aged ASCs. Nicotinamide adenine dinucleotide (NAD+) is a crucial metabolite that is involved in maintaining cellular homeostasis and stemness. The dysregulation of NAD+ levels with age has been associated with metabolic disorders and the loss of stemness. In this study, we aimed to investigate the effects of nicotinamide riboside (NR), a precursor of NAD+, on the stemness of human ASCs in cell culture. Our findings reveal that adipogenesis is accompanied by an increase in mitochondrial activity and the production of reactive oxygen species (ROS). However, treatment with NR leads to a reduction in mitochondrial activity and ROS production in ASCs. Furthermore, NR administration improves the stemness-related genes expression in ASCs and mitigates their propensity for adipocyte differentiation. These results suggest that NR treatment holds promise as a potential strategy to rejuvenate the stemness of aged ASCs. Further investigations, including in vivo evaluations using animal models and human studies, will be necessary to validate these findings and establish the clinical potential of this well-established drug for enhancing the stemness of aged stem cells.
Collapse
Affiliation(s)
| | | | | | | | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
29
|
Hieber C, Grabbe S, Bros M. Counteracting Immunosenescence-Which Therapeutic Strategies Are Promising? Biomolecules 2023; 13:1085. [PMID: 37509121 PMCID: PMC10377144 DOI: 10.3390/biom13071085] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Aging attenuates the overall responsiveness of the immune system to eradicate pathogens. The increased production of pro-inflammatory cytokines by innate immune cells under basal conditions, termed inflammaging, contributes to impaired innate immune responsiveness towards pathogen-mediated stimulation and limits antigen-presenting activity. Adaptive immune responses are attenuated as well due to lowered numbers of naïve lymphocytes and their impaired responsiveness towards antigen-specific stimulation. Additionally, the numbers of immunoregulatory cell types, comprising regulatory T cells and myeloid-derived suppressor cells, that inhibit the activity of innate and adaptive immune cells are elevated. This review aims to summarize our knowledge on the cellular and molecular causes of immunosenescence while also taking into account senescence effects that constitute immune evasion mechanisms in the case of chronic viral infections and cancer. For tumor therapy numerous nanoformulated drugs have been developed to overcome poor solubility of compounds and to enable cell-directed delivery in order to restore immune functions, e.g., by addressing dysregulated signaling pathways. Further, nanovaccines which efficiently address antigen-presenting cells to mount sustained anti-tumor immune responses have been clinically evaluated. Further, senolytics that selectively deplete senescent cells are being tested in a number of clinical trials. Here we discuss the potential use of such drugs to improve anti-aging therapy.
Collapse
Affiliation(s)
- Christoph Hieber
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
30
|
Hakim SG, von der Gracht A, Pries R, Rades D, Steller D. Protective impact of nicotinamide mononucleotide (NMN) and platelet-rich fibrin (PRF) on replicative and radiation-induced senescence of human osteoblasts. J Craniomaxillofac Surg 2023; 51:497-507. [PMID: 37438229 DOI: 10.1016/j.jcms.2023.06.008] [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: 03/09/2023] [Revised: 05/04/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023] Open
Abstract
The aim of this study was to investigate the cellular changes induced by spontaneous/replicative senescence and radiation in human osteoblasts (OBs), and the impact of cultivation with nicotinamide mononucleotide (NMN) and platelet-rich fibrin (PRF) on apoptosis, senescence-associated β-galactosidase staining (SA β-gal), and senescence-related gene expression using RT2 Profiler PCR array. The results showed that replicative OB aging follows a different pattern from that of radiation-induced cellular senescence. SA β-gal intensity score showed a significant elevation after spontaneous replicative aging of OB (agiT1) 7 days following the start of the experiment, compared with their initial control condition (T0) (T0 = 2.1 ± 0.47; agiT1 = 9.60 ± 1.56; p = 0.001). Concurrent treatment by NMN and PRF showed a protective effect on OBs undergoing replicative senescence, and reduced SA β-gal staining significantly (agiT1 = 9.60 ± 1.56; agiT1+PRF = 3.19 ± 0.52; agiT1+NMN = 3.38 ± 0.36; p < 0.001). These results provide evidence for the potential clinical implications of systematic NMN administration and local PRF application to prevent age-related bone disturbances in elderly patients.
Collapse
Affiliation(s)
- Samer G Hakim
- Department of Oral and Maxillofacial Surgery, Helios Medical Center, Schwerin, Germany; Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein (Campus Lübeck), Lübeck, Germany.
| | - Anij von der Gracht
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein (Campus Lübeck), Lübeck, Germany
| | - Ralph Pries
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Schleswig-Holstein (Campus Lübeck), Lübeck, Germany
| | - Dirk Rades
- Department of Radiation Oncology, University Hospital Schleswig-Holstein (Campus Lübeck), Lübeck, Germany
| | - Daniel Steller
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein (Campus Lübeck), Lübeck, Germany
| |
Collapse
|
31
|
Huang Y, Dou Y, Yang B, He B, Zhang X, Zhang K, Yang X. Nicotinamide mononucleotide supplementation mitigates osteopenia induced by modeled microgravity in rats. Cell Stress Chaperones 2023; 28:385-394. [PMID: 37195399 PMCID: PMC10352228 DOI: 10.1007/s12192-023-01356-7] [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: 04/30/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/18/2023] Open
Abstract
Exposure to weightlessness causes severe osteopenia, resulting in raised fracture risk. The current study aimed to investigate whether nicotinamide mononucleotide (NMN) supplementation protected against the osteopenia in hindlimb unloading (HLU) rats in vivo and modeled microgravity-induced osteoblastic dysfunction in vitro. The 3-mo-old rats were exposed to HLU and intragastrically administered NMN every 3 days (500 mg/kg body weight) for 4 weeks. NMN supplementation mitigated HLU-induced bone loss, evidenced by greater bone mass and biomechanical properties and better trabecular bone structure. NMN supplementation mitigated HLU-induced oxidative stress, evidenced by greater levels of nicotinamide adenine dinucleotide and activities of superoxide dismutase 2 and lesser malondialdehyde levels. Modeled microgravity stimulation using rotary wall vessel bioreactor in MC3T3-E1 cells inhibited osteoblast differentiation, which was reversed by NMN treatment. Furthermore, NMN treatment mitigated microgravity-induced mitochondrial impairments, evidenced by lesser reactive oxygen species generation and greater adenosine triphosphate production, mtDNA copy number, and activities of superoxide dismutase 2 and Complex I and II. Additionally, NMN promoted activation of AMP-activated protein kinase (AMPK), evidenced by greater AMPKα phosphorylation. Our research suggested that NMN supplementation attenuated osteoblastic mitochondrial impairment and mitigated osteopenia induced by modeled microgravity.
Collapse
Affiliation(s)
- Yunfei Huang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Yusheng Dou
- Department of Should and Elbow Joint, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Bo Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Baorong He
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Xuefang Zhang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Ke Zhang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Xiaobin Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China.
| |
Collapse
|
32
|
Li Y, Hu M, Xie J, Li S, Dai L. Dysregulation of histone modifications in bone marrow mesenchymal stem cells during skeletal ageing: roles and therapeutic prospects. Stem Cell Res Ther 2023; 14:166. [PMID: 37357311 DOI: 10.1186/s13287-023-03393-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 05/31/2023] [Indexed: 06/27/2023] Open
Abstract
Age-associated bone diseases such as osteoporosis (OP) are common in the elderly due to skeletal ageing. The process of skeletal ageing can be accelerated by reduced proliferation and osteogenesis of bone marrow mesenchymal stem cells (BM-MSCs). Senescence of BM-MSCs is a main driver of age-associated bone diseases, and the fate of BM-MSCs is tightly regulated by histone modifications, such as methylation and acetylation. Dysregulation of histone modifications in BM-MSCs may activate the genes related to the pathogenesis of skeletal ageing and age-associated bone diseases. Here we summarize the histone methylation and acetylation marks and their regulatory enzymes that affect BM-MSC self-renewal, differentiation and senescence. This review not only describes the critical roles of histone marks in modulating BM-MSC functions, but also underlines the potential of epigenetic enzymes as targets for treating age-associated bone diseases. In the future, more effective therapeutic approaches based on these epigenetic targets will be developed and will benefit elderly individuals with bone diseases, such as OP.
Collapse
Affiliation(s)
- Yujue Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingxing Hu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinwei Xie
- Department of Orthopedics Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuangqing Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lunzhi Dai
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
33
|
Zheng Z, Wang X, Ouyang L, Chen W, Zhang L, Cao Y. Antioxidants Improve the Proliferation and Efficacy of hUC-MSCs against H 2O 2-Induced Senescence. Antioxidants (Basel) 2023; 12:1334. [PMID: 37507874 PMCID: PMC10376626 DOI: 10.3390/antiox12071334] [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: 04/15/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Human umbilical cord mesenchymal stem cells (hUC-MSCs) are broadly applied in clinical treatment due to convenient accessibility, low immunogenicity, and the absence of any ethical issues involved. However, the microenvironment of inflammatory tissues may cause oxidative stress and induce senescence in transplanted hUC-MSCs, which will further reduce the proliferation, migration ability, and the final therapeutic effects of hUC-MSCs. Beta-nicotinamide mononucleotide (NMN) and coenzyme Q10 (CoQ10) are famous antioxidants and longevity medicines that could reduce intracellular reactive oxygen species levels by different mechanisms. In this study, hUC-MSCs were treated in vitro with NMN and CoQ10 to determine if they could reduce oxidative stress caused by hydrogen peroxide (H2O2) and recover cell functions. The effects of NMN and CoQ10 on the cell proliferation, the mRNA levels of the inflammatory cytokine TNFα and the anti-inflammatory cytokine IL10, and the differentiation and cell migration ability of hUC-MSCs before and after H2O2 treatment were investigated. The findings revealed that NMN and CoQ10 reduced H2O2-induced senescence and increased hUC-MSCs' proliferation in the late phase as passage 12 and later. The TNFα mRNA level of hUC-MSCs induced by H2O2 was significantly decreased after antioxidant treatment. NMN and CoQ10 all reduced the adipogenic differentiation ability of hUC-MSCs. CoQ10 improved the chondrogenic differentiation ability of hUC-MSCs. Furthermore, NMN was found to significantly enhance the migration ability of hUC-MSCs. Transcriptomic analysis revealed that NMN and CoQ10 both increased DNA repair ability and cyclin expression and downregulated TNF and IL-17 inflammatory signaling pathways, thereby contributing to the proliferative promotion of senecent stem cells and resistance to oxidative stress. These findings suggest that antioxidants can improve the survival and efficacy of hUC-MSCs in stem cell therapy for inflammation-related diseases.
Collapse
Affiliation(s)
- Zhaojuan Zheng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xia Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Liming Ouyang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wenxia Chen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Lixin Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yulin Cao
- Beijing Tang Yi Hui Kang Biomedical Technology Co., Ltd., Beijing 100032, China
| |
Collapse
|
34
|
Aobulikasimu A, Liu T, Piao J, Sato S, Ochi H, Okawa A, Tsuji K, Asou Y. SIRT6-PAI-1 axis is a promising therapeutic target in aging-related bone metabolic disruption. Sci Rep 2023; 13:7991. [PMID: 37198221 DOI: 10.1038/s41598-023-33297-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 04/11/2023] [Indexed: 05/19/2023] Open
Abstract
The mechanistic regulation of bone mass in aged animals is poorly understood. In this study, we examined the role of SIRT6, a longevity-associated factor, in osteocytes, using mice lacking Sirt6 in Dmp-1-expressing cells (cKO mice) and the MLO-Y4 osteocyte-like cell line. cKO mice exhibited increased osteocytic expression of Sost, Fgf23 and senescence inducing gene Pai-1 and the senescence markers p16 and Il-6, decreased serum phosphate levels, and low-turnover osteopenia. The cKO phenotype was reversed in mice that were a cross of PAI-1-null mice with cKO mice. Furthermore, senescence induction in MLO-Y4 cells increased the Fgf23 and Sost mRNA expression. Sirt6 knockout and senescence induction increased HIF-1α binding to the Fgf23 enhancer sequence. Bone mass and serum phosphate levels were higher in PAI-1-null aged mice than in wild-type mice. Therefore, SIRT6 agonists or PAI-1 inhibitors may be promising therapeutic options for aging-related bone metabolism disruptions.
Collapse
Affiliation(s)
- Alkebaier Aobulikasimu
- Department of Orthopedics Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Tao Liu
- Department of Orthopedics Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Jinying Piao
- Department of Orthopedics Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Shingo Sato
- Department of Orthopedics Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Movement Functions, Research Institute, National Rehabilitation Center for Persons With Disabilities, Tokorozawa-Shi, Saitama, Japan
| | - Atsushi Okawa
- Department of Orthopedics Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Kunikazu Tsuji
- Department of Orthopedics Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yoshinori Asou
- Department of Orthopedics Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-Ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
35
|
Cheng M, Yuan W, Moshaverinia A, Yu B. Rejuvenation of Mesenchymal Stem Cells to Ameliorate Skeletal Aging. Cells 2023; 12:998. [PMID: 37048071 PMCID: PMC10093211 DOI: 10.3390/cells12070998] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
Advanced age is a shared risk factor for many chronic and debilitating skeletal diseases including osteoporosis and periodontitis. Mesenchymal stem cells develop various aging phenotypes including the onset of senescence, intrinsic loss of regenerative potential and exacerbation of inflammatory microenvironment via secretory factors. This review elaborates on the emerging concepts on the molecular and epigenetic mechanisms of MSC senescence, such as the accumulation of oxidative stress, DNA damage and mitochondrial dysfunction. Senescent MSCs aggravate local inflammation, disrupt bone remodeling and bone-fat balance, thereby contributing to the progression of age-related bone diseases. Various rejuvenation strategies to target senescent MSCs could present a promising paradigm to restore skeletal aging.
Collapse
Affiliation(s)
- Mingjia Cheng
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Weihao Yuan
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Alireza Moshaverinia
- Section of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Bo Yu
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
36
|
Tian Y, Zhu CL, Li P, Li HR, Liu Q, Deng XM, Wang JF. Nicotinamide Mononucleotide Attenuates LPS-Induced Acute Lung Injury With Anti-Inflammatory, Anti-Oxidative and Anti-Apoptotic Effects. J Surg Res 2023; 283:9-18. [PMID: 36347171 DOI: 10.1016/j.jss.2022.09.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/23/2022] [Accepted: 09/18/2022] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Nicotinamide mononucleotide (NMN) is a nucleotide that is commonly recognized for its role as an intermediate of nicotinamide adenine dinucleotide (NAD+) biosynthesis with multiple pharmacological effects. The purpose of this study was to evaluate the protective effect of nicotinamide mononucleotide (NMN) against lipopolysaccharide (LPS)-induced acute lung injury (ALI). METHODS We investigated the effect of NMN on ALI-induced inflammatory response, oxidative stress, and cell apoptosis. The ALI mouse model was performed by injecting LPS intratracheally at a dose of 10 mg/kg in 50 μL saline. Flow cytometry was used to detect neutrophil infiltration in bronchoalveolar lavage fluid (BALF), and ELISA was used to detect the contents of inflammatory cytokines TNF-α, IL-1β and IL-6 in BALF. Oxidative stress was evaluated by determining the superoxide dismutase (SOD) activity and malondialdehyde (MDA) content in lung tissue. ROS formation was analyzed by immunofluorescence. Western blotting was performed to detect apoptotic levels and p38MAPK/NF-κB phosphorylation levels in lung tissue. RESULTS In the ALI mouse model, NMN showed a significant therapeutic effect compared to the LPS group. NMN attenuated the pathological damage and cell apoptosis in lung tissue, decreased the levels of TNF-α, IL-1β, and IL-6 in BALF, and reduced the number of total cells and neutrophils in BALF. In addition, NMN attenuated the LPS-induced elevation of dry-to-wet ratio, MDA content, p38 MAPK and p65 NF-κB phosphorylation levels, and the SOD activity was increased by NMN treatment. CONCLUSIONS In conclusion, the present study showed that NMN exerted a protective effect on LPS-induced ALI with anti-inflammatory, antioxidative, and antiapoptotic effects.
Collapse
Affiliation(s)
- Ye Tian
- Department of Anesthesiology, The Sixth Medical Centre of General Hospital of PLA, Beijing, China
| | - Cheng-Long Zhu
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Peng Li
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui-Ru Li
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qiang Liu
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Ming Deng
- Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Jia-Feng Wang
- Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
37
|
Wang C, Chen R, Zhu X, Zhang X, Lian N. METTL14 alleviates the development of osteoporosis in ovariectomized mice by upregulating m 6A level of SIRT1 mRNA. Bone 2023; 168:116652. [PMID: 36584783 DOI: 10.1016/j.bone.2022.116652] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
The purpose of this study was to investigate whether METTL14 participated in ovariectomized (OVX)-induced osteoporosis (OP) in mice by regulating the m6A level of SIRT1 mRNA. OVX was performed on mice to induce OP, and mouse bone marrow stromal cells (BMSCs) and bone marrow mononuclear macrophages (BMMs) were isolated to induce osteoblast differentiation and osteoclast differentiation, respectively. The morphology of bone trabeculae was evaluated under a micro-CT scanner. The changes in pathology of bone tissues were observed through staining using hematoxylin-eosin. The number of osteoclasts was measured by tartrate-resistant acid phosphatase staining, and the content of serum calcium, PINP, and CTX-I was tested by enzyme-linked immunosorbent assay, accompanied by the measurement of the expression of SIRT1, METTL14, osteogenic marker genes, and osteoclast marker genes. The m6A modification level of SIRT1 and the binding between METTL14 and SIRT1 were verified. In OVX mice, SIRT1 and METTL14 were downregulated. Overexpression of SIRT1 or METTL14 increased the expression of osteogenic marker genes but decreased the expression of osteoclast marker genes. Additionally, METTL14 overexpression increased m6A level of SIRT1 mRNA. Furthermore, overexpression of METTL14 promoted osteoblast differentiation and suppressed osteoclast differentiation, which were reversed by knockdown of SIRT1. METTL14 promoted osteoblast differentiation and repressed osteoclast differentiation by m6A-dependent upregulation of SIRT1 mRNA, thereby alleviating OP development.
Collapse
Affiliation(s)
- Changsheng Wang
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China.
| | - Rongsheng Chen
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China
| | - Xitian Zhu
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China
| | - Xiaobo Zhang
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China
| | - Nancheng Lian
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China
| |
Collapse
|
38
|
Lu Z, Jiang L, Lesani P, Zhang W, Li N, Luo D, Li Y, Ye Y, Bian J, Wang G, Dunstan CR, Jiang X, Zreiqat H. Nicotinamide Mononucleotide Alleviates Osteoblast Senescence Induction and Promotes Bone Healing in Osteoporotic Mice. J Gerontol A Biol Sci Med Sci 2023; 78:186-194. [PMID: 36037105 DOI: 10.1093/gerona/glac175] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Combating the accumulated senescent cells and the healing of osteoporotic bone fractures in the older remains a significant challenge. Nicotinamide mononucleotide (NMN), a precursor of NAD+, is an excellent candidate for mitigating aging-related disorders. However, it is unknown if NMN can alleviate senescent cell induction and enhance osteoporotic bone fracture healing. Here we show that NMN treatment partially reverses the effects of tumor necrosis factor-alpha (TNF-α) on human primary osteoblasts (HOBs): senescent cell induction, diminished osteogenic differentiation ability, and intracellular NAD+ and NADH levels. Mechanistically, NMN restores the mitochondrial dysfunction in HOBs induced by TNF-α evidenced by increased mitochondrial membrane potential and reduced reactive oxidative species and mitochondrial mass. NMN also increases mitophagy activity by down-regulating P62 expression and up-regulating light chain 3B-II protein expression. In addition, the cell senescence protective effects of NMN on HOBs are mitigated by a mitophagy inhibitor (Bafilomycin A1). In vivo, NMN supplementation attenuates senescent cell induction in growth plates, partially prevents osteoporosis in an ovariectomized mouse model, and accelerates bone healing in osteoporotic mice. We conclude that NMN can be a novel and promising therapeutic candidate to enhance bone fracture healing capacity in the older.
Collapse
Affiliation(s)
- ZuFu Lu
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, the University of Sydney, Sydney, New South Wales, Australia.,ARC Training Centre for Innovative BioEngineering, the University of Sydney, Sydney, New South Wales, Australia
| | - Liting Jiang
- Department of Stomatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Pooria Lesani
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, the University of Sydney, Sydney, New South Wales, Australia.,ARC Training Centre for Innovative BioEngineering, the University of Sydney, Sydney, New South Wales, Australia
| | - WenJie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine.,National Clinical Research Center for Oral diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Ning Li
- Department of Stomatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Danyang Luo
- Department of Stomatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Yusi Li
- Department of Stomatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Yulin Ye
- Department of Stomatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine.,College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Ji Bian
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, the University of Sydney, Sydney, New South Wales, Australia
| | - Guocheng Wang
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Colin R Dunstan
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, the University of Sydney, Sydney, New South Wales, Australia.,ARC Training Centre for Innovative BioEngineering, the University of Sydney, Sydney, New South Wales, Australia
| | - XinQuan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine.,National Clinical Research Center for Oral diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, the University of Sydney, Sydney, New South Wales, Australia.,ARC Training Centre for Innovative BioEngineering, the University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
39
|
NAD +-Consuming Enzymes in Stem Cell Homeostasis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:4985726. [PMID: 36819783 PMCID: PMC9931471 DOI: 10.1155/2023/4985726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/27/2022] [Accepted: 01/06/2023] [Indexed: 02/10/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a coenzyme used in redox reactions, energy metabolism, and mitochondrial biogenesis. NAD+ is also required as a cofactor by nonredox NAD+-dependent enzymes. Hundreds of enzymes that consume NAD+ have been identified. The NAD+-consuming enzymes are involved in a variety of cellular processes such as signal transduction, DNA repair, cellular senescence, and stem cell (SC) homeostasis. In this review, we discussed how different types of NAD+-consuming enzymes regulate SC functions and summarized current research on the roles of the NAD+ consumers in SC homeostasis. We hope to provide a more global and integrative insight to the mechanism and intervention of SC homeostasis via the regulation of the NAD+-consuming enzymes.
Collapse
|
40
|
Extracellular Vesicles from NMN Preconditioned Mesenchymal Stem Cells Ameliorated Myocardial Infarction via miR-210-3p Promoted Angiogenesis. Stem Cell Rev Rep 2023; 19:1051-1066. [PMID: 36696015 DOI: 10.1007/s12015-022-10499-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/26/2023]
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSCs-EVs) possess cardioprotection in acute myocardial infarction. Nevertheless, the therapeutic intervention potential and the molecular mechanism of EVs from NMN (Nicotinamide mononucleotide) preconditioned hUCMSCs (N-EVs) in acute myocardial infarction remains unknown. In the present study, EVs from hUCMSCs (M-EVs) and N-EVs were identified by electron microscopy, immunoblotting and nanoparticle tracking analysis. Compared with M-EVs, N-EVs significantly increased the proliferation, migration, and angiogenesis of HUVECs. Meanwhile, N-EVs markedly reduced apoptosis and cardiac fibrosis and promoted angiogenesis in the peri-infarct region in the MI rats. A high-throughput miRNA sequencing and qPCR methods analysis revealed that miR-210-3p was abundant in N-EVs and the expression of miR-210-3p was obviously upregulated in HUVECs after N-EVs treated. Overexpression of miR-210-3p in HUVECs significantly enhanced the tube formation, migration and proliferative capacities of HUVECs. However, downregulation of miR-210-3p in HUVECs markedly decreased the tube formation, migration and proliferative capacities of HUVECs. Furthermore, bioinformatics analysis and luciferase assays revealed that EphrinA3 (EFNA3) was a direct target of miR-210-3p. Knockdown of miR-210-3p in N-EVs significantly impaired its ability to protect the heart after myocardial infarction. Altogether, these results indicated that N-EVs promoted the infarct healing through improvement of angiogenesis by miR-210-3p via targeting the EFNA3. Created with Biorender.com.
Collapse
|
41
|
Wang H, Sun Y, Pi C, Yu X, Gao X, Zhang C, Sun H, Zhang H, Shi Y, He X. Nicotinamide Mononucleotide Supplementation Improves Mitochondrial Dysfunction and Rescues Cellular Senescence by NAD +/Sirt3 Pathway in Mesenchymal Stem Cells. Int J Mol Sci 2022; 23:ijms232314739. [PMID: 36499074 PMCID: PMC9738479 DOI: 10.3390/ijms232314739] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
In vitro expansion-mediated replicative senescence has severely limited the clinical applications of mesenchymal stem cells (MSCs). Accumulating studies manifested that nicotinamide adenine dinucleotide (NAD+) depletion is closely related to stem cell senescence and mitochondrial metabolism disorder. Promoting NAD+ level is considered as an effective way to delay aging. Previously, we have confirmed that nicotinamide mononucleotide (NMN), a precursor of NAD+, can alleviate NAD+ deficiency-induced MSC senescence. However, whether NMN can attenuate MSC senescence and its underlying mechanisms are still incompletely clear. The present study herein showed that late passage (LP) MSCs displayed lower NAD+ content, reduced Sirt3 expression and mitochondrial dysfunction. NMN supplementation leads to significant increase in intracellular NAD+ level, NAD+/ NADH ratio, Sirt3 expression, as well as ameliorated mitochondrial function and rescued senescent MSCs. Additionally, Sirt3 over-expression relieved mitochondrial dysfunction, and retrieved senescence-associated phenotypic features in LP MSCs. Conversely, inhibition of Sirt3 activity via a selective Sirt3 inhibitor 3-TYP in early passage (EP) MSCs resulted in aggravated cellular senescence and abnormal mitochondrial function. Furthermore, NMN administration also improves 3-TYP-induced disordered mitochondrial function and cellular senescence in EP MSCs. Collectively, NMN replenishment alleviates mitochondrial dysfunction and rescues MSC senescence through mediating NAD+/Sirt3 pathway, possibly providing a novel mechanism for MSC senescence and a promising strategy for anti-aging pharmaceuticals.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xu He
- Correspondence: ; Tel.: +86-135-0430-7430
| |
Collapse
|
42
|
Zhao X, Zhang M, Wang J, Ji K, Wang Y, Sun X, Xu C, Wang Q, He N, Song H, Du L, Wang F, Huang H, Liu Y, Liu Q. NMN ameliorated radiation induced damage in NRF2-deficient cell and mice via regulating SIRT6 and SIRT7. Free Radic Biol Med 2022; 193:342-353. [PMID: 36252808 DOI: 10.1016/j.freeradbiomed.2022.10.267] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/18/2022] [Accepted: 10/07/2022] [Indexed: 12/14/2022]
Abstract
Risk of cancer often increases with aging, and radiotherapy is an essential component of treatment. As for abdominal and pelvic cancer, radiotherapy always inevitably causes injury to intestines through direct DNA damage or overload of reactive oxygen species (ROS). Nuclear factor erythroid 2-related factor 2 (NRF2) has been identified as a key protective factor against ionizing-radiation induced damage through promoting DNA damage repair and antioxidant modulation. However, the level of NRF2 always decreases with aging. Here, we demonstrated that NRF2 deficiency aggravated cellular DNA damage and the intestinal pathological lesion. Overexpression of SIRT6 or SIRT7 could improve cell proliferation and protect against radiation injury in NRF2 knock-out (KO) cells by modulating oxidative-stress and DNA damage repair. Consistently, supplement of nicotinamide mononucleotide (NMN), the agonist of sirtuins, increased the level of SIRT6 and SIRT7 in NRF2 KO cells, concomitant with reduced cellular ROS level and ameliorated DNA damage. In vivo, long-term oral administration of NMN attenuated the radiation-induced injury of jejunum, increased the number of intestinal stem cells, and promoted the ability of intestinal proliferation in NRF2-/- mice. Together, our results indicated that SIRT6 and SIRT7 had involved in scavenging ROS and repairing DNA damage, and NMN could be a promising candidate for preventing radiation damage when NRF2 is lacking.
Collapse
Affiliation(s)
- Xiaotong Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Manman Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Jinhan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Kaihua Ji
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Yan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Xiaohui Sun
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Chang Xu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Qin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Huijuan Song
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Liqing Du
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Feng Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hao Huang
- Effepharm (Shanghai) Co. Ltd, No.1 Mid Wangdong Rd, Songjiang District, Shanghai, 201601, China
| | - Yang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China.
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China.
| |
Collapse
|
43
|
Liang T, Li P, Liang A, Zhu Y, Qiu X, Qiu J, Peng Y, Huang D, Gao W, Gao B. Identifying the key genes regulating mesenchymal stem cells chondrogenic differentiation: an in vitro study. BMC Musculoskelet Disord 2022; 23:985. [PMCID: PMC9664719 DOI: 10.1186/s12891-022-05958-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background
Mesenchymal stem cells (MSCs) possess the potential to differentiate into chondrocytes, which makes them an ideal source for healing cartilage defects. Here, we seek to identify the essential genes participating in MSCs chondrogenesis.
Methods
Human MSCs were induced for chondrogenesis for 7, 14, and 21 days using a high-density micromass culture system, and RNA was extracted for RNA-seq.
Results
A total of 6247 differentially expressed genes (DEGs) were identified on day 7, and 85 DEGs were identified on day 14. However, no significant DEGs was identified on day 21. The top 30 DEGs at day 7, including COL9A3, COL10A1, and CILP2, are closely related to extracellular matrix organization. While the top 30 DEGs at day 14 revealed that inflammation-related genes were enriched, including CXCL8, TLR2, and CCL20. We also conducted protein–protein interaction (PPI) networks analysis using the search tool for the retrieval of interacting genes (STRING) database and identified key hub genes, including CXCL8, TLR2, CCL20, and MMP3. The transcriptional factors were also analyzed, identifying the top 5 TFs: LEF1, FOXO1, RORA, BHLHE41, and SOX5. We demonstrated one particular TF, RORA, in promoting early MSCs chondrogenesis.
Conclusions
Taken together, our results suggested that these DEGs may have a complex effect on MSCs chondrogenesis both synergistically and solitarily.
Collapse
|
44
|
Sánchez-Ramírez E, Ung TPL, Alarcón del Carmen A, del Toro-Ríos X, Fajardo-Orduña GR, Noriega LG, Cortés-Morales VA, Tovar AR, Montesinos JJ, Orozco-Solís R, Stringari C, Aguilar-Arnal L. Coordinated metabolic transitions and gene expression by NAD+ during adipogenesis. J Biophys Biochem Cytol 2022; 221:213521. [PMID: 36197339 PMCID: PMC9538974 DOI: 10.1083/jcb.202111137] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 08/18/2022] [Accepted: 09/20/2022] [Indexed: 12/17/2022] Open
Abstract
Adipocytes are the main cell type in adipose tissue, which is a critical regulator of metabolism, highly specialized in storing energy as fat. Adipocytes differentiate from multipotent mesenchymal stromal cells (hMSCs) through adipogenesis, a tightly controlled differentiation process involving close interplay between metabolic transitions and sequential programs of gene expression. However, the specific gears driving this interplay remain largely obscure. Additionally, the metabolite nicotinamide adenine dinucleotide (NAD+) is becoming increasingly recognized as a regulator of lipid metabolism, and a promising therapeutic target for dyslipidemia and obesity. Here, we explored how NAD+ bioavailability controls adipogenic differentiation from hMSC. We found a previously unappreciated repressive role for NAD+ on adipocyte commitment, while a functional NAD+-dependent deacetylase SIRT1 appeared crucial for terminal differentiation of pre-adipocytes. Repressing NAD+ biosynthesis during adipogenesis promoted the adipogenic transcriptional program, while two-photon microscopy and extracellular flux analyses suggest that SIRT1 activity mostly relies on the metabolic switch. Interestingly, SIRT1 controls subcellular compartmentalization of redox metabolism during adipogenesis.
Collapse
Affiliation(s)
- Edgar Sánchez-Ramírez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Thi Phuong Lien Ung
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, Palaiseau, France
| | - Alejandro Alarcón del Carmen
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ximena del Toro-Ríos
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Guadalupe R. Fajardo-Orduña
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lilia G. Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Victor A. Cortés-Morales
- Mesenchymal Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, Mexico City, Mexico
| | - Armando R. Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Juan José Montesinos
- Mesenchymal Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, Mexico City, Mexico
| | - Ricardo Orozco-Solís
- Laboratorio de Cronobiología y Metabolismo, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Chiara Stringari
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, Palaiseau, France,Chiara Stringari:
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico,Correspondence to Lorena Aguilar-Arnal:
| |
Collapse
|
45
|
Metformin Facilitates Osteoblastic Differentiation and M2 Macrophage Polarization by PI3K/AKT/mTOR Pathway in Human Umbilical Cord Mesenchymal Stem Cells. Stem Cells Int 2022; 2022:9498876. [PMID: 35761829 PMCID: PMC9233575 DOI: 10.1155/2022/9498876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are the most promising multipotent stem cells that can differentiate into osteoblasts, chondrocytes, and adipocytes. This cellular flexibility contributes to widespread clinical use of MSCs in tissue repair and regeneration. The immune system is a key player in regulating bone remodeling. In recent years, the association between the immune system and bone metabolism has become an increasing focus of interest. Metformin, a glucose-lowering drug, exerts powerful impact on metabolic signaling. However, whether metformin can modulate bone metabolism or whether metformin can influence immune milieu by regulation of macrophages has not been thoroughly elucidated. Herein, we specifically explored the complex interactions between macrophages and human umbilical cord mesenchymal stem cells (UC-MSCs) in the context of metformin. Our research demonstrated that metformin not only stimulated osteogenesis of UC-MSCs but also influenced the immune system via promoting M2 but reducing M1 macrophages. Mechanically, we found that metformin-treated M2 macrophages possessed more potent osteoinductive capacity in our coculture system. Molecularly, these metformin-stimulated M2 macrophages facilitated osteogenesis via activating the PI3K/AKT/mTOR pathway. As demonstrated by using PI3K-specific inhibitor LY294002, we found that the pathway inhibitor partly reversed osteoinductive activity which was activated by coculture of metformin-treated M2 macrophages. Overall, our novel research illuminated the cooperative and synergistic effects of metformin and M2 macrophages on the dynamic balance of bone metabolism.
Collapse
|
46
|
Tower RJ, Busse E, Jaramillo J, Lacey M, Hoffseth K, Guntur AR, Simkin J, Sammarco MC. Spatial transcriptomics reveals metabolic changes underly age-dependent declines in digit regeneration. eLife 2022; 11:71542. [PMID: 35616636 PMCID: PMC9135401 DOI: 10.7554/elife.71542] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 05/18/2022] [Indexed: 12/12/2022] Open
Abstract
De novo limb regeneration after amputation is restricted in mammals to the distal digit tip. Central to this regenerative process is the blastema, a heterogeneous population of lineage-restricted, dedifferentiated cells that ultimately orchestrates regeneration of the amputated bone and surrounding soft tissue. To investigate skeletal regeneration, we made use of spatial transcriptomics to characterize the transcriptional profile specifically within the blastema. Using this technique, we generated a gene signature with high specificity for the blastema in both our spatial data, as well as other previously published single-cell RNA-sequencing transcriptomic studies. To elucidate potential mechanisms distinguishing regenerative from non-regenerative healing, we applied spatial transcriptomics to an aging model. Consistent with other forms of repair, our digit amputation mouse model showed a significant impairment in regeneration in aged mice. Contrasting young and aged mice, spatial analysis revealed a metabolic shift in aged blastema associated with an increased bioenergetic requirement. This enhanced metabolic turnover was associated with increased hypoxia and angiogenic signaling, leading to excessive vascularization and altered regenerated bone architecture in aged mice. Administration of the metabolite oxaloacetate decreased the oxygen consumption rate of the aged blastema and increased WNT signaling, leading to enhanced in vivo bone regeneration. Thus, targeting cell metabolism may be a promising strategy to mitigate aging-induced declines in tissue regeneration.
Collapse
Affiliation(s)
- Robert J Tower
- Department of Orthopaedics, Johns Hopkins University, Baltimore, United States
| | - Emily Busse
- Department of Surgery, Tulane School of Medicine, New Orleans, United States
| | - Josue Jaramillo
- Department of Surgery, Tulane School of Medicine, New Orleans, United States
| | - Michelle Lacey
- Department of Mathematics, Tulane University, New Orleans, United States
| | - Kevin Hoffseth
- Department of Biological & Agricultural Engineering, Louisiana State University, Baton Rouge, United States
| | - Anyonya R Guntur
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, United States
| | - Jennifer Simkin
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, New Orleans, United States
| | - Mimi C Sammarco
- Department of Surgery, Tulane School of Medicine, New Orleans, United States
| |
Collapse
|
47
|
Mladenova SG, Savova MS, Marchev AS, Ferrante C, Orlando G, Wabitsch M, Georgiev MI. Anti-adipogenic activity of maackiain and ononin is mediated via inhibition of PPARγ in human adipocytes. Biomed Pharmacother 2022; 149:112908. [PMID: 35367764 DOI: 10.1016/j.biopha.2022.112908] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 11/26/2022] Open
Abstract
Obesity is a global health burden for which we do not yet have effective treatments for prevention or therapy. Plants are an invaluable source of bioactive leads possessing anti-adipogenic potential. Ethnopharmacological use of Ononis spinosa L. roots (OSR) for treatment of obesity and metabolic disorders requires а scientific rationale. The current study examined the anti-adipogenic capacity of OSR and its secondary metabolites ononin (ONON) and maackiain (MACK) in human adipocytes as an in vitro model of obesity. Both ONON and MACK diminished lipid accumulation during adipocyte differentiation. Molecular docking analysis exposed the potential interactions between MACK or ONON and target regulatory adipogenic proteins. Furthermore, results from an RT-qPCR analysis disclosed significant upregulation of AMPK by MACK and ONON treatment. In addition, ONON increased SIRT1, PI3K and ACC mRNA expression, while MACK notably downregulated CEBPA, AKT, SREBP1, ACC and ADIPOQ. The protein level of PI3K, C/EBPα, PPARγ and adiponectin was reduced upon MACK treatment in a concentration-dependent manner. Similarly, ONON suppressed PI3K, PPARγ and adiponectin protein abundance. Finally, our study provides evidence that ONON exerts anti-adipogenic effect by upregulation of SIRT1 and inhibition of PI3K, PPARγ and adiponectin, while MACK induced strong inhibitory effect on adipogenesis via hampering PI3K, PPARγ/C/EBPα signaling and anti-lipogenic effect through downregulation of SREBP1 and ACC. Even though OSR does not hamper adipogenic differentiation, it could be exploited as a source of natural leads with anti-adipogenic potential. The multidirectional mechanism of action of MACK warrant further validation in the context of in vivo obesity models.
Collapse
Affiliation(s)
- Saveta G Mladenova
- BB-NCIPD Ltd., BB-National Centre of Infectious and Parasitic Diseases, Ministry of Health, 1000 Sofia, Bulgaria
| | - Martina S Savova
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Department of Biotechnology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd, 4000 Plovdiv, Bulgaria
| | - Andrey S Marchev
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Department of Biotechnology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd, 4000 Plovdiv, Bulgaria
| | - Claudio Ferrante
- Department of Pharmacy, G. d'Annunzio University, 66100 Chieti, Italy
| | - Giustino Orlando
- Department of Pharmacy, G. d'Annunzio University, 66100 Chieti, Italy
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89073 Ulm, Germany
| | - Milen I Georgiev
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Department of Biotechnology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd, 4000 Plovdiv, Bulgaria.
| |
Collapse
|
48
|
Little-Letsinger SE, Rubin J, Diekman B, Rubin CT, McGrath C, Pagnotti GM, Klett EL, Styner M. Exercise to Mend Aged-tissue Crosstalk in Bone Targeting Osteoporosis & Osteoarthritis. Semin Cell Dev Biol 2022; 123:22-35. [PMID: 34489173 PMCID: PMC8840966 DOI: 10.1016/j.semcdb.2021.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022]
Abstract
Aging induces alterations in bone structure and strength through a multitude of processes, exacerbating common aging- related diseases like osteoporosis and osteoarthritis. Cellular hallmarks of aging are examined, as related to bone and the marrow microenvironment, and ways in which these might contribute to a variety of age-related perturbations in osteoblasts, osteocytes, marrow adipocytes, chondrocytes, osteoclasts, and their respective progenitors. Cellular senescence, stem cell exhaustion, mitochondrial dysfunction, epigenetic and intracellular communication changes are central pathways and recognized as associated and potentially causal in aging. We focus on these in musculoskeletal system and highlight knowledge gaps in the literature regarding cellular and tissue crosstalk in bone, cartilage, and the bone marrow niche. While senolytics have been utilized to target aging pathways, here we propose non-pharmacologic, exercise-based interventions as prospective "senolytics" against aging effects on the skeleton. Increased bone mass and delayed onset or progression of osteoporosis and osteoarthritis are some of the recognized benefits of regular exercise across the lifespan. Further investigation is needed to delineate how cellular indicators of aging manifest in bone and the marrow niche and how altered cellular and tissue crosstalk impact disease progression, as well as consideration of exercise as a therapeutic modality, as a means to enhance discovery of bone-targeted therapies.
Collapse
Affiliation(s)
- SE Little-Letsinger
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill
| | - J Rubin
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,North Carolina Diabetes Research Center (NCDRC), University of North Carolina at Chapel Hill,Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill
| | - B Diekman
- Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill,Joint Departments of Biomedical Engineering NC State & University of North Carolina at Chapel Hill
| | - CT Rubin
- Department of Biomedical Engineering, State University of New York at Stony Brook
| | - C McGrath
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill
| | - GM Pagnotti
- Dept of Endocrine, Neoplasia, and Hormonal Disorders, University Texas MD Anderson Cancer Center, Houston
| | - EL Klett
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,Department of Nutrition, School of Public Health, University of North Carolina at Chapel Hill
| | - M Styner
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,North Carolina Diabetes Research Center (NCDRC), University of North Carolina at Chapel Hill,Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill
| |
Collapse
|
49
|
Wang S, Mi R, Cai Z, Wang Z, Zeng C, Xie Z, Li J, Ma M, Liu W, Su H, Cen S, Wu Y, Shen H. DAPK1 Interacts with the p38 isoform MAPK14, Preventing its Nuclear Translocation and Stimulation of Bone Marrow Adipogenesis. Stem Cells 2022; 40:508-522. [PMID: 35403694 DOI: 10.1093/stmcls/sxac013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/04/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Bone marrow (BM) adipose tissue (BMAT), a unique adipose depot, plays an important role in diseases such as osteoporosis and bone metastasis. Precise control of mesenchymal stem cell (MSC) differentiation is critical for BMAT formation and regeneration. Here, we show that death associated protein kinase 1 (DAPK1) negatively regulates BM adipogenesis in vitro and in vivo. Prx1 creDapk1 loxp/loxp mice showed more adipocytes in the femur than Dapk1 loxp/loxp mice. Further mechanistic analyses revealed that DAPK1 inhibits p38 mitogen-activated protein kinase (MAPK) signaling in the nucleus by binding the p38 isoform MAPK14, decreasing p38 nuclear activity, which subsequently inhibits BM adipogenesis. The inhibitory effect of DAPK1 against MAPK14 was independent of its kinase activity. In addition, the decreased DAPK1 was observed in the BM-MSCs of ageing mice. Our results reveal a previously undescribed function for DAPK1 in the regulation of adipogenesis, and may also reveal the underlying mechanism of BMAT formation in ageing.
Collapse
Affiliation(s)
- Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Rujia Mi
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Zhaopeng Cai
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Zhongyu Xie
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Jinteng Li
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Mengjun Ma
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Wenjie Liu
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Hongjun Su
- Center for Biotherapy, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Shuizhong Cen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| |
Collapse
|
50
|
Tozzi R, Masi D, Cipriani F, Contini S, Gangitano E, Spoltore ME, Barchetta I, Basciani S, Watanabe M, Baldini E, Ulisse S, Lubrano C, Gnessi L, Mariani S. Circulating SIRT1 and Sclerostin Correlates with Bone Status in Young Women with Different Degrees of Adiposity. Nutrients 2022; 14:nu14050983. [PMID: 35267956 PMCID: PMC8912833 DOI: 10.3390/nu14050983] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/28/2022] Open
Abstract
Sirtuin1 (SIRT1) and sclerostin play important roles in adipose tissue and bone metabolism. We evaluated the circulating SIRT1 and sclerostin relationship with mass and quality of bone while considering the degree of adiposity. Sixty-six premenopausal women (16 underweight, 25 normal weight and 25 with obesity), aged <50 years, were enrolled. Plasma SIRT1, sclerostin and DXA body composition (total fat mass (FM), abdominal visceral adipose tissue, lean mass, trabecular bone score (TBS) and lumbar spine and femoral neck (FN) bone mineral density (BMD)) were assessed. The patients with obesity showed the lowest SIRT1 and TBS values and the highest sclerostin concentrations; BMD increased with FM and BMI and had an inverse association with SIRT1. Sclerostin was negatively correlated with SIRT1 (ρ = −0.37, p = 0.002). When spine BMD, FN BMD and TBS were standardized for BMI, a positive correlation with SIRT1 and a negative correlation with sclerostin were seen (p < 0.005). In the regression analysis, sclerostin was the best independent, negative predictor for BMD and TBS, while SIRT1 directly predicted TBS (p < 0.05). In conclusion, blood measurement of SIRT1 and sclerostin could represent a snapshot of the bone status that, taking into account the degree of adiposity, may reduce the interference of confounding factors in the interpretation of bone health parameters.
Collapse
Affiliation(s)
- Rossella Tozzi
- Department of Molecular Medicine, “Sapienza” University of Rome, 00161 Rome, Italy;
| | - Davide Masi
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Fiammetta Cipriani
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Savina Contini
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Elena Gangitano
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Maria Elena Spoltore
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Ilaria Barchetta
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Sabrina Basciani
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Mikiko Watanabe
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Enke Baldini
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.U.)
| | - Salvatore Ulisse
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy; (E.B.); (S.U.)
| | - Carla Lubrano
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Lucio Gnessi
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
| | - Stefania Mariani
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, 00161 Rome, Italy; (D.M.); (F.C.); (S.C.); (E.G.); (M.E.S.); (I.B.); (S.B.); (M.W.); (C.L.); (L.G.)
- Correspondence: ; Tel.: +39-06499-70721; Fax: +39-06446-1450
| |
Collapse
|