1
|
Hu Y, He J, Ma Y, Ge L, Lou B, Fang X, Wang H, Xu Y. Arsenic and metabolic diseases: New insights from mesenchymal stem cells. Toxicol Appl Pharmacol 2025; 498:117299. [PMID: 40081540 DOI: 10.1016/j.taap.2025.117299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/27/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Arsenic is a common toxic metal contaminant in the environment. Humans are exposed to arsenic through drinking water, air, food, and medical treatment. Chronic exposure to arsenic is a well-documented risk factor of type 2 diabetes and a potential risk factor of osteoporosis and obesity. Mesenchymal stem cells (MSCs) are adult stem cells with multiple differentiation potential and immunomodulatory capacity. These cells have shown therapeutic potential in experimental studies of metabolic diseases by differentiating into parenchymal cells of damaged tissues, such as islet-like cells and osteoblasts, and resisting chronic inflammation. Meanwhile, when key functional genes were suppressed in MSCs, experimental animals showed metabolic disease-related changes, such as insulin resistance and obesity. Arsenic exposure inhibits the differentiation capacity of MSCs, leads to changes in the synthesis and secretion of immunomodulatory factors, and induces cellular senescence and apoptosis. Therefore, dysfunction and death of MSCs may be important pathogenesis of arsenic-related metabolic diseases. Future studies on the functional changes of MSCs in arsenic-related metabolic diseases and the role of MSCs in arsenic pathogenesis are worthwhile. In addition, the mechanism of arsenic-induced dysfunction in MSCs needs to be explored in depth.
Collapse
Affiliation(s)
- Yuxin Hu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, People's Republic of China; School of Public Health, China Medical University, Shenyang, People's Republic of China; Key Laboratory of Toxic and Biological Effects of Arsenic (China Medical University), Shenyang, Liaoning Province, People's Republic of China
| | - Jialin He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, People's Republic of China; School of Public Health, China Medical University, Shenyang, People's Republic of China; Key Laboratory of Toxic and Biological Effects of Arsenic (China Medical University), Shenyang, Liaoning Province, People's Republic of China
| | - Yue Ma
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, People's Republic of China; School of Public Health, China Medical University, Shenyang, People's Republic of China; Key Laboratory of Toxic and Biological Effects of Arsenic (China Medical University), Shenyang, Liaoning Province, People's Republic of China
| | - Lili Ge
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, People's Republic of China; School of Public Health, China Medical University, Shenyang, People's Republic of China; Key Laboratory of Toxic and Biological Effects of Arsenic (China Medical University), Shenyang, Liaoning Province, People's Republic of China
| | - Bin Lou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, People's Republic of China; School of Public Health, China Medical University, Shenyang, People's Republic of China; Key Laboratory of Toxic and Biological Effects of Arsenic (China Medical University), Shenyang, Liaoning Province, People's Republic of China
| | - Xin Fang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, People's Republic of China; School of Public Health, China Medical University, Shenyang, People's Republic of China; Key Laboratory of Toxic and Biological Effects of Arsenic (China Medical University), Shenyang, Liaoning Province, People's Republic of China
| | - Huihui Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, People's Republic of China; School of Public Health, China Medical University, Shenyang, People's Republic of China; Key Laboratory of Toxic and Biological Effects of Arsenic (China Medical University), Shenyang, Liaoning Province, People's Republic of China
| | - Yuanyuan Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, People's Republic of China; School of Public Health, China Medical University, Shenyang, People's Republic of China; Key Laboratory of Toxic and Biological Effects of Arsenic (China Medical University), Shenyang, Liaoning Province, People's Republic of China.
| |
Collapse
|
2
|
Yang G, Tan X, Zhai Q, Wang Y, Zhang X, Zhao P, Liang F, Lu J, Bao L. Plasma Lipidomics, Gut Microbiota Profile, and Phenotype of Adipose Tissue in an ApoE-/- Mouse Model of Plaque Instability. FRONT BIOSCI-LANDMRK 2025; 30:27236. [PMID: 40152393 DOI: 10.31083/fbl27236] [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: 10/30/2024] [Revised: 01/12/2025] [Accepted: 02/08/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND An appropriate animal model that can simulate the pathological process of atherosclerosis is urgently needed to improve treatment strategies. This study aimed to develop a new atherosclerosis model using ApoE-/- mice and to characterize lipidomics, gut microbiota profiles, and phenotypic alterations in adipose tissue using this model. METHODS After a 14- or 18-week high-fat diet (HFD), male ApoE-/- mice were randomly divided into four groups and treated separately with or without short-term and strong co-stimulation, including ice water bath and intraperitoneal injection of lipopolysaccharide and phenylephrine. As a control group, C57BL/6 mice were fed with conventional chow. The serum lipid levels, aortic arch pathology, adipose tissue phenotypic changes, plasma lipidomics, and 16S rDNA gene sequencing of colon feces were investigated. RESULTS The serum lipid levels were significantly lowered following extended HFD feeding for four weeks. However, co-stimulation increased serum interleukin (IL)-1β levels but did not affect serum lipid profiles. Co-stimulation revealed typical vulnerable atherosclerotic plaque characteristics and defective adipose hypertrophy associated with peroxisome proliferator-activated receptor γ (PPARγ) regulation in adipose tissue and a reduction in mitochondrial uncoupling protein 1 (UCP1) within brown adipose tissue. Plasma lipidomic analysis showed that sphingomyelin (SM), ceramide (Cer), and monohexosylceramide (HexCer) levels in plasma were significantly elevated by HFD feeding, whereas co-stimulation further elevated HexCer levels. Additionally, glycerophosphocholines (16:0/16:0, 18:2/20:4, 18:1/18:1) and HexCer (C12:1, C16:0), Cer (d18:1/16:0), and SM (C16:0) were the most sensitive to co-stimulation. Combined co-stimulation and HFD-fed increased the abundance of Firmicutes, the abundance of f_Erysipelotrichaceae, and the Firmicutes/Bacteroidota ratio but decreased the abundance of microflora promoting bile acid metabolism and short-chain fatty acids (SCFAs) in mouse feces. The results were consistent with the findings of epidemiologic atherosclerotic cardiovascular disease studies. CONCLUSIONS This study established an ApoE-/- mouse atherosclerotic vulnerable plaque model using a multi-index evaluation method. Adipogenic disorders, dysregulation of lipid metabolism at the molecular level, and increasing harmful gut microbiota are significant risk factors for vulnerable plaques, with sphingolipid metabolism receiving the most attention.
Collapse
Affiliation(s)
- Guanlin Yang
- Laboratory of Pharmacology, Zaozhuang Thoracic Hospital, 277500 Zaozhuang, Shandong, China
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Xin Tan
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Qiong Zhai
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Yuewu Wang
- School of Pharmacy, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Xuan Zhang
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Pengwei Zhao
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Fangyuan Liang
- School of Pharmacy, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Jingkun Lu
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - LiLi Bao
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| |
Collapse
|
3
|
Li P, Wang Y, Liang Y, Jiang X, Tang T, Fan X, Wang R, Yang M, Liu Y, Qi K, Zhang Y. Imbalance of early-life vitamin D intake targets ROS-mediated crosstalk between mitochondrial dysfunction and differentiation potential of MSCs associated the later obesity. Stem Cell Res Ther 2024; 15:252. [PMID: 39135105 PMCID: PMC11321190 DOI: 10.1186/s13287-024-03860-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/24/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Obesity is characterized by excessive fat accumulation, which is related with abnormal pluripotency of mesenchymal stem cells (MSCs). Recently, there is growing evidence that the disorder of maternal vitamin D (VD) intake is a well-known risk factor for long-term adverse health outcomes to their offspring. Otherwise, less is known of its repercussion and underlying mechanisms on the different differentiation potential of MSCs. METHODS Four-week-old female C57BL/6J mice were fed with different VD reproductive diets throughout the whole pregnancy and lactation. The characteristics of BMSCs from their seven-day male offspring, VDR knockdown establishment of HuMSCs and HuMSCs under the different VD interventions in vitro were confirmed by flow cytometry, RT-PCR, and immunofluorescence. The roles of VD on their mitochondrial dysfunction and differentiation potential were also investigated. Then their remaining weaned male pups were induced by administrating high-fat-diet (HFD) for 16 weeks and normal fat diet was simultaneously as controls. Their lipid accumulation and adipocytes hypertrophy were determined by histological staining and related gene expressions. RESULTS Herein, it was proved that imbalance of early-life VD intake could significantly aggravate the occurrence of obesity by inducing the adipogenesis through affecting the VD metabolism and related metabolites (P < 0.05). Moreover, abnormally maternal VD intake might be involved on the disorders of differentiation potential to inhibit the maintenance of MSCs stemness through increasing the productions of ROS, which was accompanied by impairing the expression of related genes on the adipo-osteogenic differentiation (P < 0.05). Moreover, it was along with increasing potential of adipogenic differentiation of MSCs as higher ROS in the state of VD deficiency, while excessive maternal VD status could conversely enhance the osteogenic differentiation with slightly lower ROS (P < 0.05). Furthermore, the underlying mechanisms might be involved on the mitochondria dysfunctional, especially the mitophagy, by activating the LC3b, P62 and etc. using in vivo and in vitro studies (P < 0.05). CONCLUSION These findings demonstrated that imbalance of early-life VD intake could target ROS-mediated crosstalk between mitochondrial dysfunction and differentiation potential of MSCs, which was significantly associated with the later obesity. Obviously, our results could open up an attractive modality for the benefits of suitable VD intake during the pregnancy and lactation.
Collapse
Affiliation(s)
- Ping Li
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children's Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-Li-Shi Road, Beijing, 100045, China.
| | - Yang Wang
- Department of Stem Cell & Regeneration Medicine, Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, No.27 Tai-Ping Road, Beijing, 100091, China
| | - Yueqing Liang
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children's Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-Li-Shi Road, Beijing, 100045, China
| | - Xueyi Jiang
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children's Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-Li-Shi Road, Beijing, 100045, China
| | - Tiantian Tang
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children's Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-Li-Shi Road, Beijing, 100045, China
| | - Xiuqin Fan
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children's Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-Li-Shi Road, Beijing, 100045, China
| | - Rui Wang
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children's Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-Li-Shi Road, Beijing, 100045, China
| | - Mengyi Yang
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children's Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-Li-Shi Road, Beijing, 100045, China
| | - Yuanlin Liu
- Department of Stem Cell & Regeneration Medicine, Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, No.27 Tai-Ping Road, Beijing, 100091, China
| | - Kemin Qi
- Laboratory of Nutrition and Development, Key Laboratory of Major Diseases in Children's Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No.56 Nan-Li-Shi Road, Beijing, 100045, China.
| | - Yi Zhang
- Department of Stem Cell & Regeneration Medicine, Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, No.27 Tai-Ping Road, Beijing, 100091, China.
| |
Collapse
|
4
|
Kim HY, Jang HJ, Muthamil S, Shin UC, Lyu JH, Kim SW, Go Y, Park SH, Lee HG, Park JH. Novel insights into regulators and functional modulators of adipogenesis. Biomed Pharmacother 2024; 177:117073. [PMID: 38981239 DOI: 10.1016/j.biopha.2024.117073] [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: 04/15/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
Adipogenesis is a process that differentiates new adipocytes from precursor cells and is tightly regulated by several factors, including many transcription factors and various post-translational modifications. Recently, new roles of adipogenesis have been suggested in various diseases. However, the molecular mechanisms and functional modulation of these adipogenic genes remain poorly understood. This review summarizes the regulatory factors and modulators of adipogenesis and discusses future research directions to identify novel mechanisms regulating adipogenesis and the effects of adipogenic regulators in pathological conditions. The master adipogenic transcriptional factors PPARγ and C/EBPα were identified along with other crucial regulatory factors such as SREBP, Kroxs, STAT5, Wnt, FOXO1, SWI/SNF, KLFs, and PARPs. These transcriptional factors regulate adipogenesis through specific mechanisms, depending on the adipogenic stage. However, further studies related to the in vivo role of newly discovered adipogenic regulators and their function in various diseases are needed to develop new potent therapeutic strategies for metabolic diseases and cancer.
Collapse
Affiliation(s)
- Hyun-Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; New Drug Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea.
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Seon-Wook Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea.
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea.
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea.
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
5
|
Hamamura K, Nagao M, Furukawa K. Regulation of Glycosylation in Bone Metabolism. Int J Mol Sci 2024; 25:3568. [PMID: 38612379 PMCID: PMC11011486 DOI: 10.3390/ijms25073568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Glycosylation plays a crucial role in the maintenance of homeostasis in the body and at the onset of diseases such as inflammation, neurodegeneration, infection, diabetes, and cancer. It is also involved in bone metabolism. N- and O-glycans have been shown to regulate osteoblast and osteoclast differentiation. We recently demonstrated that ganglio-series and globo-series glycosphingolipids were essential for regulating the proliferation and differentiation of osteoblasts and osteoclasts in glycosyltransferase-knockout mice. Herein, we reviewed the importance of the regulation of bone metabolism by glycoconjugates, such as glycolipids and glycoproteins, including our recent results.
Collapse
Affiliation(s)
- Kazunori Hamamura
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya 464-8650, Japan
| | - Mayu Nagao
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya 464-8650, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Aichi, Japan
| |
Collapse
|
6
|
Qin X, Wei Q, An R, Yang Y, Cai M, Han X, Mao H, Gao X. Regulation of bone and fat balance by Fructus Ligustri Lucidi in ovariectomized mice. PHARMACEUTICAL BIOLOGY 2023; 61:391-403. [PMID: 36740874 PMCID: PMC9904306 DOI: 10.1080/13880209.2023.2168019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 12/03/2022] [Accepted: 01/09/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT Fructus Ligustri Lucidi (FLL), a commonly used herb of traditional Chinese medicine (TCM), is the fruit of Ligustrum lucidum Ait. (Oleaceae). The ethanol extract of FLL is a potential candidate for preventing and treating postmenopausal osteoporosis (PMOP) by nourishing the liver and kidneys. OBJECTIVE This study determines whether an ethanol extract of FLL has anti-osteoporotic effects in ovariectomized (OVX) mice and explores the underlying mechanism. MATERIALS AND METHODS The OVX model of eight-week-old C57BL/6J female mice was taken, and ovariectomy was used as PMOP. Mice were divided into five groups: sham-operated group (n = 10), OVX group (n = 10), OVX + E2 group (n = 10; 0.039 mg/kg), OVX + FLL group (n = 10; 2 g/kg) and OVX + FLL group (n = 10; 4 g/kg). Mice were treated by gavage with FLL or CMCNa once daily for 8 weeks. We harvested uteri, femur, and tibias from mice; bone mineral density (BMD) and bone microstructure were obtained by X-ray absorptiometry and micro-CT. Furthermore, the effect of FLL on the balance of osteoblast and adipocyte differentiation was investigated using bone marrow mesenchymal stem cells (BMMSCs). RESULTS The results indicated that FLL did not affect OVX-induced estradiol reduction. Compared with OVX mice, FLL significantly increased BMD (63.54 vs. 61.96), Conn. D (86.46 vs. 57.00), and left tibial strength (13.91 vs. 11.27), decreased Tb. Sp (0.38 vs. 0.44) and body fat content (4.19% vs. 11.24%). FLL decreased osteoclast activity and enhanced RUNX2 expression; inhibited perilipin peroxisome proliferator-activated receptor gamma (PPARγ) expression and adipocyte differentiation from BMMSCs. CONCLUSIONS FLL prevented additional bone loss and improved bone microstructure in OVX mice by modulating bone and fat balance, suggesting that FLL might be a therapeutic agent for PMOP.
Collapse
Affiliation(s)
- Xiaoyan Qin
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiu Wei
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ran An
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yun Yang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mingqi Cai
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoling Han
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haoping Mao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Jang HJ, Lee YH, Dao T, Jo Y, Khim KW, Eom HJ, Lee JE, Song YJ, Choi SS, Park K, Ji H, Chae YC, Myung K, Kim H, Ryu D, Park NH, Park SH, Choi JH. Thrap3 promotes nonalcoholic fatty liver disease by suppressing AMPK-mediated autophagy. Exp Mol Med 2023; 55:1720-1733. [PMID: 37524868 PMCID: PMC10474030 DOI: 10.1038/s12276-023-01047-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/14/2023] [Accepted: 04/30/2023] [Indexed: 08/02/2023] Open
Abstract
Autophagy functions in cellular quality control and metabolic regulation. Dysregulation of autophagy is one of the major pathogenic factors contributing to the progression of nonalcoholic fatty liver disease (NAFLD). Autophagy is involved in the breakdown of intracellular lipids and the maintenance of healthy mitochondria in NAFLD. However, the mechanisms underlying autophagy dysregulation in NAFLD remain unclear. Here, we demonstrate that the hepatic expression level of Thrap3 was significantly increased in NAFLD conditions. Liver-specific Thrap3 knockout improved lipid accumulation and metabolic properties in a high-fat diet (HFD)-induced NAFLD model. Furthermore, Thrap3 deficiency enhanced autophagy and mitochondrial function. Interestingly, Thrap3 knockout increased the cytosolic translocation of AMPK from the nucleus and enhanced its activation through physical interaction. The translocation of AMPK was regulated by direct binding with AMPK and the C-terminal domain of Thrap3. Our results indicate a role for Thrap3 in NAFLD progression and suggest that Thrap3 is a potential target for NAFLD treatment.
Collapse
Affiliation(s)
- Hyun-Jun Jang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, 58245, Republic of Korea
| | - Yo Han Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Tam Dao
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, 16419, Republic of Korea
| | - Yunju Jo
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, 16419, Republic of Korea
| | - Keon Woo Khim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Hye-Jin Eom
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Ju Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yi Jin Song
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Sun Sil Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Kieun Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Haneul Ji
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Young Chan Chae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Republic of Korea
| | - Hongtae Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, 16419, Republic of Korea
| | - Neung Hwa Park
- Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan, 44033, Republic of Korea.
| | - Sung Ho Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Jang Hyun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
8
|
DeVeaux SA, Ogle ME, Vyshnya S, Chiappa NF, Leitmann B, Rudy R, Day A, Mortensen LJ, Kurtzberg J, Roy K, Botchwey EA. Characterizing human mesenchymal stromal cells' immune-modulatory potency using targeted lipidomic profiling of sphingolipids. Cytotherapy 2022; 24:608-618. [PMID: 35190267 PMCID: PMC10725732 DOI: 10.1016/j.jcyt.2021.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022]
Abstract
Cell therapies are expected to increase over the next decade owing to increasing demand for clinical applications. Mesenchymal stromal cells (MSCs) have been explored to treat a number of diseases, with some successes in early clinical trials. Despite early successes, poor MSC characterization results in lessened therapeutic capacity once in vivo. Here, we characterized MSCs derived from bone marrow (BM), adipose tissue and umbilical cord tissue for sphingolipids (SLs), a class of bioactive lipids, using liquid chromatography/tandem mass spectrometry. We found that ceramide levels differed based on the donor's sex in BM-MSCs. We detected fatty acyl chain variants in MSCs from all three sources. Linear discriminant analysis revealed that MSCs separated based on tissue source. Principal component analysis showed that interferon-γ-primed and unstimulated MSCs separated according to their SL signature. Lastly, we detected higher ceramide levels in low indoleamine 2,3-dioxygenase MSCs, indicating that sphingomyelinase or ceramidase enzymatic activity may be involved in their immune potency.
Collapse
Affiliation(s)
- S’Dravious A. DeVeaux
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Molly E. Ogle
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Sofiya Vyshnya
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Nathan F. Chiappa
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Bobby Leitmann
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA
| | - Ryan Rudy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Abigail Day
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Luke J. Mortensen
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia Institute of Technology, Atlanta, GA
| | - Edward A. Botchwey
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory, Atlanta, GA
- Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
9
|
Lee YH, Jang HJ, Kim S, Choi SS, Khim KW, Eom HJ, Hyun J, Shin KJ, Chae YC, Kim H, Park J, Park NH, Woo CY, Hong CH, Koh EH, Nam D, Choi JH. Hepatic MIR20B promotes nonalcoholic fatty liver disease by suppressing PPARA. eLife 2021; 10:70472. [PMID: 34964438 PMCID: PMC8758141 DOI: 10.7554/elife.70472] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation and imbalances in lipid metabolism in the liver. Although nuclear receptors (NRs) play a crucial role in hepatic lipid metabolism, the underlying mechanisms of NR regulation in NAFLD remain largely unclear. Methods Using network analysis and RNA-seq to determine the correlation between NRs and microRNA in human NAFLD patients, we revealed that MIR20B specifically targets PPARA. MIR20B mimic and anti-MIR20B were administered to human HepG2 and Huh-7 cells and mouse primary hepatocytes as well as high fat diet (HFD)- or methionine-deficient diet (MCD)-fed mice to verify the specific function of MIR20B in NAFLD. We tested the inhibition of the therapeutic effect of a PPARα agonist, fenofibrate, by Mir20b and the synergic effect of combination of fenofibrate with anti-Mir20b in NAFLD mouse model. Results We revealed that MIR20B specifically targets PPARA through miRNA regulatory network analysis of nuclear receptor genes in NAFLD. The expression of MIR20B was upregulated in free fatty acid (FA)-treated hepatocytes and the livers of both obesity-induced mice and NAFLD patients. Overexpression of MIR20B significantly increased hepatic lipid accumulation and triglyceride levels. Furthermore, MIR20B significantly reduced FA oxidation and mitochondrial biogenesis by targeting PPARA. In Mir20b-introduced mice, the effect of fenofibrate to ameliorate hepatic steatosis was significantly suppressed. Finally, inhibition of Mir20b significantly increased FA oxidation and uptake, resulting in improved insulin sensitivity and a decrease in NAFLD progression. Moreover, combination of fenofibrate and anti-Mir20b exhibited the synergic effect on improvement of NAFLD in MCD-fed mice. Conclusions Taken together, our results demonstrate that the novel MIR20B targets PPARA, plays a significant role in hepatic lipid metabolism, and present an opportunity for the development of novel therapeutics for NAFLD. Funding This research was funded by Korea Mouse Phenotyping Project (2016M3A9D5A01952411), the National Research Foundation of Korea (NRF) grant funded by the Korea government (2020R1F1A1061267, 2018R1A5A1024340, NRF-2021R1I1A2041463, 2020R1I1A1A01074940), and the Future-leading Project Research Fund (1.210034.01) of UNIST.
Collapse
Affiliation(s)
- Yo Han Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hyun-Jun Jang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Sounkou Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Sun Sil Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Keon Woo Khim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hye-Jin Eom
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jimin Hyun
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Kyeong Jin Shin
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Young Chan Chae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hongtae Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jiyoung Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Neung Hwa Park
- Department of Internal Medicine, Ulsan University Hospital, Ulsan, Republic of Korea
| | - Chang-Yun Woo
- Department of Internal Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Chung Hwan Hong
- Department of Medical Science, Asan Medical Center, Seoul, Republic of Korea
| | - Eun Hee Koh
- Department of Internal Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Dougu Nam
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jang Hyun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
10
|
Husson H, Bukanov NO, Moreno S, Smith MM, Richards B, Zhu C, Picariello T, Park H, Wang B, Natoli TA, Smith LA, Zanotti S, Russo RJ, Madden SL, Klinger KW, Modur V, Ibraghimov-Beskrovnaya O. Correction of cilia structure and function alleviates multi-organ pathology in Bardet-Biedl syndrome mice. Hum Mol Genet 2021; 29:2508-2522. [PMID: 32620959 PMCID: PMC7471507 DOI: 10.1093/hmg/ddaa138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/22/2020] [Accepted: 07/01/2020] [Indexed: 12/21/2022] Open
Abstract
Bardet–Biedl syndrome (BBS) is a pleiotropic autosomal recessive ciliopathy affecting multiple organs. The development of potential disease-modifying therapy for BBS will require concurrent targeting of multi-systemic manifestations. Here, we show for the first time that monosialodihexosylganglioside accumulates in Bbs2−/− cilia, indicating impairment of glycosphingolipid (GSL) metabolism in BBS. Consequently, we tested whether BBS pathology in Bbs2−/− mice can be reversed by targeting the underlying ciliary defect via reduction of GSL metabolism. Inhibition of GSL synthesis with the glucosylceramide synthase inhibitor Genz-667161 decreases the obesity, liver disease, retinal degeneration and olfaction defect in Bbs2−/− mice. These effects are secondary to preservation of ciliary structure and signaling, and stimulation of cellular differentiation. In conclusion, reduction of GSL metabolism resolves the multi-organ pathology of Bbs2−/− mice by directly preserving ciliary structure and function towards a normal phenotype. Since this approach does not rely on the correction of the underlying genetic mutation, it might translate successfully as a treatment for other ciliopathies.
Collapse
Affiliation(s)
- Hervé Husson
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Nikolay O Bukanov
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Sarah Moreno
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Mandy M Smith
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | | | - Cheng Zhu
- Translational Sciences, Sanofi, Framingham, MA 01701, USA
| | - Tyler Picariello
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Hyejung Park
- Pre-Development Sciences, Sanofi, Waltham, MA 02451, USA
| | - Bing Wang
- Pre-Development Sciences, Sanofi, Waltham, MA 02451, USA
| | - Thomas A Natoli
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Laurie A Smith
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Stefano Zanotti
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Ryan J Russo
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | | | | | - Vijay Modur
- Rare Diseases Development, Sanofi, Cambridge, MA 02142, USA
| | | |
Collapse
|
11
|
Rozenfeld PA, Crivaro AN, Ormazabal M, Mucci JM, Bondar C, Delpino MV. Unraveling the mystery of Gaucher bone density pathophysiology. Mol Genet Metab 2021; 132:76-85. [PMID: 32782168 DOI: 10.1016/j.ymgme.2020.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 01/18/2023]
Abstract
Gaucher disease (GD) is caused by pathogenic mutations in GBA1, the gene that encodes the lysosomal enzyme β-glucocerebrosidase. Despite the existence of a variety of specific treatments for GD, they cannot completely reverse bone complications. Many studies have evidenced the impairment in bone tissue of GD, and molecular mechanisms of bone density alterations in GD are being studied during the last years and different reports emphasized its efforts trying to unravel why and how bone tissue is affected. The cause of skeletal density affection in GD is a matter of debates between research groups. and there are two opposing hypotheses trying to explain reduced bone mineral density in GD: increased bone resorption versus impaired bone formation. In this review, we discuss the diverse mechanisms of bone alterations implicated in GD revealed until the present, along with a presentation of normal bone physiology and its regulation. With this information in mind, we discuss effectiveness of specific therapies, introduce possible adjunctive therapies and present a novel model for GD-associated bone density pathogenesis. Under the exposed evidence, we may conclude that both sides of the balance of remodeling process are altered. In GD the observed osteopenia/osteoporosis may be the result of contribution of both reduced bone formation and increased bone resorption.
Collapse
Affiliation(s)
- P A Rozenfeld
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina.
| | - A N Crivaro
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina
| | - M Ormazabal
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina
| | - J M Mucci
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina
| | - C Bondar
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina
| | - M V Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Universidad de Buenos Aires, CONICET, Av. Córdoba 2351, (C1120ABG), Buenos Aires, Argentina
| |
Collapse
|
12
|
Molecular Mechanisms and Emerging Therapeutics for Osteoporosis. Int J Mol Sci 2020; 21:ijms21207623. [PMID: 33076329 PMCID: PMC7589419 DOI: 10.3390/ijms21207623] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 12/15/2022] Open
Abstract
Osteoporosis is the most common chronic metabolic bone disease. It has been estimated that more than 10 million people in the United States and 200 million men and women worldwide have osteoporosis. Given that the aging population is rapidly increasing in many countries, osteoporosis could become a global challenge with an impact on the quality of life of the affected individuals. Osteoporosis can be defined as a condition characterized by low bone density and increased risk of fractures due to the deterioration of the bone architecture. Thus, the major goal of treatment is to reduce the risk for fractures. There are several treatment options, mostly medications that can control disease progression in risk groups, such as postmenopausal women and elderly men. Recent studies on the basic molecular mechanisms and clinical implications of osteoporosis have identified novel therapeutic targets. Emerging therapies targeting novel disease mechanisms could provide powerful approaches for osteoporosis management in the future. Here, we review the etiology of osteoporosis and the molecular mechanism of bone remodeling, present current pharmacological options, and discuss emerging therapies targeting novel mechanisms, investigational treatments, and new promising therapeutic approaches.
Collapse
|
13
|
Mitrofanova A, Drexler Y, Merscher S, Fornoni A. Role of Sphingolipid Signaling in Glomerular Diseases: Focus on DKD and FSGS. JOURNAL OF CELLULAR SIGNALING 2020; 1:56-69. [PMID: 32914148 PMCID: PMC7480905 DOI: 10.33696/signaling.1.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sphingolipids are well-recognized as major players in the pathogenesis of many human diseases, including chronic kidney disease. The kidney is a very sensitive organ to alterations in sphingolipid metabolism. The critical issues to be addressed in this review relate to the role of sphingolipids and enzymes involved in sphingolipid metabolism in the pathogenesis of glomerular diseases with a special focus on podocytes, a key cellular component of the glomerular filtration barrier. Among several sphingolipids, we will highlight the role of ceramide, sphingosine, sphingosine-1-phosphate and ceramide-1-phosphate. Additionally, we will summarize the current knowledge with regard to the use of sphingolipids as therapeutic agents for the treatment of podocyte injury in kidney disease.
Collapse
Affiliation(s)
- Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Yelena Drexler
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
14
|
Crivaro A, Bondar C, Mucci JM, Ormazabal M, Feldman RA, Delpino MV, Rozenfeld PA. Gaucher disease-associated alterations in mesenchymal stem cells reduce osteogenesis and favour adipogenesis processes with concomitant increased osteoclastogenesis. Mol Genet Metab 2020; 130:274-282. [PMID: 32536424 DOI: 10.1016/j.ymgme.2020.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/02/2020] [Accepted: 06/02/2020] [Indexed: 01/18/2023]
Abstract
Gaucher disease (GD) is caused by pathogenic mutations in GBA1, the gene that encodes the lysosomal enzyme β-glucocerebrosidase. Until now, treatments for GD cannot completely reverse bone problems. The aim of this work was to evaluate the potential of MSCs from GD patients (GD MSCs) to differentiate towards the osteoblast (GD Ob) and adipocyte (GD Ad) lineages, and their role in osteoclastogenesis. We observed that GD Ob exhibited reduced mineralization, collagen deposition and alkaline phosphatase activity (ALP), as well as decreased gene expression of RUNX2, COLA1 and ALP. We also evaluated the process of osteoclastogenesis and observed that conditioned media from GD MSCs supernatants induced an increase in the number of osteoclasts. In this model, osteoclastogenesis was induced by RANKL and IL-1β. Furthermore, results showed that in GD MSCs there was a promotion in NLRP3 and PPAR-γ gene expression. Adipogenic differentiation revealed that GD Ad had an increase in PPAR-γ and a reduced RUNX2 gene expression, promoting adipocyte differentiation. In conclusion, our results show that GD MSCs exhibited deficient GD Ob differentiation and increased adipogenesis. In addition, we show that GD MSCs promoted increased osteoclastogenesis through RANKL and IL-1β. These changes in GD MSCs are likely to contribute to skeletal imbalance observed in GD patients.
Collapse
Affiliation(s)
- A Crivaro
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina
| | - C Bondar
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina
| | - J M Mucci
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina
| | - M Ormazabal
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina
| | - R A Feldman
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Hospital de Clínicas "José de San Martín", Facultad de Medicina, CONICET-Universidad de Buenos Aires, Paraguay 2155, (C1121ABG), Buenos Aires, Argentina
| | - M V Delpino
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - P A Rozenfeld
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Universidad Nacional de La Plata, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, Bv. 120 N(o)1489 (1900), La Plata, Argentina.
| |
Collapse
|