1
|
Herichová I, Reis R, Vanátová D. Differences in the role of Gper1 in colorectal cancer progression depending on sex. Oncol Lett 2025; 29:305. [PMID: 40321663 PMCID: PMC12046377 DOI: 10.3892/ol.2025.15051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/13/2025] [Indexed: 05/08/2025] Open
Abstract
To evaluate the role of 17β-oestradiol (E2) in the sex-dependent progression of colorectal cancer (CRC), the present study focused on E2 signalling mediated via the nuclear receptors [oestrogen receptor (ESR)1 and ESR2] and the membrane G protein-coupled oestrogen receptor 1 (Gper1) in males and females diagnosed with CRC. This study also investigated Gper1 signalling in the CRC cell lines DLD1 and LoVo, which differ in the p53 pathway. In cancer tissue, Gper1 becomes by far the most abundant E2 receptor due to an increase in Gper1 and a decrease in ESR2 expression. These changes are more prominent in males than in females. More pronounced differences in Gper1 expression between cancer and adjacent tissues were observed in males in lower stages compared with those in higher stages of disease and females. High expression of Gper1 was associated with worse survival in males without nodal involvement but not in females. The expression of E2 receptors in the CRC cell lines DLD1 and LoVo resembles that of human cancer tissue. Silencing of Gper1 (siGper1) caused an increase in the rate of metabolism in LoVo cells with wild-type tp53. In DLD1 cells with the mutated form of tp53, siGper1 did not exert this effect. High levels of Gper1 were associated with worse survival and could contribute to sex-dependent changes in the CRC prognosis. Tumour suppressor effects of Gper1 were, at least to some extent, dependent on signalling downstream of p53, which was more frequently deficient in males than in females. Overall, this suggests that up-regulation of Gper1 (or administration of a Gper1 agonist) would be more beneficial for patients with wild-type tp53.
Collapse
Affiliation(s)
- Iveta Herichová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215 Bratislava, Slovak Republic
| | - Richard Reis
- First Surgery Department, University Hospital, Comenius University in Bratislava, 81107 Bratislava, Slovak Republic
| | - Denisa Vanátová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215 Bratislava, Slovak Republic
| |
Collapse
|
2
|
Chuang SC, Chou YS, Lin YS, Chang JK, Chen CH, Ho ML. Cyclooxygenase-2 negatively regulates osteogenic differentiation in murine bone marrow mesenchymal stem cells via the FOXO3a/p27kip1 pathway. Bone Joint Res 2025; 14:407-419. [PMID: 40335058 PMCID: PMC12058311 DOI: 10.1302/2046-3758.145.bjr-2024-0262.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/09/2025] Open
Abstract
Aims Cyclooxygenase-2 (COX-2) is an enzyme that synthesizes prostaglandins from arachidonic acid. Previous reports have indicated that COX-2 is constitutively expressed in osteogenic cells instead of being expressed only after pathogenic induction, and that it facilitates osteoblast proliferation via PTEN/Akt/p27kip1 signalling. However, the role of COX-2 in osteogenic differentiation of murine bone marrow mesenchymal stromal cells (BMSCs) remains controversial. In this study, we investigated the function of COX-2 in the osteogenic differentiation of BMSCs. Methods COX-2 inhibitor, COX-2 overexpression vector, and p27kip1 small interfering RNA (siRNA) were used to evaluate the role of COX-2 in osteogenic differentiation and related signalling pathways in BMSCs. Results We found that the messenger RNA (mRNA) and protein levels of COX-2 decreased gradually during osteogenic differentiation. Inhibition of COX-2 activity promoted FOXO3a and p27kip1 expression and simultaneously enhanced osteogenesis, as indicated by increased osteogenic gene expression and mineralization in BMSCs. Furthermore, when p27kip1 was silenced, the suppressive effects of COX-2 on osteogenesis were reversed. It demonstrated that the negative regulatory effect of COX-2 on osteogenesis was mediated by p27kip1. In addition, our results showed that overexpression of COX-2 reduced the mRNA and protein levels of FOXO3a and p27kip1, and thus attenuated osteogenic gene expression. These results indicate that COX-2 negatively regulates osteogenic differentiation by reducing the expression of osteogenic genes via the FOXO3a/p27kip1 signalling pathway. Conclusion Together with the findings from previous and current studies, these results indicate that COX-2 has a different role in proliferation versus differentiation during osteogenesis via FOXO3a/p27kip1 signalling in osteoblasts or BMSCs.
Collapse
Affiliation(s)
- Shu-Chun Chuang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ya-Shuan Chou
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Je-Ken Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopaedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopaedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- PhD Program in Biomedical Engineering, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Materials Engineering, College of Engineering, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Mei-Ling Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physiology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
3
|
Ning Q, Li M, Liao Z, Chen E, Liu H, Liang Y, Chen Y, Li Y, Huang L. LncRNA MRF targeting FSHR inhibits the osteogenic differentiation of BMSCs and bone defect repair through the regulation of the cAMP-PKA-CREB signaling pathway. Stem Cell Res Ther 2025; 16:200. [PMID: 40264197 PMCID: PMC12016372 DOI: 10.1186/s13287-025-04291-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs), known for their ability to differentiate into osteoblasts, play a pivotal role in bone metabolism. In our previous investigations, we identified a novel long non-coding RNA (lncRNA) named MCP1 Regulatory Factor (MRF), which exhibits significant involvement in immune regulation of BMSCs. Moreover, we observed noticeable expression changes of MRF during the osteogenic differentiation of BMSCs. However, the exact role and underlying mechanism of MRF in the osteogenic differentiation of BMSCs remain elusive. METHODS QRT-PCR analysis was employed to assess the expression levels of MRF. RNA interference and overexpression plasmids were utilized to modulate MRF expression, allowing for the observation of changes in the osteogenic differentiation capacity of BMSCs. Downstream pathways involved in the MRF-mediated regulation of BMSCs' osteogenic differentiation were predicted using transcriptome sequencing. The functionality of MRF in vivo was validated through a mouse tibial drilling defect model. RESULTS In patients with osteoporosis, there is a notable increase in the expression of MRF within BMSCs. During the osteogenic differentiation of BMSCs, the MRF expression progressively decreases. The knockdown of MRF significantly enhances the osteogenic differentiation of BMSCs, promoting an increased expression of bone-related proteins such as RUNX2, ALP, and COL1A1. Transcriptome sequencing and western blot indicated that cAMP/PKA/CREB signaling pathway was significantly activated after lncRNA-MRF knockdown. Moreover, in the mouse tibial drilling defect model, MRF knockdown significantly promotes ossification in vivo. CONCLUSIONS MRF modulates the cAMP/PKA/CREB signaling pathway via the follicle stimulating hormone receptor (FSHR), thereby influencing the ossification differentiation of BMSCs. Our research suggests that MRF may serve as a potential target for bone-related disorders.
Collapse
Affiliation(s)
- Qing Ning
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Ming Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Zhuangyao Liao
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Enming Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Huatao Liu
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Yuwei Liang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Yuanquan Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Yuxi Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
| | - Lin Huang
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
4
|
Caviness PC, Lazarenko OP, Blackburn ML, Chen JR. Sex-dependent effect of GPR109A gene deletion in myeloid cells on bone development in mice. RESEARCH SQUARE 2025:rs.3.rs-6206075. [PMID: 40235504 PMCID: PMC11998753 DOI: 10.21203/rs.3.rs-6206075/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Blueberry metabolite-derived phenolic acids are thought to suppress bone resorption via interactions with the G protein-coupled receptor 109A (GPR109A). Previously, global GPR109A knockout (GPR109A ⁻/⁻ ) mice exhibited increased bone mass and a diminished bone-protective response to phenolic acids. While GPR109A is highly expressed in osteoclast precursor macrophages, its role in bone development remains unclear. To address this, we generated a myeloid cell-specific GPR109A knockout (GPR109A flox/flox /LysM-Cre⁺; CKO) mouse model and assessed bone phenotypes in male and female mice at 35 days, 3 months, 6 months, and 12 months using µCT. At 35 days, CKO males showed significantly improved tibia and vertebrae µCT parameters compared to controls (f/f, Cre⁺). However, at later time points (6 and 12 months), Cre recombinase effects were observed, with Cre⁺ males exhibiting similar bone parameters to CKO mice. In contrast, female CKO mice displayed significantly improved µCT parameters at 6 and 12 months. Notably, 12-month-old Cre⁺ males exhibited altered bone mechanical properties, while females did not. Gene expression analysis revealed increased Interferon regulatory factor 8 (Irf8), an osteoclastogenesis suppressor, in female CKO mice. These findings suggest that GPR109A regulates bone resorption through osteoclastogenic pathways in a sex-specific manner.
Collapse
|
5
|
Zhang XH, Li JX, Wu XX, Zhang Q, Tian M, Yang SQ, Liu D, Yang XQ. PABPN1 functions as a downstream gene of CREB to inhibit the proliferation of preadipocytes. Anim Biosci 2025; 38:41-53. [PMID: 39210800 PMCID: PMC11725739 DOI: 10.5713/ab.24.0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/03/2023] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE This study was conducted to reveal the role of nuclear poly(A) binding protein 1 (PABPN1) in the proliferation of preadipocytes, and to reveal the relationship between PABPN1 and cAMP response element (CRE)-binding protein (CREB) in the regulation of preadipocyte proliferation. METHODS Vectors overexpressing and siRNAs against PABPN1/CREB were transiently transfected into both porcine preadipocytes and mouse 3T3-L1 cells. Preadipocyte proliferation was measured with cell counting kit-8, 5-ethynyl-2'-deoxyuridine, real-time quantitative polymerase chain reaction, Western blotting, and flow cytometry analyses. Additionally, the transcriptional regulation of CREB on PABPN1 were analyzed with dual-luciferase reporter gene and electrophoretic mobility shift assay. RESULTS Overexpression of PABPN1 inhibits, and knockdown of PABPN1 promotes, the proliferation of both porcine preadipocytes and 3T3-L1 cell lines. PABPN1 overexpression increased, while knockdown decreased, the cell population in the G0/G1 phase. These indicates that PABPN1 repressed preadipocyte proliferation by inhibiting cell cycle progress. Additionally, it was revealed that CREB regulated the expression of PABPN1 through binding to the promoter and that CREB inhibited preadipocyte proliferation by repressed cell cycle progress. Furthermore, we showed that PABPN1 functions as a downstream gene of CREB to regulate the proliferation of preadipocytes. CONCLUSION PABPN1 inhibits preadipocyte proliferation by suppressing the cell cycle. We also found that CREB could promote PABPN1 expression by binding to a motif in the promoter. Further analysis confirmed that PABPN1 functions as a downstream gene of CREB to regulate the proliferation of preadipocytes. These results suggest that the CREB/PABPN1 axis plays a role in the regulation of preadipocyte proliferation, which will contribute to further revealing the mechanism of fat accumulation.
Collapse
Affiliation(s)
- Xiao-Han Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Jia-Xin Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Xiao-Xu Wu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Qian Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Ming Tian
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086,
China
| | - Si-Qi Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086,
China
| | - Xiu-Qin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| |
Collapse
|
6
|
Lu L, Zhang Q, Aladelokun O, Berardi D, Shen X, Marin A, Garcia-Milian R, Roper J, Khan SA, Johnson CH. Asparagine synthetase and G-protein coupled estrogen receptor are critical responders to nutrient supply in KRAS mutant colorectal cancer. Int J Cancer 2025; 156:52-68. [PMID: 39039782 PMCID: PMC11537827 DOI: 10.1002/ijc.35104] [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: 06/13/2023] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/24/2024]
Abstract
Survival differences exist in colorectal cancer (CRC) patients by sex and disease stage. However, the potential molecular mechanism(s) are not well understood. Here we show that asparagine synthetase (ASNS) and G protein-coupled estrogen receptor-1 (GPER1) are critical sensors of nutrient depletion and linked to poorer outcomes for females with CRC. Using a 3D spheroid model of isogenic SW48 KRAS wild-type (WT) and G12A mutant (MT) cells grown under a restricted nutrient supply, we found that glutamine depletion inhibited cell growth in both cell lines, whereas ASNS and GPER1 expression were upregulated in KRAS MT versus WT. Estradiol decreased growth in KRAS WT but had no effect on MT cells. Selective GPER1 and ASNS inhibitors suppressed cell proliferation with increased caspase-3 activity of MT cells under glutamine depletion condition particularly in the presence of estradiol. In a clinical colon cancer cohort from The Cancer Genome Atlas, both high GPER1 and ASNS expression were associated with poorer overall survival for females only in advanced stage tumors. These results suggest KRAS MT cells have mechanisms in place that respond to decreased nutrient supply, typically observed in advanced tumors, by increasing the expression of ASNS and GPER1 to drive cell growth. Furthermore, KRAS MT cells are resistant to the protective effects of estradiol under nutrient deplete conditions. The findings indicate that GPER1 and ASNS expression, along with the interaction between nutrient supply and KRAS mutations shed additional light on the mechanisms underlying sex differences in metabolism and growth in CRC, and have clinical implications in the precision management of KRAS mutant CRC.
Collapse
Affiliation(s)
- Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT 06520-8034 USA
| | - Qian Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
- Department of Colorectal Surgery, Second Affiliated Hospital Harbin Medical University, Heilongjiang Academy of Medical Science, Harbin, Heilongjiang Province 150086, China
| | - Oladimeji Aladelokun
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| | - Domenica Berardi
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| | - Xinyi Shen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| | - Audrey Marin
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| | - Rolando Garcia-Milian
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
- Bioinformatics Support Program, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jatin Roper
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, NC 27710, USA
| | - Sajid A. Khan
- Section of Surgical Oncology, Department of Surgery, Yale School of Medicine, Yale Cancer Center, Yale University, New Haven, CT 06510, USA
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| |
Collapse
|
7
|
Hsu SH, Chen LR, Chen KH. Primary Osteoporosis Induced by Androgen and Estrogen Deficiency: The Molecular and Cellular Perspective on Pathophysiological Mechanisms and Treatments. Int J Mol Sci 2024; 25:12139. [PMID: 39596206 PMCID: PMC11593909 DOI: 10.3390/ijms252212139] [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: 10/06/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Primary osteoporosis is closely linked to hormone deficiency, which disrupts the balance of bone remodeling. It affects postmenopausal women but also significantly impacts older men. Estrogen can promote the production of osteoprotegerin, a decoy receptor for RANKL, thereby preventing RANKL from activating osteoclasts. Furthermore, estrogen promotes osteoblast survival and function via activation of the Wnt signaling pathway. Likewise, androgens play a critical role in bone metabolism, primarily through their conversion to estrogen in men. Estrogen deficiency accelerates bone resorption through a rise in pro-inflammatory cytokines (IL-1, IL-6, TNF-α) and RANKL, which promote osteoclastogenesis. In the classic genomic pathway, estrogen binds to estrogen receptors in the cytoplasm, forming a complex that migrates to the nucleus and binds to estrogen response elements on DNA, regulating gene transcription. Androgens can be defined as high-affinity ligands for the androgen receptor; their combination can serve as a ligand-inducible transcription factor. Hormone replacement therapy has shown promise but comes with associated risks and side effects. In contrast, the non-genomic pathway involves rapid signaling cascades initiated at the cell membrane, influencing cellular functions without directly altering gene expression. Therefore, the ligand-independent actions and rapid signaling pathways of estrogen and androgen receptors can be harnessed to develop new drugs that provide bone protection without the side effects of traditional hormone therapies. To manage primary osteoporosis, other pharmacological treatments (bisphosphonates, teriparatide, RANKL inhibitors, sclerostin inhibitors, SERMs, and calcitonin salmon) can ameliorate osteoporosis and improve BMD via actions on different pathways. Non-pharmacological treatments include nutritional support and exercise, as well as the dietary intake of antioxidants and natural products. The current study reviews the processes of bone remodeling, hormone actions, hormone receptor status, and therapeutic targets of primary osteoporosis. However, many detailed cellular and molecular mechanisms underlying primary osteoporosis seem complicated and unexplored and warrant further investigation.
Collapse
Affiliation(s)
- Shao-Heng Hsu
- Department of Medical Education, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, New Taipei City 231, Taiwan;
| | - Li-Ru Chen
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei 104, Taiwan;
- Department of Mechanical Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Kuo-Hu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, New Taipei City 231, Taiwan
- School of Medicine, Tzu-Chi University, Hualien 970, Taiwan
| |
Collapse
|
8
|
Liu Q, Li J, Li Y, Cheng M, Zhang H, Ma B. Estrogen Regulates Ca 2+ to Promote Mitochondrial Function Through G-Protein-Coupled Estrogen Receptors During Oocyte Maturation. Biomolecules 2024; 14:1430. [PMID: 39595606 PMCID: PMC11591592 DOI: 10.3390/biom14111430] [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: 09/04/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Estrogen is a steroid hormone that plays a key role in regulating many physiological processes, such as follicle activation and development and oocyte maturation in mammals. Ca2+ is crucial in oogenesis, oocyte maturation, ovulation, and fertilization. However, the mechanism by which estrogen regulates Ca2+ during oocyte maturation in mice has not been reported. This study revealed that Ca2+ levels in oocytes significantly increase during the 4-12 h period in vitro. Oocytes treated with 0.1 µM estrogen and 1 µM G1, a G-protein-coupled estrogen receptor (GPER) agonist, showed significantly increased Ca2+ levels, while treatment with 1 µM G15, an antagonist of GPER, significantly decreased Ca2+ levels. Notably, estrogen regulates Ca2+ in oocytes through the GPER pathway and promotes the expression of the Ca2+-producing protein EPAC1. In addition, estrogen alleviates the inhibitory effect of the Ca2+ chelator BAPTA-AM during oocyte maturation by promoting Ca2+ production. Furthermore, estrogen can promote the expression of the mitochondrial generation-associated protein SIRT1 through the GPER pathway, alleviate mitochondrial oxidative damage caused by BAPTA-AM, and restore the mitochondrial membrane potential level. Collectively, this study demonstrates that estrogen can regulate Ca2+ through the GPER-EPAC1 pathway and promote the expression of SIRT1, which promotes oocyte mitochondrial function during oocyte maturation.
Collapse
Affiliation(s)
- Qingyang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Jingmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Yanxue Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Ming Cheng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Hui Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Q.L.); (J.L.); (Y.L.); (M.C.)
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| |
Collapse
|
9
|
Shi V, Morgan EF. Estrogen and estrogen receptors mediate the mechanobiology of bone disease and repair. Bone 2024; 188:117220. [PMID: 39106937 PMCID: PMC11392539 DOI: 10.1016/j.bone.2024.117220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024]
Abstract
It is well understood that the balance of bone formation and resorption is dependent on both mechanical and biochemical factors. In addition to cell-secreted cytokines and growth factors, sex hormones like estrogen are critical to maintaining bone health. Although the direct osteoprotective function of estrogen and estrogen receptors (ERs) has been reported extensively, evidence that estrogen signaling also has a role in mediating the effects of mechanical loading on maintenance of bone mass and healing of bone injuries has more recently emerged. Recent studies have underscored the role of estrogen and ERs in many pathways of bone mechanosensation and mechanotransduction. Estrogen and ERs have been shown to augment integrin-based mechanotransduction as well as canonical Wnt/b-catenin, RhoA/ROCK, and YAP/TAZ pathways. Estrogen and ERs also influence the mechanosensitivity of not only osteocytes but also osteoblasts, osteoclasts, and marrow stromal cells. The current review will highlight these roles of estrogen and ERs in cellular mechanisms underlying bone mechanobiology and discuss their implications for management of osteoporosis and bone fractures. A greater understanding of the mechanisms behind interactions between estrogen and mechanical loading may be crucial to addressing the shortcomings of current hormonal and pharmaceutical therapies. A combined therapy approach including high-impact exercise therapy may mitigate adverse side effects and allow an effective long-term solution for the prevention, treatment, and management of bone fragility in at-risk populations. Furthermore, future implications to novel local delivery mechanisms of hormonal therapy for osteoporosis treatment, as well as the effects on bone health of applications of sex hormone therapy outside of bone disease, will be discussed.
Collapse
Affiliation(s)
- Vivian Shi
- Boston University, Department of Biomedical Engineering, 44 Cummington St, Boston 02215, MA, USA; Center for Multiscale and Translational Mechanobiology, Boston University, 44 Cummington St, Boston 02215, MA, USA
| | - Elise F Morgan
- Boston University, Department of Biomedical Engineering, 44 Cummington St, Boston 02215, MA, USA; Center for Multiscale and Translational Mechanobiology, Boston University, 44 Cummington St, Boston 02215, MA, USA.
| |
Collapse
|
10
|
Wanionok NE, Colareda GA, Fernandez JM. In vitro effects and mechanisms of Humulus lupulus extract on bone marrow progenitor cells and endothelial cells. Mol Cell Endocrinol 2024; 592:112328. [PMID: 38996835 DOI: 10.1016/j.mce.2024.112328] [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: 03/16/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Osteoporosis is the most common metabolic bone disorder and is associated with a high incidence of fractures. Angiogenesis and adequate blood flow are important during bone repair and maintenance. Estrogens play a key role in bone formation, in the prevention of bone resorption and vasculature maintenance. Hormone replacement therapy (HRT) has been used with great benefits for bone fracture prevention but has been linked to the development of serious important side effects, including cancer and stroke. Phytoestrogens are an attractive alternative to HRT because their chemical structure is similar to estradiol but, they could behave as selective modulators: acting as antagonists of estrogen receptors in the breast and endometrium and as agonists in the vascular endothelium and bone. Hops contain a wide variety of phytoestrogens that have individually been shown to possess estrogenic activity by either blocking or mimicking. In this study we have to evaluate the in vitro effects and mechanisms of action of hops extracts on the osteogenic and adipogenic capacity of bone marrow progenitor cells (BMPCs), and the angiogenic potential of EA.hy926 endothelial cells. We show that hops extracts increase the proliferative capacity of BMPCs and promote their osteogenic differentiation while decreasing their pro-osteoclastogenic capacity; and that these effects are mediated by the MAPK pathway. Additionally, hops extracts prevent the adipogenic differentiation of BMPCs and promote endothelial cell activity, by mechanisms also partially mediated by MAPK.
Collapse
Affiliation(s)
- Nahuel E Wanionok
- Laboratorio de Investigaciones en Osteopatias y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP)-Centro de Investigaciones Científicas (CIC), Calle 47 y 115, La Plata, (1900), Argentina
| | - Germán A Colareda
- Farmacología-GFEYEC, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), Calle 47 y 115, La Plata, (1900), Argentina
| | - Juan M Fernandez
- Laboratorio de Investigaciones en Osteopatias y Metabolismo Mineral (LIOMM), Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP)-Centro de Investigaciones Científicas (CIC), Calle 47 y 115, La Plata, (1900), Argentina.
| |
Collapse
|
11
|
Luo L, Peng B, Xiong L, Wang B, Wang L. Ginsenoside Re promotes proliferation of murine bone marrow mesenchymal stem cells in vitro through estrogen-like action. In Vitro Cell Dev Biol Anim 2024; 60:996-1008. [PMID: 39256290 DOI: 10.1007/s11626-024-00969-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/09/2024] [Indexed: 09/12/2024]
Abstract
Ginsenoside Re (GS-Re) is a major saponin monomer found in Panax ginseng Meyer. It has been shown to exhibit a wide range of biological and pharmacological activities. This study aimed to investigate the effect of GS-Re on the proliferation of murine bone marrow-derived MSCs in vitro and to assess whether its effect is dependent on the estrogen receptor-mediated signal transduction. CFU colony formation assay, cell counting, and colorimetric MTT test were employed to examine effects of GS-Re on the in vitro proliferation of MSCs and the mechanisms of the underlying effect were detected by flow cytometric analysis, immunofluorescence staining for BrdU, and Western blotting. GS-Re dose-dependently promoted the in vitro proliferation of murine bone marrow-derived MSCs over a range of concentrations of 0.5 ~ 20 µmol/L, and this effect approached the maximal level at 10 µmol/L. Increases in the expression level of phosphorylated extracellular signal-regulated kinases 1/2 (p-ERK1/2) were observed in the passaged MSCs treated with 10 µmol/L of GS-Re. These effects of GS-Re on the MSCs were significantly counteracted by the addition of ICI 182, 780 (an estrogen receptor antagonist) to the culture media. We concluded that GS-Re is able to exert a proliferation-promoting effect on murine bone marrow-derived mesenchymal stem cells in vitro, and its action is involved in the estrogen receptor-mediated signaling.
Collapse
Affiliation(s)
- Linzi Luo
- Department of Endoscopic Diagnosis and Treatment, Hunan Chest Hospital, Changsha, China
| | - Bin Peng
- School of Life Science, Hunan Normal University, Changsha, China
| | - Lei Xiong
- School of Life Science, Hunan Normal University, Changsha, China
| | - Baohe Wang
- School of Life Science, Hunan Normal University, Changsha, China
| | - Linghao Wang
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
12
|
Komori T. Regulation of Skeletal Development and Maintenance by Runx2 and Sp7. Int J Mol Sci 2024; 25:10102. [PMID: 39337587 PMCID: PMC11432631 DOI: 10.3390/ijms251810102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Runx2 (runt related transcription factor 2) and Sp7 (Sp7 transcription factor 7) are crucial transcription factors for bone development. The cotranscription factor Cbfb (core binding factor beta), which enhances the DNA-binding capacity of Runx2 and stabilizes the Runx2 protein, is necessary for bone development. Runx2 is essential for chondrocyte maturation, and Sp7 is partly involved. Runx2 induces the commitment of multipotent mesenchymal cells to osteoblast lineage cells and enhances the proliferation of osteoprogenitors. Reciprocal regulation between Runx2 and the Hedgehog, fibroblast growth factor (Fgf), Wnt, and parathyroid hormone-like hormone (Pthlh) signaling pathways and Dlx5 (distal-less homeobox 5) plays an important role in these processes. The induction of Fgfr2 (Fgf receptor 2) and Fgfr3 expression by Runx2 is important for the proliferation of osteoblast lineage cells. Runx2 induces Sp7 expression, and Runx2+ osteoprogenitors become Runx2+Sp7+ preosteoblasts. Sp7 induces the differentiation of preosteoblasts into osteoblasts without enhancing their proliferation. In osteoblasts, Runx2 is required for bone formation by inducing the expression of major bone matrix protein genes, including Col1a1 (collagen type I alpha 1), Col1a2, Spp1 (secreted phosphoprotein 1), Ibsp (integrin binding sialoprotein), and Bglap (bone gamma carboxyglutamate protein)/Bglap2. Bglap/Bglap2 (osteocalcin) regulates the alignment of apatite crystals parallel to collagen fibrils but does not function as a hormone that regulates glucose metabolism, testosterone synthesis, and muscle mass. Sp7 is also involved in Co1a1 expression and regulates osteoblast/osteocyte process formation, which is necessary for the survival of osteocytes and the prevention of cortical porosity. SP7 mutations cause osteogenesis imperfecta in rare cases. Runx2 is an important pathogenic factor, while Runx1, Runx3, and Cbfb are protective factors in osteoarthritis development.
Collapse
Affiliation(s)
- Toshihisa Komori
- Department of Molecular Tumor Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
13
|
Chen L, Xu T, Lou J, Zhang T, Wu S, Xie R, Xu J. The beneficial roles and mechanisms of estrogens in immune health and infection disease. Steroids 2024; 207:109426. [PMID: 38685461 DOI: 10.1016/j.steroids.2024.109426] [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: 10/22/2023] [Revised: 02/28/2024] [Accepted: 04/21/2024] [Indexed: 05/02/2024]
Abstract
Multiple epidemiologic studies have revealed that gender is considered one of the important factors in the frequency and severity of certain infectious diseases, in which estrogens may play a vital role. There is growing evidence that estrogens as female sex hormone can modulate multiple biological functions outside of the reproductive system, such as in brain and cardiovascular system. However, it is largely unknown about the roles and mechanisms of estrogens/estrogen receptors in immune health and infection disease. Thence, by reading a lot of literature, we summarized the regulatory mechanisms of estrogens/estrogen receptors in immune cells and their roles in certain infectious diseases with gender differences. Therefore, estrogens may have therapeutic potentials to prevent and treat these infectious diseases, which needs further clinical investigation.
Collapse
Affiliation(s)
- Lan Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ting Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Lou
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ting Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Sheng Wu
- Department of Gastroenterology, Liupanshui People's Hospital, Liupanshui City 553000, Guizhou Province, China
| | - Rui Xie
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Jingyu Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
14
|
He W, Zhang S, Qi Z, Liu W. Unveiling the potential of estrogen: Exploring its role in neuropsychiatric disorders and exercise intervention. Pharmacol Res 2024; 204:107201. [PMID: 38704108 DOI: 10.1016/j.phrs.2024.107201] [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: 12/21/2023] [Revised: 04/01/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Neuropsychiatric disorders shorten human life spans through multiple ways and become major threats to human health. Exercise can regulate the estrogen signaling, which may be involved in depression, Alzheimer's disease (AD) and Parkinson's disease (PD), and other neuropsychiatric disorders as well in their sex differences. In nervous system, estrogen is an important regulator of cell development, synaptic development, and brain connectivity. Therefore, this review aimed to investigate the potential of estrogen system in the exercise intervention of neuropsychiatric disorders to better understand the exercise in neuropsychiatric disorders and its sex specific. Exercise can exert a protective effect in neuropsychiatric disorders through regulating the expression of estrogen and estrogen receptors, which are involved in neuroprotection, neurodevelopment, and neuronal glucose homeostasis. These processes are mediated by the downstream factors of estrogen signaling, including N-myc downstream regulatory gene 2 (Ndrg2), serotonin (5-HT), delta like canonical Notch ligand 1 (DLL1), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), etc. In addition, exercise can act on the estrogen response element (ERE) fragment in the genes of estrogenic downstream factors like β-amyloid precursor protein cleavase 1 (BACE1). However, there are few studies on the relationship between exercise, the estrogen signaling pathway, and neuropsychiatric disorders. Hence, we review how the estrogen signaling mediates the mechanism of exercise intervention in neuropsychiatric disorders. We aim to provide a theoretical perspective for neuropsychiatric disorders affecting female health and provide theoretical support for the design of exercise prescriptions.
Collapse
Affiliation(s)
- Wenke He
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; College of Physical Education and Health, East China Normal University, Shanghai 200241,China
| | - Sen Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; College of Physical Education and Health, East China Normal University, Shanghai 200241,China
| | - Zhengtang Qi
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; College of Physical Education and Health, East China Normal University, Shanghai 200241,China.
| | - Weina Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; College of Physical Education and Health, East China Normal University, Shanghai 200241,China.
| |
Collapse
|
15
|
Suzuki H, Fujiwara Y, Ariyani W, Amano I, Ishii S, Ninomiya AK, Sato S, Takaoka A, Koibuchi N. 17β-Estradiol (E2) Activates Matrix Mineralization through Genomic/Nongenomic Pathways in MC3T3-E1 Cells. Int J Mol Sci 2024; 25:4727. [PMID: 38731947 PMCID: PMC11083456 DOI: 10.3390/ijms25094727] [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: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Estrogen plays an important role in osteoporosis prevention. We herein report the possible novel signaling pathway of 17β-estradiol (E2) in the matrix mineralization of MC3T3-E1, an osteoblast-like cell line. In the culture media-containing stripped serum, in which small lipophilic molecules such as steroid hormones including E2 were depleted, matrix mineralization was significantly reduced. However, the E2 treatment induced this. The E2 effects were suppressed by ICI182,780, the estrogen receptor (ER)α, and the ERβ antagonist, as well as their mRNA knockdown, whereas Raloxifene, an inhibitor of estrogen-induced transcription, and G15, a G-protein-coupled estrogen receptor (GPER) 1 inhibitor, had little or no effect. Furthermore, the E2-activated matrix mineralization was disrupted by PMA, a PKC activator, and SB202190, a p38 MAPK inhibitor, but not by wortmannin, a PI3K inhibitor. Matrix mineralization was also induced by the culture media from the E2-stimulated cell culture. This effect was hindered by PMA or heat treatment, but not by SB202190. These results indicate that E2 activates the p38 MAPK pathway via ERs independently from actions in the nucleus. Such activation may cause the secretion of certain signaling molecule(s), which inhibit the PKC pathway. Our study provides a novel pathway of E2 action that could be a therapeutic target to activate matrix mineralization under various diseases, including osteoporosis.
Collapse
Affiliation(s)
- Hiraku Suzuki
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Gunma, Japan; (H.S.); (Y.F.); (W.A.); (I.A.); (S.I.); (A.K.N.)
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Hokkaido, Japan; (S.S.); (A.T.)
| | - Yuki Fujiwara
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Gunma, Japan; (H.S.); (Y.F.); (W.A.); (I.A.); (S.I.); (A.K.N.)
| | - Winda Ariyani
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Gunma, Japan; (H.S.); (Y.F.); (W.A.); (I.A.); (S.I.); (A.K.N.)
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Gunma, Japan; (H.S.); (Y.F.); (W.A.); (I.A.); (S.I.); (A.K.N.)
| | - Sumiyasu Ishii
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Gunma, Japan; (H.S.); (Y.F.); (W.A.); (I.A.); (S.I.); (A.K.N.)
| | - Ayane Kate Ninomiya
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Gunma, Japan; (H.S.); (Y.F.); (W.A.); (I.A.); (S.I.); (A.K.N.)
| | - Seiichi Sato
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Hokkaido, Japan; (S.S.); (A.T.)
- Molecular Medical Biochemistry Unit, Biological Chemistry and Engineering Course, Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-0815, Hokkaido, Japan
| | - Akinori Takaoka
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Hokkaido, Japan; (S.S.); (A.T.)
- Molecular Medical Biochemistry Unit, Biological Chemistry and Engineering Course, Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-0815, Hokkaido, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi 371-8511, Gunma, Japan; (H.S.); (Y.F.); (W.A.); (I.A.); (S.I.); (A.K.N.)
| |
Collapse
|
16
|
Piñon-Teal WL, Ogilvie JM. G protein-coupled estrogen receptor expression in postnatal developing mouse retina. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1331298. [PMID: 38984123 PMCID: PMC11182193 DOI: 10.3389/fopht.2024.1331298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/13/2024] [Indexed: 07/11/2024]
Abstract
Introduction Estrogen has emerged as a multifaceted signaling molecule in the retina, playing an important role in neural development and providing neuroprotection in adults. It interacts with two receptor types: classical estrogen receptors (ERs) alpha and beta, and G protein-coupled estrogen receptor (Gper). Gper differs from classical ERs in structure, localization, and signaling. Here we provide the first report of the temporal and spatial properties of Gper transcript and protein expression in the developing and mature mouse retina. Methods We applied qRT-PCR to determine Gper transcript expression in wild type mouse retina from P0-P21. Immunohistochemistry and Western blot were used to determine Gper protein expression and localization at the same time points. Results Gper expression showed a 6-fold increase during postnatal development, peaking at P14. Relative total Gper expression exhibited a significant decrease during retinal development, although variations emerged in the timing of changes among different forms of the protein. Gper immunoreactivity was seen in retinal ganglion cells (RGCs) throughout development and also in somas in the position of horizontal cells at early time points. Immunoreactivity was observed in the cytoplasm and Golgi at all time points, in the nucleus at early time points, and in RGC axons as the retina matured. Discussion In conclusion, our study illuminates the spatial and temporal expression patterns of Gper in the developing mouse retina and provides a vital foundation for further investigations into the role of Gper in retinal development and degeneration.
Collapse
Affiliation(s)
| | - Judith Mosinger Ogilvie
- Department of Biology, Saint Louis University, St. Louis, MO, United States
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
17
|
Liu Z, Li S, Chen S, Sheng J, Li Z, Lv T, Yu W, Fan Y, Wang J, Liu W, Hu S, Jin J. YAP-mediated GPER signaling impedes proliferation and survival of prostate epithelium in benign prostatic hyperplasia. iScience 2024; 27:109125. [PMID: 38420594 PMCID: PMC10901089 DOI: 10.1016/j.isci.2024.109125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/21/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) occurs when there is an imbalance between the proliferation and death of prostate cells, which is regulated tightly by estrogen signaling. However, the role of G protein-coupled estrogen receptor (GPER) in prostate cell survival remains ambiguous. In this study, we observed that prostates with epithelial hyperplasia showed increased yes-associated protein 1 (YAP) expression and decreased levels of estrogen and GPER. Blocking YAP through genetic or drug interventions led to reduced proliferation and increased apoptosis in the prostate epithelial cells. Interestingly, GPER agonists produced similar effects. GPER activation enhanced the phosphorylation and degradation of YAP, which was crucial for suppressing cell proliferation and survival. The Gαs/cAMP/PKA/LATS pathway, downstream of GPER, transmitted signals that facilitated YAP inhibition. This study investigated the interaction between GPER and YAP in the prostate epithelial cells and its contribution to BPH development. It lays the groundwork for future research on developing BPH treatments.
Collapse
Affiliation(s)
- Zhifu Liu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Senmao Li
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Shengbin Chen
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Jindong Sheng
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
- Department of Gynaecological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zheng Li
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Tianjing Lv
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Wei Yu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Yu Fan
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Jinlong Wang
- Department of Urology, Tibet Autonomous Region People's Hospital, Lhasa 850000, China
| | - Wei Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen 518036, China
- Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Shuai Hu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Jie Jin
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| |
Collapse
|
18
|
Feng Y, Wang H, Xu S, Huang J, Pei Q, Wang Z. The detection of Gper1 as an important gene promoting jawbone regeneration in the context of estrogen deficiency. Bone 2024; 180:116990. [PMID: 38141748 DOI: 10.1016/j.bone.2023.116990] [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: 07/07/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Numerous studies have demonstrated that estrogen deficiency inhibit the proliferation and differentiation of pre-osteoblasts in skeleton by affecting osteogenic signaling, lead to decreased bone mass and impaired regeneration. To explore the mechanisms maintaining bone regeneration under estrogen deficiency, we randomly selected 1102 clinical cases, in which female patients aged between 18 and 75 have underwent tooth extraction in Stomatological Hospital of Tongji University, there is little difference in the healing effect of extraction defects, suggesting that to some extent, the regeneration of jawbone is insensitive to the decreased estrogen level. To illuminate the mechanisms promoting jawbone regeneration under estrogen deficiency, a tooth extraction defect model was established in the maxilla of female rats who underwent ovariectomy (OVX) or sham surgery, and jawbone marrow stromal cells (BMSCs) were isolated for single-cell sequencing. Further quantitative PCR, RNA interference, alizarin red staining, immunohistochemistry and western blotting experiments demonstrated that in the context of ovariectomy, maxillary defects promoted G protein-coupled estrogen receptor 1 (Gper1) expression, stimulate downstream cAMP/PKA/pCREB signaling, and facilitate cell proliferation, and thus provided sufficient progenitors for osteogenesis and enhanced the regeneration capacity of the jawbone. Correspondingly, the heterozygous deletion of the Gper1 gene attenuated the phosphorylation of CREB, led to decreased cell proliferation, and impaired the restoration of maxillary defects. This study demonstrates the importance of Gper1 in maintaining jawbone regeneration, especially in the context of estrogen deficiency.
Collapse
Affiliation(s)
- Yuan Feng
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China
| | - Haicheng Wang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China
| | - Shuyu Xu
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China
| | - Jie Huang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China
| | - Qingguo Pei
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China
| | - Zuolin Wang
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, No.399 Middle Yanchang Road, Shanghai 200072, PR China.
| |
Collapse
|
19
|
Ren Z, Yang X, Ku T, Liu QS, Liang J, Zhou Q, Faiola F, Jiang G. Perfluorinated iodine alkanes promote the differentiation of mouse embryonic stem cells by regulating estrogen receptor signaling. J Environ Sci (China) 2024; 137:443-454. [PMID: 37980029 DOI: 10.1016/j.jes.2023.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 11/20/2023]
Abstract
Investigating the development toxicity of perfluorinated iodine alkanes (PFIs) is critical, given their estrogenic effects through binding with estrogen receptors (ERs). In the present study, two PFIs, including dodecafluoro-1,6-diiodohexane (PFHxDI) and tridecafluorohexyl iodide (PFHxI), with binding preference to ERα and ERβ, respectively, were selected to evaluate their effects on proliferation and differentiation of the mouse embryonic stem cells (mESCs). The results revealed that, similar to E2, 50 µmol/L PFHxDI accelerated the cell proliferation of the mESCs. The PFI stimulation at the exposure concentrations of 2-50 µmol/L promoted the differentiation of the mESCs as characterized by the upregulation of differentiation-related biomarkers (i.e., Otx2 and Dnmt3β) and downregulation of pluripotency genes (i.e., Oct4, Nanog, Sox2, Prdm14 and Rex1). Comparatively, PFHxDI exhibited higher induction effect on the differentiation of the mESCs than did PFHxI. The tests on ER signaling indicated that both PFI compounds induced exposure concentration-dependent expressions of ER signaling-related biomarkers (i.e., ERα, ERβ and Caveolin-1) in the mESCs, and the downstream ER responsive genes (i.e., c-fos, c-myc and c-jun) well responded to PFHxI stimulation. The role of ER in PFI-induced effects on the mESCs was further validated by the antagonistic experiments using an ER inhibitor (ICI). The findings demonstrated that PFIs triggered ER signaling, and perturbed the differentiation program of the mESCs, causing the potential health risk during early stage of development.
Collapse
Affiliation(s)
- Zhihua Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Tingting Ku
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jiefeng Liang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Sino-Danish, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
20
|
Heo G, Lee SH, Kim JD, Lee GH, Sim JM, Zhou D, Guo J, Cui XS. GRP78 acts as a cAMP/PKA signaling modulator through the MC4R pathway in porcine embryonic development. FASEB J 2023; 37:e23274. [PMID: 37917004 DOI: 10.1096/fj.202301356r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/23/2023] [Accepted: 10/08/2023] [Indexed: 11/03/2023]
Abstract
Glucose-regulated protein 78 (GRP78) binds to and stabilizes melanocortin 4 receptor (MC4R), which activates protein kinase A (PKA) by regulating G proteins. GRP78 is primarily used as a marker for endoplasmic reticulum stress; however, its other functions have not been well studied. Therefore, in this study, we aimed to investigate the function of GRP78 during porcine embryonic development. The developmental quality of porcine embryos, expression of cell cycle proteins, and function of mitochondria were evaluated by inhibiting the function of GRP78. Porcine oocytes were activated to undergo parthenogenesis, and blastocysts were obtained after 7 days of in vitro culture. GRP78 function was inhibited by adding 20 μM HA15 to the in vitro culture medium. The inhibition in GRP78 function led to a decrease in G proteins release, which subsequently downregulated the cyclic adenosine monophosphate (cAMP)/PKA pathway. Ultimately, inhibition of GRP78 function induced the inhibition of CDK1 and cyclin B expression and disruption of the cell cycle. In addition, inhibition of GRP78 function regulated DRP1 and SIRT1 expression, resulting in mitochondrial dysfunction. This study provides new insights into the role of GRP78 in porcine embryonic development, particularly its involvement in the regulation of the MC4R pathway and downstream cAMP/PKA signaling. The results suggest that the inhibition of GRP78 function in porcine embryos by HA15 treatment may have negative effects on embryo quality and development. This study also demonstrated that GRP78 plays a crucial role in the functioning of MC4R, which releases the G protein during porcine embryonic development.
Collapse
Affiliation(s)
- Geun Heo
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Song-Hee Lee
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Ji-Dam Kim
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Gyu-Hyun Lee
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Jae-Min Sim
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Dongjie Zhou
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Jing Guo
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
21
|
Yang S, Jia Y, Wu Z, Fu B, Zhou S, Pires LV, Cheng JC, Fang L. Activation of G protein-coupled estrogen receptor stimulates placental human chorionic gonadotropin expression through PKA-CREB signaling. Mol Cell Endocrinol 2023; 577:112033. [PMID: 37506871 DOI: 10.1016/j.mce.2023.112033] [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: 05/17/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
The placenta-secreted human chorionic gonadotropin (hCG) is a hormone that plays a critical role in inducing ovarian progesterone production, which is required for maintaining normal pregnancy. The bioavailability of hCG depends on the expression of the beta-subunit of hCG (hCG-β) which is encoded by the chorionic gonadotropin beta (CGB) gene. G protein-coupled estrogen receptor (GPER) is a membrane estrogen receptor involved in non-genomic estrogen signaling. Estradiol (E2) has been shown to stimulate hCG production. However, the role of the GPER in regulating CGB expression remains unknown. In the present study, our results revealed that treatment with G1 upregulated CGB expression in two human choriocarcinoma cell lines, BeWo and JEG-3, and primary human cytotrophoblast cells. In addition, G1 treatment activated the cAMP-response element binding protein (CREB). Using a pharmacological inhibitor and siRNA-mediated knockdown approach, we showed that the stimulatory effect of G1 on CGB expression is mediated by the protein kinase A (PKA)-CREB signaling pathway. This study increases the understanding of the role of GPER in the human placenta. In addition, our results provide important insights into the molecular mechanisms that mediate hCG expression, which may lead to the development of alternative therapeutic approaches for treating placental diseases.
Collapse
Affiliation(s)
- Sizhu Yang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Jia
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ze Wu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bingxin Fu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shenghui Zhou
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Leticia Vicosa Pires
- Department of Gynaecology and Obstetrics, Federal University of Health Sciences of Porto Alegre, Rio Grande do Sul, Brazil
| | - Jung-Chien Cheng
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
22
|
Tirado-Garibay AC, Falcón-Ruiz EA, Ochoa-Zarzosa A, López-Meza JE. GPER: An Estrogen Receptor Key in Metastasis and Tumoral Microenvironments. Int J Mol Sci 2023; 24:14993. [PMID: 37834441 PMCID: PMC10573234 DOI: 10.3390/ijms241914993] [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: 08/30/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
Estrogens and their role in cancer are well-studied, and some cancer types are classified in terms of their response to them. In recent years, a G protein-coupled estrogen receptor (GPER) has been described with relevance in cancer. GPER is a pleiotropic receptor with tissue-specific activity; in normal tissues, its activation is related to correct development and homeostasis, while in cancer cells, it can be pro- or anti-tumorigenic. Also, GPER replaces estrogen responsiveness in estrogen receptor alpha (ERα)-lacking cancer cell lines. One of the most outstanding activities of GPER is its role in epithelial-mesenchymal transition (EMT), which is relevant for metastasis development. In addition, the presence of this receptor in tumor microenvironment cells contributes to the phenotypic plasticity required for the dissemination and maintenance of tumors. These characteristics suggest that GPER could be a promising therapeutic target for regulating cancer development. This review focuses on the role of GPER in EMT in tumorigenic and associated cells, highlighting its role in relation to the main hallmarks of cancer and possible therapeutic options.
Collapse
Affiliation(s)
| | | | | | - Joel E. López-Meza
- Centro Multidisciplinario de Estudios en Biotecnología—FMVZ, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58893, Mexico; (A.C.T.-G.); (E.A.F.-R.); (A.O.-Z.)
| |
Collapse
|
23
|
Bourebaba L, Zyzak M, Sikora M, Serwotka-Suszczak A, Mularczyk M, Al Naem M, Marycz K. Sex Hormone-Binding Globulin (SHBG) Maintains Proper Equine Adipose-Derived Stromal Cells (ASCs)' Metabolic Functions and Negatively Regulates their Basal Adipogenic Potential. Stem Cell Rev Rep 2023; 19:2251-2273. [PMID: 37402098 PMCID: PMC10579166 DOI: 10.1007/s12015-023-10580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Sex hormone binding globulin (SHBG) deteriorated expression has been recently strongly correlated to increased level of circulating pro-inflammatory cytokines and insulin resistance, which are typical manifestations of equine metabolic syndrome (EMS). Despite previous reports demonstrated the potential therapeutic application of SHBG for liver-related dysfunctions, whether SHBG might modulate equine adipose-derived stem/stromal cells (EqASCs) metabolic machinery remains unknown. Therefore, we evaluated for the first time the impact of SHBG protein on metabolic changes in ASCs isolated from healthy horses. METHODS Beforehand, SHBG protein expression has been experimentally lowered using a predesigned siRNA in EqASCs to verify its metabolic implications and potential therapeutic value. Then, apoptosis profile, oxidative stress, mitochondrial network dynamics and basal adipogenic potential have been evaluated using various molecular and analytical techniques. RESULTS The SHBG knockdown altered the proliferative and metabolic activity of EqASCs, while dampening basal apoptosis via Bax transcript suppression. Furthermore, the cells treated with siRNA were characterized by senescent phenotype, accumulation of reactive oxygen species (ROS), nitric oxide, as well as decreased mitochondrial potential that was shown by mitochondrial membrane depolarization and lower expression of key mitophagy factors: PINK, PARKIN and MFN. The addition of SHBG protein reversed the impaired and senescent phenotype of EMS-like cells that was proven by enhanced proliferative activity, reduced apoptosis resistance, lower ROS accumulation and greater mitochondrial dynamics, which is proposed to be related to a normalization of Bax expression. Crucially, SHBG silencing enhanced the expression of key pro-adipogenic effectors, while decreased the abundance of anti-adipogenic factors namely HIF1-α and FABP4. The addition of exogenous SHBG further depleted the expression of PPARγ and C/EBPα and restored the levels of FABP4 and HIF1-α evoking a strong inhibitory potential toward ASCs adipogenesis. CONCLUSION Herein, we provide for the first time the evidence that SHBG protein in importantly involved in various key metabolic pathways governing EqASCs functions, and more importantly we showed that SHBG negatively affect the basal adipogenic potential of tested ASCs through a FABP4-dependant pathway, and provide thus new insights for the development of potential anti-obesity therapeutic approach in both animals and humans.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Magdalena Zyzak
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Mateusz Sikora
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Anna Serwotka-Suszczak
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Malwina Mularczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland.
- Department of Veterinary Medicine and Epidemiology, Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
24
|
Lee I, Doepner M, Weissenrieder J, Majer AD, Mercado S, Estell A, Natale CA, Sung PJ, Foskett JK, Carroll MP, Ridky TW. LNS8801 inhibits Acute Myeloid Leukemia by Inducing the Production of Reactive Oxygen Species and Activating the Endoplasmic Reticulum Stress Pathway. CANCER RESEARCH COMMUNICATIONS 2023; 3:1594-1606. [PMID: 37599786 PMCID: PMC10438922 DOI: 10.1158/2767-9764.crc-22-0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/24/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
Despite recent therapeutic advances, the 5-year survival rate for adults with acute myeloid leukemia (AML) is poor and standard-of-care chemotherapy is associated with significant toxicity, highlighting the need for new therapeutic approaches. Recent work from our group and others established that the G protein-coupled estrogen receptor (GPER) is tumor suppressive in melanoma and other solid tumors. We performed a preliminary screen of human cancer cell lines from multiple malignancies and found that LNS8801, a synthetic pharmacologic agonist of GPER currently in early phase clinical trials, promoted apoptosis in human AML cells. Using human AML cell lines and primary cells, we show that LNS8801 inhibits human AML in preclinical in vitro models, while not affecting normal mononuclear cells. Although GPER is broadly expressed in normal and malignant myeloid cells, this cancer-specific LNS8801-induced inhibition appeared to be independent of GPER signaling. LNS8801 induced AML cell death primarily through a caspase-dependent apoptosis pathway. This was independent of secreted classical death receptor ligands, and instead required induction of reactive oxygen species (ROS) and activation of endoplasmic reticulum (ER) stress response pathways including IRE1α. These studies demonstrate a novel activity of LNS8801 in AML cells and show that targeting ER stress with LNS8801 may be a useful therapeutic approach for AML. Significance Previous work demonstrated that LNS8801 inhibits cancer via GPER activation, especially in solid tumors. Here we show that LNS8801 inhibits AML via GPER-independent mechanisms that include ROS induction and ER activation.
Collapse
Affiliation(s)
- Inyoung Lee
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Miriam Doepner
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jillian Weissenrieder
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ariana D. Majer
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sophia Mercado
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Angela Estell
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher A. Natale
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Linnaeus Therapeutics, Haddonfield, New Jersey
| | - Pamela J. Sung
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New Jersey
| | - J. Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd W. Ridky
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Pakdel F. The Role of Estrogen Receptors in Health and Disease. Int J Mol Sci 2023; 24:11354. [PMID: 37511113 PMCID: PMC10378944 DOI: 10.3390/ijms241411354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Many biological and physiological events, including growth, development, and metabolism of reproductive and non-reproductive tissues in men and women, are regulated by estrogens and estrogen receptors (ERs) [...].
Collapse
Affiliation(s)
- Farzad Pakdel
- Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, EHESP, Inserm, Université de Rennes, F-35000 Rennes, France
| |
Collapse
|
26
|
Bai J, Li H, Wang L, Shi Y, Su X, Xu C, Guo Q, Feng J, Sun X, Cheng Y, Kang J, Wen J, Wu F. Effect of silk fibroin scaffold loaded with 17-β estradiol on the proliferation and differentiation of BMSCs. Regen Ther 2023; 23:76-83. [PMID: 37131535 PMCID: PMC10149272 DOI: 10.1016/j.reth.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/16/2023] [Accepted: 03/09/2023] [Indexed: 05/04/2023] Open
Abstract
In this study, different concentrations of 17-β estradiol silk fibroin (SF)porous scaffolds (SFPS) were prepared using freeze-drying technique, with a hope for optimal concentration and apply it locally to the bone defect area. In this study, the porous scaffold morphology structure was characterized by SEM, FTIR and universal capacity testing machines, and the in vitro cytocompatibility and biological activity of scaffold materials were studied by cell adhesion, viability and proliferation experiments. The results showed that SFPS boasts better physicochemical properties, while 17-β estradiol SF scaffolds with low concentrations of 10-10 mol/L and 10-12 mol/L had more growth and proliferation of SF scaffolds with higher concentrations, and 10-10 mol/L was the optimal concentration of 17-β estradiol SFPS, which was more conducive to cell adhesion and proliferation. On the other hand, after osteogenesis induction of BMSCs inoculated on 17-β estradiol SFPS at different concentrations, it was found that the expression of alkaline phosphatase in BMSCs on different concentrations of 17-β estradiol porous scaffolds was not large. No conflict of interest exits in the submission of this manuscript.
Collapse
Affiliation(s)
- Juan Bai
- Department of Prosthodontics, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Haotian Li
- First Clinical Medical College of Shanxi Medical University, Taiyuan, 030001, China
| | - Lu Wang
- Department of Prosthodontics, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Yue Shi
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xiaomin Su
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Changzhen Xu
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Qiaoling Guo
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jing Feng
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xilin Sun
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Yitong Cheng
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jie Kang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jiayu Wen
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Feng Wu
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| |
Collapse
|
27
|
Lu L, Zhang Q, Shen X, Zhen P, Marin A, Garcia-Milian R, Roper J, Khan SA, Johnson CH. Asparagine synthetase and G-protein coupled estrogen receptor are critical responders to nutrient supply in KRAS mutant colorectal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539577. [PMID: 37205388 PMCID: PMC10187315 DOI: 10.1101/2023.05.05.539577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The nutrient status of the tumor microenvironment has major impacts on cell growth. Under nutrient depletion, asparagine synthetase (ASNS)-mediated asparagine production increases to sustain cell survival. G protein-coupled estrogen receptor-1 (GPER1) signaling converges via cAMP/PI3K/AKT with KRAS signaling to regulate ASNS expression. However, the role of GPER1 in CRC progression is still debated, and the effect of nutrient supply on both ASNS and GPER1 relative to KRAS genotype is not well understood. Here, we modeled a restricted nutrient supply by eliminating glutamine from growing cancer cells in a 3D spheroid model of human female SW48 KRAS wild-type (WT) and KRAS G12A mutant (MT) CRC cells, to examine effects on ASNS and GPER1 expression. Glutamine depletion significantly inhibited cell growth in both KRAS MT and WT cells; however, ASNS and GPER1 were upregulated in KRAS MT compared to WT cells. When nutrient supply was adequate, ASNS and GPER1 were not altered between cell lines. The impact of estradiol, a ligand for GPER1, was examined for any additional effects on cell growth. Under glutamine deplete conditions, estradiol decreased the growth of KRAS WT cells but had no effect on KRAS MT cells; estradiol had no additive or diminutive effect on the upregulation of ASNS or GPER1 between the cell lines. We further examined the association of GPER1 and ASNS levels with overall survival in a clinical colon cancer cohort of The Cancer Genome Atlas. Both high GPER1 and ASNS expression associated with poorer overall survival for females only in advanced stage tumors. These findings suggest that KRAS MT cells have mechanisms in place that respond to decreased nutrient supply, typically observed in advanced tumors, by increasing the expression of ASNS and GPER1 to drive cell growth. Furthermore, KRAS MT cells are resistant to the protective effects of estradiol under nutrient deplete conditions. ASNS and GPER1 may therefore be potential therapeutic targets that can be exploited to manage and control KRAS MT CRC.
Collapse
Affiliation(s)
- Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT 06520-8034 USA
| | - Qian Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
- Department of Colorectal Surgery, Second Affiliated Hospital Harbin Medical University, Heilongjiang Academy of Medical Science, Harbin, Heilongjiang Province 150086, China
| | - Xinyi Shen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| | - Pinyi Zhen
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| | - Audrey Marin
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| | - Rolando Garcia-Milian
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
- Bioinformatics Support Program, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jatin Roper
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, NC 27710, USA
| | - Sajid A Khan
- Section of Surgical Oncology, Department of Surgery, Yale School of Medicine, Yale Cancer Center, Yale University, New Haven, CT 06510, USA
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| |
Collapse
|
28
|
Chronic GPER activation prompted the proliferation of ileal stem cell in ovariectomized mice depending on Paneth cell-derived Wnt3. Clin Sci (Lond) 2023; 137:109-127. [PMID: 36503938 DOI: 10.1042/cs20220392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Menopausal women often face long-term estrogen treatment. G protein-coupled estrogen receptor (GPER) expressed in intestinal crypt was activated by estrogen therapy, but it was unclear whether chronic GPER activation during menopause had an effect on intestinal stem cells (ISCs). We tested the effect of chronic GPER activation on ISCs of ovariectomized (OVX) mice by injection of the selective GPER agonist G-1 for 28 days, or G-1 stimulation of organoids derived from crypts of OVX mice. G-1 up-regulated crypt depth, the number of Ki67+, bromodeoxyuridine+ cells and Olfm4+ ISCs, and the expression of ISCs marker genes (Lgr5, Olfm4 and Axin2). G-1 administration promoted organoid growth, increased the number of EdU+ cells per organoid and protein expression of Cyclin D1 and cyclin B1 in organoids. After G-1 treatment in vivo or in vitro, Paneth cell-derived Wnt3, Wnt3 effector β-catenin and Wnt target genes c-Myc and Cyclin D1 increased in ileum or organoids. Once blocking the secretion of Wnt3 from Paneth cells, the effects of G-1 on organoids growth, ISCs marker genes and Wnt/β-catenin signaling were abolished. G-1 did not affect the number of Paneth cells in ex vivo organoids, while activated Mmp7/cryptdin program in Paneth cells, promoted their maturation, and increased the expression of lysozyme protein. G-1 pretreatment in OVX mice inhibited radiation-induced ISCs proliferation injury and enhanced the resistance of mice to intestinal injury. In conclusion, chronic GPER activation prompted the Wnt3 synthesis in Paneth cells, thus increased the proliferation of ISCs via activation of Wnt3/β-catenin signaling in OVX mice.
Collapse
|
29
|
Railean V, Buszewski B. Flow Cytometry - Sophisticated Tool for Basic Research or/and Routine Diagnosis; Impact of the Complementarity in Both Pre- as Well as Clinical Studies. Crit Rev Anal Chem 2022; 54:2087-2109. [PMID: 36576036 DOI: 10.1080/10408347.2022.2154596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Flow cytometry is a sophisticated technology used widely in both basic research and as a routine tool in clinical diagnosis. The technology has progressed from single parameter detection in the 1970s and 1980s to high end multicolor analysis, with currently 30 parameters detected simultaneously, allowing the identification and purification of rare subpopulations of cells of interest. Flow cytometry continues to evolve and expand to facilitate the investigation of new diagnostic and therapeutic avenues. The present review gives an overview of basic theory and instrumentation, presents and compares the advantages and disadvantages of conventional, spectral and imaging flow cytometry as well as mass cytometry. Current methodologies and applications in both research, pre- and clinical settings are discussed, as well as potential limitations and future evolution. This finding encourages the reader to promote such relationship between basic science, diagnosis and multidisciplinary approach since the standard methods have limitations (e.g., in differentiating the cells after staining). Moreover, such path inspires future cytometry specialists develop new/alternative frontiers between pre- and clinical diagnosis and be more flexible in designing the study for both human as well as veterinary medicine.
Collapse
Affiliation(s)
- Viorica Railean
- Department of Infectious, Invasive Diseases and Veterinary Administration, Institute of Veterinary Medicine, Toruń, Poland
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Torun, Poland
| | - Bogusław Buszewski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Torun, Poland
- Department of Environmental Chemistry and Bioanalysis, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
30
|
Chou HC, Lin SY, Chou LY, Ho ML, Chuang SC, Cheng TL, Kang L, Lin YS, Wang YH, Wei CW, Chen CH, Wang CZ. Ablation of Discoidin Domain Receptor 1 Provokes an Osteopenic Phenotype by Regulating Osteoblast/Osteocyte Autophagy and Apoptosis. Biomedicines 2022; 10:biomedicines10092173. [PMID: 36140274 PMCID: PMC9496360 DOI: 10.3390/biomedicines10092173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/10/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Discoidin domain receptor 1 (DDR1) is a collagen receptor that belongs to the receptor tyrosine kinase family. We have previously shown that DDR1 plays a crucial role during bone development, resulting in dwarfism and a short stature in osteoblast-specific knockout mice (OKO mice). However, the detailed pathophysiological effects of DDR1 on bone development throughout adulthood have remained unclear. This study aims to identify how DDR1 regulates osteoblast and osteocyte functions in vivo and in vitro during bone development in adulthood. The metabolic changes in bone tissues were analyzed using Micro-CT and immunohistochemistry staining (IHC) in vivo; the role of DDR1 in regulating osteoblasts was examined in MC3T3-E1 cells in vitro. The Micro-CT analysis results demonstrated that OKO mice showed a 10% reduction in bone-related parameters from 10 to 14 weeks old and a significant reduction in cortical thickness and diameter compared with flox/flox control mice (FF) mice. These results indicated that DDR1 knockout in OKO mice exhibiting significant bone loss provokes an osteopenic phenotype. The IHC staining revealed a significant decrease in osteogenesis-related genes, including RUNX2, osteocalcin, and osterix. We noted that DDR1 knockout significantly induced osteoblast/osteocyte apoptosis and markedly decreased autophagy activity in vivo. Additionally, the results of the gain- and loss-of-function of the DDR1 assay in MC3T3-E1 cells indicated that DDR1 can regulate the osteoblast differentiation through activating autophagy by regulating the phosphorylation of the mechanistic target of rapamycin (p-mTOR), light chain 3 (LC3), and beclin-1. In conclusion, our study highlights that the ablation of DDR1 results in cancellous bone loss by regulating osteoblast/osteocyte autophagy. These results suggest that DDR1 can act as a potential therapeutic target for managing cancellous bone loss.
Collapse
Affiliation(s)
- Hsin-Chiao Chou
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Sung-Yen Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City 80145, Taiwan
| | - Liang-Yin Chou
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Mei-Ling Ho
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Shu-Chun Chuang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tsung-Lin Cheng
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City 80145, Taiwan
| | - Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yan-Hsiung Wang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chun-Wang Wei
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City 80145, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Ph.D. Program in Biomedical Engineering, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan
- Correspondence: (C.-H.C.); (C.-Z.W.); Tel.: +886-7-3209209 (C.-H.C.); +886-7-3121101 (ext. 2140) (C.-Z.W.)
| | - Chau-Zen Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan
- Correspondence: (C.-H.C.); (C.-Z.W.); Tel.: +886-7-3209209 (C.-H.C.); +886-7-3121101 (ext. 2140) (C.-Z.W.)
| |
Collapse
|
31
|
Shen X, Jain A, Aladelokun O, Yan H, Gilbride A, Ferrucci LM, Lu L, Khan SA, Johnson CH. Asparagine, colorectal cancer, and the role of sex, genes, microbes, and diet: A narrative review. Front Mol Biosci 2022; 9:958666. [PMID: 36090030 PMCID: PMC9453556 DOI: 10.3389/fmolb.2022.958666] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/21/2022] [Indexed: 02/05/2023] Open
Abstract
Asparagine (Asn) and enzymes that catalyze the metabolism of Asn have been linked to the regulation and propagation of colorectal cancer (CRC). Increased Asn and asparagine synthetase (ASNS) expression, both contribute to CRC progression and metastasis. In contradistinction, L-asparaginase (ASNase) which breaks down Asn, exhibits an anti-tumor effect. Metabolic pathways such as KRAS/PI3K/AKT/mTORC1 signaling and high SOX12 expression can positively regulate endogenous Asn production. Conversely, the tumor suppressor, TP53, negatively impacts ASNS, thus limiting Asn synthesis and reducing tumor burden. Asn abundance can be altered by factors extrinsic to the cancer cell such as diet, the microbiome, and therapeutic use of ASNase. Recent studies have shown that sex-related factors can also influence the regulation of Asn, and high Asn production results in poorer prognosis for female CRC patients but not males. In this narrative review, we critically review studies that have examined endogenous and exogenous modulators of Asn bioavailability and summarize the key metabolic networks that regulate Asn metabolism. We also provide new hypotheses regarding sex-related influences on Asn, including the involvement of the sex-steroid hormone estrogen and estrogen receptors. Further, we hypothesize that sex-specific factors that influence Asn metabolism can influence clinical outcomes in CRC patients.
Collapse
Affiliation(s)
- Xinyi Shen
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, United States
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Abhishek Jain
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Oladimeji Aladelokun
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Hong Yan
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Austin Gilbride
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Leah M. Ferrucci
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Sajid A. Khan
- Division of Surgical Oncology, Department of Surgery, Yale University School of Medicine, New Haven, CT, United States
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, United States
| |
Collapse
|
32
|
Livshits G, Kalinkovich A. Targeting chronic inflammation as a potential adjuvant therapy for osteoporosis. Life Sci 2022; 306:120847. [PMID: 35908619 DOI: 10.1016/j.lfs.2022.120847] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/07/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022]
Abstract
Systemic, chronic, low-grade inflammation (SCLGI) underlies the pathogenesis of various widespread diseases. It is often associated with bone loss, thus connecting chronic inflammation to the pathogenesis of osteoporosis. In postmenopausal women, osteoporosis is accompanied by SCLGI development, likely owing to estrogen deficiency. We propose that SCGLI persistence in osteoporosis results from failed inflammation resolution, which is mainly mediated by specialized, pro-resolving mediators (SPMs). In corroboration, SPMs demonstrate encouraging therapeutic effects in various preclinical models of inflammatory disorders, including bone pathology. Since numerous data implicate gut dysbiosis in osteoporosis-associated chronic inflammation, restoring balanced microbiota by supplementing probiotics and prebiotics could contribute to the efficient resolution of SCGLI. In the present review, we provide evidence for this hypothesis and argue that efficient SCGLI resolution may serve as a novel approach for treating osteoporosis, complementary to traditional anti-osteoporotic medications.
Collapse
Affiliation(s)
- Gregory Livshits
- Adelson School of Medicine, Ariel University, Ariel 4077625, Israel; Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel.
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel
| |
Collapse
|
33
|
Xu KJ, Loganathan N, Belsham DD. Bisphenol S induces Agrp expression through GPER1 activation and alters transcription factor expression in immortalized hypothalamic neurons: A mechanism distinct from BPA-induced upregulation. Mol Cell Endocrinol 2022; 552:111630. [PMID: 35569583 DOI: 10.1016/j.mce.2022.111630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/09/2022] [Accepted: 03/25/2022] [Indexed: 11/22/2022]
Abstract
The increasing prevalence of obesity around the world has brought concern upon ubiquitously present obesogenic environmental compounds, such as bisphenol A (BPA). Increasingly tightened regulations on the industrial use of BPA have prompted a transition to a structurally similar alternative, bisphenol S (BPS). BPS displays endocrine-disrupting behaviours similar to those of BPA and increases body weight, food intake and the hypothalamic expression of Agrp in vivo. However, the mechanisms behind this deleterious effect are unclear. Here, we report an increase in the mRNA level of Agrp at 4 h following BPS treatment in immortalized murine hypothalamic cell lines of embryonic and adult origin (mHypoE-41, mHypoA-59). BPS-induced changes in the expression of transcription factors and estrogen receptors that occurred concurrently with Agrp upregulation demonstrated similarities to BPA-induced changes, however, there were also changes that were unique to BPS. Specifically, while Chop, Atf3, Atf4, Atf6, Klf4, and Creb1 were upregulated and Gper1 was downregulated by both BPA and BPS, Esr1 mRNA levels were upregulated and Foxo1 and Stat3 levels remained unchanged by BPS. Finally, inhibition of GPER1 by G15 prevented BPS-mediated Agrp upregulation, independent of Atf3 and Klf4 upregulation. Overall, our results demonstrate the ability of BPS to increase Agrp mRNA expression through GPER1 signaling and to alter transcription factor expression in hypothalamic neurons, further elucidating the endocrine-disrupting potential of this alternative industrial chemical.
Collapse
Affiliation(s)
- Katherine J Xu
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Neruja Loganathan
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
34
|
Zhao Y, Liu H, Fan M, Miao Y, Zhao X, Wei Q, Ma B. G protein-coupled receptor 30 mediates cell proliferation of goat mammary epithelial cells via MEK/ERK&PI3K/AKT signaling pathway. Cell Cycle 2022; 21:2027-2037. [PMID: 35659445 DOI: 10.1080/15384101.2022.2083708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The mammary gland of mammals possesses the specific function of synthesizing, secreting, and delivering milk. Notably, mammary epithelial cells are considered to be central to control the expansion and remodeling of mammary gland into a milk-secretory organ. And the biological function of mammary gland is mainly regulated by the endocrine system, especially for estrogen. G protein-coupled receptor 30 (GPR30), an estrogen membrane receptor, mediates estrogen-induced functions of physiology and pathophysiology. However, the relationship between estrogen/GPR30 signaling and proliferation of goat mammary epithelial cells (gMECs) is still unclear. Herein, estrogen promoted cell proliferation than control, as evidence by upregulation of cell numbers, BrdU-positive cell counts, and cell viability. Of note, these activities were all obviously reduced by treatment with GPR30 antagonist G15, yet GPR30 agonist G1 increased cell proliferation than control. Further, GPR30 silencing inhibited cell proliferation than negative control. This inhibition was accompanied by a G2/M phase arrest and downregulation of cell cycle regulators. Meanwhile, estrogen increased the phosphorylation of ERK1/2 and AKT. Further, the protein level of p-ERK1/2 and p-AKT was enhanced by GPR30 agonist G1 but inhibited by GPR30 antagonist G15 and GPR30 silencing. Importantly, MEK inhibitor and PI3K inhibitor decreased the expression of cell cycle regulators, and repressed estrogen-induced and G1-driven promotion of cell proliferation, suggesting that estrogen regulated cell proliferation of gMECs through mechanisms involving cell cycle, dependent of GPR30 and MEK/ERK and PI3K/AKT signaling pathway. This may provide a strong theoretical basis for researching estrogen sustained-release drugs promoting breast development and improving lactation performance.Abbreviations: gMECs, goat mammary epithelial cells; E2, 17β-estradiol; GPR30, G protein-coupled receptor 30; shRNA, small hairpin RNA; CDK, cyclin-dependent kinase; PI3K, phosphatidylinositol 3-kinase; AKT, proteinkinase B; MAPK, mitogen-activated protein kinase; MEK, mitogen-activated protein kinase kinase; ERK1/2, extracellular signal-regulated kinase 1/2.
Collapse
Affiliation(s)
- Ying Zhao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haokun Liu
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingzhen Fan
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuyang Miao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoe Zhao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qing Wei
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Baohua Ma
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
35
|
Ba H, Chen M, Li C. Cross-Species Analysis Reveals Co-Expressed Genes Regulating Antler Development in Cervidae. Front Genet 2022; 13:878078. [PMID: 35664330 PMCID: PMC9157503 DOI: 10.3389/fgene.2022.878078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Antlers constitute an interesting model for basic research in regenerative biology. Despite decades of being studied, much is still unknown about the genes related to antler development. Here, we utilized both the genome and antlerogenic periosteum (AP) transcriptome data of four deer species to reveal antler-related genes through cross-species comparative analysis. The results showed that the global gene expression pattern matches the status of antler phenotypes, supporting the fact that the genes expressed in the AP may be related to antler phenotypes. The upregulated genes of the AP in three-antlered deer showed evidence of co-expression, and their protein sequences were highly conserved. These genes were growth related and likely participated in antler development. In contrast, the upregulated genes in antler-less deer (Chinese water deer) were involved mainly in organismal death and growth failure, possibly related to the loss of antlers during evolution. Overall, this study demonstrates that the co-expressed genes in antlered deer may regulate antler development.
Collapse
Affiliation(s)
- Hengxing Ba
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
- Jilin Provincial Key Laboratory of Deer Antler Biology, Changchun, China
| | - Min Chen
- School of Life Sciences, Institute of Eco-Chongming (IEC), East China Normal University, Shanghai, China
- Yangtze Delta Estuarine Wetland Ecosystem Observation and Research Station, Ministry of Education & Shanghai Science and Technology Committee, Shanghai, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
- Jilin Provincial Key Laboratory of Deer Antler Biology, Changchun, China
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| |
Collapse
|
36
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
37
|
G protein-coupled estrogen receptor 1 mediates proliferation and adipogenic differentiation of goat adipose-derived stem cells through ERK1/2-NF-κB signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2022; 54:494-503. [PMID: 35607957 PMCID: PMC9828292 DOI: 10.3724/abbs.2022031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Adipose tissue formation and moderate fat deposition are important for the production performance and eating quality of livestock meats. The self-renewal and adipogenic differentiation of adipose-derived stem cells are responsible for the formation and development of adipose tissue. In addition, estrogen targeting G protein-coupled estrogen receptor 1 (GPER1) has been reported to modulate cell proliferation and differentiation during tissue and organ development. However, the potential correlation among estrogen, GPER1, proliferation, and adipogenic differentiation in goat adipose-derived stem cells (gADSCs) is still unclear. Herein, we demonstrated that 17β-estradiol enhances the proliferative ability of gADSCs, indicated by the increased cell number and cell viability, accompanied by up-regulated expressions of cyclin D1 and PCNA. Meanwhile, the adipogenic differentiation is promoted by 17β-estradiol, supported by higher ccumulation of intracellular lipids and increased expressions of PPARγ, ACC, and FABP4. Notably, these activities are all obviously reduced by administration with GPER1 antagonist G15, but GPER1 agonist G1 enhances cell proliferation and adipogenic differentiation. Moreover, GPER1 silencing diminishes cell proliferation and adipogenic differentiation. In parallel, 17β-estradiol elevates the protein level of nuclear p-p65. Furthermore, the phosphorylation of p65 is enhanced by G1 but inhibited by G15 and GPER1 silencing. In addition, the phosphorylation of p65 is mediated by ERK1/2, suggesting that estrogen targeting GPER1 regulates cell proliferation and adipogenic differentiation of gADSCs through the ERK1/2-NF-κB signaling pathway. This study may provide a strong theoretical basis for improving meat quality, flavor, and cold resistance of livestock.
Collapse
|
38
|
Li H, Li M, Ran X, Cui J, Wei F, Yi G, Chen W, Luo X, Chen Z. The Role of Zinc in Bone Mesenchymal Stem Cell Differentiation. Cell Reprogram 2022; 24:80-94. [PMID: 35172118 DOI: 10.1089/cell.2021.0137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Zinc is an essential trace element for bone growth and bone homeostasis in the human body. Bone mesenchymal stem cells (BMSCs) are multipotent progenitors existing in the bone marrow stroma with the capability of differentiating along multiple lineage pathways. Zinc plays a paramount role in BMSCs, which can be spurred differentiating into osteoblasts, chondrocytes, or adipocytes, and modulates the formation and activity of osteoclasts. The expression of related genes also changed during the differentiation of various cell phenotypes. Based on the important role of zinc in BMSC differentiation, using zinc as a therapeutic approach for bone remodeling will be a promising method. This review explores the role of zinc ion in the differentiation of BMSCs into various cell phenotypes and outlines the existing research on their molecular mechanism.
Collapse
Affiliation(s)
- Huiyun Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Muzhe Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xun Ran
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Juncheng Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Fu Wei
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Guoliang Yi
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Wei Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xuling Luo
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhiwei Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
39
|
Jin Z, Zhang Y, Li J, Lv S, Zhang L, Feng Y. Endometriosis stem cell sources and potential therapeutic targets: literature review and bioinformatics analysis. Regen Med 2021; 16:949-962. [PMID: 34585967 DOI: 10.2217/rme-2021-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The stem cell origin theory of endometriosis (EMS) is a significant area of new research but the sources of this have yet to be adequately summarized. Existing treatments for EMS are commonly associated with a high recurrence rate; consequently, there is an urgent need to develop new therapeutic measures for the future treatment of this disease from the view of stem cells and gene therapy. Recently, we described the evidence for the potential sources of EMS stem cells and other key molecules participating in the establishment of lesions, and predict the miRNAs that target these key genes via bioinformatics analysis for further research. This review highlights the origin of EMS stem cells and potential therapy targets.
Collapse
Affiliation(s)
- Zhe Jin
- The Second Affiliated Hospital of Nanchang University, Jiangxi, 330006, China.,The Second Clinical Medical School of Nanchang University, Jiangxi, 330031, China
| | - Yize Zhang
- The Second Affiliated Hospital of Nanchang University, Jiangxi, 330006, China.,The Second Clinical Medical School of Nanchang University, Jiangxi, 330031, China
| | - Jingyi Li
- School of Public Health of Nanchang University, Jiangxi, 330031, China
| | - Sidi Lv
- The Second Affiliated Hospital of Nanchang University, Jiangxi, 330006, China.,The Second Clinical Medical School of Nanchang University, Jiangxi, 330031, China
| | - Lixia Zhang
- The First Hospital of Handan City, Hebei, 056004, China
| | - Ying Feng
- The Second Affiliated Hospital of Nanchang University, Jiangxi, 330006, China
| |
Collapse
|
40
|
Chou YS, Chuang SC, Chen CH, Ho ML, Chang JK. G-Protein-Coupled Estrogen Receptor-1 Positively Regulates the Growth Plate Chondrocyte Proliferation in Female Pubertal Mice. Front Cell Dev Biol 2021; 9:710664. [PMID: 34490260 PMCID: PMC8417792 DOI: 10.3389/fcell.2021.710664] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Estrogen enhances long bone longitudinal growth during early puberty. Growth plate chondrocytes are the main cells that contribute to long bone elongation. The role of G-protein-coupled estrogen receptor-1 (GPER-1) in regulating growth plate chondrocyte function remains unclear. In the present study, we generated chondrocyte-specific GPER-1 knockout (CKO) mice to investigate the effect of GPER-1 in growth plate chondrocytes. In control mice, GPER-1 was highly expressed in the growth plates of 4- and 8-week-old mice, with a gradual decline through 12 to 16 weeks. In CKO mice, the GPER-1 expression in growth plate chondrocytes was significantly lower than that in the control mice (80% decrease). The CKO mice also showed a decrease in body length (crown-rump length), body weight, and the length of tibias and femurs at 8 weeks. More importantly, the cell number and thickness of the proliferative zone of the growth plate, as well as the thickness of primary spongiosa and length of metaphysis plus diaphysis in tibias of CKO mice, were significantly decreased compared with those of the control mice. Furthermore, there was also a considerable reduction in the number of proliferating cell nuclear antigens and Ki67-stained proliferating chondrocytes in the tibia growth plate in the CKO mice. The chondrocyte proliferation mediated by GPER-1 was further demonstrated via treatment with a GPER-1 antagonist in cultured epiphyseal cartilage. This study demonstrates that GPER-1 positively regulates chondrocyte proliferation at the growth plate during early puberty and contributes to the longitudinal growth of long bones.
Collapse
Affiliation(s)
- Ya-Shuan Chou
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Chun Chuang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopaedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopaedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan.,Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan.,Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Je-Ken Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopaedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopaedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| |
Collapse
|
41
|
Lei P, Qian H, Zhang T, Lei T, Hu Y, Chen C, Zhou K. Porous tantalum structure integrated on Ti6Al4V base by Laser Powder Bed Fusion for enhanced bony-ingrowth implants: In vitro and in vivo validation. Bioact Mater 2021; 7:3-13. [PMID: 34430760 PMCID: PMC8367833 DOI: 10.1016/j.bioactmat.2021.05.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023] Open
Abstract
Despite the widespread application of Ti6Al4V and tantalum (Ta) in orthopedics, bioinertia and high cost limit their further applicability, respectively, and tremendous efforts have been made on the Ti6Al4V-Ta alloy and Ta coating to address these drawbacks. However, the scaffolds obtained are unsatisfactory. In this study, novel high-interface-strength Ti6Al4V-based porous Ta scaffolds were successfully manufactured using Laser Powder Bed Fusion for the first time, in which porous Ta was directly manufactured on a solid Ti6Al4V substrate. Mechanical testing revealed that the novel scaffolds were biomechanically compatible, and the interfacial bonding strength was as high as 447.5 MPa. In vitro biocompatibility assay, using rat bone marrow mesenchymal stem cells (r-BMSCs), indicated that the novel scaffolds were biocompatible. Alkaline phosphatase and mineralized nodule determination demonstrated that the scaffolds favored the osteogenic differentiation of r-BMSCs. Moreover, scaffolds were implanted into rabbits with femur bone defects, and imaging and histological evaluation identified considerable new bone formation and bone ingrowth, suggesting that the scaffolds were well integrated with the host bone. Overall, these results demonstrated good mechanical compatibility, biocompatibility, and osteointegration performance of the novel Ti6Al4V-based porous Ta scaffold, which possesses great potential for orthopedic clinical applications.
Collapse
Affiliation(s)
- Pengfei Lei
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Orthopedic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, China.,State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Hu Qian
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Taomei Zhang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Ting Lei
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yihe Hu
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Orthopedic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, China
| | - Chao Chen
- Department of Orthopedic Surgery, Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha 410008, China.,State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Kechao Zhou
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| |
Collapse
|
42
|
Kalinkovich A, Livshits G. Biased and allosteric modulation of bone cell-expressing G protein-coupled receptors as a novel approach to osteoporosis therapy. Pharmacol Res 2021; 171:105794. [PMID: 34329703 DOI: 10.1016/j.phrs.2021.105794] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 12/16/2022]
Abstract
On the cellular level, osteoporosis (OP) is a result of imbalanced bone remodeling, in which osteoclastic bone resorption outcompetes osteoblastic bone formation. Currently available OP medications include both antiresorptive and bone-forming drugs. However, their long-term use in OP patients, mainly in postmenopausal women, is accompanied by severe side effects. Notably, the fundamental coupling between bone resorption and formation processes underlies the existence of an undesirable secondary outcome that bone anabolic or anti-resorptive drugs also reduce bone formation. This drawback requires the development of anti-OP drugs capable of selectively stimulating osteoblastogenesis and concomitantly reducing osteoclastogenesis. We propose that the application of small synthetic biased and allosteric modulators of bone cell receptors, which belong to the G-protein coupled receptors (GPCR) family, could be the key to resolving the undesired anti-OP drug selectivity. This approach is based on the capacity of these GPCR modulators, unlike the natural ligands, to trigger signaling pathways that promote beneficial effects on bone remodeling while blocking potentially deleterious effects. Under the settings of OP, an optimal anti-OP drug should provide fine-tuned regulation of downstream effects, for example, intermittent cyclic AMP (cAMP) elevation, preservation of Ca2+ balance, stimulation of osteoprotegerin (OPG) and estrogen production, suppression of sclerostin secretion, and/or preserved/enhanced canonical β-catenin/Wnt signaling pathway. As such, selective modulation of GPCRs involved in bone remodeling presents a promising approach in OP treatment. This review focuses on the evidence for the validity of our hypothesis.
Collapse
Affiliation(s)
- Alexander Kalinkovich
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel
| | - Gregory Livshits
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel; Adelson School of Medicine, Ariel University, Ariel 4077625, Israel.
| |
Collapse
|
43
|
Abstract
Estrogen plays important roles in bone homeostasis throughout a person's life, including longitudinal bone growth, bone healing, and adaptation to mechanical forces. Estrogen exerts its action by binding to its multiple receptors in the cell membrane and cytoplasm. Until now at least three estrogen receptors (ER) have been reported: ER alpha (ERα), ER beta (ERβ), and G-protein coupled estrogen receptor 1 (GPER1) also known as GP30. Recently it has been observed that estrogen crosstalk with other signaling pathways helping to understand its wide effects in bone homeostasis. Abrupt loss of estrogen production experienced by menopausal women is associated with the rapid loss of bone mass ultimately leading to osteoporosis. The detrimental results during its absence with aging and the increased life expectancy of current and future generations make it of high importance to fully understand its mechanism of action. This review article aims to update on (1) the molecular mechanism of action of estrogen in the skeletal system, (2) ERs expression in different bone cells, (3) recent reported ER mutations resulting in pathological human conditions, and (4) role of estrogen signaling during bone healing.
Collapse
Affiliation(s)
- Nuria Lara-Castillo
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, 650 East 25th Street, Kansas City, MO 64110, USA
| |
Collapse
|
44
|
A novel transcript variant of human G-protein coupled estrogen receptor. Mol Biol Rep 2021; 48:2979-2983. [PMID: 33677756 DOI: 10.1007/s11033-021-06242-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/18/2021] [Indexed: 10/22/2022]
Abstract
The G-protein coupled estrogen receptor (GPER) mediates short-term non-genomic effects of estrogen in diverse cell types and tissues. According to the NCBI nucleotide database, three variants of GPER are known. They are NM_001505.2 (GPER-v2), NM_001039966.1 (GPER-v3), and NM_001098201.1 (GPER-v4). Investigations on GPER expression are key to understand its physiological and pathological roles. However, most studies on GPER mRNA expression have considered total GPER mRNA expression regardless of the individual variants. The present study is motivated by a novel transcript observed in the UCSC Genome Browser (uc010ksd.1), which is annotated as GPER. The novel variant is similar to the known transcript variants of GPER in terms of the protein-coding sequence and the 3'UTR. However, it has a unique 5'UTR, which distinguishes it from other GPER variants. Using primers specific for uc010ksd.1, we have performed RT-PCR to show that the novel GPER transcript (hereafter referred to as GPER-v5) is expressed in human cancer cell lines, such as MCF-7, SW-620, COLO-205, and HT-29. Preliminary evidences indicate that GPER-v5 is a novel GPER mRNA variant. The expression of GPER-v5 in primary cells and tissues should be investigated before probing into its role and relevance in physiological and pathological conditions.
Collapse
|