1
|
Wang Y, Yu X, Sun F, Fu Y, Hu T, Shi Q, Man Q. METTL14 Mediates Glut3 m6A methylation to improve osteogenesis under oxidative stress condition. Redox Rep 2025; 30:2435241. [PMID: 39737912 DOI: 10.1080/13510002.2024.2435241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025] Open
Abstract
OBJECTIVES Bone remodeling imbalance contributes to osteoporosis. Though current medications enhance osteoblast involvement in bone formation, the underlying pathways remain unclear. This study was aimed to explore the pathways involved in bone formation by osteoblasts, we investigate the protective role of glycolysis and N6-methyladenosine methylation (m6A) against oxidative stress-induced impairment of osteogenesis in MC3T3-E1 cells. METHODS We utilized a concentration of 200 μM hydrogen peroxide (H2O2) to establish an oxidative damage model of MC3T3-E1 cells. Subsequently, we examined the alterations in the m6A methyltransferases (METTL3, METTL14), glucose transporter proteins (GLUT1, GLUT3) and validated m6A methyltransferase overexpression in vitro and in an osteoporosis model. The osteoblast differentiation and osteogenesis-related molecules and serum bone resorption markers were measured by biochemical analysis, Alizarin Red S staining, Western blot and ELISA. RESULTS H2O2 treatment inhibited glycolysis and osteoblast differentiation in MC3T3-E1 cells. However, when METTL14 was overexpressed, these changes induced by H2O2 could be mitigated. Our findings indicate that METTL14 promotes GLUT3 expression via YTHDF1, leading to the modulation of various parameters in the H2O2-induced model. Similar positive effects of METTL14 on osteogenesis were observed in an ovariectomized mouse osteoporosis model. DISCUSSION METTL14 could serve as a potential therapeutic approach for enhancing osteoporosis treatment.
Collapse
Affiliation(s)
- Ying Wang
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xueying Yu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Fenyong Sun
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yan Fu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Tingting Hu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qiqing Shi
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qiuhong Man
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Strutynska NA, Balatskyi VV, B Strutynskyi R, Goshovska YV, Mys LA, Luchkova AY, Denysova MV, Korkach YP, Strutynskyi VR, Piven OO, Dobrzyn P, Sagach VF. Pyridoxal-5-phosphate mitigates age-related metabolic imbalances in the rat heart through the H 2S/AKT/GSK3β signaling axis. Mitochondrion 2025:102001. [PMID: 39755161 DOI: 10.1016/j.mito.2024.102001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 12/09/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
Pyridoxal-5-phosphate (PLP) enhances the synthesis of endogenous hydrogen sulfide, a potent regulator of cell metabolism. We used 24-month-old rats to investigate the PLP mitoprotective function in the aging heart. We demonstrated improvement of mitochondrial bioenergetic functions, inhibition of mPTP opening after PLP administration. Moreover, PLP treatment increased glucose consumption and utilization, decreased lipid transport into the cells, but increased fatty acid β-oxidation, providing sufficient energy. An ECG study showed a significant improvement in cardiac function in PLP-treated old rats. Our data suggest that PLP may exert its effect through the H2S/AKT/GSK3β axis with further targeting of the Sirt1/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Nataliia A Strutynska
- The Department of Blood Circulation of Bogomoletz Institute of Physiology of the National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine.
| | - Volodymyr V Balatskyi
- The Laboratory of Molecular Medical Biochemistry of Nencki Institute of Experimental Biology, Polish Academy of Sciences. Address: 3 Pasteur Str., Warsaw 02-093, Poland
| | - Ruslan B Strutynskyi
- The Department of General and Molecular Pathophysiology of Bogomoletz Institute of Physiology of the National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine
| | - Yulia V Goshovska
- The Department of Blood Circulation of Bogomoletz Institute of Physiology of the National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine
| | - Lidiia A Mys
- The Department of Blood Circulation of Bogomoletz Institute of Physiology of the National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine
| | - Alina Yu Luchkova
- The Department of Blood Circulation of Bogomoletz Institute of Physiology of the National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine
| | - Maiia V Denysova
- The Department of Blood Circulation of Bogomoletz Institute of Physiology of the National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine
| | - Yuliia P Korkach
- The Department of Blood Circulation of Bogomoletz Institute of Physiology of the National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine
| | - Vladyslav R Strutynskyi
- The Department of Immunophysiology of Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine
| | - Oksana O Piven
- The Laboratory of Molecular Medical Biochemistry of Nencki Institute of Experimental Biology, Polish Academy of Sciences. Address: 3 Pasteur Str., Warsaw 02-093, Poland; The Department of Human Genetics of Institute of Molecular Biology and Genetics of the National Academy of Sciences of Ukraine. Address: 150 Akad. Zabolotnogo Str., Kyiv 03680, Ukraine.
| | - Pawel Dobrzyn
- The Laboratory of Molecular Medical Biochemistry of Nencki Institute of Experimental Biology, Polish Academy of Sciences. Address: 3 Pasteur Str., Warsaw 02-093, Poland
| | - Vadym F Sagach
- The Department of Blood Circulation of Bogomoletz Institute of Physiology of the National Academy of Sciences of Ukraine, Kyiv, Ukraine. Address: 4, Bogomoletz Str., Kyiv 01024, Ukraine
| |
Collapse
|
3
|
Liu H, Negoita F, Brook M, Sakamoto K, Morton NM. Quantification of persulfidation on specific proteins: are we nearly there yet? Essays Biochem 2024; 68:467-478. [PMID: 39290133 PMCID: PMC11625863 DOI: 10.1042/ebc20230095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024]
Abstract
Hydrogen sulfide (H2S) played a pivotal role in the early evolution of life on Earth before the predominance of atmospheric oxygen. The legacy of a persistent role for H2S in life's processes recently emerged through its discovery in modern biochemistry as an endogenous cellular signalling modulator involved in numerous biological processes. One major mechanism through which H2S signals is protein cysteine persulfidation, an oxidative post-translational modification. In recent years, chemoproteomic technologies have been developed to allow the global scanning of protein persulfidation targets in mammalian cells and tissues, providing a powerful tool to elucidate the broader impact of altered H2S in organismal physiological health and human disease states. While hundreds of proteins were confirmed to be persulfidated by global persulfidome methodologies, the targeting of specific proteins of interest and the investigation of further mechanistic studies are still underdeveloped due to a lack of stringent specificity of the methods and the inherent instability of persulfides. This review provides an overview of the processes of endogenous H2S production, oxidation, and signalling and highlights the application and limitations of current persulfidation labelling approaches for investigation of this important evolutionarily conserved biological switch for protein function.
Collapse
Affiliation(s)
- Hongling Liu
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, U.K
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Florentina Negoita
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Matthew Brook
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, U.K
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Nicholas M Morton
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, U.K
- Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, School of Science and Technology, Nottingham Trent University, NG11 8NS, U.K
| |
Collapse
|
4
|
Jiang L, Wu Y, Xu Z, Hou M, Chen S, Cheng C, Hu D, Lu D, Zhu X, Li C. Harnessing hydrogen sulfide in injectable hydrogels that guide the immune response and osteoclastogenesis balance for osteoporosis treatment. Mater Today Bio 2024; 29:101338. [PMID: 39649246 PMCID: PMC11625156 DOI: 10.1016/j.mtbio.2024.101338] [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: 07/20/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 12/10/2024] Open
Abstract
Elevated levels of oxidative stress, inflammation, and a dysregulated osteoclastogenesis balance frequently characterize the microenvironment of osteoporosis, which impedes the processes of healing and repair. Existing treatment approaches are limited in scope and rely primarily on factors and drugs. An injectable hydrogel designed for the ROS-responsive release of H2S gas is presented in this study. The first network of the hydrogel comprises sodium alginate (SA-SATO) chelated with S-aroylthiooxime (SATO) and an H2S-generating group, while the second network is composed of photocrosslinkable PEGDA. Through the integration of Cys-releasing microspheres that are reactive with ROS, a composite hydrogel was developed that exhibited advantageous mechanical characteristics and biosafety. The composite hydrogel effectively promoted osteogenic differentiation of mesenchymal stem cells, modulated macrophage polarization, decreased inflammatory responses, and halted cell apoptosis, as evidenced by in vitro experiments. Additionally, it released H2S gas and mitigated excess ROS in cells. The efficacy of the composite hydrogel in promoting bone defect repair and regeneration in an osteoporotic model was further validated by in vivo findings. In summary, the composite hydrogel exhibits potential as a viable approach to address osteoporotic bone defects by harmonizing osteogenesis and osteoclast activity, modulating the microenvironment of bone injuries, and reducing inflammation. Consequently, it presents a viable strategy for the efficient repair of bone defects.
Collapse
Affiliation(s)
- Lianghua Jiang
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, China
| | - Yubin Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Zonghan Xu
- Department of Orthopedics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, 215008, China
| | - Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Shayang Chen
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, China
| | - Chao Cheng
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, China
| | - Dan Hu
- Department of Orthopedics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, 215008, China
| | - Daming Lu
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Chong Li
- Department of Orthopedics, Affiliated Kunshan Hospital of Jiangsu University, Jiangsu, 215300, China
| |
Collapse
|
5
|
Pieles O, Morsczeck C. The Role of Protein Kinase C During the Differentiation of Stem and Precursor Cells into Tissue Cells. Biomedicines 2024; 12:2735. [DOI: 10.3390/biomedicines12122735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Protein kinase C (PKC) plays an essential role during many biological processes including development from early embryonic stages until the terminal differentiation of specialized cells. This review summarizes the current knowledge about the involvement of PKC in molecular processes during the differentiation of stem/precursor cells into tissue cells with a particular focus on osteogenic, adipogenic, chondrogenic and neuronal differentiation by using a comprehensive approach. Interestingly, studies examining the overall role of PKC, or one of its three isoform groups (classical, novel and atypical PKCs), often showed controversial results. A discrete observation of distinct isoforms demonstrated that the impact on differentiation differs highly between the isoforms, and that during a certain process, the influence of only some isoforms is crucial, while others are less important. In particular, PKCβ inhibits, and PKCδ strongly supports osteogenesis, whereas it is the other way around for adipogenesis. PKCε is another isoform that overwhelmingly supports adipogenic differentiation. In addition, PKCα plays an important role in chondrogenesis, while neuronal differentiation has been positively associated with numerous isoforms including classical, novel and atypical PKCs. In a cellular context, various upstream mediators, like the canonical and non-canonical Wnt pathways, endogenously control PKC activity and thus, their activity interferes with the influence of PKC on differentiation. Downstream of PKC, several proteins and pathways build the molecular bridge between the enzyme and the control of differentiation, of which only a few have been well characterized so far. In this context, PKC also cooperates with other kinases like Akt or protein kinase A (PKA). Furthermore, PKC is capable of directly phosphorylating transcription factors with pivotal function for a certain developmental process. Ultimately, profound knowledge about the role of distinct PKC isoforms and the involved signaling pathways during differentiation constitutes a promising tool to improve the use of stem cells in regenerative therapies by precisely manipulating the activity of PKC or downstream effectors.
Collapse
Affiliation(s)
- Oliver Pieles
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Christian Morsczeck
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
6
|
Arciero I, Buonvino S, Melino S. Slow H 2S-Releasing Donors and 3D Printable Arrays Cellular Models in Osteo-Differentiation of Mesenchymal Stem Cells for Personalized Therapies. Biomolecules 2024; 14:1380. [PMID: 39595557 PMCID: PMC11592188 DOI: 10.3390/biom14111380] [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: 09/24/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The effects of the hydrogen sulfide (H2S) slow-releasing donor, named GSGa, a glutathione-conjugate water-soluble garlic extract, on human mesenchymal stem cells (hMSCs) in both bidimensional (2D) and three-dimensional (3D) cultures were investigated, demonstrating increased expression of the antioxidant enzyme HO-1 and decreased expression of the pro-inflammatory cytokine interleukin-6 (IL-6). The administration of the H2S donor can therefore increase the expression of antioxidant enzymes, which may have potential therapeutic applications in osteoarthritis (OA). Moreover, GSGa was able to promote the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), but not of cardiac mesenchymal stem cells (cMSCs) in a 2D culture system. This result highlights the varying sensitivity of hMSCs to the H2S donor GSGa, suggesting that the induction of osteogenic differentiation in stem cells by chemical factors is dependent on the tissue of origin. Additionally, a 3D-printable mesenchymal stem cells-bone matrix array (MSCBM), designed to closely mimic the stiffness of bone tissue, was developed to serve as a versatile tool for evaluating the effects of drugs and stem cells on bone repair in chronic diseases, such as OA. We demonstrated that the osteogenic differentiation process in cMSCs can be induced just by simulating bone stiffness in a 3D system. The expression of osteocalcin, RUNX2, and antioxidant enzymes was also assessed after treating MSCs with GSGa and/or increasing the stiffness of the culture environment. The printability of the array may enable better customization of the cavities, enabling an accurate replication of real bone defects. This could optimize the BM array to mimic bone defects not only in terms of stiffness, but also in terms of shape. This culture system may enable a rapid screening of antioxidant and anti-inflammatory compounds, facilitating a more personalized approach to regenerative therapy.
Collapse
Affiliation(s)
- Ilaria Arciero
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy;
| | - Silvia Buonvino
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| | - Sonia Melino
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
7
|
Liu C, Cyphert EL, Stephen SJ, Wang B, Morales AL, Nixon JC, Natsoulas NR, Garcia M, Carmona PB, Vill AC, Donnelly E, Brito IL, Vashishth D, Hernandez CJ. Microbiome-induced increases and decreases in bone matrix strength can be initiated after skeletal maturity. J Bone Miner Res 2024; 39:1621-1632. [PMID: 39348436 PMCID: PMC11523134 DOI: 10.1093/jbmr/zjae157] [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: 01/05/2024] [Revised: 07/25/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Abstract
Recent studies in mice have indicated that the gut microbiome can regulate bone tissue strength. However, prior work involved modifications to the gut microbiome in growing animals and it is unclear if the same changes in the microbiome, applied later in life, would change matrix strength. Here we changed the composition of the gut microbiome before and/or after skeletal maturity (16 weeks of age) using oral antibiotics (ampicillin + neomycin). Male and female mice (n = 143 total, n = 12-17/group/sex) were allocated into five study groups: (1) Unaltered, (2) Continuous (dosing 4-24 weeks of age), (3) Delayed (dosing only 16-24 weeks of age), (4) Initial (dosing 4-16 weeks of age, suspended at 16 weeks), and (5) Reconstituted (dosing from 4-16 weeks following by fecal microbiota transplant from Unaltered donors). Animals were euthanized at 24 weeks of age. In males, bone matrix strength in the femur was 25%-35% less than expected by geometry in mice from the Continuous (p = 0.001), Delayed (p = 0.005), and Initial (p = 0.040) groups as compared to Unaltered. Reconstitution of the gut microbiota led to a bone matrix strength similar to Unaltered animals (p = 0.929). In females, microbiome-induced changes in bone matrix strength followed the same trend as males but were not significantly different, demonstrating a sex-dependent response of bone matrix to the gut microbiota. Minor differences in chemical composition of bone matrix were observed with Raman spectroscopy. Our findings indicate that microbiome-induced impairment of bone matrix in males can be initiated and/or reversed after skeletal maturity. The portion of the femoral cortical bone formed after skeletal maturity (16 weeks) was small; suggesting that microbiome-induced changes in bone matrix occurred without osteoblast/osteoclast turnover through a yet unidentified mechanism. These findings provide evidence that the mechanical properties of bone matrix can be altered in the adult skeleton.
Collapse
Affiliation(s)
- Chongshan Liu
- Departments of Orthopaedic Surgery and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, United States
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Erika L Cyphert
- Departments of Orthopaedic Surgery and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, United States
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Samuel J Stephen
- Shirley Ann Jackson, PhD Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Bowen Wang
- Shirley Ann Jackson, PhD Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Angie L Morales
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Jacob C Nixon
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Nicholas R Natsoulas
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Matthew Garcia
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | | | - Albert C Vill
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Eve Donnelly
- Department of Material Science and Engineering, Cornell University, Ithaca, NY, United States
- Reseach Division, Hospital for Special Surgery, New York, NY, United States
| | - Ilana L Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Deepak Vashishth
- Shirley Ann Jackson, PhD Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Rensselaer - Icahn School of Medicine at Mount Sinai Center for Engineering and Precision Medicine, New York, NY, United States
| | - Christopher J Hernandez
- Departments of Orthopaedic Surgery and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
8
|
Lyu Z, Yuan G, Zhang Y, Zhang F, Liu Y, Li Y, Li G, Wang Y, Zhang M, Hu Y, Guo Y, Liu D. Anaerostipes caccae CML199 enhances bone development and counteracts aging-induced bone loss through the butyrate-driven gut-bone axis: the chicken model. MICROBIOME 2024; 12:215. [PMID: 39438898 PMCID: PMC11495078 DOI: 10.1186/s40168-024-01920-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND The gut microbiota is a key regulator of bone metabolism. Investigating the relationship between the gut microbiota and bone remodeling has revealed new avenues for the treatment of bone-related disorders. Despite significant progress in understanding gut microbiota-bone interactions in mammals, research on avian species remains limited. Birds have unique bone anatomy and physiology to support egg-laying. However, whether and how the gut microbiota affects bone physiology in birds is still unknown. In this study, we utilized laying hens as a research model to analyze bone development patterns, elucidate the relationships between bone and the gut microbiota, and mine probiotics with osteomodulatory effects. RESULTS Aging led to a continuous increase in bone mineral density in the femur of laying hens. The continuous deposition of medullary bone in the bone marrow cavity of aged laying hens led to significant trabecular bone loss and weakened bone metabolism. The cecal microbial composition significantly shifted before and after sexual maturity, with some genera within the class Clostridia potentially linked to postnatal bone development in laying hens. Four bacterial strains associated with bone development, namely Blautia coccoides CML164, Fournierella sp002159185 CML151, Anaerostipes caccae CML199 (ANA), and Romboutsia lituseburensis CML137, were identified and assessed in chicks with low bacterial loads and chicken primary osteoblasts. Among these, ANA demonstrated the most significant promotion of bone formation both in vivo and in vitro, primarily attributed to butyrate in its fermentation products. A long-term feeding experiment of up to 72 weeks confirmed that ANA enhanced bone development during sexual maturity by improving the immune microenvironment of the bone marrow in laying hens. Dietary supplementation of ANA for 50 weeks prevented excessive medullary bone deposition and mitigated aging-induced trabecular bone loss. CONCLUSIONS These findings highlight the beneficial effects of ANA on bone physiology, offering new perspectives for microbial-based interventions for bone-related disorders in both poultry and possibly extending to human health. Video Abstract.
Collapse
Affiliation(s)
- Zhengtian Lyu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Gaoxiang Yuan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuying Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Fengwenhui Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yan Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yifan Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guang Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ying Wang
- Sichuan Tieqilishi Industrial Co., Ltd, Mianyang, 621010, China
| | - Ming Zhang
- Sichuan Tieqilishi Industrial Co., Ltd, Mianyang, 621010, China
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Dan Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
9
|
Kushram P, Bose S. Improving Biological Performance of 3D-Printed Scaffolds with Garlic-Extract Nanoemulsions. ACS APPLIED MATERIALS & INTERFACES 2024; 16:48955-48968. [PMID: 39196793 DOI: 10.1021/acsami.4c05588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Complex bone diseases such as osteomyelitis, osteosarcoma, and osteoporosis often cause critical-size bone defects that the body cannot self-repair and require an advanced bone graft material to repair. We have fabricated 3D-printed tricalcium phosphate bone scaffolds functionalized with garlic extract (GE). GE was encapsulated in a nanoemulsion (GE-NE) to enhance bioavailability and stability. GE-NE showed ∼73% drug encapsulation efficiency, with an average particle size of 158 nm and a zeta potential of -14.2 mV. Release of GE-NEs from the scaffold displayed a controlled and biphasic release profile at both acidic and physiological mediums. Results from the osteosarcoma study show that GE-NE demonstrated ∼88% reduction in cancer cell growth while exhibiting no cytotoxicity toward bone-forming cells. Interaction for the functionalized scaffold with Gram-positive Staphylococcus aureus and Gram-negative Pseudomonas aeruginosa showed a substantial reduction in bacteria growth by more than 90% compared to the unfunctionalized scaffold. These findings demonstrate the potential of GE-NEs-treated porous scaffolds to treat bone-related diseases, particularly for non-load bearing applications.
Collapse
Affiliation(s)
- Priya Kushram
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| |
Collapse
|
10
|
Yuan Z, Li J, He K, Sun Z, Luo G, Liu H, Dong J, Zhou C, Cui H, Fan C. Endogenous hydrogen sulfide accelerated trauma-induced heterotopic ossification through the Ca 2+/ERK pathway-enhanced aberrant osteogenic activity. Redox Biol 2024; 75:103265. [PMID: 39003920 PMCID: PMC11298937 DOI: 10.1016/j.redox.2024.103265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Unveiling of the mechanism involved in the occurrence and development of trauma-induced heterotopic ossification (tHO) is highly demanding due to current ineffective clinical treatment for it. Previous studies proposed that hydrogen sulfide (H2S) was vital for fate determination of stem cells, suggesting a potential role in the regulation of tHO development. In the current study, We found that expression of metabolic enzyme within sulfur conversion pathway was enhanced after tendon injury, leading to H2S accumulation within the tHO region. Increased production of endogenous H2S was shown to promote aberrant osteogenic activity of tendon-derived stem cells (TDSCs), which accelerated tHO formation. The inhibition of metabolic enzyme of H2S production or directly absorption of H2S could abolished osteogenic induction of TDSCs and the formation of tHO. Mechanistically, through RNA sequencing combined with rescue experiments, we demonstrated that activation of Ca2+/ERK pathway was the downstream molecular event of H2S-induced osteogenic commitment of TDSCs and tHO. For treatment strategy exploration, zine oxide nanoparticles (ZnO) as an effective H2S elimination material was validated to ideally halt the tHO formation in this study. Furthermore, in terms of chirality of nanoparticles, D-ZnO or L-ZnO nanoparticles showed superiority over R-ZnO nanoparticles in both clearing of H2S and inhibition of tHO. Our study not only revealed the mechanism of tHO through the endogenous gas signaling event from a new perspective, but also presented a applicable platform for elimination of the inordinate gas production, thus aiding the development of clinical treatment for tHO.
Collapse
Affiliation(s)
- Zhengqiang Yuan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Juehong Li
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kuangyu He
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ziyang Sun
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Gang Luo
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hang Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jinlei Dong
- Department of Orthopaedics Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, PR China
| | - Chao Zhou
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Haomin Cui
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China; Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
11
|
Zhu P, Tao H, Chen K, Chu M, Wang Q, Yang X, Zhou J, Yang H, Geng D. TRPA1 aggravates osteoclastogenesis and osteoporosis through activating endoplasmic reticulum stress mediated by SRXN1. Cell Death Dis 2024; 15:624. [PMID: 39191723 DOI: 10.1038/s41419-024-07018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Osteoporosis (OP) is a disorder of bone remodeling caused by an imbalance between bone resorption by osteoclasts and bone formation by osteoblasts. Therefore, inhibiting excessive osteoclast activity is one of the promising strategies for treating OP. A major transient receptor potential cation channel, known as transient receptor potential ankyrin 1 (TRPA1), was found to alleviate joint pain and cartilage degeneration in osteoarthritis. However, little research has focused on TRPA1 function in OP. As a result, this study aimed to explore the TRPA1 characteristics and its potential therapeutic function during osteoclastogenesis. The TRPA1 expression gradually increased in the osteoclast differentiation process; however, its suppression with small interfering RNA and an inhibitor (HC030031) significantly controlled the osteoclast count and the expression of osteoclast characteristic genes. Its suppression also inhibited endoplasmic reticulum (ER) stress-related pancreatic ER kinase (PERK) pathways. An ER stress inhibitor (thapsigargin) reversed the down-regulated levels of ER stress and osteoclast differentiation by suppressing TRPA1. Transcriptome sequencing results demonstrated that TRPA1 negatively regulated reactive oxygen species (ROS) and significantly increased the expression of an antioxidant gene, SRXN1. The osteoclast differentiation and the levels of ER stress were enhanced with SRXN1 inhibition. Finally, TRPA1 knockdown targeting macrophages by adeno-associated virus-9 could relieve osteoclast differentiation and osteopenia in ovariectomized mice. In summary, silencing TRPA1 restrained osteoclast differentiation through ROS-mediated down-regulation of ER stress via inhibiting PERK pathways. The study also indicated that TRPA1 might become a prospective treatment target for OP.
Collapse
Affiliation(s)
- Pengfei Zhu
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Huaqiang Tao
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Kai Chen
- Department of Orthopedics, Hai'an People's Hospital, Hai'an, 226600, Jiangsu, China
| | - Miao Chu
- Department of Orthopedics, Yixing People's Hospital, Yixing, 214200, Jiangsu, China
| | - Qiufei Wang
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People's Hospital of Changshu City, Changshu, 215500, Jiangsu, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, 215008, Jiangsu, China.
| | - Jun Zhou
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
12
|
Chen S, Zhao X, Zhou L. The Application of Hydrogen Sulfide Fluorescent Probe in Food Preservation, Detection and Evaluation. Molecules 2024; 29:3973. [PMID: 39203051 PMCID: PMC11356813 DOI: 10.3390/molecules29163973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
This work primarily reviewed the response mechanism of fluorescent probes for H2S detection in foodstuffs in recent years, as well as the methodologies employed for detecting foodstuffs. Firstly, the significance of studying H2S gas as an important signaling molecule is introduced. Subsequently, a review of the response mechanism of the scientific community on how to detect H2S in foodstuffs samples by fluorescent probe technology is carried out. Secondly, the methods commonly used for detecting foodstuffs samples are discussed, including the test strip method and the spiking recovery methods. Nevertheless, despite the significant advancements in this field, there remain some research gaps. Finally, the article identifies the remaining issues that require further attention in current research and proposes avenues for future investigation. More importantly, this work identifies the current limitations of research in this field and proposes future applications of fluorescent probes for H2S in assessing food freshness and determining food spoilage. Therefore, this review will provide robust technical support for the protection of consumer health and the advancement of the sustainable development of the food industry and also put forward some new ideas and suggestions for future research.
Collapse
Affiliation(s)
- Sitong Chen
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| | - Xiongjie Zhao
- College of Chemistry and Biological Engineering, Hunan University of Science and Engineering, Yongzhou 425199, China
| | - Liyi Zhou
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China
| |
Collapse
|
13
|
Liu H, Zheng Y, Li F, Geng B, Liao F. Endoplasmic reticulum protein of 57 kDa sulfhydration promotes intestinal calcium absorption to attenuate primary osteoporosis. Nitric Oxide 2024; 149:32-40. [PMID: 38830571 DOI: 10.1016/j.niox.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/05/2024]
Abstract
Endogenous hydrogen sulfide (H2S) plays an important role in bone metabolism. However, the exact role of H2S in intestinal calcium and phosphorus absorption and its potential in preventing and treating primary osteoporosis remains unknown. Therefore, this study aimed to investigate the potential of H2S in promoting intestinal calcium and phosphorus absorption and alleviating primary osteoporosis. We measured the apparent absorptivity of calcium, femoral bone density, expression and sulfhydration of the duodenal endoplasmic reticulum protein of 57 kDa (ERp57), duodenal cystathionine γ-lyase (CSE) expression, and serum H2S content in adult and old CSE-knockout and wild-type mice. We also assessed intracellular reactive oxygen species (ROS) and Ca2+ content in CSE-overexpressing or knockout intestinal epithelial cell (IEC)-6 cells. In senile mice, CSE knockout decreased endogenous H2S, ERp57 sulfhydration, and intestinal calcium absorption and worsened osteoporosis, which were partially reversed by GYY4137, an H2S donor. CSE overexpression in IEC-6 cells increased ERp57 sulfhydration, protein kinase A and C activity, and intracellular Ca2+, whereas CSE knockout exerted the opposite effects. Furthermore, hydrogen peroxide (H2O2) stimulation had similar effects as in CSE knockout, which were reversed by pretreatment with sodium hydrosulfide before H2O2 stimulation and restored by DL-dithiothreitol. These findings suggest that H2S attenuates primary osteoporosis by preventing ROS-induced ERp57 damage in intestinal epithelial cells by enhancing ERp57 activity and promoting intestinal calcium absorption, thereby aiding in developing therapeutic interventions to prevent osteoporosis.
Collapse
Affiliation(s)
- Huifang Liu
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China; Department of Rehabilitation Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Yang Zheng
- Department of Orthopedics, Fourth Medical Center of PLA General Hospital, Beijing, 100048, PR China
| | - Fuming Li
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Bin Geng
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, 102308, PR China
| | - Feng Liao
- Department of Orthopedics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, PR China.
| |
Collapse
|
14
|
Huang T, Zeng Y, Li C, Zhou Z, Xu J, Wang L, Yu DG, Wang K. Application and Development of Electrospun Nanofiber Scaffolds for Bone Tissue Engineering. ACS Biomater Sci Eng 2024; 10:4114-4144. [PMID: 38830819 DOI: 10.1021/acsbiomaterials.4c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Nanofiber scaffolds have gained significant attention in the field of bone tissue engineering. Electrospinning, a straightforward and efficient technique for producing nanofibers, has been extensively researched. When used in bone tissue engineering scaffolds, electrospun nanofibers with suitable surface properties promote new bone tissue growth and enhance cell adhesion. Recent advancements in electrospinning technology have provided innovative approaches for scaffold fabrication in bone tissue engineering. This review comprehensively examines the utilization of electrospun nanofibers in bone tissue engineering scaffolds and evaluates the relevant literature. The review begins by presenting the fundamental principles and methodologies of electrospinning. It then discusses various materials used in the production of electrospun nanofiber scaffolds for bone tissue engineering, including natural and synthetic polymers, as well as certain inorganic materials. The challenges associated with these materials are also described. The review focuses on novel electrospinning techniques for scaffold construction in bone tissue engineering, such as multilayer nanofibers, multifluid electrospinning, and the integration of electrospinning with other methods. Recent advancements in electrospinning technology have enabled the fabrication of precisely aligned nanofiber scaffolds with nanoscale architectures. These innovative methods also facilitate the fabrication of biomimetic structures, wherein bioactive substances can be incorporated and released in a controlled manner for drug delivery purposes. Moreover, they address issues encountered with traditional electrospun nanofibers, such as mechanical characteristics and biocompatibility. Consequently, the development and implementation of novel electrospinning technologies have revolutionized scaffold fabrication for bone tissue engineering.
Collapse
Affiliation(s)
- Tianyue Huang
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - YuE Zeng
- Department of Neurology, RuiJin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chaofei Li
- Department of General Surgery, RuiJin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhengqing Zhou
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - Jie Xu
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - Lean Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - Deng-Guang Yu
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| | - Ke Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology 516 Jungong Road, Shanghai 200093, China
| |
Collapse
|
15
|
Wang K, Rong F, Peng H, Yuan Z, Huo J, Liu P, Ding R, Yan C, Liu G, Wang T, Li P. Infection Microenvironment-Responsive Coating on Titanium Surfaces for On-Demand Release of Therapeutic Gas and Antibiotic. Adv Healthc Mater 2024; 13:e2304510. [PMID: 38532711 DOI: 10.1002/adhm.202304510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/10/2024] [Indexed: 03/28/2024]
Abstract
Aseptic loosening and bacterial infection pose significant challenges in the clinical application of titanium (Ti) orthopedic implants, which are primarily caused by insufficient osseointegration and bacterial contamination. To address these issues, a responsive coating on Ti surface is constructed, which achieves enhanced osseointegration and infection elimination by on-demand release of therapeutic gas hydrogen sulfide (H2S) and antibiotic. TiO2 nanotubes (TNT) are anodized on the Ti surface to enhance its bioactivity and serve as reservoirs for the antibiotic. An infection microenvironment-responsive macromolecular H2S donor layer is coated on top of TNT to inhibit premature leakage of antibiotic. This layer exhibits a sustained release of low-dosage H2S, which is capable of promoting the osteogenic differentiation and migration of cells. Moreover, the compactness of the macromolecular H2S donor layer could be broken by bacterial invasion, leading to rapid antibiotic release thus preventing infection. In vitro antibacterial experiments validates significant antibacterial activity of the coating against both Gram-negative (Escherichia coli) and Gram-positive bacteria (Staphylococcus aureus). Crucially, this coating effectively suppresses implant-associated infection with 98.7% antibacterial efficiency in a rat femoral bone defect model, mitigates inflammation at the defect site and promotes osseointegration of the Ti orthopedic implant.
Collapse
Affiliation(s)
- Kun Wang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Fan Rong
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Haowei Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Chongqing Innovation Center, Northwestern Polytechnical University, Chongqing, 401135, China
| | - Jingjing Huo
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Pengxiang Liu
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Rui Ding
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Cuiping Yan
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Guming Liu
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| | - Tengjiao Wang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
- Chongqing Innovation Center, Northwestern Polytechnical University, Chongqing, 401135, China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China
| |
Collapse
|
16
|
Hansdah K, Lui JC. Emerging Insights into the Endocrine Regulation of Bone Homeostasis by Gut Microbiome. J Endocr Soc 2024; 8:bvae117. [PMID: 38957653 PMCID: PMC11215793 DOI: 10.1210/jendso/bvae117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Indexed: 07/04/2024] Open
Abstract
Gut microbiota plays an important role in the regulation of bone homeostasis and bone health. Recent studies showed that these effects could be mediated through microbial metabolites released by the microbiota like short-chain fatty acids, metabolism of endogenous molecules such as bile acids, or a complex interplay between microbiota, the endocrine system, and the immune system. Importantly, some studies showed a reciprocal relationship between the endocrine system and gut microbiota. For instance, postmenopausal estrogen deficiency could lead to dysbiosis of the gut microbiota, which could in turn affect various immune response and bone remodeling. In addition, evidence showed that shift in the indigenous gut microbiota caused by antibiotics treatment may also impact normal skeletal growth and maturation. In this mini-review, we describe recent findings on the role of microbiome in bone homeostasis, with a particular focus on molecular mechanisms and their interactions with the endocrine and immune system. We will also discuss the recent findings on estrogen deficiency and microbiota dysbiosis, and the clinical implications for the development of new therapeutic strategies for osteoporosis and other bone disorders.
Collapse
Affiliation(s)
- Kirtal Hansdah
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Julian C Lui
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Ma J, Yang P, Zhou Z, Song T, Jia L, Ye X, Yan W, Sun J, Ye T, Zhu L. GYY4137-induced p65 sulfhydration protects synovial macrophages against pyroptosis by improving mitochondrial function in osteoarthritis development. J Adv Res 2024:S2090-1232(24)00223-6. [PMID: 38844123 DOI: 10.1016/j.jare.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is the most common arthritis that is characterized by the progressive synovial inflammation and loss of articular cartilage. Although GYY4137 is a novel and slow-releasing hydrogen sulfide (H2S) donor with potent anti-inflammatory properties that may modulate the progression of OA, its underlying mechanism remains unclear. OBJECTIVES In this study, we validated the protective role of GYY4137 against OA pathological courses and elucidated its underlying regulatory mechanisms. METHODS Cell transfection, immunofluorescence staining, EdU assay, transmission electron microscopy, mitochondrial membrane potential measurement, electrophoretic mobility shift assay, sulfhydration assay, qPCR and western blot assays were performed in the primary mouse chondrocytes or the mouse macrophage cell line raw 264.7 for in vitro study. DMM-induced OA mice model and Macrophage-specific p65 knockout (p65f/f LysM-CreERT2) mice on the C57BL/6 background were used for in vivo study. RESULTS We found that GYY4137 can alleviate OA progress by suppressing synovium pyroptosis in vivo. Moreover, our in vitro data revealed that GYY4137 attenuates inflammation-induced NLRP3 and caspase-1 activation and results in a decrease of IL-1β production in macrophages. Mechanistically, GYY4137 increased persulfidation of NF-kB p65 in response to inflammatory stimuli that results in a decrease of cellular reactive oxygen species (ROS) accumulation and ameliorates mitochondrial dysfunctions. Using site-directed mutagenesis, we showed that H2S persulfidates cysteine38 in p65 protein and hampers p65 transcriptional activity, and p65 mutant impaired macrophage responses to GYY4137. CONCLUSION These findings suggest a mechanism by which GYY4137 through redox modification of p65 participates in inhibiting NLRP3 activation by OA to regulate inflammatory responses. Thus, we propose that GYY4137 represents a promising novel therapeutic strategy for the treatment of OA.
Collapse
Affiliation(s)
- Jun Ma
- Department of Orthepaedics, Naval Medical Center of PLA, Naval Medical University, Shanghai, China; Department of Health Statistics, Naval Medical University, Shanghai, China; Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Peng Yang
- Department of Orthepaedics, Naval Medical Center of PLA, Naval Medical University, Shanghai, China; Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhibin Zhou
- Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China; Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Tengfei Song
- Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Liang Jia
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Xiaofei Ye
- Department of Health Statistics, Naval Medical University, Shanghai, China
| | - Wei Yan
- Department of Orthepaedics, Naval Medical Center of PLA, Naval Medical University, Shanghai, China
| | - Jiuyi Sun
- Department of Orthepaedics, Naval Medical Center of PLA, Naval Medical University, Shanghai, China.
| | - Tianwen Ye
- Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Lei Zhu
- Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
18
|
Zuo Y, Liu HT, Lin LB, Yue RZ, Liu HH, Wang HW, Wang L, Hou RL, Liu WZ, Li CZ, Wang JZ, Li P, Yin YL. A new metal ion chelator attenuates human tau accumulation-induced neurodegeneration and memory deficits in mice. Exp Neurol 2024; 373:114657. [PMID: 38141802 DOI: 10.1016/j.expneurol.2023.114657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/16/2023] [Indexed: 12/25/2023]
Abstract
Neuronal neurofibrillary tangles containing Tau hyperphosphorylation proteins are a typical pathological marker of Alzheimer's disease (AD). The level of tangles in neurons correlates positively with severe dementia. However, how Tau induces cognitive dysfunction is still unknown, which leads to a lack of effective treatments for AD. Metal ions deposition occurs with tangles in AD brain autopsy. Reduced metal ion can improve the pathology of AD. To explore whether abnormally phosphorylated Tau causes metal ion deposition, we overexpressed human full-length Tau (hTau) in the hippocampal CA3 area of mice and primary cultured hippocampal neurons (CPHN) and found that Tau accumulation induced iron deposition and activated calcineurin (CaN), which dephosphorylates glycogen synthase kinase 3 beta (GSK3β), mediating Tau hyperphosphorylation. Simultaneous activation of CaN dephosphorylates cyclic-AMP response binding protein (CREB), leading to synaptic deficits and memory impairment, as shown in our previous study; this seems to be a vicious cycle exacerbating tauopathy. In the current study, we developed a new metal ion chelator that displayed a significant inhibitory effect on Tau phosphorylation and memory impairment by chelating iron ions in vivo and in vitro. These findings provide new insight into the mechanism of memory impairment induced by Tau accumulation and develop a novel potential treatment for tauopathy in AD.
Collapse
Affiliation(s)
- Yue Zuo
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China; School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang 453003, China
| | - Hui-Ting Liu
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Lai-Biao Lin
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Rui-Zhu Yue
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Huan-Huan Liu
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China; The Second Affiliated Hospital, Xinxiang Medical University, Xinxiang 453003, China
| | - Hong-Wei Wang
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Lu Wang
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ruan-Ling Hou
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Wei-Zhen Liu
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Chang-Zheng Li
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China; School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang 453003, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Li
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang 453003, China; Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, China.
| | - Ya-Ling Yin
- School of Basic Medical Sciences, Sino-UK Joint Laboratory of Brain Function and Injury, Henan International Key Laboratory for Non-invasive Neuromodulation, Department of Physiology and Pathology, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
19
|
Zhang X, Zhou W, Xi W. Advancements in incorporating metal ions onto the surface of biomedical titanium and its alloys via micro-arc oxidation: a research review. Front Chem 2024; 12:1353950. [PMID: 38456182 PMCID: PMC10917964 DOI: 10.3389/fchem.2024.1353950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/31/2024] [Indexed: 03/09/2024] Open
Abstract
The incorporation of biologically active metallic elements into nano/micron-scale coatings through micro-arc oxidation (MAO) shows significant potential in enhancing the biological characteristics and functionality of titanium-based materials. By introducing diverse metal ions onto titanium implant surfaces, not only can their antibacterial, anti-inflammatory and corrosion resistance properties be heightened, but it also promotes vascular growth and facilitates the formation of new bone tissue. This review provides a thorough examination of recent advancements in this field, covering the characteristics of commonly used metal ions and their associated preparation parameters. It also highlights the diverse applications of specific metal ions in enhancing osteogenesis, angiogenesis, antibacterial efficacy, anti-inflammatory and corrosion resistance properties of titanium implants. Furthermore, the review discusses challenges faced and future prospects in this promising area of research. In conclusion, the synergistic approach of micro-arc oxidation and metal ion doping demonstrates substantial promise in advancing the effectiveness of biomedical titanium and its alloys, promising improved outcomes in medical implant applications.
Collapse
Affiliation(s)
- Xue’e Zhang
- Jiangxi Province Key Laboratory of Oral Biomedicine, School of Stomatology, Jiangxi Medical College, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, China
| | - Wuchao Zhou
- Jiangxi Province Key Laboratory of Oral Biomedicine, The Affiliated Stomatological Hospital, Jiangxi Medical College, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, China
| | - Weihong Xi
- Jiangxi Province Key Laboratory of Oral Biomedicine, The Affiliated Stomatological Hospital, Jiangxi Medical College, Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Stevens CM, Jain SK. Vitamin D/Bone Mineral Density and Triglyceride Paradoxes Seen in African Americans: A Cross-Sectional Study and Review of the Literature. Int J Mol Sci 2024; 25:1305. [PMID: 38279305 PMCID: PMC10816015 DOI: 10.3390/ijms25021305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Vitamin D is known to have a positive effect on bone health. Despite the greater frequency of vitamin D deficiency in African Americans (AA), they have a higher bone mineral density (BMD) compared to whites, demonstrating a disconnect between BMD and vitamin D levels in AA. Another intriguing relationship seen in AA is the triglyceride (TG) paradox, an unusual phenomenon in which a normal TG status is observed even when patients house conditions known to be characterized by high TG levels, such as Type II diabetes. To the best of our knowledge, no study has examined whether these two paradoxical relationships exist simultaneously in AA subjects with Type II diabetes. In this study, we compared levels of blood markers, including HbA1c, TG, and vitamin D, measured as serum 25-hydroxyvitamin D [25(OH)VD] µM/mL, [25(OH)VD]/TG, calcium, and BMD in AA (n = 56) and white (n = 26) subjects with Type II diabetes to see whether these relationships exist concurrently. We found that AA subjects had significantly lower TG and [25(OH)VD] levels and a significantly higher BMD status compared to white subjects, even when the ages, BMI, duration of diabetes, HbA1c, and calcium levels were similar between the two groups. This demonstrates that these two paradoxical relationships exist simultaneously in Type II diabetic AA subjects. In addition to these findings, we discuss the current hypotheses in the literature that attempt to explain why these two intriguing relationships exist. This review also discusses four novel hypotheses, such as altered circulating levels and the potential role of estrogen and hydrogen sulfide on BMD and HMG-CoA reductase as a possible contributor to the TG paradox in AA subjects. This manuscript demonstrates that there are still many unanswered questions regarding these two paradoxical relationships and further research is needed to determine why they exist and how they can be implemented to improve healthcare.
Collapse
Affiliation(s)
| | - Sushil K. Jain
- Department of Pediatrics, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71103, USA;
| |
Collapse
|
21
|
Liu C, Cyphert EL, Stephen SJ, Wang B, Morales AL, Nixon JC, Natsoulas NR, Garcia M, Blazquez Carmona P, Vill AC, Donnelly EL, Brito IL, Vashishth D, Hernandez CJ. Microbiome-induced Increases and Decreases in Bone Tissue Strength can be Initiated After Skeletal Maturity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.574074. [PMID: 38260539 PMCID: PMC10802367 DOI: 10.1101/2024.01.03.574074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Recent studies in mice have indicated that the gut microbiome can regulate bone tissue strength. However, prior work involved modifications to the gut microbiome in growing animals and it is unclear if the same changes in the microbiome, applied later in life, would change matrix strength. Here we changed the composition of the gut microbiome before and/or after skeletal maturity (16 weeks of age) using oral antibiotics (ampicillin + neomycin). Male and female mice (n=143 total, n=12-17/group/sex) were allocated into five study groups:1) Unaltered, 2) Continuous (dosing 4-24 weeks of age), 3) Delayed (dosing only 16-24 weeks of age), 4) Initial (dosing 4-16 weeks of age, suspended at 16 weeks), and 5) Reconstituted (dosing from 4-16 weeks following by fecal microbiota transplant from Unaltered donors). Animals were euthanized at 24 weeks of age. In males, bone matrix strength in the femur was 25-35% less than expected from geometry in mice from the Continuous (p= 0.001), Delayed (p= 0.005), and Initial (p=0.040) groups as compared to Unaltered. Reconstitution of the gut microbiota, however, led to a bone matrix strength similar to Unaltered animals (p=0.929). In females, microbiome-induced changes in bone matrix strength followed the same trend as males but were not significantly different, demonstrating sex-related differences in the response of bone matrix to the gut microbiota. Minor differences in chemical composition of bone matrix were observed (Raman spectroscopy). Our findings indicate that microbiome-induced impairment of bone matrix in males can be initiated and/or reversed after skeletal maturity. The portion of the femoral cortical bone formed after skeletal maturity (16 weeks) is small; however, this suggests that microbiome-induced changes in bone matrix occur without osteoblast/osteoclast turnover using an, as of yet unidentified mechanism. These findings add to evidence that the mechanical properties of bone matrix can be altered in the adult skeleton.
Collapse
Affiliation(s)
- C Liu
- Departments of Orthopaedic Surgery and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - E L Cyphert
- Departments of Orthopaedic Surgery and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - S J Stephen
- Shirley Ann Jackson, PhD Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - B Wang
- Shirley Ann Jackson, PhD Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - A L Morales
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - J C Nixon
- Departments of Orthopaedic Surgery and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- Shirley Ann Jackson, PhD Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla, Spain
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Material Science and Engineering, Cornell University, Ithaca, NY, USA
- Reseach Division, Hospital for Special Surgery, New York, NY, USA
- Rensselaer - Icahn School of Medicine at Mount Sinai Center for Engineering and Precision Medicine, New York, NY
- Chan Zuckerberg Biohub San Francisco, CA, US
| | - N R Natsoulas
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - M Garcia
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | | - A C Vill
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - E L Donnelly
- Department of Material Science and Engineering, Cornell University, Ithaca, NY, USA
- Reseach Division, Hospital for Special Surgery, New York, NY, USA
| | - I L Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - D Vashishth
- Shirley Ann Jackson, PhD Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
- Rensselaer - Icahn School of Medicine at Mount Sinai Center for Engineering and Precision Medicine, New York, NY
| | - C J Hernandez
- Departments of Orthopaedic Surgery and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, CA, US
| |
Collapse
|
22
|
Ou Q, Qiao X, Li Z, Niu L, Lei F, Cheng R, Xie T, Yang N, Liu Y, Fu L, Yang J, Mao X, Kou X, Chen C, Shi S. Apoptosis releases hydrogen sulfide to inhibit Th17 cell differentiation. Cell Metab 2024; 36:78-89.e5. [PMID: 38113886 DOI: 10.1016/j.cmet.2023.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/04/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
Over 50 billion cells undergo apoptosis each day in an adult human to maintain immune homeostasis. Hydrogen sulfide (H2S) is also required to safeguard the function of immune response. However, it is unknown whether apoptosis regulates H2S production. Here, we show that apoptosis-deficient MRL/lpr (B6.MRL-Faslpr/J) and Bim-/- (B6.129S1-Bcl2l11tm1.1Ast/J) mice exhibit significantly reduced H2S levels along with aberrant differentiation of Th17 cells, which can be rescued by the additional H2S. Moreover, apoptotic cells and vesicles (apoVs) express key H2S-generating enzymes and generate a significant amount of H2S, indicating that apoptotic metabolism is an important source of H2S. Mechanistically, H2S sulfhydrates selenoprotein F (Sep15) to promote signal transducer and activator of transcription 1 (STAT1) phosphorylation and suppress STAT3 phosphorylation, leading to the inhibition of Th17 cell differentiation. Taken together, this study reveals a previously unknown role of apoptosis in maintaining H2S homeostasis and the unique role of H2S in regulating Th17 cell differentiation via sulfhydration of Sep15C38.
Collapse
Affiliation(s)
- Qianmin Ou
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhengshi Li
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
| | - Luhan Niu
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
| | - Fangcao Lei
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
| | - Ruifeng Cheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100101, China
| | - Ting Xie
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ning Yang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China
| | - Yao Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China
| | - Ling Fu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100101, China
| | - Jing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100101, China
| | - Xueli Mao
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
| | - Xiaoxing Kou
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China; Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Songtao Shi
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China; Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China.
| |
Collapse
|
23
|
Zhou YK, Han CS, Zhu ZL, Chen P, Wang YM, Lin S, Chen LJ, Zhuang ZM, Zhou YH, Yang RL. M2 exosomes modified by hydrogen sulfide promoted bone regeneration by moesin mediated endocytosis. Bioact Mater 2024; 31:192-205. [PMID: 37593496 PMCID: PMC10429289 DOI: 10.1016/j.bioactmat.2023.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023] Open
Abstract
Bone defects caused by trauma or tumor led to high medical costs and poor life quality for patients. The exosomes, micro vesicles of 30-150 nm in diameter, derived from macrophages manipulated bone regeneration. However, the role of hydrogen sulfide (H2S) in the biogenesis and function of exosomes and its effects on bone regeneration remains elusive. In this study, we used H2S slow releasing donor GYY4137 to stimulate macrophages and found that H2S promoted the polarization of M2 macrophages to increase bone regeneration of MSCs in vitro and in vivo. Moreover, we developed the H2S pre-treated M2 macrophage exosomes and found these exosomes displayed significantly higher capacity to promote bone regeneration in calvarial bone defects by re-establishing the local immune microenvironment. Mechanically, H2S treatment altered the protein profile of exosomes derived from M2 macrophages. One of the significantly enriched exosomal proteins stimulated by H2S, moesin protein, facilitated the exosomes endocytosis into MSCs, leading to activated the β-catenin signaling pathway to promote osteogenic differentiation of MSCs. In summary, H2S pretreated M2 exosomes promoted the bone regeneration of MSCs via facilitating exosomes uptake by MSCs and activate β-catenin signaling pathway. This study not only provides new strategies for promoting bone regeneration, but also provides new insights for the effect and mechanism of exosomes internalization.
Collapse
Affiliation(s)
- Yi-kun Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Chun-shan Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Zi-lu Zhu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Peng Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yi-ming Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Shuai Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Liu-jing Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Zi-meng Zhuang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yan-heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Rui-li Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| |
Collapse
|
24
|
Waldbaum JD, Xhumari J, Akinsuyi OS, Arjmandi B, Anton S, Roesch LFW. Association between Dysbiosis in the Gut Microbiota of Primary Osteoporosis Patients and Bone Loss. Aging Dis 2023; 14:2081-2095. [PMID: 37199579 PMCID: PMC10676803 DOI: 10.14336/ad.2023.0425] [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: 01/15/2023] [Accepted: 04/25/2023] [Indexed: 05/19/2023] Open
Abstract
In recent decades, gut microbiome research has experienced significant growth, driven by technological advances that enable quantifying bacterial taxa with greater precision. Age, diet, and living environment have emerged as three key factors influencing gut microbes. Dysbiosis, resulting from alterations in these factors, may lead to changes in bacterial metabolites that regulate pro- and anti-inflammatory processes and consequently impact bone health. Restoration of a healthy microbiome signature could mitigate inflammation and potentially reduce bone loss associated with osteoporosis or experienced by astronauts during spaceflight. However, current research is hindered by contradictory findings, insufficient sample sizes, and inconsistency in experimental conditions and controls. Despite progress in sequencing technology, defining a healthy gut microbiome across global populations remains elusive. Challenges persist in identifying accurate gut bacterial metabolics, specific taxa, and their effects on host physiology. We suggest greater attention be directed towards this issue in Western countries as the cost of treating osteoporosis in the United States reaches billions of dollars annually, with expenses projected to continue rising.
Collapse
Affiliation(s)
- Julien D.H. Waldbaum
- Department of Microbiology and Cell Science, College of Agriculture and Life Sciences, University of Florida, Florida, USA.
| | - Jessica Xhumari
- Department of Microbiology and Cell Science, College of Agriculture and Life Sciences, University of Florida, Florida, USA.
| | - Oluwamayowa S. Akinsuyi
- Department of Microbiology and Cell Science, College of Agriculture and Life Sciences, University of Florida, Florida, USA.
| | - Bahram Arjmandi
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, Florida, USA.
| | - Stephen Anton
- Department of Physiology and Aging, College of Public Health and Health Professions, College of Medicine, University of Florida, Florida, USA.
| | - Luiz Fernando Wurdig Roesch
- Department of Microbiology and Cell Science, College of Agriculture and Life Sciences, University of Florida, Florida, USA.
| |
Collapse
|
25
|
Zhang N, Zhou Z, Huang Y, Wang G, Tang Z, Lu J, Wang C, Ni X. Reduced hydrogen sulfide production contributes to adrenal insufficiency induced by hypoxia via modulation of NLRP3 inflammasome activation. Redox Rep 2023; 28:2163354. [PMID: 36661247 PMCID: PMC9869992 DOI: 10.1080/13510002.2022.2163354] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Objective: Adrenocortical responsiveness is critical for maintaining glucocorticoids production and homeostasis during stress. We sought to investigate adrenocortical responsiveness during hypoxia in mice and the mechanisms responsible for the regulation of adrenal responsiveness.Methods: (1) Adult male WT mice were randomly divided into four groups: normoxia, hypoxia (24h), hypoxia (72h), hypoxia (72h) + GYY4137(hydrogen sulfide (H2S) donor, 133mmol/kg/day); (2) WT mice were randomly divided into four groups: sham, adrenalectomy (ADX), sham+hypoxia, ADX+hypoxia; (3) Cse-/- mice were randomly divided into two groups: Cse-/-, Cse-/- +GYY4137.Results: The circulatory level of corticosteroid induced by ACTH stimulation was significantly reduced in the mice with hypoxia compared with control mice. The mortality rate induced by lipopolysaccharide (LPS) increased during hypoxia. Cystathionine-γ-lyase (CSE) expression was significantly reduced in adrenal glands during hypoxia. GYY4137 treatment significantly increased adrenal responsiveness and attenuated NLRP3 inflammasome activation in mice treated by hypoxia and Cse-/- mice. Furthermore, The sulfhydrated level of PSMA7 in adrenal gland was decreased in the mice with hypoxia and Cse-/- mice. PSMA7 was S-sulfhydrated at cysteine 70. Blockage of S-sulfhydration of PSMA7 increased NLRP3 expression in adrenocortical cells.Conclusion: Reduced H2S production mediated hypo-adrenocortical responsiveness and NLRP3 inflammasome activation via PAMA7 S-sulfhydration during hypoxia.
Collapse
Affiliation(s)
- Ningning Zhang
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China,Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Zhan Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China
| | - Yan Huang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Gang Wang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Zhengshan Tang
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China
| | - Jianqiang Lu
- The Key Laboratory of Exercise and Health Sciences of Ministry of Education, School of Kinesiology, Shanghai University of Sport, Shanghai, People’s Republic of China
| | - Changnan Wang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China, Changnan Wang Department of Physiology, Navy Medical University, Shanghai200433, People’s Republic of China; Xin Ni
| | - Xin Ni
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China,Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China, Changnan Wang Department of Physiology, Navy Medical University, Shanghai200433, People’s Republic of China; Xin Ni
| |
Collapse
|
26
|
Bose S, Robertson SF, Vu AA. Garlic extract enhances bioceramic bone scaffolds through upregulating ALP & BGLAP expression in hMSC-monocyte co-culture. BIOMATERIALS ADVANCES 2023; 154:213622. [PMID: 37742556 DOI: 10.1016/j.bioadv.2023.213622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 08/15/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023]
Abstract
Bone homeostasis is predicated by osteoblast and osteoclast cell cycles where gene expressions are responsible for their differentiation from human mesenchymal stem cells (hMSC) and monocytes, respectively. The pro-osteogenic potential of an hMSC-monocyte co-culture can be measured through complementary DNA (mRNA synthesis) within the nucleus, known as quantitative polymerase chain reaction (qPCR). Through this technique, the effects of garlic extract (allicin) release from calcium phosphate bone scaffolds on gene expression of bone forming and bone remodeling cells was explored. Results show this complex biomaterial system enhances hMSC differentiation through the upregulation of bone-forming proteins. Osteoblastic gene markers alkaline phosphatase (ALP) and osteocalcin (BGLAP), are respectively upregulated by 3-fold and 1.6-fold by day 14. These mature osteoblasts then upregulate the receptor activator of nuclear factor-kB ligand (RANKL) which recruits osteoclast cells, as captured by a nearly 2-fold higher osteoclast expression of tartrate-resistance acid-phosphatase (ACP5). This also activates antagonist osteoprotegerin (OPG) expression in osteoblasts, decreasing osteoclast resorption potential and ACP5 expression by day 21. The pro-osteogenic environment with garlic extract release is further quantified by a 4× increase in phosphatase activity and visibly captured in immunofluorescent tagged confocal images. Also corroborated by enhanced collagen formation in a preliminary in vivo rat distal femur model, this work collectively reveals how garlic extract can enhance bioceramic scaffolds for bone tissue regenerative applications.
Collapse
Affiliation(s)
- Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States.
| | - Samuel F Robertson
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| | - Ashley A Vu
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| |
Collapse
|
27
|
Lyu Z, Hu Y, Guo Y, Liu D. Modulation of bone remodeling by the gut microbiota: a new therapy for osteoporosis. Bone Res 2023; 11:31. [PMID: 37296111 PMCID: PMC10256815 DOI: 10.1038/s41413-023-00264-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/01/2023] [Accepted: 04/18/2023] [Indexed: 06/12/2023] Open
Abstract
The gut microbiota (GM) plays a crucial role in maintaining the overall health and well-being of the host. Recent studies have demonstrated that the GM may significantly influence bone metabolism and degenerative skeletal diseases, such as osteoporosis (OP). Interventions targeting GM modification, including probiotics or antibiotics, have been found to affect bone remodeling. This review provides a comprehensive summary of recent research on the role of GM in regulating bone remodeling and seeks to elucidate the regulatory mechanism from various perspectives, such as the interaction with the immune system, interplay with estrogen or parathyroid hormone (PTH), the impact of GM metabolites, and the effect of extracellular vesicles (EVs). Moreover, this review explores the potential of probiotics as a therapeutic approach for OP. The insights presented may contribute to the development of innovative GM-targeted therapies for OP.
Collapse
Affiliation(s)
- Zhengtian Lyu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
28
|
Mesfin FM, Manohar K, Shelley WC, Brokaw JP, Liu J, Ma M, Markel TA. Stem cells as a therapeutic avenue for active and long-term complications of Necrotizing Enterocolitis. Semin Pediatr Surg 2023; 32:151311. [PMID: 37276782 PMCID: PMC10330659 DOI: 10.1016/j.sempedsurg.2023.151311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal intestinal disease associated with significant morbidity and mortality. Although decades of research have been dedicated to understanding the pathogenesis of NEC and developing therapies, it remains the leading cause of death among neonatal gastrointestinal diseases. Mesenchymal stem cells (MSCs) have garnered significant interest recently as potential therapeutic agents for the treatment of NEC. They have been shown to rescue intestinal injury and reduce the incidence and severity of NEC in various preclinical animal studies. MSCs and MSC-derived organoids and tissue engineered small intestine (TESI) have shown potential for the treatment of long-term sequela of NEC such as short bowel syndrome, neurodevelopmental delay, and chronic lung disease. Although the advances made in the use of MSCs are promising, further research is needed prior to the widespread use of these cells for the treatment of NEC.
Collapse
Affiliation(s)
- Fikir M Mesfin
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Krishna Manohar
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - W Christopher Shelley
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John P Brokaw
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jianyun Liu
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Troy A Markel
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA.
| |
Collapse
|
29
|
Liu F, Yuan L, Li L, Yang J, Liu J, Chen Y, Zhang J, Lu Y, Yuan Y, Cheng J. S-sulfhydration of SIRT3 combats BMSC senescence and ameliorates osteoporosis via stabilizing heterochromatic and mitochondrial homeostasis. Pharmacol Res 2023; 192:106788. [PMID: 37146925 DOI: 10.1016/j.phrs.2023.106788] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 04/23/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Senescence of bone marrow mesenchymal stem cells (BMSCs) is one of the leading causes of osteoporosis. SIRT3, an essential NAD-dependent histone deacetylase, is highly correlated with BMSC senescence-mediated bone degradation and mitochondrial/heterochromatic disturbance. S-sulfhydration of cysteine residues favorably enhances SIRT3 activity by forming persulfides. Nevertheless, the underlying molecular mechanism of SIRT3 S-sulfhydration on mitochondrial/heterochromatic homeostasis involved in BMSC senescence remains unknown. Here, we demonstrated that CBS and CSE, endogenous hydrogen sulfide synthases, are downregulated with BMSC senescence. Exogenous H2S donor NaHS-mediated SIRT3 augmentation rescued the senescent phenotypes of BMSCs. Conversely, SIRT3 deletion accelerated oxidative stress-induced BMSC senescence through mitochondrial dysfunction and the detachment of the heterochromatic protein H3K9me3 from the nuclear envelope protein Lamin B1. H2S-mediated SIRT3 S-sulfhydration modification rescued the disorganized heterochromatin and fragmented mitochondria induced by the S-sulfhydration inhibitor dithiothreitol, thus leading to elevated osteogenic capacity and preventing BMSC senescence. The antisenescence effect of S-sulfhydration modification on BMSCs was abolished when the CXXC sites of the SIRT3 zinc finger motif were mutated. In vivo, aged mice-derived BMSCs pretreated with NaHS were orthotopically transplanted to the ovariectomy-induced osteoporotic mice, and we proved that SIRT3 ameliorates bone loss by inhibiting BMSC senescence. Overall, our study for the first time indicates a novel role of SIRT3 S-sulfhydration in stabilizing heterochromatin and mitochondrial homeostasis in counteracting BMSC senescence, providing a potential target for the treatment of degenerative bone diseases.
Collapse
Affiliation(s)
- Fei Liu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Longhui Yuan
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lan Li
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingchao Yang
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Younan Chen
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhang
- Core Facility of West China Hospital, Sichuan University, Chengdu P.R. China
| | - Yanrong Lu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujia Yuan
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jingqiu Cheng
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Mesfin FM, Manohar K, Hunter CE, Shelley WC, Brokaw JP, Liu J, Ma M, Markel TA. Stem cell derived therapies to preserve and repair the developing intestine. Semin Perinatol 2023; 47:151727. [PMID: 36964032 PMCID: PMC10133028 DOI: 10.1016/j.semperi.2023.151727] [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] [Indexed: 03/26/2023]
Abstract
Stem cell research and the use of stem cells in therapy have seen tremendous growth in the last two decades. Neonatal intestinal disorders such as necrotizing enterocolitis, Hirschsprung disease, and gastroschisis have high morbidity and mortality and limited treatment options with varying success rates. Stem cells have been used in several pre-clinical studies to address various neonatal disorders with promising results. Stem cell and patient population selection, timing of therapy, as well as safety and quality control are some of the challenges that must be addressed prior to the widespread clinical application of stem cells. Further research and technological advances such as the use of cell delivery technology can address these challenges and allow for continued progress towards clinical translation.
Collapse
Affiliation(s)
- Fikir M Mesfin
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Krishna Manohar
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Chelsea E Hunter
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - W Christopher Shelley
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - John P Brokaw
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Jianyun Liu
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY
| | - Troy A Markel
- Department of Surgery, Section of Pediatric Surgery, Indiana University School of Medicine, Indianapolis, IN; Riley Hospital for Children at Indiana University Health, Indianapolis, IN.
| |
Collapse
|
31
|
Liu P, An Y, Zhu T, Tang S, Huang X, Li S, Fu F, Chen J, Xuan K. Mesenchymal stem cells: Emerging concepts and recent advances in their roles in organismal homeostasis and therapy. Front Cell Infect Microbiol 2023; 13:1131218. [PMID: 36968100 PMCID: PMC10034133 DOI: 10.3389/fcimb.2023.1131218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/03/2023] [Indexed: 03/11/2023] Open
Abstract
Stem cells play a crucial role in re-establishing homeostasis in the body, and the search for mechanisms by which they interact with the host to exert their therapeutic effects remains a key question currently being addressed. Considering their significant regenerative/therapeutic potential, research on mesenchymal stem cells (MSCs) has experienced an unprecedented advance in recent years, becoming the focus of extensive works worldwide to develop cell-based approaches for a variety of diseases. Initial evidence for the effectiveness of MSCs therapy comes from the restoration of dynamic microenvironmental homeostasis and endogenous stem cell function in recipient tissues by systemically delivered MSCs. The specific mechanisms by which the effects are exerted remain to be investigated in depth. Importantly, the profound cell-host interplay leaves persistent therapeutic benefits that remain detectable long after the disappearance of transplanted MSCs. In this review, we summarize recent advances on the role of MSCs in multiple disease models, provide insights into the mechanisms by which MSCs interact with endogenous stem cells to exert therapeutic effects, and refine the interconnections between MSCs and cells fused to damaged sites or differentiated into functional cells early in therapy.
Collapse
Affiliation(s)
- Peisheng Liu
- The College of Life Science, Northwest University, Xi’an, Shaanxi, China
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yongqian An
- Department of Stomatology, 962 Hospital of People's Liberation Army of China, Harbin, Heilongjiang, China
| | - Ting Zhu
- The College of Life Science, Northwest University, Xi’an, Shaanxi, China
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Siyuan Tang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University, Xi’an, Shaanxi, China
- School of Basic Medicine, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xiaoyao Huang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Shijie Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Fei Fu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Ji Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Oral Implantology, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
- *Correspondence: Ji Chen, ; Kun Xuan,
| | - Kun Xuan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, China
- *Correspondence: Ji Chen, ; Kun Xuan,
| |
Collapse
|
32
|
Sun J, Wang W, Hu X, Zhang X, Zhu C, Hu J, Ma R. Local delivery of gaseous signaling molecules for orthopedic disease therapy. J Nanobiotechnology 2023; 21:58. [PMID: 36810201 PMCID: PMC9942085 DOI: 10.1186/s12951-023-01813-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Over the past decade, a proliferation of research has used nanoparticles to deliver gaseous signaling molecules for medical purposes. The discovery and revelation of the role of gaseous signaling molecules have been accompanied by nanoparticle therapies for their local delivery. While most of them have been applied in oncology, recent advances have demonstrated their considerable potential in diagnosing and treating orthopedic diseases. Three of the currently recognized gaseous signaling molecules, nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), are highlighted in this review along with their distinctive biological functions and roles in orthopedic diseases. Moreover, this review summarizes the progress in therapeutic development over the past ten years with a deeper discussion of unresolved issues and potential clinical applications.
Collapse
Affiliation(s)
- Jiaxuan Sun
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Wenzhi Wang
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xianli Hu
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xianzuo Zhang
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Jinming Hu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Ruixiang Ma
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
33
|
Titanium dioxide nanotubes increase purinergic receptor P2Y6 expression and activate its downstream PKCα-ERK1/2 pathway in bone marrow mesenchymal stem cells under osteogenic induction. Acta Biomater 2023; 157:670-682. [PMID: 36442823 DOI: 10.1016/j.actbio.2022.11.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/25/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Titanium dioxide (TiO2) nanotubes can improve the osseointegration of pure titanium implants, but this exact mechanism has not been fully elucidated. The purinergic receptor P2Y6 is expressed in bone marrow mesenchymal stem cells (BMSCs) and participates in the regulation of bone metabolism. However, it is unclear as to whether P2Y6 is involved in the osteogenic differentiation of BMSCs induced by TiO2 nanotubes. TiO2 nanotubes were prepared on the surface of titanium specimens using the anodizing method and characterized their features. Quantitative reverse transcriptase polymerase chain reaction and western blotting were used to detect the expression of P2Y6, markers of osteogenic differentiation, and PKCα-ERK1/2. A rat femoral defect model was established to evaluate the osseointegration effect of TiO2 nanotubes combined with P2Y6 agonists. The results showed that the average inner diameter of the TiO2 nanotubes increased with an increase in voltage (voltage range of 30-90V), and the expression of P2Y6 in BMSCs could be upregulated by TiO2 nanotubes in osteogenic culture. Inhibition of P2Y6 expression partially inhibited the osteogenic effect of TiO2 nanotubes and downregulated the activity of the PKCα-ERK1/2 pathway. When using in vitro and in vivo experiments, the osteogenic effect of TiO2 nanotubes when combined with P2Y6 agonists was more pronounced. TiO2 nanotubes promoted the P2Y6 expression of BMSCs during osteogenic differentiation and promoted osteogenesis by activating the PKCα-ERK1/2 pathway. The combined application of TiO2 nanotubes and P2Y6 agonists may be an effective new strategy to improve the osseointegration of titanium implants. STATEMENT OF SIGNIFICANCE: Titanium dioxide (TiO2) nanotubes can improve the osseointegration of pure titanium implants, but this exact mechanism has not been fully elucidated. The purinergic receptor P2Y6 is expressed in bone marrow mesenchymal stem cells (BMSCs) and participates in the regulation of bone metabolism. However, it is unclear as to whether P2Y6 is involved in the osteogenic differentiation of BMSCs induced by TiO2 nanotubes. For the first time, this study revealed the relationship between TiO2 nanotubes and purine receptor P2Y6, and further explored its mode of action, which may provide clues as to the regulatory role of TiO2 nanotubes on osteogenic differentiation of BMSCs. These findings will help to develop novel methods for guiding material design and biosafety evaluation of nano implants.
Collapse
|
34
|
Fraser D, Ganesan SM. Microbiome, alveolar bone, and metabolites: Connecting the dots. FRONTIERS IN DENTAL MEDICINE 2023. [DOI: 10.3389/fdmed.2022.1074339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The oral microbiome (OM) is a diverse and dynamic collection of species, separated from alveolar bone by the oral mucosa. Pathogenic shifts in the OM (dysbiosis) during periodontitis are associated with an inflammatory response in the oral mucosa that drives alveolar bone resorption. Alveolar bone is also affected by metabolic disorders such as osteoporosis. Accumulating evidence has linked another microbial community, the gut microbiome (GM), to systemic bone metabolism and osteoporosis. Underlying this connection is the biologic activity of metabolites, byproducts of host and bacterial activity. Limited evidence also suggests that metabolites in the oral cavity signal between the OM and immune system, influencing both alveolar bone homeostasis and pathologic bone destruction in periodontitis. While the oral cavity and gut are connected through the gastrointestinal tract, dissimilar roles for known metabolites between these two niches exemplify the difficulty in translating knowledge on gut-derived metabolites and bone metabolism to alveolar bone. Integrated metabolomic, transcriptomic, and metagenomic approaches hold promise for resolving these challenges and identifying novel metabolites which impact alveolar bone health. Further interrogation through mechanistic testing in pre-clinical models and carefully controlled clinical studies have potential to lead toward translation of these discoveries into meaningful therapies.
Collapse
|
35
|
On-demand therapeutic delivery of hydrogen sulfide aided by biomolecules. J Control Release 2022; 352:586-599. [PMID: 36328076 DOI: 10.1016/j.jconrel.2022.10.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
Hydrogen sulfide (H2S), known as the third gasotransmitter, exerts various physiological functions including cardiac protection, angiogenesis, anti-inflammatory, and anti-cancer capability. Given its promising therapeutic potential as well as severe perniciousness if improper use, the sustained and tunable H2S delivery systems are highly required for H2S-based gas therapy with enhanced bioactivity and reduced side effects. To this end, a series of stimuli-responsive compounds capable of releasing H2S (termed H2S donors) have been designed over the past two decades to mimic the endogenous generation of H2S and elucidate the biological functions. Further to improve the stability of H2S donors and achieve the targeted delivery, various delivery systems have been constructed. In this review, we focus on the recent advances of an emerging subset, biomolecular-based H2S delivery systems, which combine H2S donors with biomolecular vectors including polysaccharide, peptide, and protein. We demonstrated their basic structures, building strategies, and therapeutic applications respectively to unfold their inherent merits endued by biomolecules including biocompatibility, biodegradability as well as expansibility. The varied development potentials of biomolecular-based H2S delivery systems based on their specific properties are also discussed. At the end, brief future outlooks and upcoming challenges are presented as well.
Collapse
|
36
|
Nassar SZ, Abdelmonsif DA, Ali RG, Badae NM. Sodium hydrosulfide and bone marrow derived mesenchymal stem cells combined therapy for bleomycin induced pulmonary fibrosis in rats: Implication of micro RNA-21 and Lnc GAS5. Life Sci 2022; 309:120988. [PMID: 36155181 DOI: 10.1016/j.lfs.2022.120988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022]
Abstract
AIMS Pulmonary fibrosis (PF) is considered as an end stage for many lung diseases. Mesenchymal stem cells (MSC) as regenerative therapy have become a remarkably valuable therapeutic strategy in different diseases. Hydrogen sulfide has been recently introduced into the medical field for its antifibrotic properties in addition to enhancement of MSC stemness and function. The aim of the present study was to investigate the ability of BM-MSC in combination with NaHS to attenuate Bleomycin induced pulmonary fibrosis was studied in rats. A special emphasis was given to miR-21 and GAS5 as important players in the development of PF. MAIN METHODS PF was induced in 32 Wistar male rats by single endotracheal injection of bleomycin, those were randomly divided into four groups (8 rats each): (untreated PF group) - (PF + MSC) treated group- (PF + NaHS treated group) - PF + combined (NAHS + MSC) treated group. KEY FINDINGS Induction of PF was associated with increased miR-21 and decreased lncRNA-GAS5 expression. Treatment with either NaHS or BM-MSC leads to an inhibitory effect on pulmonary fibrosis as evidenced by improvement of histopathological studies, pulmonary function tests, reduction of inflammatory and fibrotic markers like Hydroxyproline, TNF α, TGF-β and caspase -3 together with downregulation miR-21 and increase lncRNA-GAS5 expression. SIGNIFICANCE The current work revealed the inhibitory effect of combined NaHS and BM-MSC on pulmonary fibrosis with concomitant modulation of miR-21 and lncRNA-GAS5 expression.
Collapse
Affiliation(s)
- Seham Z Nassar
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Doaa A Abdelmonsif
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt; Molecular Biology and Nanomedicine Labs, Centre of Excellence for Regenerative Medicine Research & Applications, University of Alexandria, Alexandria, Egypt
| | - Rania Gaber Ali
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Noha Mohamed Badae
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
37
|
Li S, Lee DJ, Kim HY, Kim JY, Jung YS, Jung HS. Unraveled roles of Cav1.2 in proliferation and stemness of ameloblastoma. Cell Biosci 2022; 12:145. [PMID: 36057617 PMCID: PMC9440535 DOI: 10.1186/s13578-022-00873-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022] Open
Abstract
Background Transcriptome analysis has been known as a functional tool for cancer research recently. Mounting evidence indicated that calcium signaling plays several key roles in cancer progression. Despite numerous studies examining calcium signaling in cancer, calcium signaling studies in ameloblastoma are limited. Results In the present study, comparative transcriptome profiling of two representative odontogenic lesions, ameloblastoma and odontogenic keratocyst, revealed that Cav1.2 (CACNA1C, an L-type voltage-gated calcium channel) is strongly enriched in ameloblastoma. It was confirmed that the Ca2+ influx in ameloblastoma cells is mainly mediated by Cav1.2 through L-type voltage-gated calcium channel agonist and blocking reagent treatment. Overexpression and knockdown of Cav1.2 showed that Cav1.2 is directly involved in the regulation of the nuclear translocation of nuclear factor of activated T cell 1 (NFATc1), which causes cell proliferation. Furthermore, a tumoroid study indicated that Cav1.2-dependent Ca2+ entry is also associated with the maintenance of stemness of ameloblastoma cells via the enhancement of Wnt/β-catenin signaling activity. Conclusion In conclusion, Cav1.2 regulates the NFATc1 nuclear translocation to enhance ameloblastoma cell proliferation. Furthermore, Cav1.2 dependent Ca2+ influx contributes to the Wnt/β-catenin activity for the ameloblastoma cell stemness and tumorigenicity. Our fundamental findings could have a major impact in the fields of oral maxillofacial surgery, and genetic manipulation or pharmacological approaches to Cav1.2 can be considered as new therapeutic options. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00873-9.
Collapse
|
38
|
Gilbert AK, Newton TD, Hettiaratchi MH, Pluth MD. Reactive sulfur and selenium species in the regulation of bone homeostasis. Free Radic Biol Med 2022; 190:148-157. [PMID: 35940516 PMCID: PMC9893879 DOI: 10.1016/j.freeradbiomed.2022.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 02/04/2023]
Abstract
Reactive oxygen species (ROS) are important modulators of physiological signaling and play important roles in bone tissue regulation. Both reactive sulfur species (RSS) and reactive selenium species (RSeS) are involved in ROS signaling, and recent work suggests RSS and RSeS involvement in the regulation of bone homeostasis. For example, RSS can promote osteogenic differentiation and decrease osteoclast activity and differentiation, and the antioxidant activity of RSeS play crucial roles in balancing bone remodeling. Here, we outline current research progress on the application of RSS and RSeS in bone disease and regeneration. Focusing on these investigations, we highlight different methods, tools, and sources of RSS and RSeS, and we also highlight future opportunities for delivery of RSS and RSeS in biological environments relating to bone.
Collapse
Affiliation(s)
- Annie K Gilbert
- Department of Chemistry and Biochemistry, Materials Science Institute, Knight Campus for Accelerating Scientific Impact, Institute of Molecular Biology, University of Oregon, Eugene, OR, 97403, United States
| | - Turner D Newton
- Department of Chemistry and Biochemistry, Materials Science Institute, Knight Campus for Accelerating Scientific Impact, Institute of Molecular Biology, University of Oregon, Eugene, OR, 97403, United States
| | - Marian H Hettiaratchi
- Department of Chemistry and Biochemistry, Materials Science Institute, Knight Campus for Accelerating Scientific Impact, Institute of Molecular Biology, University of Oregon, Eugene, OR, 97403, United States.
| | - Michael D Pluth
- Department of Chemistry and Biochemistry, Materials Science Institute, Knight Campus for Accelerating Scientific Impact, Institute of Molecular Biology, University of Oregon, Eugene, OR, 97403, United States.
| |
Collapse
|
39
|
Roda B, Zhang N, Gambari L, Grigolo B, Eller-Vainicher C, Gennari L, Zappi A, Giordani S, Marassi V, Zattoni A, Reschiglian P, Grassi F. Optimization of a Monobromobimane (MBB) Derivatization and RP-HPLC-FLD Detection Method for Sulfur Species Measurement in Human Serum after Sulfur Inhalation Treatment. Antioxidants (Basel) 2022; 11:antiox11050939. [PMID: 35624802 PMCID: PMC9138032 DOI: 10.3390/antiox11050939] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Hydrogen sulfide (H2S) is a widely recognized gasotransmitter, with key roles in physiological and pathological processes. The accurate quantification of H2S and reactive sulfur species (RSS) may hold important implications for the diagnosis and prognosis of diseases. However, H2S species quantification in biological matrices is still a challenge. Among the sulfide detection methods, monobromobimane (MBB) derivatization coupled with reversed phase high-performance liquid chromatography (RP-HPLC) is one of the most reported. However, it is characterized by a complex preparation and time-consuming process, which may alter the actual H2S level; moreover, a quantitative validation has still not been described. (2) Methods: We developed and validated an improved analytical protocol for the MBB RP-HPLC method. MBB concentration, temperature and sample handling were optimized, and the calibration method was validated using leave-one-out cross-validation and tested in a clinical setting. (3) Results: The method shows high sensitivity and allows the quantification of H2S species, with a limit of detection of 0.5 µM. Finally, it can be successfully applied in measurements of H2S levels in the serum of patients subjected to inhalation with vapors rich in H2S. (4) Conclusions: These data demonstrate that the proposed method is precise and reliable for measuring H2S species in biological matrices and can be used to provide key insights into the etiopathogenesis of several diseases and sulfur-based treatments.
Collapse
Affiliation(s)
- Barbara Roda
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (N.Z.); (A.Z.); (S.G.); (V.M.); (A.Z.); (P.R.)
- byFlow SRL, 40129 Bologna, Italy
- Correspondence: (B.R.); (F.G.)
| | - Nan Zhang
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (N.Z.); (A.Z.); (S.G.); (V.M.); (A.Z.); (P.R.)
| | - Laura Gambari
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (L.G.); (B.G.)
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (L.G.); (B.G.)
| | - Cristina Eller-Vainicher
- Unit of Endocrinology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy;
| | - Alessandro Zappi
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (N.Z.); (A.Z.); (S.G.); (V.M.); (A.Z.); (P.R.)
| | - Stefano Giordani
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (N.Z.); (A.Z.); (S.G.); (V.M.); (A.Z.); (P.R.)
| | - Valentina Marassi
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (N.Z.); (A.Z.); (S.G.); (V.M.); (A.Z.); (P.R.)
- byFlow SRL, 40129 Bologna, Italy
| | - Andrea Zattoni
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (N.Z.); (A.Z.); (S.G.); (V.M.); (A.Z.); (P.R.)
- byFlow SRL, 40129 Bologna, Italy
| | - Pierluigi Reschiglian
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (N.Z.); (A.Z.); (S.G.); (V.M.); (A.Z.); (P.R.)
- byFlow SRL, 40129 Bologna, Italy
| | - Francesco Grassi
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (L.G.); (B.G.)
- Correspondence: (B.R.); (F.G.)
| |
Collapse
|
40
|
Zhou X, Liu J, Zheng Y, Zhang Z, Wu Y, Yang W, Liu J, Huang Y, Yi Y, Zhao Z, Xiao H, Mo X, Wang J. SM22α-lineage niche cells regulate intramembranous bone regeneration via PDGFRβ-triggered hydrogen sulfide production. Cell Rep 2022; 39:110750. [PMID: 35508129 DOI: 10.1016/j.celrep.2022.110750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/02/2022] [Accepted: 04/06/2022] [Indexed: 02/08/2023] Open
Abstract
Bone stromal cells are critical for bone homeostasis and regeneration. Growing evidence suggests that non-stem bone niche cells support bone homeostasis and regeneration via paracrine mechanisms, which remain to be elucidated. Here, we show that physiologically quiescent SM22α-lineage stromal cells expand after bone injury to regulate diverse processes of intramembranous bone regeneration. The majority of SM22α-lineage cells neither act as stem cells in vivo nor show their expression patterns. Dysfunction of SM22α-lineage niche cells induced by loss of platelet-derived growth factor receptor β (PDGFRβ) impairs bone repair. We further show that PDGFRβ-triggered hydrogen sulfide (H2S) generation in SM22α-lineage niche cells facilitates osteogenesis and angiogenesis and suppresses overactive osteoclastogenesis. Collectively, these data demonstrate that non-stem SM22α-lineage niche cells support the niche for bone regeneration with a PDGFRβ/H2S-dependent regulatory mechanism. Our findings provide further insight into non-stem bone stromal niche cell populations and niche-regulation strategy for bone repair.
Collapse
Affiliation(s)
- Xueman Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jin Liu
- Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yingcheng Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yange Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenke Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaqi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanmei Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yating Yi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hengyi Xiao
- Laboratory of Aging Research, State Key Laboratory of Biotherapy & National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xianming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Jun Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
41
|
Ascenção K, Szabo C. Emerging roles of cystathionine β-synthase in various forms of cancer. Redox Biol 2022; 53:102331. [PMID: 35618601 PMCID: PMC9168780 DOI: 10.1016/j.redox.2022.102331] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
The expression of the reverse transsulfuration enzyme cystathionine-β-synthase (CBS) is markedly increased in many forms of cancer, including colorectal, ovarian, lung, breast and kidney, while in other cancers (liver cancer and glioma) it becomes downregulated. According to the clinical database data in high-CBS-expressor cancers (e.g. colon or ovarian cancer), high CBS expression typically predicts lower survival, while in the low-CBS-expressor cancers (e.g. liver cancer), low CBS expression is associated with lower survival. In the high-CBS expressing tumor cells, CBS, and its product hydrogen sulfide (H2S) serves as a bioenergetic, proliferative, cytoprotective and stemness factor; it also supports angiogenesis and epithelial-to-mesenchymal transition in the cancer microenvironment. The current article reviews the various tumor-cell-supporting roles of the CBS/H2S axis in high-CBS expressor cancers and overviews the anticancer effects of CBS silencing and pharmacological CBS inhibition in various cancer models in vitro and in vivo; it also outlines potential approaches for biomarker identification, to support future targeted cancer therapies based on pharmacological CBS inhibition.
Collapse
|
42
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
43
|
Kwack KH, Lee HW. Clinical Potential of Dental Pulp Stem Cells in Pulp Regeneration: Current Endodontic Progress and Future Perspectives. Front Cell Dev Biol 2022; 10:857066. [PMID: 35478967 PMCID: PMC9035692 DOI: 10.3389/fcell.2022.857066] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022] Open
Abstract
Dental caries is a common disease that not only destroys the rigid structure of the teeth but also causes pulp necrosis in severe cases. Once pulp necrosis has occurred, the most common treatment is to remove the damaged pulp tissue, leading to a loss of tooth vitality and increased tooth fragility. Dental pulp stem cells (DPSCs) isolated from pulp tissue exhibit mesenchymal stem cell-like characteristics and are considered ideal candidates for regenerating damaged dental pulp tissue owing to their multipotency, high proliferation rate, and viability after cryopreservation. Importantly, DPSCs do not elicit an allogeneic immune response because they are non-immunogenic and exhibit potent immunosuppressive properties. Here, we provide an up-to-date review of the clinical applicability and potential of DPSCs, as well as emerging trends in the regeneration of damaged pulp tissue. In addition, we suggest the possibility of using DPSCs as a resource for allogeneic transplantation and provide a perspective for their clinical application in pulp regeneration.
Collapse
Affiliation(s)
- Kyu Hwan Kwack
- Department of Dentistry, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Hyeon-Woo Lee
- Department of Pharmacology, School of Dentistry, Graduate School, Institute of Oral Biology, Kyung Hee University, Seoul, South Korea
- *Correspondence: Hyeon-Woo Lee,
| |
Collapse
|
44
|
Hine C, Treviño-Villarreal JH, Mejia P, Longchamp A, Brace LE, Harputlugil E, Mitchell SJ, Yang J, Guan Y, Maciejewski JP, Jha BK, Mitchell JR. Sulfur Amino Acid Supplementation Abrogates Protective Effects of Caloric Restriction for Enhancing Bone Marrow Regrowth Following Ionizing Radiation. Nutrients 2022; 14:nu14071529. [PMID: 35406143 PMCID: PMC9002760 DOI: 10.3390/nu14071529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/30/2022] [Accepted: 04/02/2022] [Indexed: 02/01/2023] Open
Abstract
Radiation therapy damages and depletes total bone marrow (BM) cellularity, compromising safety and limiting effective dosing. Aging also strains total BM and BM hematopoietic stem and progenitor cell (HSPC) renewal and function, resulting in multi-system defects. Interventions that preserve BM and BM HSPC homeostasis thus have potential clinical significance. Here, we report that 50% calorie restriction (CR) for 7-days or fasting for 3-days prior to irradiation improved mouse BM regrowth in the days and weeks post irradiation. Specifically, one week of 50% CR ameliorated loss of total BM cellularity post irradiation compared to ad libitum-fed controls. CR-mediated BM protection was abrogated by dietary sulfur amino acid (i.e., cysteine, methionine) supplementation or pharmacological inhibition of sulfur amino acid metabolizing and hydrogen sulfide (H2S) producing enzymes. Up to 2-fold increased proliferative capacity of ex vivo-irradiated BM isolated from food restricted mice relative to control mice indicates cell autonomy of the protective effect. Pretreatment with H2S in vitro was sufficient to preserve proliferative capacity by over 50% compared to non-treated cells in ex vivo-irradiated BM and BM HSPCs. The exogenous addition of H2S inhibited Ten eleven translocation 2 (TET2) activity in vitro, thus providing a potential mechanism of action. Short-term CR or fasting therefore offers BM radioprotection and promotes regrowth in part via altered sulfur amino acid metabolism and H2S generation, with translational implications for radiation treatment and aging.
Collapse
Affiliation(s)
- Christopher Hine
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Correspondence:
| | - J. Humberto Treviño-Villarreal
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Service of Endocrinology, Department of Internal Medicine, University Hospital and School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey N.L. 64460, Mexico
| | - Pedro Mejia
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Alban Longchamp
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lear E. Brace
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Eylul Harputlugil
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Sarah J. Mitchell
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland
| | - Jie Yang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA;
| | - Yihong Guan
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - Babal K. Jha
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - James R. Mitchell
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland
| |
Collapse
|
45
|
Gupta R, Sahu M, Tripathi R, Ambasta RK, Kumar P. Protein S-sulfhydration: Unraveling the prospective of hydrogen sulfide in the brain, vasculature and neurological manifestations. Ageing Res Rev 2022; 76:101579. [PMID: 35124235 DOI: 10.1016/j.arr.2022.101579] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) and hydrogen polysulfides (H2Sn) are essential regulatory signaling molecules generated by the entire body, including the central nervous system. Researchers have focused on the classical H2S signaling from the past several decades, whereas the last decade has shown the emergence of H2S-induced protein S-sulfhydration signaling as a potential therapeutic approach. Cysteine S-persulfidation is a critical paradigm of post-translational modification in the process of H2S signaling. Additionally, studies have shown the cross-relationship between S-sulfhydration and other cysteine-induced post-translational modifications, namely nitrosylation and carbonylation. In the central nervous system, S-sulfhydration is involved in the cytoprotection through various signaling pathways, viz. inflammatory response, oxidative stress, endoplasmic reticulum stress, atherosclerosis, thrombosis, and angiogenesis. Further, studies have demonstrated H2S-induced S-sulfhydration in regulating different biological processes, such as mitochondrial integrity, calcium homeostasis, blood-brain permeability, cerebral blood flow, and long-term potentiation. Thus, protein S-sulfhydration becomes a crucial regulatory molecule in cerebrovascular and neurodegenerative diseases. Herein, we first described the generation of intracellular H2S followed by the application of H2S in the regulation of cerebral blood flow and blood-brain permeability. Further, we described the involvement of S-sulfhydration in different biological and cellular functions, such as inflammatory response, mitochondrial integrity, calcium imbalance, and oxidative stress. Moreover, we highlighted the importance of S-sulfhydration in cerebrovascular and neurodegenerative diseases.
Collapse
|
46
|
Song S, Guo Y, Yang Y, Fu D. Advances in pathogenesis and therapeutic strategies for osteoporosis. Pharmacol Ther 2022; 237:108168. [PMID: 35283172 DOI: 10.1016/j.pharmthera.2022.108168] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 02/07/2023]
Abstract
Osteoporosis, is the most common bone disorder worldwide characterized by low bone mineral density, leaving affected bones vulnerable to fracture. Bone homeostasis depends on the precise balance between bone resorption by osteoclasts and bone matrix formation by mesenchymal lineage osteoblasts, and involves a series of complex and highly regulated steps. Bone homeostasis will be disrupted when the speed of bone resorption is faster than bone formation. Based on various regulatory mechanisms of bone homeostasis, a series of drugs targeting osteoporosis have emerged in clinical practice, including bisphosphonates, selective estrogen receptor modulators, calcitonin, molecular-targeted drugs and so on. However, many drugs have major adverse effects or are unsuitable for long-term use. Therefore, it is very urgent to find more effective therapeutic drugs based on the new pathogenesis of osteoporosis. In this review, we summarize novel mechanisms involved in the pathological process of osteoporosis, including the roles of gut microbiome, autophagy, iron balance and cellular senescence. Based on the above pathological mechanism, we found promising drugs for osteoporosis treatment, such as: probiotics, alpha-ketoglutarate, senolytics and hydrogen sulfide. This new finding may provide an important basis for elucidating the complex pathological mechanisms of osteoporosis and provide promising drugs for clinical osteoporosis treatment.
Collapse
Affiliation(s)
- Shasha Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, PR China
| | - Yuanyuan Guo
- Department of Pharmacy, Liyuan Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei 430077, PR China
| | - Yuehua Yang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Dehao Fu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China.
| |
Collapse
|
47
|
Glucoraphanin Increases Intracellular Hydrogen Sulfide (H2S) Levels and Stimulates Osteogenic Differentiation in Human Mesenchymal Stromal Cell. Nutrients 2022; 14:nu14030435. [PMID: 35276794 PMCID: PMC8837953 DOI: 10.3390/nu14030435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/11/2022] Open
Abstract
Osteopenia and osteoporosis are among the most prevalent consequences of ageing, urging the promotion of healthy nutritional habits as a tool in preventing bone fractures. Glucosinolates (GLSs) are organosulfur compounds considered relatively inert precursors of reactive derivatives isothiocyanates (ITCs). Recent evidence suggests that GLSs may exert biological properties based on their capacity to release hydrogen sulfide (H2S). H2S-donors are known to exert anabolic function on bone cells. Here, we investigated whether a GLS, glucoraphanin (GRA) obtained from Tuscan black kale, promotes osteogenesis in human mesenchymal stromal cells (hMSCs). H2S release in buffer and intracellular H2S levels were detected by amperometric measurements and fluorimetric/cytofluorimetric analyses, respectively. Alizarin red staining assay and real-time PCR were performed to evaluate mineral apposition and mRNA expression of osteogenic genes. Using an in vitro cell culture model, our data demonstrate a sulforaphane (SFN)-independent osteogenic stimulation of GRA in hMSCs, at least partially attributable to H2S release. In particular, GRA upregulated the expression of osteogenic genes and enhanced mineral apposition while increasing intracellular concentrations of H2S. Overall, this study suggests the feasibility of using cruciferous derivatives as natural alternatives to chemical H2S-donors as adjuvant therapies in the treatment of bone-wasting diseases.
Collapse
|
48
|
Pozzi G, Gobbi G, Masselli E, Carubbi C, Presta V, Ambrosini L, Vitale M, Mirandola P. Buffering Adaptive Immunity by Hydrogen Sulfide. Cells 2022; 11:cells11030325. [PMID: 35159135 PMCID: PMC8834412 DOI: 10.3390/cells11030325] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 02/06/2023] Open
Abstract
T cell-mediated adaptive immunity is designed to respond to non-self antigens and pathogens through the activation and proliferation of various T cell populations. T helper 1 (Th1), Th2, Th17 and Treg cells finely orchestrate cellular responses through a plethora of paracrine and autocrine stimuli that include cytokines, autacoids, and hormones. Hydrogen sulfide (H2S) is one of these mediators able to induce/inhibit immunological responses, playing a role in inflammatory and autoimmune diseases, neurological disorders, asthma, acute pancreatitis, and sepsis. Both endogenous and exogenous H2S modulate numerous important cell signaling pathways. In monocytes, polymorphonuclear, and T cells H2S impacts on activation, survival, proliferation, polarization, adhesion pathways, and modulates cytokine production and sensitivity to chemokines. Here, we offer a comprehensive review on the role of H2S as a natural buffer able to maintain over time a functional balance between Th1, Th2, Th17 and Treg immunological responses.
Collapse
Affiliation(s)
- Giulia Pozzi
- Anatomy Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.P.); (G.G.); (C.C.); (V.P.); (L.A.); (M.V.)
| | - Giuliana Gobbi
- Anatomy Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.P.); (G.G.); (C.C.); (V.P.); (L.A.); (M.V.)
| | - Elena Masselli
- Anatomy Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.P.); (G.G.); (C.C.); (V.P.); (L.A.); (M.V.)
- University Hospital of Parma, AOU-PR, Via Gramsci 14, 43126 Parma, Italy
- Correspondence: (E.M.); (P.M.)
| | - Cecilia Carubbi
- Anatomy Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.P.); (G.G.); (C.C.); (V.P.); (L.A.); (M.V.)
| | - Valentina Presta
- Anatomy Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.P.); (G.G.); (C.C.); (V.P.); (L.A.); (M.V.)
| | - Luca Ambrosini
- Anatomy Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.P.); (G.G.); (C.C.); (V.P.); (L.A.); (M.V.)
| | - Marco Vitale
- Anatomy Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.P.); (G.G.); (C.C.); (V.P.); (L.A.); (M.V.)
- University Hospital of Parma, AOU-PR, Via Gramsci 14, 43126 Parma, Italy
- Italian Foundation for the Research in Balneology, Via Po 22, 00198 Rome, Italy
| | - Prisco Mirandola
- Anatomy Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.P.); (G.G.); (C.C.); (V.P.); (L.A.); (M.V.)
- Correspondence: (E.M.); (P.M.)
| |
Collapse
|
49
|
Gambari L, Grigolo B, Grassi F. Dietary organosulfur compounds: Emerging players in the regulation of bone homeostasis by plant-derived molecules. Front Endocrinol (Lausanne) 2022; 13:937956. [PMID: 36187121 PMCID: PMC9521401 DOI: 10.3389/fendo.2022.937956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
The progressive decline of bone mass and the deterioration of bone microarchitecture are hallmarks of the bone aging. The resulting increase in bone fragility is the leading cause of bone fractures, a major cause of disability. As the frontline pharmacological treatments for osteoporosis suffer from low patients' adherence and occasional side effects, the importance of diet regimens for the prevention of excessive bone fragility has been increasingly recognized. Indeed, certain diet components have been already associated to a reduced fracture risk. Organosulfur compounds are a broad class of molecules containing sulfur. Among them, several molecules of potential therapeutic interest are found in edible plants belonging to the Allium and Brassica botanical genera. Polysulfides derived from Alliaceae and isothiocyanates derived from Brassicaceae hold remarkable nutraceutical potential as anti-inflammatory, antioxidants, vasorelaxant and hypolipemic. Some of these effects are linked to the ability to release the gasotrasmitter hydrogen sulfide (H2S). Recent preclinical studies have investigated the effect of organosulfur compounds in bone wasting and metabolic bone diseases, revealing a strong potential to preserve skeletal health by exerting cytoprotection and stimulating the bone forming activity by osteoblasts and attenuating bone resorption by osteoclasts. This review is intended for revising evidence from preclinical and epidemiological studies on the skeletal effects of organosulfur molecules of dietary origin, with emphasis on the direct regulation of bone cells by plant-derived polysulfides, glucosinolates and isothiocyanates. Moreover, we highlight the potential molecular mechanisms underlying the biological role of these compounds and revise the importance of the so-called 'H2S-system' on the regulation of bone homeostasis.
Collapse
|
50
|
Popović T, Amidžić L, Čeko M, Marković S, Škrbić R. Effect of hydrogen sulphide containing mineral water on experimental osteoporosis in rats. SCRIPTA MEDICA 2022. [DOI: 10.5937/scriptamed53-41462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background/Aim: Sulphur mineral water is widely used in the treatment of musculoskeletal diseases. Hydrogen sulphide is an important regulator of bone metabolism and its application in the treatment of osteoporosis is intensively researched. The aim of this study was to analyse biochemical and histological effects of H2S containing mineral water of "Mlječanica" spring on ovariectomy-induced experimental osteoporosis in rats. Methods: In this experiment a 14-week-old Wistar female rats were used. The animals undergone bilateral ovariectomy (OVX groups) as an experimental model for oestrogen-deficient osteoporosis. After six weeks, animals were divided into control and the experimental group. Rats from the experimental group treated with H2S (SW group) containing mineral water ad libitum during five weeks. Biochemical parameters for monitoring sulphur water effects were concentration in serum of osteocalcin, alkaline phosphatase, calcium and phosphorus. Histological analyses of the left tibia coloured with haematoxylin-eosin were carried out. Results: Regarding the biochemical parameters, a statistically significant increase was observed in the OVX group for osteocalcin, alkaline phosphatase calcium and phosphorus compared to the sham-operated (CNT) group (p < 0.01). In SW + OVX, alkaline phosphatase was statistically significantly decreased (p < 0.01) and serum osteocalcin and phosphorus increased (p < 0.01). Calcium values were increased without significance. In the OVX + SW group, histological analyses showed numerous osteoblasts along the trabecular endosteum and the growth of young chondrocytes in the central bone zone and their migration to the peripheral parts. Conclusion: Drinking the H2S containing "Mlječanica" mineral water has led to decreased alkaline phosphatase, increased osteocalcin and phosphorus concentration in serum and stimulated the bone reparation in osteoporotic rats.
Collapse
|