1
|
Luo Y, Liu B, Qiu Y, Li L, Yang F, Zhang C, Wang J. Divalent metal ions enhance bone regeneration through modulation of nervous systems and metabolic pathways. Bioact Mater 2025; 47:432-447. [PMID: 40034410 PMCID: PMC11872643 DOI: 10.1016/j.bioactmat.2025.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
The divalent metal cations promote new bone formation through modulation of sensory and sympathetic nervous systems (SNS) activities. In addition, acetylcholine (Ach), as a chief neurotransmitter released by the parasympathetic nervous system (PNS), also affects bone remodeling, so it is of worth to investigate if the divalent cations influence PNS activity. Of note, these cations are key co-enzymes modulating glucose metabolism. Aerobic glycolysis rather than oxidative phosphorylation favors osteogenesis of mesenchymal stem cells (MSCs), so it is of interest to study the effects of these cations on glucose metabolic pathway. Prior to biological function assessment, the tolerance limits of the divalent metal cations (Mg2+, Zn2+, and Ca2+) and their combinations were profiled. In terms of direct effects, these divalent cations potentially enhanced migration and adhesion capability of MSCs through upregulating Tgf-β1 and Integrin-β1 levels. Interestingly, the divalent cations alone did not influence osteogenesis and aerobic glycolysis of undifferentiated MSCs. However, once the osteogenic differentiation of MSCs was initiated by neurotransmitters or osteogenic differentiation medium, the osteogenesis of MSCs could be significantly promoted by the divalent cations, which was accompanied by the improved aerobic glycolysis. In terms of indirect effects, the divalent cations significantly upregulated levels of sensory nerve derived CGRP, PNS produced choline acetyltransferase and type H vessels, while significantly tuned down sympathetic activity in the defect zone in rats, thereby contributing to significantly increased bone formation relative to the control group. Together, the divalent cations favor bone regeneration via modulation of sensory-autonomic nervous systems and promotion of aerobic glycolysis-driven osteogenesis of MSCs after osteogenic initiation by neurotransmitters.
Collapse
Affiliation(s)
- Ying Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Baoyi Liu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, Liaoning, China
| | - Yashi Qiu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Lichen Li
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Fan Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, Liaoning, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
2
|
Liu M, Dudley SC. Magnesium Homeostasis and Magnesium Transporters in Human Health. Nutrients 2025; 17:920. [PMID: 40077788 PMCID: PMC11901764 DOI: 10.3390/nu17050920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Magnesium (Mg2+) used to be considered only as a passive cation associated with ATP, but this special issue reinforces the idea that Mg2+ has many more roles [...].
Collapse
Affiliation(s)
- Man Liu
- Cardiovascular Division, Department of Medicine, The Lillehei Heart Institute, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA;
| | | |
Collapse
|
3
|
Zhang J, Ge Q, Du T, Kuang Y, Fan Z, Jia X, Gu W, Chen Z, Wei Z, Shen B. SPHK1/S1PR1/PPAR-α axis restores TJs between uroepithelium providing new ideas for IC/BPS treatment. Life Sci Alliance 2025; 8:e202402957. [PMID: 39578076 PMCID: PMC11584326 DOI: 10.26508/lsa.202402957] [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: 07/24/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) represents a chronic, aseptic inflammatory bladder condition with an unclear etiology and few therapeutic options. A composite barrier structure composed of the uroepithelium and glycosaminoglycan layer forms on the bladder's inner surface to block urine and other harmful substances. Dysfunction of this barrier may initiate the pathogenesis of IC/BPS. Sphingosine-1-phosphate (S1P) plays a crucial role in forming tight junctions. Perfusion of S1P into the bladder restored uroepithelial tight junctions in mice with cyclophosphamide-induced acute cystitis and ameliorated symptoms of the lower urinary tract. Mice lacking sphingosine kinase 1 (SHPK1) exhibited more severe bladder injuries and dysfunction. Concurrent in vitro experiments elucidated S1P's protective effects and its role as a primary messenger through SPHK1 and S1P receptor 1 (S1PR1) knockdown. This study identifies a novel mechanism whereby S1P binding to S1PR1 activates the PPAR-α pathway, thereby enhancing cholesterol transport and restoring tight junctions between uroepithelial cells. These findings elucidate the regulatory role of S1P in the bladder epithelial barrier and highlight a promising therapeutic target for IC/BPS.
Collapse
Affiliation(s)
- Junjie Zhang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Qingyu Ge
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Tianpeng Du
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Yuhao Kuang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Zongyao Fan
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Xinyi Jia
- Respiratory Department, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjin Gu
- Nanjing Medical University, Nanjing, China
| | - Zhengsen Chen
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Zhongqing Wei
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Baixin Shen
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Abidin BM, Rios FJ, Montezano AC, Touyz RM. Transient receptor potential melastatin 7 cation channel, magnesium and cell metabolism in vascular health and disease. Acta Physiol (Oxf) 2025; 241:e14282. [PMID: 39801180 DOI: 10.1111/apha.14282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 12/07/2024] [Accepted: 01/01/2025] [Indexed: 01/30/2025]
Abstract
Preserving the balance of metabolic processes in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), is crucial for optimal vascular function and integrity. ECs are metabolically active and depend on aerobic glycolysis to efficiently produce energy for their essential functions, which include regulating vascular tone. Impaired EC metabolism is linked to endothelial damage, increased permeability and inflammation. Metabolic alterations in VSMCs also contribute to vascular dysfunction in atherosclerosis and hypertension. Magnesium (Mg2+) is the second most abundant intracellular divalent cation and influences molecular processes that regulate vascular function, including vasodilation, vasoconstriction, and release of vasoactive substances. Mg2+ is critically involved in maintaining cellular homeostasis and metabolism since it is an essential cofactor for ATP, nucleic acids and hundreds of enzymes involved in metabolic processes. Low Mg2+ levels have been linked to endothelial dysfunction, increased vascular tone, vascular inflammation and arterial remodeling. Growing evidence indicates an important role for the transient receptor potential melastatin-subfamily member 7 (TRPM7) cation channel in the regulation of Mg2+ homeostasis in EC and VSMCs. In the vasculature, TRPM7 deficiency leads to impaired endothelial function, increased vascular contraction, phenotypic switching of VSMCs, inflammation and fibrosis, processes that characterize the vascular phenotype in hypertension. Here we provide a comprehensive overview on TRPM7/Mg2+ in the regulation of vascular function and how it influences EC and VSMC metabolism such as glucose and energy homeostasis, redox regulation, phosphoinositide signaling, and mineral metabolism. The putative role of TRPM7/Mg2+ and altered cellular metabolism in vascular dysfunction and hypertension is also discussed.
Collapse
Affiliation(s)
- Belma Melda Abidin
- Cardiovascular Health Across the Life Span, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Francisco J Rios
- Cardiovascular Health Across the Life Span, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Augusto C Montezano
- Cardiovascular Health Across the Life Span, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Rhian M Touyz
- Cardiovascular Health Across the Life Span, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Family Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Shen Z, Zhu Y, Yan X, Wu G. Hypomagnesemia is Associated with the Skull Computed Tomography Black Hole Sign in Patients with Spontaneous Intracerebral Hemorrhage. World Neurosurg 2025; 193:483-491. [PMID: 39293736 DOI: 10.1016/j.wneu.2024.09.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVES The evaluation of hypomagnesemia's significance in predicting the presence of the black hole sign in patients with intracranial hemorrhage is currently under investigation. METHODS The study included 261 patients with cerebral hemorrhage who underwent initial skull computed tomography within 24 hours of admission. Sixty-nine patients (26.4%) exhibited hypomagnesemia in the initial laboratory examinations. The black hole sign was observed in 123 patients (referred to as the black hole sign group, which includes patients with and without hypomagnesemia), while the remaining 138 patients (nonblack hole sign group) did not exhibit this feature. The values of hypomagnesemia were assessed through multivariable logistic regression analyses. RESULTS The black hole sign occurred in 45 of the 69 (65.2%) patients with hypomagnesemia, and in 78 of the 192 (40.6%) patients without hypomagnesemia. In the black hole sign group, hypomagnesemia was observed in 45 patients (36.6%). However, only 24 patients (19.5%) from the normal magnesium concentration group exhibited hypomagnesemia. The sensitivity, specificity, and positive and negative predictive values of hypomagnesemia for predicting the black hole sign were 69.9%, 82.5%, 36.6%, and 82.8%, respectively. The odds ratios for hypomagnesemia, smoking history, and hypokalemia in predicting the presence of the black hole sign were 2.74, 1.971, and 1.629, correspondingly. CONCLUSIONS The presence of hypomagnesemia may serve as a predictive factor for the black hole sign and rebleeding in patients with intracerebral hemorrhage, thereby providing valuable guidance for clinical treatment.
Collapse
Affiliation(s)
- Zhengkui Shen
- Department of Emergency Intensive Care Unit, The Mingguang City People's Hospital, Chuzhou, China; Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
| | - Yan Zhu
- Department of Neurological Rehabilitation, Shanghai Second Rehabilitation Hospital, Shanghai, China
| | - Xiaotong Yan
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Guofeng Wu
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
6
|
Peters K, Staehlke S, Rebl H, Jonitz-Heincke A, Hahn O. Impact of Metal Ions on Cellular Functions: A Focus on Mesenchymal Stem/Stromal Cell Differentiation. Int J Mol Sci 2024; 25:10127. [PMID: 39337612 PMCID: PMC11432215 DOI: 10.3390/ijms251810127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Metals play a crucial role in the human body, especially as ions in metalloproteins. Essential metals, such as calcium, iron, and zinc are crucial for various physiological functions, but their interactions within biological networks are complex and not fully understood. Mesenchymal stem/stromal cells (MSCs) are essential for tissue regeneration due to their ability to differentiate into various cell types. This review article addresses the effects of physiological and unphysiological, but not directly toxic, metal ion concentrations, particularly concerning MSCs. Overloading or unbalancing of metal ion concentrations can significantly impair the function and differentiation capacity of MSCs. In addition, excessive or unbalanced metal ion concentrations can lead to oxidative stress, which can affect viability or inflammation. Data on the effects of metal ions on MSC differentiation are limited and often contradictory. Future research should, therefore, aim to clarify the mechanisms by which metal ions affect MSC differentiation, focusing on aspects such as metal ion interactions, ion concentrations, exposure duration, and other environmental conditions. Understanding these interactions could ultimately improve the design of biomaterials and implants to promote MSC-mediated tissue regeneration. It could also lead to the development of innovative therapeutic strategies in regenerative medicine.
Collapse
Affiliation(s)
- Kirsten Peters
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Susanne Staehlke
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Henrike Rebl
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Anika Jonitz-Heincke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Strasse 142, 18057 Rostock, Germany;
| | - Olga Hahn
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| |
Collapse
|
7
|
Fu C, Huang L, Lian C, Yue J, Lin P, Xu L, Lai W, Gao C, Li C, Long Y. Effects of long-term magnesium L-threonate supplementation on neuroinflammation, demyelination and blood-brain barrier integrity in mice with neuromyelitis optica spectrum disorder. Brain Res 2024; 1846:149234. [PMID: 39260790 DOI: 10.1016/j.brainres.2024.149234] [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: 06/18/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
In clinical practice, we found cerebrospinal fluid magnesium concentration significantly lower in neuromyelitis optica spectrum disorder (NMOSD) patients compared to controls with non-autoimmune encephalitis neurological diseases. To investigate the effects and potential mechanisms of long-term magnesium supplementation on neuroinflammation, demyelination, and blood-brain barrier (BBB) integrity in NMOSD, we used two models: (1) NMOSD mouse model, which was induced by intraperitoneal injection of purified NMO-IgG to experimental autoimmune encephalomyelitis (EAE) mice, and (2) cultured human cerebral microvascular endothelial cells/D3 (hCMEC/D3). In the NMOSD mouse model, Magnesium L-threonate (MgT) pretreatment alleviated NMO-IgG-induced effects, including AQP4 loss, leukocyte infiltration, astrocyte and microglia activation, demyelination, decreased tight junction (TJ) protein expression, and neurological deficits. In vitro, MgT pretreatment ameliorated NMO-IgG induced damage to TJ protein expression in a (transient receptor potential melastatin 7) TRPM7-dependent manner. Magnesium supplementation shows potential protective effects against NMOSD, suggesting it may be a novel therapeutic approach for this condition. The beneficial effects appear to be mediated through preservation of blood-brain barrier integrity and reduction of neuroinflammation and demyelination.
Collapse
Affiliation(s)
- Congcong Fu
- Department of Neurology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, 8 Huaying Road, Baiyun District, Guangzhou 510440, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China
| | - Lu Huang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China; Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260, Guangdong Province, China
| | - Chun Lian
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China; Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260, Guangdong Province, China
| | - Jiajia Yue
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China; Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260, Guangdong Province, China
| | - Peihao Lin
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China; Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260, Guangdong Province, China
| | - Lufen Xu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China; Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260, Guangdong Province, China
| | - Wendong Lai
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China; Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260, Guangdong Province, China
| | - Cong Gao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China; Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260, Guangdong Province, China
| | - Chuo Li
- Department of Neurology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, 8 Huaying Road, Baiyun District, Guangzhou 510440, China.
| | - Youming Long
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260 Guangdong Province, China; Institute of Neuroscience and The Second Affiliated Hospital of Guangzhou Medical University, 250# Changgang Road, Guangzhou 510260, Guangdong Province, China.
| |
Collapse
|
8
|
Dai Y, Wu J, Wang J, Wang H, Guo B, Jiang T, Cai Z, Han J, Zhang H, Xu B, Zhou X, Wang C. Magnesium Ions Promote the Induction of Immunosuppressive Bone Microenvironment and Bone Repair through HIF-1α-TGF-β Axis in Dendritic Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311344. [PMID: 38661278 DOI: 10.1002/smll.202311344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/22/2024] [Indexed: 04/26/2024]
Abstract
The effect of immunoinflammation on bone repair during the recovery process of bone defects needs to be further explored. It is reported that Mg2+ can promote bone repair with immunoregulatory effect, but the underlying mechanism on adaptive immunity is still unclear. Here, by using chitosan and hyaluronic acid-coated Mg2+ (CSHA-Mg) in bone-deficient mice, it is shown that Mg2+ can inhibit the activation of CD4+ T cells and increase regulatory T cell formation by inducing immunosuppressive dendritic cells (imDCs). Mechanistically, Mg2+ initiates the activation of the MAPK signaling pathway through TRPM7 channels on DCs. This process subsequently induces the downstream HIF-1α expression, a transcription factor that amplifies TGF-β production and inhibits the effective T cell function. In vivo, knock-out of HIF-1α in DCs or using a HIF-1α inhibitor PX-478 reverses inhibition of bone inflammation and repair promotion upon Mg2+-treatment. Moreover, roxadustat, which stabilizes HIF-1α protein expression, can significantly promote immunosuppression and bone repair in synergism with CSHA-Mg. Thus, the findings identify a key mechanism for DCs and its HIF-1α-TGF-β axis in the induction of immunosuppressive bone microenvironment, providing potential targets for bone regeneration.
Collapse
Affiliation(s)
- Yuya Dai
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Jinhui Wu
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Junyou Wang
- State-Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Haoze Wang
- Nation Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Bingqing Guo
- Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213000, China
| | - Tao Jiang
- Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213000, China
| | - Zhuyun Cai
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Junjie Han
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Haoyu Zhang
- Nation Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Bangzhe Xu
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Xuhui Zhou
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Ce Wang
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| |
Collapse
|
9
|
Rezzani R, Favero G, Gianò M, Pinto D, Labanca M, van Noorden CJ, Rinaldi F. Transient Receptor Potential Channels in the Healthy and Diseased Blood-Brain Barrier. J Histochem Cytochem 2024; 72:199-231. [PMID: 38590114 PMCID: PMC11020746 DOI: 10.1369/00221554241246032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
The large family of transient receptor potential (TRP) channels are integral membrane proteins that function as environmental sensors and act as ion channels after activation by mechanical (touch), physical (heat, pain), and chemical stimuli (pungent compounds such as capsaicin). Most TRP channels are localized in the plasma membrane of cells but some of them are localized in membranes of organelles and function as intracellular Ca2+-ion channels. TRP channels are involved in neurological disorders but their precise role(s) and relevance in these disorders are not clear. Endothelial cells of the blood-brain barrier (BBB) express TRP channels such as TRP vanilloid 1-4 and are involved in thermal detection by regulating BBB permeability. In neurological disorders, TRP channels in the BBB are responsible for edema formation in the brain. Therefore, drug design to modulate locally activity of TRP channels in the BBB is a hot topic. Today, the application of TRP channel antagonists against neurological disorders is still limited.
Collapse
Affiliation(s)
- Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Gaia Favero
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
| | - Marzia Gianò
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, Milan, Italy
| | - Mauro Labanca
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, Milan, Italy
| |
Collapse
|
10
|
Ablooglu AJ, Chen WS, Xie Z, Desai A, Paul S, Lack JB, Scott LA, Eisch AR, Dudek AZ, Parikh SM, Druey KM. Intrinsic endothelial hyperresponsiveness to inflammatory mediators drives acute episodes in models of Clarkson disease. J Clin Invest 2024; 134:e169137. [PMID: 38502192 DOI: 10.1172/jci169137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
Clarkson disease, or monoclonal gammopathy-associated idiopathic systemic capillary leak syndrome (ISCLS), is a rare, relapsing-remitting disorder featuring the abrupt extravasation of fluids and proteins into peripheral tissues, which in turn leads to hypotensive shock, severe hemoconcentration, and hypoalbuminemia. The specific leakage factor(s) and pathways in ISCLS are unknown, and there is no effective treatment for acute flares. Here, we characterize an autonomous vascular endothelial defect in ISCLS that was recapitulated in patient-derived endothelial cells (ECs) in culture and in a mouse model of disease. ISCLS-derived ECs were functionally hyperresponsive to permeability-inducing factors like VEGF and histamine, in part due to increased endothelial nitric oxide synthase (eNOS) activity. eNOS blockade by administration of N(γ)-nitro-l-arginine methyl ester (l-NAME) ameliorated vascular leakage in an SJL/J mouse model of ISCLS induced by histamine or VEGF challenge. eNOS mislocalization and decreased protein phosphatase 2A (PP2A) expression may contribute to eNOS hyperactivation in ISCLS-derived ECs. Our findings provide mechanistic insights into microvascular barrier dysfunction in ISCLS and highlight a potential therapeutic approach.
Collapse
Affiliation(s)
- Ararat J Ablooglu
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Wei-Sheng Chen
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Zhihui Xie
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Abhishek Desai
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Subrata Paul
- Integrative Data Sciences Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Justin B Lack
- Integrative Data Sciences Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Linda A Scott
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - A Robin Eisch
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| | - Arkadiusz Z Dudek
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samir M Parikh
- Division of Nephrology, Departments of Internal Medicine and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kirk M Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, and
| |
Collapse
|
11
|
Kudryavtseva O, Lyngsø KS, Jensen BL, Dimke H. Nitric oxide, endothelium-derived hyperpolarizing factor, and smooth muscle-dependent mechanisms contribute to magnesium-dependent vascular relaxation in mouse arteries. Acta Physiol (Oxf) 2024; 240:e14096. [PMID: 38258597 DOI: 10.1111/apha.14096] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/27/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024]
Abstract
AIM Magnesium (Mg2+ ) is a vasorelaxant. The underlying physiological mechanisms driving this vasorelaxation remain unclear. Studies were designed to test the hypothesis that multiple signaling pathways including nitric oxide (NO) and endothelium-derived hyperpolarizing factor (EDHF) in endothelial cells as well as Ca2+ antagonization and TRPM7 channels in vascular smooth muscle cells mediate Mg2+ -dependent vessel relaxation. METHODS To uncover these mechanisms, force development was measured ex vivo in aorta rings from mice using isometric wire myography. Concentration responses to Mg2+ were studied in intact and endothelium-denuded aortas. Key findings were confirmed in second-order mesenteric resistance arteries perfused ex vivo using pressure myography. Effects of Mg2+ on NO formation were measured in Chinese Hamster Ovary (CHO) cells, isolated mesenteric vessels, and mouse urine. RESULTS Mg2+ caused a significant concentration-dependent relaxation of aorta rings. This relaxation was attenuated significantly in endothelium-denuded aortas. The endothelium-dependent portion was inhibited by NO and cGMP blockade but not by cyclooxygenase inhibition. Mg2+ stimulated local NO formation in CHO cells and isolated mesenteric vessels without changing urinary NOx levels. High extracellular Mg2+ augmented acetylcholine-induced relaxation. SKCa and IKCa channel blockers apamin and TRAM34 inhibited Mg2+ -dependent relaxation. The endothelium-independent relaxation in aorta rings was inhibited by high extracellular Ca2+ . Combined blockade of NO, SKCa , and IKCa channels significantly reduced Mg2+ -dependent dilatation in mesenteric resistance vessels. CONCLUSIONS In mouse conductance and resistance arteries Mg2+ -induced relaxation is contributed by endothelial NO formation, EDHF pathways, antagonism of Ca2+ in smooth muscle cells, and additional unidentified mechanisms.
Collapse
Affiliation(s)
- Olga Kudryavtseva
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Kristina S Lyngsø
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
12
|
Liu L, Wang F, Song W, Zhang D, Lin W, Yin Q, Wang Q, Li H, Yuan Q, Zhang S. Magnesium promotes vascularization and osseointegration in diabetic states. Int J Oral Sci 2024; 16:10. [PMID: 38296940 PMCID: PMC10831079 DOI: 10.1038/s41368-023-00271-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 02/02/2024] Open
Abstract
Diabetes has long been considered a risk factor in implant therapy and impaired wound healing in soft and hard oral tissues. Magnesium has been proved to promote bone healing under normal conditions. Here, we elucidate the mechanism by which Mg2+ promotes angiogenesis and osseointegration in diabetic status. We generated a diabetic mice model and demonstrated the alveolar bone healing was compromised, with significantly decreased angiogenesis. We then developed Mg-coating implants with hydrothermal synthesis. These implants successfully improved the vascularization and osseointegration in diabetic status. Mechanically, Mg2+ promoted the degradation of Kelch-like ECH-associated protein 1 (Keap1) and the nucleation of nuclear factor erythroid 2-related factor 2 (Nrf2) by up-regulating the expression of sestrin 2 (SESN2) in endothelial cells, thus reducing the elevated levels of oxidative stress in mitochondria and relieving endothelial cell dysfunction under hyperglycemia. Altogether, our data suggested that Mg2+ promoted angiogenesis and osseointegration in diabetic mice by regulating endothelial mitochondrial metabolism.
Collapse
Affiliation(s)
- Linfeng Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Feiyu Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Song
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Danting Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Yin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qian Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hanwen Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Li W, Lv BM, Quan Y, Zhu Q, Zhang HY. Associations between Serum Mineral Nutrients, Gut Microbiota, and Risk of Neurological, Psychiatric, and Metabolic Diseases: A Comprehensive Mendelian Randomization Study. Nutrients 2024; 16:244. [PMID: 38257137 PMCID: PMC10818407 DOI: 10.3390/nu16020244] [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: 11/29/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Recent observational studies have reported associations between serum mineral nutrient levels, gut microbiota composition, and neurological, psychiatric, and metabolic diseases. However, the causal effects of mineral nutrients on gut microbiota and their causal associations with diseases remain unclear and require further investigation. This study aimed to identify the associations between serum mineral nutrients, gut microbiota, and risk of neurological, psychiatric, and metabolic diseases using Mendelian randomization (MR). We conducted an MR study using the large-scale genome-wide association study (GWAS) summary statistics of 5 serum mineral nutrients, 196 gut microbes at the phylum, order, family, and genus levels, and a variety of common neurological, psychiatric, and metabolic diseases. Initially, the independent causal associations of mineral nutrients and gut microbiota with diseases were examined by MR. Subsequently, the causal effect of mineral nutrients on gut microbiota was estimated to investigate whether specific gut microbes mediated the association between mineral nutrients and diseases. Finally, we performed sensitivity analyses to assess the robustness of the study results. After correcting for multiple testing, we identified a total of 33 causal relationships among mineral nutrients, gut microbiota, and diseases. Specifically, we found 4 causal relationships between 3 mineral nutrition traits and 3 disease traits, 15 causal associations between 14 gut microbiota traits and 6 disease traits, and 14 causal associations involving 4 mineral nutrition traits and 15 gut microbiota traits. Meanwhile, 118 suggestive associations were identified. The current study reveals multiple causal associations between serum mineral nutrients, gut microbiota, risk of neurological, psychiatric, and metabolic diseases, and potentially provides valuable insights for subsequent nutritional therapies.
Collapse
Affiliation(s)
- Wang Li
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
| | - Bo-Min Lv
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
- Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
| | - Qiang Zhu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
- Key Laboratory of Smart Farming for Agricultural Animals, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
| |
Collapse
|
14
|
Wu J, Cheng X, Wu J, Chen J, Pei X. The development of magnesium-based biomaterials in bone tissue engineering: A review. J Biomed Mater Res B Appl Biomater 2024; 112:e35326. [PMID: 37861271 DOI: 10.1002/jbm.b.35326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/15/2023] [Accepted: 08/23/2023] [Indexed: 10/21/2023]
Abstract
Bone regeneration is a vital clinical challenge in massive or complicated bone defects. Recently, bone tissue engineering has come to the fore to meet the demand for bone repair with various innovative materials. However, the reported materials usually cannot satisfy the requirements, such as ideal mechanical and osteogenic properties, as well as biocompatibility at the same time. Mg-based biomaterials have considerable potential in bone tissue engineering owing to their excellent mechanical strength and biosafety. Moreover, the biocompatibility and osteogenic activity of Mg-based biomaterials have been the research focuses in recent years. The main limitation faced in the applications of Mg-based biomaterials is rapid degradation, which can produce excessive Mg2+ and hydrogen, affecting the healing of the bone defect. In order to overcome the limitations, researchers have explored several ways to improve the properties of Mg-based biomaterials, including alloying, surface modification with coatings, and synthesizing other composite materials to control the degradation rate upon implantation. This article reviewed the osteogenic mechanism and requirement for appropriate degradation rate and focused on current progress in the biomedical use of Mg-based biomaterials to inspire more clinical applications of Mg in bone regeneration in the future.
Collapse
Affiliation(s)
- Jiaxin Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinting Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jicenyuan Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Ding Z, Cheng W, Liu L, Xu G, Lu Q, Kaplan DL. Nanosized Silk-Magnesium Complexes for Tissue Regeneration. Adv Healthc Mater 2023; 12:e2300887. [PMID: 37317936 DOI: 10.1002/adhm.202300887] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/12/2023] [Indexed: 06/16/2023]
Abstract
Metal ions provide multifunctional signals for cell and tissue functions, including regeneration. Inspired by metal-organic frameworks (MOFs), nanosized silk protein aggregates with a high negative charge density are used to form stable silk-magnesium ion complexes. Magnesium ions (Mg ions) are added directly to silk nanoparticle solutions, inducing gelation through the formation of silk-Mg coordination complexes. The Mg ions are released slowly from the nanoparticles through diffusion, with sustained release via tuning the degradation or dissolution of the nanosized silk aggregates. Studies in vitro reveal a dose-dependent influence of Mg ions on angiogenic and anti-inflammatory functions. Silk-Mg ion complexes in the form of hydrogels also stimulate tissue regeneration with a reduced formation of scar tissue in vivo, suggesting potential utility in tissue regeneration.
Collapse
Affiliation(s)
- Zhaozhao Ding
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123, P. R. China
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, 215123, P. R. China
| | - Weinan Cheng
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai, 200233, P. R. China
- Department of Orthopedics, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, P. R. China
| | - Lutong Liu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, 215123, P. R. China
| | - Gang Xu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Lianyungang, 222061, P. R. China
| | - Qiang Lu
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123, P. R. China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
16
|
Mohebi F, Ostadhadi S, Vaziri MS, Hassanzadeh M, Koochakkhani S, Azarkish F, Farshidi H, Eftekhar E. The effect of magnesium sulfate on gene expression and serum level of inflammatory cytokines in coronary artery disease patients. Inflammopharmacology 2023; 31:2421-2430. [PMID: 37665448 DOI: 10.1007/s10787-023-01328-4] [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: 04/16/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023]
Abstract
PURPOSE To evaluate the effect of oral magnesium sulfate (MgSO4) on the gene expression and serum levels of inflammatory cytokines including TNF-α, IL-18, IL-1β, IL-6, and IFN-γ in patients with moderate coronary artery disease (CAD). METHODS 60 CAD patients were selected based on angiography findings and were randomly divided into two groups that received 300 mg/day MgSO4 (n = 30) or placebo (n = 30) for 3 months. Gene expression and serum levels of inflammatory cytokines were assessed. RESULTS After 3 months of intervention, gene expression and serum levels of IL-18 and TNF-α in the MgSO4 group were significantly less than the placebo group (P < 0.05). However, no significant difference in gene expression and serum levels of IL-1β, IL-6, and IFN-γ was observed between the two groups (P > 0.05). In addition, within group analysis demonstrate that Mg-treatment significantly decrease serum level of TNF-α and IL-18 as compared to pretreatment. CONCLUSION The results of our study demonstrate that 3-month magnesium sulfate administration (300 mg/day) to CAD patients could significantly decrease serum concentration and gene expression levels of IL-18 and TNF-α. Our findings support the potential beneficial effect of magnesium supplementation on alleviating CAD complications through modulating inflammatory cytokines.
Collapse
Affiliation(s)
- Fatemeh Mohebi
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Samane Ostadhadi
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Sadegh Vaziri
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Marziyeh Hassanzadeh
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Shabnaz Koochakkhani
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fariba Azarkish
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hossein Farshidi
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ebrahim Eftekhar
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
17
|
Liu M, Dudley SC. Beyond Ion Homeostasis: Hypomagnesemia, Transient Receptor Potential Melastatin Channel 7, Mitochondrial Function, and Inflammation. Nutrients 2023; 15:3920. [PMID: 37764704 PMCID: PMC10536927 DOI: 10.3390/nu15183920] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/02/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
As the second most abundant intracellular divalent cation, magnesium (Mg2+) is essential for cell functions, such as ATP production, protein/DNA synthesis, protein activity, and mitochondrial function. Mg2+ plays a critical role in heart rhythm, muscle contraction, and blood pressure. A significant decline in Mg2+ intake has been reported in developed countries because of the increased consumption of processed food and filtered/deionized water, which can lead to hypomagnesemia (HypoMg). HypoMg is commonly observed in cardiovascular diseases, such as heart failure, hypertension, arrhythmias, and diabetic cardiomyopathy, and HypoMg is a predictor for cardiovascular and all-cause mortality. On the other hand, Mg2+ supplementation has shown significant therapeutic effects in cardiovascular diseases. Some of the effects of HypoMg have been ascribed to changes in Mg2+ participation in enzyme activity, ATP stabilization, enzyme kinetics, and alterations in Ca2+, Na+, and other cations. In this manuscript, we discuss new insights into the pathogenic mechanisms of HypoMg that surpass previously described effects. HypoMg causes mitochondrial dysfunction, oxidative stress, and inflammation. Many of these effects can be attributed to the HypoMg-induced upregulation of a Mg2+ transporter transient receptor potential melastatin 7 channel (TRMP7) that is also a kinase. An increase in kinase signaling mediated by HypoMg-induced TRPM7 transcriptional upregulation, independently of any change in Mg2+ transport function, likely seems responsible for many of the effects of HypoMg. Therefore, Mg2+ supplementation and TRPM7 kinase inhibition may work to treat the sequelae of HypoMg by preventing increased TRPM7 kinase activity rather than just altering ion homeostasis. Since many diseases are characterized by oxidative stress or inflammation, Mg2+ supplementation and TRPM7 kinase inhibition may have wider implications for other diseases by acting to reduce oxidative stress and inflammation.
Collapse
Affiliation(s)
- Man Liu
- Cardiovascular Division, Department of Medicine, The Lillehei Heart Institute, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA;
| | | |
Collapse
|
18
|
Fujita K, Shindo Y, Katsuta Y, Goto M, Hotta K, Oka K. Intracellular Mg 2+ protects mitochondria from oxidative stress in human keratinocytes. Commun Biol 2023; 6:868. [PMID: 37620401 PMCID: PMC10449934 DOI: 10.1038/s42003-023-05247-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Reactive oxygen species (ROS) are harmful for the human body, and exposure to ultraviolet irradiation triggers ROS generation. Previous studies have demonstrated that ROS decrease mitochondrial membrane potential (MMP) and that Mg2+ protects mitochondria from oxidative stress. Therefore, we visualized the spatio-temporal dynamics of Mg2+ in keratinocytes (a skin component) in response to H2O2 (a type of ROS) and found that it increased cytosolic Mg2+ levels. H2O2-induced responses in both Mg2+ and ATP were larger in keratinocytes derived from adults than in keratinocytes derived from newborns, and inhibition of mitochondrial ATP synthesis enhanced the H2O2-induced Mg2+ response, indicating that a major source of Mg2+ was dissociation from ATP. Simultaneous imaging of Mg2+ and MMP revealed that larger Mg2+ responses corresponded to lower decreases in MMP in response to H2O2. Moreover, Mg2+ supplementation attenuated H2O2-induced cell death. These suggest the potential of Mg2+ as an active ingredient to protect skin from oxidative stress.
Collapse
Affiliation(s)
- Keigo Fujita
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Yutaka Shindo
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan
- School of Frontier Engineering, Kitasato University, Sagamihara, Japan
| | - Yuji Katsuta
- MIRAI Technology Institute, Shiseido Co. Ltd., Yokohama, Japan
| | - Makiko Goto
- MIRAI Technology Institute, Shiseido Co. Ltd., Yokohama, Japan
| | - Kohji Hotta
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Kotaro Oka
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan.
- School of Frontier Engineering, Kitasato University, Sagamihara, Japan.
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo, Japan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan.
| |
Collapse
|
19
|
Locatelli L, Fedele G, Maier JA. The Role of Txnip in Mediating Low-Magnesium-Driven Endothelial Dysfunction. Int J Mol Sci 2023; 24:ijms24098351. [PMID: 37176057 PMCID: PMC10179684 DOI: 10.3390/ijms24098351] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Magnesium deficiency is associated with a greater risk of developing cardiovascular diseases since this cation is fundamental in regulating vascular function. This clinical evidence is sustained by in vitro studies showing that culturing endothelial cells in low concentrations of magnesium promotes the acquisition of a pro-oxidant and pro-inflammatory phenotype. Here, we show that the increase in reactive oxygen species in endothelial cells in low-magnesium-containing medium is due to the upregulation of the pro-oxidant protein thioredoxin interacting protein (TXNIP), with a consequent accumulation of lipid droplets and increase in endothelial permeability through the downregulation and relocalization of junctional proteins. Silencing TXNIP restores the endothelial barrier and lipid content. Because (i) mitochondria serve multiple roles in shaping cell function, health and survival and (ii) mitochondria are the main intracellular stores of magnesium, it is of note that no significant alterations were detected in their morphology and dynamics in our experimental model. We conclude that TXNIP upregulation contributes to low-magnesium-induced endothelial dysfunction in vitro.
Collapse
Affiliation(s)
- Laura Locatelli
- Department of Biomedical and Clinical Sciences, Università di Milano, Via GB Grassi 74, 20157 Milano, Italy
| | - Giorgia Fedele
- Department of Biomedical and Clinical Sciences, Università di Milano, Via GB Grassi 74, 20157 Milano, Italy
| | - Jeanette A Maier
- Department of Biomedical and Clinical Sciences, Università di Milano, Via GB Grassi 74, 20157 Milano, Italy
| |
Collapse
|
20
|
The TRPM7 channel reprograms cellular glycolysis to drive tumorigenesis and angiogenesis. Cell Death Dis 2023; 14:183. [PMID: 36878949 PMCID: PMC9988972 DOI: 10.1038/s41419-023-05701-7] [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: 11/14/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023]
Abstract
Cancer or endothelial cells preferably catabolize glucose through aerobic glycolysis rather than oxidative phosphorylation. Intracellular ionic signaling has been shown to regulate glucose metabolism, but the underlying ion channel has yet to be identified. RNA-seq, metabolomics and genetic assay revealed that the TRPM7 channel regulated cellular glycolysis. Deletion of TRPM7 suppressed cancer cell glycolysis and reduced the xenograft tumor burden. Deficiency of endothelial TRPM7 inhibited postnatal retinal angiogenesis in mice. Mechanistically, TRPM7 transcriptionally regulated the solute carrier family 2 member 3 (SLC2A3, also known as GLUT3) via Ca2+ influx-induced calcineurin activation. Furthermore, CREB-regulated transcription coactivator 2 (CRTC2) and CREB act downstream of calcineurin to relay Ca2+ signal to SLC2A3 transcription. Expression of the constitutively active CRTC2 or CREB in TRPM7 knockout cell normalized glycolytic metabolism and cell growth. The TRPM7 channel represents a novel regulator of glycolytic reprogramming. Inhibition of the TRPM7-dependent glycolysis could be harnessed for cancer therapy.
Collapse
|
21
|
Ren B, Liu R, He Q, Wu T, Song L, Wang H, Gu J. Stimulus-Responsive Zwitterionic Prodrug Delivery System with Sustained Release of Hydrogen Sulfide for Protective Aortic Dissection. ACS APPLIED MATERIALS & INTERFACES 2023; 15:9099-9109. [PMID: 36759500 DOI: 10.1021/acsami.2c21460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Aortic dissection (AD) is one of the most frequent types of aortic disease with extremely poor prognosis. The biological signaling gas hydrogen sulfide (H2S) has exhibited protective effects in various types of cardiovascular diseases. However, as a toxic, colorless gas, the application of H2S is immensely hampered due to the lack of ideal donors. In this article, a drug delivery system with a H2S donor has been prepared. Meanwhile, the donor could be deposed in a cysteine-containing environment to generate H2S. The results indicate that the H2S donor polymer nanomicelles mitigated the processive transformation of smooth muscle cells effectively in a proper concentration range, which may play a protective role in aortic dissection. In animal experiments, the sustained-release H2S donor stimulated in the presence of cysteine was found to demonstrate beneficial effects in a murine model of aortic dissection and would likely become a potential target of H2S therapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Bibo Ren
- Department of Cardiovascular surgery, West China Hospital, Sichuan University, Chengdu 610065, P. R. China
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Ruiqi Liu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu 610065, P. R. China
| | - Qian He
- Department of Emergency, West China Hospital, Sichuan University, Chengdu 610065, P. R. China
| | - Tongyi Wu
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, P. R. China
| | - Lei Song
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, P. R. China
| | - Haibo Wang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Jun Gu
- Department of Cardiovascular surgery, West China Hospital, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
22
|
Liu Q, Li S, Qiu Y, Zhang J, Rios FJ, Zou Z, Touyz RM. Cardiovascular toxicity of tyrosine kinase inhibitors during cancer treatment: Potential involvement of TRPM7. Front Cardiovasc Med 2023; 10:1002438. [PMID: 36818331 PMCID: PMC9936099 DOI: 10.3389/fcvm.2023.1002438] [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: 07/25/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) are a class of membrane spanning cell-surface receptors that transmit extracellular signals through the membrane to trigger diverse intracellular signaling through tyrosine kinases (TKs), and play important role in cancer development. Therapeutic approaches targeting RTKs such as vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor (EGFR), and platelet-derived growth factor receptor (PDGFR), and TKs, such as c-Src, ABL, JAK, are widely used to treat human cancers. Despite favorable benefits in cancer treatment that prolong survival, these tyrosine kinase inhibitors (TKIs) and monoclonal antibodies targeting RTKs are also accompanied by adverse effects, including cardiovascular toxicity. Mechanisms underlying TKI-induced cardiovascular toxicity remain unclear. The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed chanzyme consisting of a membrane-based ion channel and intracellular α-kinase. TRPM7 is a cation channel that regulates transmembrane Mg2+ and Ca2+ and is involved in a variety of (patho)physiological processes in the cardiovascular system, contributing to hypertension, cardiac fibrosis, inflammation, and atrial arrhythmias. Of importance, we and others demonstrated significant cross-talk between TRPM7, RTKs, and TK signaling in different cell types including vascular smooth muscle cells (VSMCs), which might be a link between TKIs and their cardiovascular effects. In this review, we summarize the implications of RTK inhibitors (RTKIs) and TKIs in cardiovascular toxicities during anti-cancer treatment, with a focus on the potential role of TRPM7/Mg2+ as a mediator of RTKI/TKI-induced cardiovascular toxicity. We also describe the important role of TRPM7 in cancer development and cardiovascular diseases, and the interaction between TRPM7 and RTKs, providing insights for possible mechanisms underlying cardiovascular disease in cancer patients treated with RTKI/TKIs.
Collapse
Affiliation(s)
- Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuran Qiu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayu Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Francisco J. Rios
- Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Zhiguo Zou
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Zhiguo Zou ✉
| | - Rhian M. Touyz
- Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada,*Correspondence: Rhian M. Touyz ✉
| |
Collapse
|
23
|
Coman AE, Ceasovschih A, Petroaie AD, Popa E, Lionte C, Bologa C, Haliga RE, Cosmescu A, Slănină AM, Bacușcă AI, Șorodoc V, Șorodoc L. The Significance of Low Magnesium Levels in COVID-19 Patients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020279. [PMID: 36837480 PMCID: PMC9965430 DOI: 10.3390/medicina59020279] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/22/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023]
Abstract
Magnesium is the fourth most common mineral in the human body and the second richest intracellular cation. This element is necessary for many physiological reactions, especially in the cardiovascular and respiratory systems. COVID-19 is an infectious disease caused by SARS-CoV-2. The majority of people who become ill as a result of COVID-19 have mild-to-moderate symptoms and recover without specific treatment. Moreover, there are people who develop severe forms of COVID-19, which require highly specialized medical assistance. Magnesium deficiency may play a role in the pathophysiology of infection with SARS-CoV-2. The primary manifestation of COVID-19 remains respiratory, but the virus can spread to other organs and tissues, complicating the clinical picture and culminating in multiorgan failure. The key mechanisms involved in the disease include direct viral cytotoxicity, endothelial dysfunction, and exaggerated release of inflammatory cytokines. The aim of this review was to summarize the available data regarding the role of magnesium in COVID-19 patients and its particularities in different clinical settings.
Collapse
Affiliation(s)
- Adorata Elena Coman
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Alexandr Ceasovschih
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Correspondence: (A.C.); (C.L.)
| | - Antoneta Dacia Petroaie
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Elena Popa
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Cătălina Lionte
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (A.C.); (C.L.)
| | - Cristina Bologa
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Raluca Ecaterina Haliga
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Adriana Cosmescu
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Ana Maria Slănină
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Agnes Iacinta Bacușcă
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Victorița Șorodoc
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laurențiu Șorodoc
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
24
|
Globig P, Madurawala R, Willumeit-Römer R, Martini F, Mazzoni E, Luthringer-Feyerabend BJ. Mg-based materials diminish tumor spreading and cancer metastases. Bioact Mater 2023; 19:594-610. [PMID: 35600975 PMCID: PMC9108521 DOI: 10.1016/j.bioactmat.2022.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/03/2022] [Accepted: 05/03/2022] [Indexed: 11/26/2022] Open
Abstract
Cancer metastases are the most common causes of cancer-related deaths. The formation of secondary tumors at different sites in the human body can impair multiple organ function and dramatically decrease the survival of the patients. In this stage, it is difficulty to treat tumor growth and spreading due to arising therapy resistances. Therefore, it is important to prevent cancer metastases and to increase subsequent cancer therapy success. Cancer metastases are conventionally treated with radiation or chemotherapy. However, these treatments elicit lots of side effects, wherefore novel local treatment approaches are currently discussed. Recent studies already showed anticancer activity of specially designed degradable magnesium (Mg) alloys by reducing the cancer cell proliferation. In this work, we investigated the impact of these Mg-based materials on different steps of the metastatic cascade including cancer cell migration, invasion, and cancer-induced angiogenesis. Both, Mg and Mg–6Ag reduced cell migration and invasion of osteosarcoma cells in coculture with fibroblasts. Furthermore, the Mg-based materials used in this study diminished the cancer-induced angiogenesis. Endothelial cells incubated with conditioned media obtained from these Mg and Mg–6Ag showed a reduced cell layer permeability, a reduced proliferation and inhibited cell migration. The tube formation as a last step of angiogenesis was stimulated with the presence of Mg under normoxia and diminished under hypoxia. Magnesium (Mg)-based material degradation decrease cell migration and invasion of an osteosarcoma coculture. Mg-based material degradation products reduce cancer-induced angiogenesis at an early stage. These materials may reduce secondary tumor formation and metastases.
Collapse
|
25
|
Li X, Dai B, Guo J, Zhu Y, Xu J, Xu S, Yao Z, Chang L, Li Y, He X, Chow DHK, Zhang S, Yao H, Tong W, Ngai T, Qin L. Biosynthesized Bandages Carrying Magnesium Oxide Nanoparticles Induce Cortical Bone Formation by Modulating Endogenous Periosteal Cells. ACS NANO 2022; 16:18071-18089. [PMID: 36108267 DOI: 10.1021/acsnano.2c04747] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Bone grafting is frequently conducted to treat bone defects caused by trauma and tumor removal, yet with significant medical and socioeconomic burdens. Space-occupying bone substitutes remain challenging in the control of osteointegration, and meanwhile activation of endogenous periosteal cells by using non-space-occupying implants to promote new bone formation becomes another therapeutic strategy. Here, we fabricated a magnesium-based artificial bandage with optimal micropatterns for activating periosteum-associated biomineralization. Collagen was self-assembled on the surface of magnesium oxide nanoparticles embedded electrospun fibrous membranes as a hierarchical bandage structure to facilitate the integration with periosteum in situ. After the implantation on the surface of cortical bone in vivo, magnesium ions were released to generate a pro-osteogenic immune microenvironment by activating the endogenous periosteal macrophages into M2 phenotype and, meanwhile, promote blood vessel formation and neurite outgrowth. In a cortical bone defect model, magnesium-based artificial bandage guided the surrounding newly formed bone tissue to cover the defected area. Taken together, our study suggests that the strategy of stimulating bone formation can be achieved with magnesium delivery to periosteum in situ and the proposed periosteal bandages act as a bioactive media for accelerating bone healing.
Collapse
Affiliation(s)
- Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Yuwei Zhu
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Shunxiang Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Zhi Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Ye Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Xuan He
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Shian Zhang
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| |
Collapse
|
26
|
Oost LJ, Tack CJ, de Baaij JHF. Hypomagnesemia and Cardiovascular Risk in Type 2 Diabetes. Endocr Rev 2022; 44:357-378. [PMID: 36346820 PMCID: PMC10166267 DOI: 10.1210/endrev/bnac028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/22/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Hypomagnesemia is tenfold more common in individuals with type 2 diabetes (T2D), compared to the healthy population. Factors that are involved in this high prevalence are low Mg2+ intake, gut microbiome composition, medication use and presumably genetics. Hypomagnesemia is associated with insulin resistance, which subsequently increases the risk to develop T2D or deteriorates glycaemic control in existing diabetes. Mg2+ supplementation decreases T2D associated features like dyslipidaemia and inflammation; which are important risk factors for cardiovascular disease (CVD). Epidemiological studies have shown an inverse association between serum Mg2+ and the risk to develop heart failure (HF), atrial fibrillation (AF) and microvascular disease in T2D. The potential protective effect of Mg2+ on HF and AF may be explained by reduced oxidative stress, fibrosis and electrical remodeling in the heart. In microvascular disease, Mg2+ reduces the detrimental effects of hyperglycemia and improves endothelial dysfunction. Though, clinical studies assessing the effect of long-term Mg2+ supplementation on CVD incidents are lacking and gaps remain on how Mg2+ may reduce CVD risk in T2D. Despite the high prevalence of hypomagnesemia in people with T2D, routine screening of Mg2+ deficiency to provide Mg2+ supplementation when needed is not implemented in clinical care as sufficient clinical evidence is lacking. In conclusion, hypomagnesemia is common in people with T2D and is both involved as cause, probably through molecular mechanisms leading to insulin resistance, and consequence and is prospectively associated with development of HF, AF and microvascular complications. Whether long-term supplementation of Mg2+ is beneficial, however, remains to be determined.
Collapse
Affiliation(s)
- Lynette J Oost
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
27
|
Yuan Z, Wan Z, Gao C, Wang Y, Huang J, Cai Q. Controlled magnesium ion delivery system for in situ bone tissue engineering. J Control Release 2022; 350:360-376. [PMID: 36002052 DOI: 10.1016/j.jconrel.2022.08.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
Abstract
Magnesium cation (Mg2+) has been an emerging therapeutic agent for inducing vascularized bone regeneration. However, the therapeutic effects of current magnesium (Mg) -containing biomaterials are controversial due to the concentration- and stage-dependent behavior of Mg2+. Here, we first provide an overview of biochemical mechanism of Mg2+ in various concentrations and suggest that 2-10 mM Mg2+in vitro may be optimized. This review systematically summarizes and discusses several types of controlled Mg2+ delivery systems based on polymer-Mg composite scaffolds and Mg-containing hydrogels, as well as their design philosophy and several parameters that regulate Mg2+ release. Given that the continuous supply of Mg2+ may prevent biomineral deposition in the later stage of bone regeneration and maturation, we highlight the controlled delivery of Mg2+ based dual- or multi-ions system, especially for the hierarchical therapeutic ion release system, which shows enhanced biomineralization. Finally, the remaining challenges and perspectives of Mg-containing biomaterials for future in situ bone tissue engineering are discussed as well.
Collapse
Affiliation(s)
- Zuoying Yuan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Zhuo Wan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Chenyuan Gao
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yue Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China.
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China..
| |
Collapse
|
28
|
Nahalka J. 1-L Transcription in Alzheimer's Disease. Curr Issues Mol Biol 2022; 44:3533-3551. [PMID: 36005139 PMCID: PMC9406503 DOI: 10.3390/cimb44080243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease is a very complex disease and better explanations and models are needed to understand how neurons are affected and microglia are activated. A new model of Alzheimer's disease is presented here, the β-amyloid peptide is considered an important RNA recognition/binding peptide. 1-L transcription revealed compatible sequences with AAUAAA (PAS signal) and UUUC (class III ARE rich in U) in the Aβ peptide, supporting the peptide-RNA regulatory model. When a hypothetical model of fibril selection with the prionic character of amyloid assemblies is added to the peptide-RNA regulatory model, the downregulation of the PI3K-Akt pathway and the upregulation of the PLC-IP3 pathway are well explained. The model explains why neurons are less protected from inflammation and why microglia are activated; why mitochondria are destabilized; why the autophagic flux is destabilized; and why the post-transcriptional attenuation of the axonal signal "noise" is interrupted. For example, the model suggests that Aβ peptide may post-transcriptionally control ELAVL2 (ELAV-like RNA binding protein 2) and DCP2 (decapping mRNA protein 2), which are known to regulate RNA processing, transport, and stability.
Collapse
Affiliation(s)
- Jozef Nahalka
- Institute of Chemistry, Centre for Glycomics, Slovak Academy of Sciences, Dubravska Cesta 9, SK-84538 Bratislava, Slovakia;
- Institute of Chemistry, Centre of Excellence for White-Green Biotechnology, Slovak Academy of Sciences, Trieda Andreja Hlinku 2, SK-94976 Nitra, Slovakia
| |
Collapse
|
29
|
Peraçoli JC, Silva PB, Neves HM, Borges VTM, Abbade JF, Costa RAA, Batista FRG, Peraçoli MTS, Romão-Veiga M. Modulatory effect of two regimens of magnesium sulfate on the systemic inflammatory response in pregnant women with imminent eclampsia. Pregnancy Hypertens 2022; 29:46-53. [PMID: 35728369 DOI: 10.1016/j.preghy.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/30/2022] [Accepted: 06/12/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE This study compared the modulatory effect of two intravenous magnesium sulfate (MgSO4) regimens on the systemic inflammatory response in pregnant women diagnosed with imminent eclampsia. STUDY DESIGN In a single-blind cross-sectional study, 33 women were allocated according to the Zuspan (n = 16) and Sibai (n = 17) MgSO4 regimens, and treated for 24 h. Blood samples were collected pre-administration of the loading dose, at 24 h of the maintenance dose of MgSO4, and at 48 h, when patients were without treatment. Plasma was used to determine interleukin (IL)-1 beta (IL-1β), IL-6, IL-10, tumor necrosis factor-alpha (TNF-α), heat shock protein (Hsp70), and heme oxygenase-1 (HO-1) by ELISA. RESULTS The treatment with the Zuspan's regimen didn't change plasma concentrations of TNF-α, IL-10, and Hsp70 in the three-time points studied. However, it decreased IL-1β at 24 h and 48 h and IL-6 at 48 h, and increased HO-1 concentration at 48 h. On the other hand, compared to the pre-treatment period, Sibai's regimen induced a significant decrease in TNF-α, IL-1β, IL-6, and Hsp70, while increased HO-1 levels both at 24 h and 48 h and, IL-10 concentration at 48 h. CONCLUSIONS Sibai's regimen determined an early and efficient immunoregulatory effect on systemic inflammatory response in preeclampsia, suggesting that the maintenance dose of two grams of MgSO4 was better than one gram in the treatment of imminent eclampsia.
Collapse
Affiliation(s)
- José C Peraçoli
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Patricia B Silva
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Haroldo M Neves
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Vera T M Borges
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Joelcio F Abbade
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Roberto A A Costa
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Fernanda R G Batista
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Maria T S Peraçoli
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil; Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil
| | - Mariana Romão-Veiga
- Department of Gynecology and Obstetrics, Botucatu Medical School and Department of Chemistry and Biological Sciences, Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, SP, Brazil.
| |
Collapse
|
30
|
Brokesh AM, Cross LM, Kersey AL, Murali A, Richter C, Gregory CA, Singh I, Gaharwar AK. Dissociation of nanosilicates induces downstream endochondral differentiation gene expression program. SCIENCE ADVANCES 2022; 8:eabl9404. [PMID: 35476448 PMCID: PMC9045714 DOI: 10.1126/sciadv.abl9404] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/11/2022] [Indexed: 05/16/2023]
Abstract
Bioactive materials harness the body's innate regenerative potential by directing endogenous progenitor cells to facilitate tissue repair. Dissolution products of inorganic biomaterials provide unique biomolecular signaling for tissue-specific differentiation. Inorganic ions (minerals) are vital to biological processes and play crucial roles in regulating gene expression patterns and directing cellular fate. However, mechanisms by which ionic dissolution products affect cellular differentiation are not well characterized. We demonstrate the role of the inorganic biomaterial synthetic two-dimensional nanosilicates and its ionic dissolution products on human mesenchymal stem cell differentiation. We use whole-transcriptome sequencing (RNA-sequencing) to characterize the contribution of nanosilicates and its ionic dissolution products on endochondral differentiation. Our study highlights the modulatory role of ions in stem cell transcriptome dynamics by regulating lineage-specific gene expression patterns. This work paves the way for leveraging biochemical characteristics of inorganic biomaterials to direct cellular processes and promote in situ tissue regeneration.
Collapse
Affiliation(s)
- Anna M. Brokesh
- Department of Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Lauren M. Cross
- Department of Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Anna L. Kersey
- Department of Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Aparna Murali
- Department of Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Christopher Richter
- Department of Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Carl A. Gregory
- Department of Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77807-3260, USA
| | - Irtisha Singh
- Department of Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77807-3260, USA
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX 77843, USA
- Department of Material Science and Engineering, Dwight Look College of Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
31
|
Qiu H, Shen R, Chen L, Pandey S, Sun J, Deng H. Low Serum Magnesium Levels Are Associated With Hemorrhagic Transformation After Mechanical Thrombectomy in Patients With Acute Ischemic Stroke. Front Neurol 2022; 13:831232. [PMID: 35401415 PMCID: PMC8984269 DOI: 10.3389/fneur.2022.831232] [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: 01/04/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Objective In patients with acute ischemic stroke (AIS), hemorrhagic transformation (HT) is a major complication after mechanical thrombectomy (MT). This study aimed to investigate the relationship between serum magnesium levels and HT after MT. Methods We collected 199 cases of consecutive AIS that received MT due to acute anterior circulation occlusions in our institution between January 2017 and January 2020. Baseline serum magnesium was obtained from all patients on admission before MT. The patients were divided into two groups based on the occurrence of HT. Univariate and multivariate analyses were performed to investigate whether magnesium was an independent predictor of HT. The receiver operating characteristic (ROC) curve and area under the curve (AUC) were determined. Results Of the 199 enrolled patients, 40 (20.1%) presented with HT, and 12 (6%) developed symptomatic intracranial hemorrhage (sICH). Patients with HT had lower serum magnesium levels compared to those without HT (0.76 [0.69–0.80] vs. 0.84 [0.80–0.90], p < 0.001). The multivariate logistic analysis showed that the serum magnesium level (odds ratio, [OR]: 0.000, 95% confidence interval [CI]: 0.000–0.001, p < 0.001) was significantly associated with the occurrence of HT. The ROC curve analysis revealed that the serum magnesium level could predict HT with an AUC of.820 (95% CI: 0.750–0.891 p < 0.001). Serum magnesium ≤ 0.80 mmol/L could predict HT with a sensitivity of 79.2% and a specificity of 70.0%. Of interest, the serum magnesium level was not associated with HT when the baseline of serum magnesium was higher than the cut-off value (0.80 mmol/L) in the subgroup analysis. Conclusions Lower baseline serum magnesium levels (<0.80 mmol/L) on admission are associated with increased risk of HT in AIS patients receiving MT.
Collapse
Affiliation(s)
- Huijia Qiu
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Rui Shen
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Liuwei Chen
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Sajan Pandey
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Jiping Sun
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Haoyu Deng
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart and Lung Innovation, St.Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
32
|
Chen A, Jin J, Cheng S, Liu Z, Yang C, Chen Q, Liang W, Li K, Kang D, Ouyang Z, Yao C, Bai X, Li Q, Jin D, Huang B. mTORC1 induces plasma membrane depolarization and promotes preosteoblast senescence by regulating the sodium channel Scn1a. Bone Res 2022; 10:25. [PMID: 35256591 PMCID: PMC8901653 DOI: 10.1038/s41413-022-00204-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 12/18/2022] Open
Abstract
Senescence impairs preosteoblast expansion and differentiation into functional osteoblasts, blunts their responses to bone formation-stimulating factors and stimulates their secretion of osteoclast-activating factors. Due to these adverse effects, preosteoblast senescence is a crucial target for the treatment of age-related bone loss; however, the underlying mechanism remains unclear. We found that mTORC1 accelerated preosteoblast senescence in vitro and in a mouse model. Mechanistically, mTORC1 induced a change in the membrane potential from polarization to depolarization, thus promoting cell senescence by increasing Ca2+ influx and activating downstream NFAT/ATF3/p53 signaling. We further identified the sodium channel Scn1a as a mediator of membrane depolarization in senescent preosteoblasts. Scn1a expression was found to be positively regulated by mTORC1 upstream of C/EBPα, whereas its permeability to Na+ was found to be gated by protein kinase A (PKA)-induced phosphorylation. Prosenescent stresses increased the permeability of Scn1a to Na+ by suppressing PKA activity and induced depolarization in preosteoblasts. Together, our findings identify a novel pathway involving mTORC1, Scn1a expression and gating, plasma membrane depolarization, increased Ca2+ influx and NFAT/ATF3/p53 signaling in the regulation of preosteoblast senescence. Pharmaceutical studies of the related pathways and agents might lead to novel potential treatments for age-related bone loss.
Collapse
Affiliation(s)
- Ajuan Chen
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jian Jin
- Department of Spine Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shasha Cheng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zezheng Liu
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Cheng Yang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qingjing Chen
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wenquan Liang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Kai Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dawei Kang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhicong Ouyang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Chenfeng Yao
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Qingchu Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dadi Jin
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| | - Bin Huang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
33
|
Dong Y, Chen S, Liu TL, Li J. Materials and Interface Designs of Waterproof Field-Effect Transistor Arrays for Detection of Neurological Biomarkers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106866. [PMID: 35023615 PMCID: PMC8930526 DOI: 10.1002/smll.202106866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/05/2021] [Indexed: 06/14/2023]
Abstract
The continuous, real-time, and concurrent detection of multiple biomarkers in bodily fluids is of high significance for advanced healthcare. While active, semiconductor-based biochemical sensing platforms provide levels of functionality exceeding those of their conventional passive counterparts, the stability of the active biosensors in the liquid environment for continuous operation remains a challenging topic. This work reports the development of a class of flexible and waterproof field-effect transistor arrays for multiplexed biochemical sensing. In this design, monolithic, ultrathin, dense, and low defect nanomembranes consisting of monocrystalline Si and thermally grown SiO2 simultaneously serve as high-performance backplane electronics for signal transduction and stable biofluid barriers with high structural integrity due to the high formation temperature. Coupling the waterproof transistors with various ion-selective membranes through the gate electrode allows for sensitive and selective detection of multiple ions as biomarkers for traumatic brain injury. The study also demonstrates a similar encapsulation structure which enables the design of waterproof amperometric sensors based on this materials strategy and integration scheme. Overall, key advantages in flexibility, stability, and multifunctionality highlight the potential of using such electronic sensing platforms for concurrent, continuous detection of various neurological biomarkers, proving a promising approach for early diagnosis and intervention of chronic diseases.
Collapse
Affiliation(s)
- Yan Dong
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Shulin Chen
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Tzu-Li Liu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Jinghua Li
- Department of Materials Science and Engineering, Chronic Brain Injury Program, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
34
|
A Review of Risk Factors and Predictors for Hemorrhagic Transformation in Patients with Acute Ischemic Stroke. Int J Vasc Med 2021; 2021:4244267. [PMID: 34912581 PMCID: PMC8668348 DOI: 10.1155/2021/4244267] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Acute ischemic strokes (AIS) and hemorrhagic strokes lead to disabling neuropsychiatric and cognitive deficits. A serious and fatal complication of AIS is the occurrence of hemorrhagic transformation (HT). HT is cerebral bleeding that occurs after an ischemic event in the infarcted areas. This review summarises how specific risk factors such as demographic factors like age, gender, and race/ethnicity, comorbidities including essential hypertension, atrial fibrillation, diabetes mellitus, congestive heart failure, and ischemic heart disease along with predictors like higher NIHSS score, larger infarction size, cardioembolic strokes, systolic blood pressure/pulse pressure variability, higher plasma glucose levels, and higher body temperature during ischemic event, lower low-density lipoprotein and total cholesterol, early ischemic changes on imaging modalities, and some rare causes make an individual more susceptible to developing HT. We also discuss few other risk factors such as the role of blood-brain barrier, increased arterial stiffness, and globulin levels in patients postreperfusion using thrombolysis and mechanical thrombectomy. In addition, we discuss the implications of dual antiplatelet therapy and the length of treatment in reference to the incidence of developing HT. Current research into inflammatory mediators and biomarkers such as Cyclooxygenase-2, matrix metalloproteinases, and soluble ST2 and their potential role as treatment options for HT is also briefly discussed. Finally, this review calls for more research into use of dual antiplatelet and the timing of antiplatelet and anticoagulant use in reference to hemorrhagic transformation.
Collapse
|
35
|
Yong KW, Janmaleki M, Pachenari M, Mitha AP, Sanati-Nezhad A, Sen A. Engineering a 3D human intracranial aneurysm model using liquid-assisted injection molding and tuned hydrogels. Acta Biomater 2021; 136:266-278. [PMID: 34547516 DOI: 10.1016/j.actbio.2021.09.022] [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: 05/05/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/30/2022]
Abstract
Physiologically relevant intracranial aneurysm (IA) models are crucially required to facilitate testing treatment options for IA. Herein, we report the development of a new in vitro tissue-engineered platform, which recapitulates the microenvironment, structure, and cellular complexity of native human IA. A new modified liquid-assisted injection molding technique was developed to fabricate a three-dimensional hollow IA model with clinically relevant IA dimensions within a mechanically tuned Gelatin Methacryloyl (GelMA) hydrogel. An endothelium lining was created inside the IA model by culturing human umbilical vein endothelial cells over pre-cultured human brain vascular smooth muscle cells. These cellularized IA models were subjected to medium perfusion at flow rates between 6.3 and 15.75 mL/min for inducing biomimetic vessel wall shear stress (10-25 dyn/cm2) to the cells for ten days. Both cell types maintained their secretome profiles and showed more than 96% viability, demonstrating the biocompatibility of the hydrogel during perfusion cell culture at such flow rates. Based on the characterized viscoelastic properties of the GelMA hydrogel and with the aid of a fluid-structure interaction model, the capability of the IA model in predicting the response of the IA to different fluid flow profiles was mathematically shown. With physiologically relevant behavior, our developed in vitro human IA model could allow researchers to better understand the pathophysiology and treatment of IA. STATEMENT OF SIGNIFICANCE: A three-dimensional intracranial aneurysm (IA) tissue model recapitulating the microenvironment, structure, and cellular complexity of native human IA was developed. • An endothelium lining was created inside the IA model over pre-cultured human brain vascular smooth muscle cells over at least 10-day successful culture. • The cells maintained their secretome profiles, demonstrating the biocompatibility of hydrogel during a long-term perfusion cell culture. • The IA model showed its capability in predicting the response of IA to different fluid flow profiles. • The cells in the vessel region behaved differently from cells in the aneurysm region due to alteration in hemodynamic shear stress. • The IA model could allow researchers to better understand the pathophysiology and treatment options of IA.
Collapse
|
36
|
Du Preez S, Cabanas H, Staines D, Marshall-Gradisnik S. Potential Implications of Mammalian Transient Receptor Potential Melastatin 7 in the Pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:10708. [PMID: 34682454 PMCID: PMC8535478 DOI: 10.3390/ijerph182010708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 11/25/2022]
Abstract
The transient receptor potential (TRP) superfamily of ion channels is involved in the molecular mechanisms that mediate neuroimmune interactions and activities. Recent advancements in neuroimmunology have identified a role for TRP cation channels in several neuroimmune disorders including amyotropic lateral sclerosis, multiple sclerosis, and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). ME/CFS is a debilitating disorder with an obscure aetiology, hence considerable examination of its pathobiology is warranted. Dysregulation of TRP melastatin (TRPM) subfamily members and calcium signalling processes are implicated in the neurological, immunological, cardiovascular, and metabolic impairments inherent in ME/CFS. In this review, we present TRPM7 as a potential candidate in the pathomechanism of ME/CFS, as TRPM7 is increasingly recognized as a key mediator of physiological and pathophysiological mechanisms affecting neurological, immunological, cardiovascular, and metabolic processes. A focused examination of the biochemistry of TRPM7, the role of this protein in the aforementioned systems, and the potential of TRPM7 as a molecular mechanism in the pathophysiology of ME/CFS will be discussed in this review. TRPM7 is a compelling candidate to examine in the pathobiology of ME/CFS as TRPM7 fulfils several key roles in multiple organ systems, and there is a paucity of literature reporting on its role in ME/CFS.
Collapse
Affiliation(s)
- Stanley Du Preez
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast 4215, Australia; (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Menzies Health Institute Queensland, Griffith University, Gold Coast 4215, Australia;
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast 4215, Australia
- School of Medicine and Dentistry, Griffith University, Gold Coast 4215, Australia
| | - Helene Cabanas
- Consortium Health International for Myalgic Encephalomyelitis, Menzies Health Institute Queensland, Griffith University, Gold Coast 4215, Australia;
- Institut de Recherche Saint Louis, Université de Paris, INSERM U944 and CNRS UMR 7212, Hôpital Saint Louis, APHP, 75010 Paris, France
| | - Donald Staines
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast 4215, Australia; (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Menzies Health Institute Queensland, Griffith University, Gold Coast 4215, Australia;
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Gold Coast 4215, Australia; (D.S.); (S.M.-G.)
- Consortium Health International for Myalgic Encephalomyelitis, Menzies Health Institute Queensland, Griffith University, Gold Coast 4215, Australia;
| |
Collapse
|
37
|
Locatelli L, Maier JAM. Cytoskeletal Remodeling Mimics Endothelial Response to Microgravity. Front Cell Dev Biol 2021; 9:733573. [PMID: 34568340 PMCID: PMC8458731 DOI: 10.3389/fcell.2021.733573] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/13/2021] [Indexed: 12/26/2022] Open
Abstract
Mechanical cues contribute to the maintenance of a healthy endothelium, which is essential for vascular integrity. Indeed endothelial cells are mechanosensors that integrate the forces in the form of biochemical signals. The cytoskeleton is fundamental in sensing mechanical stimuli and activating specific signaling pathways. Because the cytoskeleton is very rapidly remodeled in endothelial cells exposed to microgravity, we investigated whether the disruption of actin polymerization by cytochalasin D in 1g condition triggers and orchestrates responses similar to those occurring in micro- and macro-vascular endothelial cells upon gravitational unloading. We focused our attention on the effect of simulated microgravity on stress proteins and transient receptor potential melastatin 7 (TRPM7), a cation channel that acts as a mechanosensor and modulates endothelial cell proliferation and stress response. Simulated microgravity downregulates TRPM7 in both cell types. However, 24 h of treatment with cytochalasin D decreases the amounts of TRPM7 only in macrovascular endothelial cells, suggesting that the regulation and the role of TRPM7 in microvascular cells are more complex than expected. The 24 h culture in the presence of cytochalasin D mimics the effect of simulated microgravity in modulating stress response in micro- and macro-vascular endothelial cells. We conclude that cytoskeletal disruption might mediate some effects of microgravity in endothelial cells.
Collapse
Affiliation(s)
- Laura Locatelli
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milan, Italy
| | - Jeanette A. M. Maier
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milan, Italy
- Interdisciplinary Centre for Nanostructured Materials and Interfaces, Università di Milano, Milan, Italy
| |
Collapse
|
38
|
Hou Z, Xiang M, Chen N, Cai X, Zhang B, Luo R, Yang L, Ma X, Zhou L, He F, Yu H, Wang Y. The biological responses and mechanisms of endothelial cells to magnesium alloy. Regen Biomater 2021; 8:rbab017. [PMID: 34211729 PMCID: PMC8240605 DOI: 10.1093/rb/rbab017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/22/2021] [Accepted: 04/05/2021] [Indexed: 12/03/2022] Open
Abstract
Due to its good biocompatibility and degradability, magnesium alloy (Mg alloy) has shown great promise in cardiovascular stent applications. Rapid stent re-endothelialization is derived from migrated and adhered endothelial cells (ECs), which is an effective way to reduce late thrombosis and inhibit hyperplasia. However, fundamental questions regarding Mg alloy affecting migration and adhesion of ECs are not fully understood. Here, we evaluated the effects of Mg alloy on the ECs proliferation, adhesion and migration. A global gene expression profiling of ECs co-culturing with Mg alloy was conducted, and the adhesion- and migration-related genes were examined. We found that Mg alloy had no adverse effects on ECs viability but significantly affected ECs migration and adhesion. Co-cultured with Mg alloy extract, ECs showed contractive adhesion morphology and decreased motility, which was supported by the down-regulation of adhesion-related genes (Paxillin and Vinculin) and migration-related genes (RAC 1, Rho A and CDC 42). Accordingly, the re-endothelialization of Mg alloy stent was inhibited in vivo. Our results may provide new inspiration for improving the broad application of Mg alloy stents.
Collapse
Affiliation(s)
- Zhe Hou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Maolong Xiang
- College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Nuoya Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xiao Cai
- The Fourth People's Hospital of Chengdu, Chengdu 610036, China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xiaoyi Ma
- Beijing Key Laboratory of Cardiac Drug Device Technology and Evidence Based Medicine, Beijing 100021, China
| | - Lifeng Zhou
- Beijing Key Laboratory of Cardiac Drug Device Technology and Evidence Based Medicine, Beijing 100021, China
| | - Fugui He
- Beijing Key Laboratory of Cardiac Drug Device Technology and Evidence Based Medicine, Beijing 100021, China
| | - Hongchi Yu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
39
|
Xu L, Willumeit-Römer R, Luthringer-Feyerabend BJC. Mesenchymal Stem Cell and Oxygen Modulate the Cocultured Endothelial Cells in the Presence of Magnesium Degradation Products. ACS APPLIED BIO MATERIALS 2021; 4:2398-2407. [DOI: 10.1021/acsabm.0c01289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Lei Xu
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | - Regine Willumeit-Römer
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | | |
Collapse
|
40
|
Bagheri S, Saboury AA. What role do metals play in Alzheimer's disease? JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2021. [DOI: 10.1007/s13738-021-02181-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
41
|
Dominguez LJ, Gea A, Ruiz-Estigarribia L, Sayón-Orea C, Fresán U, Barbagallo M, Ruiz-Canela M, Martínez-González MA. Low Dietary Magnesium and Overweight/Obesity in a Mediterranean Population: A Detrimental Synergy for the Development of Hypertension. The SUN Project. Nutrients 2020; 13:125. [PMID: 33396318 PMCID: PMC7824180 DOI: 10.3390/nu13010125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/20/2022] Open
Abstract
Hypertension is the strongest independent modifiable risk factor for cardiovascular disease. We aimed to investigate the association of magnesium intake with incident hypertension in a Mediterranean population, and the potential modification of this association by body mass index BMI. We assessed 14,057 participants of the SUN (Seguimiento Universidad de Navarra) prospective cohort (67.0% women) initially free of hypertension. At baseline, a validated 136-item food frequency questionnaire was administered. We used Cox models adjusted for multiple socio-demographic, anthropometric, and lifestyle factors, and prevalent conditions present at baseline. Among a mean 9.6 years of follow-up we observed 1406 incident cases of medically diagnosed hypertension. An inverse association in multivariable-adjusted models was observed for progressively higher magnesium intake up to 500 mg/d vs. intake < 200 mg/d, which was greater among those with a BMI > 27 kg/m2. Lean participants with magnesium intake < 200 mg/d vs. >200 mg/d also had a higher risk of incident hypertension. Adherence to the Mediterranean diet did not modify these associations. In conclusion, dietary magnesium intake < 200 mg/d was independently associated with a higher risk of developing hypertension in a Mediterranean cohort, stronger for overweight/obese participants. Our results emphasize the importance of encouraging the consumption of magnesium-rich foods (vegetables, nuts, whole cereals, legumes) in order to prevent hypertension.
Collapse
Affiliation(s)
- Ligia J. Dominguez
- Geriatric Unit, Department of Internal Medicine and Geriatrics, University of Palermo, 90127 Palermo, Italy;
| | - Alfredo Gea
- Department of Preventive Medicine and Public Health, University of Navarra-IDISNA, 31008 Pamplona, Spain; (A.G.); (L.R.-E.); (C.S.-O.); (M.R.-C.); (M.A.M.-G.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Liz Ruiz-Estigarribia
- Department of Preventive Medicine and Public Health, University of Navarra-IDISNA, 31008 Pamplona, Spain; (A.G.); (L.R.-E.); (C.S.-O.); (M.R.-C.); (M.A.M.-G.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carmen Sayón-Orea
- Department of Preventive Medicine and Public Health, University of Navarra-IDISNA, 31008 Pamplona, Spain; (A.G.); (L.R.-E.); (C.S.-O.); (M.R.-C.); (M.A.M.-G.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Public Health Institute, 31003 Navarra, Spain
| | - Ujue Fresán
- eHealth Group, ISGlobal, 08036 Barcelona, Spain;
| | - Mario Barbagallo
- Geriatric Unit, Department of Internal Medicine and Geriatrics, University of Palermo, 90127 Palermo, Italy;
| | - Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, University of Navarra-IDISNA, 31008 Pamplona, Spain; (A.G.); (L.R.-E.); (C.S.-O.); (M.R.-C.); (M.A.M.-G.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel A. Martínez-González
- Department of Preventive Medicine and Public Health, University of Navarra-IDISNA, 31008 Pamplona, Spain; (A.G.); (L.R.-E.); (C.S.-O.); (M.R.-C.); (M.A.M.-G.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
42
|
Lavanderos B, Silva I, Cruz P, Orellana-Serradell O, Saldías MP, Cerda O. TRP Channels Regulation of Rho GTPases in Brain Context and Diseases. Front Cell Dev Biol 2020; 8:582975. [PMID: 33240883 PMCID: PMC7683514 DOI: 10.3389/fcell.2020.582975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological and neuropsychiatric disorders are mediated by several pathophysiological mechanisms, including developmental and degenerative abnormalities caused primarily by disturbances in cell migration, structural plasticity of the synapse, and blood-vessel barrier function. In this context, critical pathways involved in the pathogenesis of these diseases are related to structural, scaffolding, and enzymatic activity-bearing proteins, which participate in Ca2+- and Ras Homologs (Rho) GTPases-mediated signaling. Rho GTPases are GDP/GTP binding proteins that regulate the cytoskeletal structure, cellular protrusion, and migration. These proteins cycle between GTP-bound (active) and GDP-bound (inactive) states due to their intrinsic GTPase activity and their dynamic regulation by GEFs, GAPs, and GDIs. One of the most important upstream inputs that modulate Rho GTPases activity is Ca2+ signaling, positioning ion channels as pivotal molecular entities for Rho GTPases regulation. Multiple non-selective cationic channels belonging to the Transient Receptor Potential (TRP) family participate in cytoskeletal-dependent processes through Ca2+-mediated modulation of Rho GTPases. Moreover, these ion channels have a role in several neuropathological events such as neuronal cell death, brain tumor progression and strokes. Although Rho GTPases-dependent pathways have been extensively studied, how they converge with TRP channels in the development or progression of neuropathologies is poorly understood. Herein, we review recent evidence and insights that link TRP channels activity to downstream Rho GTPase signaling or modulation. Moreover, using the TRIP database, we establish associations between possible mediators of Rho GTPase signaling with TRP ion channels. As such, we propose mechanisms that might explain the TRP-dependent modulation of Rho GTPases as possible pathways participating in the emergence or maintenance of neuropathological conditions.
Collapse
Affiliation(s)
- Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Octavio Orellana-Serradell
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
43
|
de Borst MH, de Baaij JHF. Low serum magnesium as a risk factor for peripheral artery disease in chronic kidney disease: an open verdict. Nephrol Dial Transplant 2020; 35:1831-1833. [PMID: 32494818 PMCID: PMC7643669 DOI: 10.1093/ndt/gfaa115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 02/01/2023] Open
Affiliation(s)
- Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
44
|
Krzywoszyńska K, Witkowska D, Świątek-Kozłowska J, Szebesczyk A, Kozłowski H. General Aspects of Metal Ions as Signaling Agents in Health and Disease. Biomolecules 2020; 10:biom10101417. [PMID: 33036384 PMCID: PMC7600656 DOI: 10.3390/biom10101417] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
This review focuses on the current knowledge on the involvement of metal ions in signaling processes within the cell, in both physiological and pathological conditions. The first section is devoted to the recent discoveries on magnesium and calcium-dependent signal transduction-the most recognized signaling agents among metals. The following sections then describe signaling pathways where zinc, copper, and iron play a key role. There are many systems in which changes in intra- and extra-cellular zinc and copper concentrations have been linked to important downstream events, especially in nervous signal transduction. Iron signaling is mostly related with its homeostasis. However, it is also involved in a recently discovered type of programmed cell death, ferroptosis. The important differences in metal ion signaling, and its disease-leading alterations, are also discussed.
Collapse
Affiliation(s)
- Karolina Krzywoszyńska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Correspondence: (K.K.); (D.W.); Tel.: +48-77-44-23-549 (K.K); +48-77-44-23-548 (D.W.)
| | - Danuta Witkowska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Correspondence: (K.K.); (D.W.); Tel.: +48-77-44-23-549 (K.K); +48-77-44-23-548 (D.W.)
| | - Jolanta Świątek-Kozłowska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
| | - Agnieszka Szebesczyk
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
| | - Henryk Kozłowski
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Faculty of Chemistry, University of Wrocław, 14 F. Joliot-Curie St., 50-383 Wrocław, Poland
| |
Collapse
|
45
|
Cheng Z, Huang X, Muse FM, Xia L, Zhan Z, Lin X, Cao Y, Han Z. Low Serum Magnesium Levels Are Associated With Hemorrhagic Transformation After Thrombolysis in Acute Ischemic Stroke. Front Neurol 2020; 11:962. [PMID: 32982953 PMCID: PMC7492199 DOI: 10.3389/fneur.2020.00962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 07/24/2020] [Indexed: 11/25/2022] Open
Abstract
Background: In patients with acute ischemic stroke, hemorrhagic transformation is a major complication after intravenous thrombolysis. This study aimed to investigate the relationship between serum magnesium levels and hemorrhagic transformation (HT) after thrombolytic therapy. Methods: We retrospectively analyzed data from 242 patients who received thrombolytic therapy at the Second Affiliated Hospital of the Wenzhou Medical University in China. Baseline serum magnesium levels were measured before intravenous thrombolysis, and the occurrence of HT was evaluated using computed tomography images reviewed within 24–36 h after therapy. The relationship between serum magnesium levels and HT was examined using multivariate logistic regression, subgroup analysis, and restricted cubic spline models. Results: Of the 242 included patients, 43 (17.8%) developed HT. Patients with HT had significant lower serum magnesium levels than those without HT (0.81 ± 0.08 vs. 0.85 ± 0.08 mmol/L, p = 0.007). Multivariable logistic regression analysis indicated that patients with higher serum magnesium levels had lower risk of HT (OR per 0.1-mmol/L increase 0.43, 95% CI 0.27–0.73, p = 0.002). However, this association did not persist when baseline levels of serum magnesium were higher than the median value (0.85 mmol/L) in subgroup analysis (OR per 0.1-mmol/L increase 0.58, 95% CI 0.14–2.51, p = 0.47). This threshold effect was also observed in the restricted cubic spline model when serum magnesium levels were above 0.88 mmol/L. No association between symptomatic HT and serum magnesium levels was observed in our study (OR per 0.1-mmol/L increase 0.52, 95% CI 0.25–1.11, p = 0.092). Conclusions: Lower serum magnesium levels in patients with ischemic stroke are associated with an increased risk of HT after intravenous thrombolysis, but perhaps only when serum magnesium is below a certain minimal concentration.
Collapse
Affiliation(s)
- Zicheng Cheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyan Huang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Farah Mohamed Muse
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingfan Xia
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhenxiang Zhan
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianda Lin
- Department of Neurology, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| | - Yungang Cao
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhao Han
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
46
|
A Review of the Action of Magnesium on Several Processes Involved in the Modulation of Hematopoiesis. Int J Mol Sci 2020; 21:ijms21197084. [PMID: 32992944 PMCID: PMC7582682 DOI: 10.3390/ijms21197084] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022] Open
Abstract
Magnesium (Mg2+) is an essential mineral for the functioning and maintenance of the body. Disturbances in Mg2+ intracellular homeostasis result in cell-membrane modification, an increase in oxidative stress, alteration in the proliferation mechanism, differentiation, and apoptosis. Mg2+ deficiency often results in inflammation, with activation of inflammatory pathways and increased production of proinflammatory cytokines by immune cells. Immune cells and others that make up the blood system are from hematopoietic tissue in the bone marrow. The hematopoietic tissue is a tissue with high indices of renovation, and Mg2+ has a pivotal role in the cell replication process, as well as DNA and RNA synthesis. However, the impact of the intra- and extracellular disturbance of Mg2+ homeostasis on the hematopoietic tissue is little explored. This review deals specifically with the physiological requirements of Mg2+ on hematopoiesis, showing various studies related to the physiological requirements and the effects of deficiency or excess of this mineral on the hematopoiesis regulation, as well as on the specific process of erythropoiesis, granulopoiesis, lymphopoiesis, and thrombopoiesis. The literature selected includes studies in vitro, in animal models, and in humans, giving details about the impact that alterations of Mg2+ homeostasis can have on hematopoietic cells and hematopoietic tissue.
Collapse
|
47
|
Maier JA, Pickering G, Giacomoni E, Cazzaniga A, Pellegrino P. Headaches and Magnesium: Mechanisms, Bioavailability, Therapeutic Efficacy and Potential Advantage of Magnesium Pidolate. Nutrients 2020; 12:nu12092660. [PMID: 32878232 PMCID: PMC7551876 DOI: 10.3390/nu12092660] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/13/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
Magnesium deficiency may occur for several reasons, such as inadequate intake or increased gastrointestinal or renal loss. A large body of literature suggests a relationship between magnesium deficiency and mild and moderate tension-type headaches and migraines. A number of double-blind randomized placebo-controlled trials have shown that magnesium is efficacious in relieving headaches and have led to the recommendation of oral magnesium for headache relief in several national and international guidelines. Among several magnesium salts available to treat magnesium deficiency, magnesium pidolate may have high bioavailability and good penetration at the intracellular level. Here, we discuss the cellular and molecular effects of magnesium deficiency in the brain and the clinical evidence supporting the use of magnesium for the treatment of headaches and migraines.
Collapse
Affiliation(s)
- Jeanette A. Maier
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, 20157 Milano, Italy;
- Correspondence:
| | - Gisele Pickering
- Department of Clinical Pharmacology, University Hospital and Inserm 1107 Fundamental and Clinical Pharmacology of Pain, Medical Faculty, F-63000 Clermont-Ferrand, France;
| | - Elena Giacomoni
- Sanofi Consumer Health Care, 20158 Milan, Italy; (E.G.); (P.P.)
| | - Alessandra Cazzaniga
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università di Milano, 20157 Milano, Italy;
| | | |
Collapse
|
48
|
Zhou K, Li Y, Zhang L, Jin L, Yuan F, Tan J, Yuan G, Pei J. Nano-micrometer surface roughness gradients reveal topographical influences on differentiating responses of vascular cells on biodegradable magnesium. Bioact Mater 2020; 6:262-272. [PMID: 32913933 PMCID: PMC7451920 DOI: 10.1016/j.bioactmat.2020.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/05/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022] Open
Abstract
Distinctively directing endothelial cells (ECs) and smooth muscle cells (SMCs), potentially by surface topography cue, is of central importance for enhancing bioefficacy of vascular implants. For the first time, surface gradients with a broad range of nano-micrometer roughness are developed on Mg, a promising next-generation biodegradable metal, to carry out a systematic study on the response of ECs and SMCs. Cell adhesion, spreading, and proliferation are quantified along gradients by high-throughput imaging, illustrating drastic divergence between ECs and SMCs, especially in highly rough regions. The profound role of surface topography overcoming the biochemical cue of released Mg2+ is unraveled at different roughness ranges for ECs and SMCs. Further insights into the underlying regulatory mechanism are gained at subcellular and gene levels. Our work enables high-efficient exploration of optimized surface morphology for modulating favored cell selectivity of promoting ECs and suppressing SMCs, providing a potential strategy to achieve rapid endothelialization for Mg. Surface topography stimuli was engineered on Mg with varying roughness gradients. Ridge/valley network feature on Mg overperforms the influence of Mg2+. Optimized roughness facilitates proliferation of ECs while suppressing SMCs.
Collapse
Affiliation(s)
- Ke Zhou
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite, School of Materials Science & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Yutong Li
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite, School of Materials Science & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Lei Zhang
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite, School of Materials Science & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Liang Jin
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite, School of Materials Science & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Feng Yuan
- Department of Biomedical Engineering, University of Texas at Austin, TX, 78712, USA
| | - Jinyun Tan
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Guangyin Yuan
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite, School of Materials Science & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jia Pei
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite, School of Materials Science & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| |
Collapse
|
49
|
Runnels LW, Komiya Y. TRPM6 and TRPM7: Novel players in cell intercalation during vertebrate embryonic development. Dev Dyn 2020; 249:912-923. [PMID: 32315468 DOI: 10.1002/dvdy.182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 12/16/2022] Open
Abstract
A common theme in organogenesis is how the final structure of organs emerge from epithelial tube structures, with the formation of the neural tube being one of the best examples. Two types of cell movements co-occur during neural tube closure involving the migration of cells toward the midline of the embryo (mediolateral intercalation or convergent extension) as well as the deep movement of cells from inside the embryo to the outside of the lateral side of the neural plate (radial intercalation). Failure of either type of cell movement will prevent neural tube closure, which can produce a range of neural tube defects (NTDs), a common congenital disease in humans. Numerous studies have identified signaling pathways that regulate mediolateral intercalation during neural tube closure. Less understood are the pathways that govern radial intercalation. Using the Xenopus laevis system, our group reported the identification of transient receptor potential (TRP) channels, TRPM6 and TRPM7, and the Mg2+ ion they conduct, as novel and key factors regulating both mediolateral and radial intercalation during neural tube closure. Here we broadly discuss tubulogenesis and cell intercalation from the perspective of neural tube closure and the respective roles of TRPM7 and TRPM6 in this critical embryonic process.
Collapse
Affiliation(s)
- Loren W Runnels
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Yuko Komiya
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
- Faculty of Industrial Science and Technology, Tokyo University of Science, Yamakoshi-gun, Hokkaido, Japan
| |
Collapse
|
50
|
Cockerill I, Su Y, Bitten R, Cloarec B, Aouadi S, Zhu D, Young ML. Salt Preform Texturing of Absorbable Zn Substrates for Bone-implant Applications. JOM (WARRENDALE, PA. : 1989) 2020; 72:1902-1909. [PMID: 33737795 PMCID: PMC7962799 DOI: 10.1007/s11837-019-03971-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Surface roughness is an important factor in improving the bone-implant contact area to enhance bone regeneration, yet this aspect has not been applied to absorbable metals. Textured zinc surfaces with varying degrees of surface roughness were produced using a salt-preform method with fine- and coarse-grained salts and compared to a polished control sample. The resulting surfaces were characterized by scanning electron microscopy (SEM), surface roughness, corrosion rates, and in vitro cytotoxicity. The resulting textured surfaces exhibit micron-sized cavities and increased roughness consistent with the initial salt particle size. The corrosion rate was shown to accelerate significantly as compared to the polished control sample, and pre-osteoblasts displayed healthy morphologies on the textures. The results confirm textured zinc surfaces support cell adhesion and can be used to control the corrosion rate. This study represents an important intermediate step that can be applied to porous absorbable metal scaffolds for bone-implant applications.
Collapse
Affiliation(s)
- Irsalan Cockerill
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76210, USA
- Department of Materials Science and Engineering, University of North Texas, Denton, TX 76210, USA
| | - Yingchao Su
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76210, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Reid Bitten
- Department of Materials Science and Engineering, University of North Texas, Denton, TX 76210, USA
| | - Benjamin Cloarec
- Department of Materials Science and Engineering, University of North Texas, Denton, TX 76210, USA
- Department of Physical Measurements, University of Rouen, Rouen, France
| | - Samir Aouadi
- Department of Materials Science and Engineering, University of North Texas, Denton, TX 76210, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76210, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Marcus L Young
- Department of Materials Science and Engineering, University of North Texas, Denton, TX 76210, USA
| |
Collapse
|