1
|
Flanagan D. Horizontal Alveolar Ridge Splitting and Expansion. J ORAL IMPLANTOL 2024; 50:200-210. [PMID: 38624042 DOI: 10.1563/aaid-joi-d-23-00186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
When considering placing dental implants in atrophic edentulous sites, there may be inadequate site width and little or no vertical bone loss. Any of several surgical procedures can augment these sites. Extracortical augmentation is done by applying graft material against the cortical bone. This technique expects progenitor cells to migrate outside the bony ridge's confines and form new bone. Another method entails ridge splitting and expansion to create space for osteogenesis and, when possible, implant placement. This may be a better method for horizontal ridge augmentation. The ridge is split, separating the facial and lingual cortices for a complete bone fracture. The patient's osseous cells can then migrate into the created space from the exposed medullary bone to form bone. The technique can be preferably performed flapless so the intact periosteum maintains a blood supply to ensure appropriate healing.
Collapse
|
2
|
Ma Y, Wang S, Wang H, Chen X, Shuai Y, Wang H, Mao Y, He F. Mesenchymal stem cells and dental implant osseointegration during aging: from mechanisms to therapy. Stem Cell Res Ther 2023; 14:382. [PMID: 38124153 PMCID: PMC10734190 DOI: 10.1186/s13287-023-03611-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
Dental implants are widely used to replace missing teeth, providing patients with unparalleled levels of effectiveness, convenience, and affordability. The biological basis for the clinical success of dental implants is osseointegration. Bone aging is a high-risk factor for the reduced osseointegration and survival rates of dental implants. In aged individuals, mesenchymal stem cells (MSCs) in the bone marrow show imbalanced differentiation with a reduction in osteogenesis and an increase in adipogenesis. This leads to impaired osseointegration and implant failure. This review focuses on the molecular mechanisms underlying the dysfunctional differentiation of aged MSCs, which primarily include autophagy, transcription factors, extracellular vesicle secretion, signaling pathways, epigenetic modifications, microRNAs, and oxidative stress. Furthermore, this review addresses the pathological changes in MSCs that affect osseointegration and discusses potential therapeutic interventions to enhance osseointegration by manipulating the mechanisms underlying MSC aging.
Collapse
Affiliation(s)
- Yang Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Siyuan Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Hui Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiaoyu Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yi Shuai
- Nanjing Jinling Hospital: East Region Military Command General Hospital, Nanjing, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| | - Yingjie Mao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| | - Fuming He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Lima LTFD, Ganzella FADO, Cardoso GC, Pires VDS, Chequin A, Santos GL, Braun-Prado K, Galindo CM, Braz Junior O, Molento MB, Acco A, Adami ER, Costa ET, Cavichiolo Franco CR, Klassen G, Ramos EADS. l-carvone decreases breast cancer cells adhesion, migration, and invasion by suppressing FAK activation. Chem Biol Interact 2023; 378:110480. [PMID: 37059214 DOI: 10.1016/j.cbi.2023.110480] [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: 03/09/2021] [Revised: 03/24/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
Breast cancer is one of the most common types of cancer in the world and current therapeutic strategies present severe drawbacks. l-carvone (CRV), a monoterpene found in Mentha spicata (spearmint), has been reported to have potent anti-inflammatory activity. Here, we examined the role of CRV in breast cancer cell adhesion, migration and invasion in vitro and how this component could suppress the growth of Ehrlich carcinoma-bearing mice. In vivo, treatment with CRV significantly decreased tumor growth, increased tumor necrosis area, and reduced the expression of VEGF and HIF-1α in Ehrlich carcinoma-bearing mice. Furthermore, the anticancer efficacy of CRV was similar to currently used chemotherapy (Methotrexate), and the combination of CRV with MTX potentiated the chemotherapy effects. Further mechanistic investigation in vitro revealed that CRV modulates the interaction of breast cancer cells with the extracellular matrix (ECM) by disrupting focal adhesion, which was shown by scanning electron microscopy (SEM) and immunofluorescence. Moreover, CRV caused a decrease in β1-integrin expression and inhibited focal adhesion kinase (FAK) activation. FAK is one of the most important downstream activators of several metastatic processes, including MMP-2 mediated invasion and HIF-1α/VEGF angiogenesis stimulus, both of which were found to be reduced in MDA-MB-231 cells exposed to CRV. Our results provide new insight about targeting β1-integrin/FAK signaling pathway with CRV, which could be a new potential agent in the treatment of breast cancer.
Collapse
Affiliation(s)
- Lucas Trevisan França de Lima
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | | | - Gabriela Casani Cardoso
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Verônica Dos Santos Pires
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Andressa Chequin
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Giulia Luiza Santos
- Molecular Oncology Center, Research and Education Institute, Hospital Sirio-Libanes, São Paulo, SP, Brazil
| | - Karin Braun-Prado
- Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | | | - Odair Braz Junior
- Pos-graduate Program of Cellular and Molecular Biology, Federal University of Parana, Curitiba, PR, Brazil
| | - Marcelo Beltrão Molento
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Veterinary Medicine, Federal University of Parana, Curitiba, PR, Brazil
| | - Alexandra Acco
- Pos-graduate Program of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - Eliana Rezende Adami
- Pos-graduate Program of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - Erico Tosoni Costa
- Molecular Oncology Center, Research and Education Institute, Hospital Sirio-Libanes, São Paulo, SP, Brazil
| | | | - Giseli Klassen
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Edneia Amancio de Souza Ramos
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil.
| |
Collapse
|
4
|
Dunlap B, Patterson GT, Kumar S, Vyavahare S, Mishra S, Isales C, Fulzele S. Vitamin C supplementation for the treatment of osteoarthritis: perspectives on the past, present, and future. Ther Adv Chronic Dis 2021; 12:20406223211047026. [PMID: 34729150 PMCID: PMC8543556 DOI: 10.1177/20406223211047026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/31/2021] [Indexed: 11/21/2022] Open
Abstract
According to the US Centers for Disease Control and Prevention (CDC), an estimated 14% of adults in the United States have either been diagnosed with osteoarthritis (OA) or have symptoms suggestive of the disease. The CDC also points out that the incidence of OA has been gradually increasing over the past 30 years. What is more worrisome is that this trend is going to accelerate due to the aging demographics of the United States and the increasing prevalence of obesity seen in the country. The need for better preventive treatments and efficacious therapeutics are direly needed to combat this public health crisis. Among the possible treatments being hypothesized, antioxidant supplementation has become one of the most widely studied over the past decade due to its ability to attenuate reactive oxygen species (ROS) formation within chondrocytes, a critical step in the pathogenesis of this disease. Vitamin C has emerged as among the most promising of the antioxidant group, with many animal and human studies having been conducted in recent years. Although many of the studies have shown encouraging results in terms of preventing OA, others have reached opposite conclusions, thus making the data controversial. However, after reviewing several of these studies, we hypothesize that certain parameters may not have been properly considered during data collection. In the end, more randomized placebo-controlled trials in humans are desperately needed in order to fully understand whether vitamin C therapy is efficacious in treating and/or preventing OA.
Collapse
Affiliation(s)
- Burton Dunlap
- The University of Tennessee Health Science Center, Chattanooga, TN, USA
| | | | - Sandeep Kumar
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Sagar Vyavahare
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Samarth Mishra
- Department of the College of Science and Mathematics, Augusta University, Augusta, GA, USA
| | - Carlos Isales
- Department of Orthopaedics, Augusta University, Augusta, GA, USA
- Department of Medicine, Augusta University, Augusta, GA, USA
- Center for Healthy Aging, Augusta University, Augusta, GA, USA
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Augusta University, Augusta, GA 30904, USA
- Department of Orthopaedics, Augusta University, Augusta, GA, USA
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
- Center for Healthy Aging, Augusta University, Augusta, GA, USA
| |
Collapse
|
5
|
MicroRNA124 and microRNA21-5p regulate migration, proliferation and differentiation of rat bone marrow mesenchymal stem cells. Biosci Rep 2021; 40:226597. [PMID: 33026076 PMCID: PMC7584812 DOI: 10.1042/bsr20193531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 08/29/2020] [Accepted: 10/02/2020] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that can be a useful source of cells for the treatment of many diseases, including neurologic diseases. The curative effect of MSCs relies mostly on cell’s capacity of migration, proliferation and differentiation. MicroRNAs (miRNAs) are small non-coding RNAs that play important roles on regulating various cell behaviors. Here, we report that miRNA-124 (miR124) and miRNA-21-5p (miR21-5p) display different regulatory roles on migration, proliferation and neuron differentiation of MSCs. MiR124 was shown greatly promoting MSCs migration and neuronal differentiation. MiR21-5p could significantly enhance the proliferation and neuronal differentiation ability of MSCs. MiR124 and miR21-5p synergistically promote differentiation of MSCs into neurons. Collectively, miR124 and miR21-5p can functionally regulate cell migration, proliferation and neuronal differentiation of MSCs. Therefore, miR124 and miR21-5p may be promising tools to improve transplantation efficiency for neural injury.
Collapse
|
6
|
Patterson T, Isales CM, Fulzele S. Low level of Vitamin C and dysregulation of Vitamin C transporter might be involved in the severity of COVID-19 Infection. Aging Dis 2021; 12:14-26. [PMID: 33532123 PMCID: PMC7801272 DOI: 10.14336/ad.2020.0918] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/18/2020] [Indexed: 01/02/2023] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been spreading around the world at an exponential pace, leading to millions of individuals developing the associated disease called COVID-19. Due to the novel nature and the lack of immunity within humans, there has been a collective global effort to find effective treatments against the virus. This has led the scientific community to repurpose Food and Drug Administration (FDA) approved drugs with known safety profiles. Of the many possible drugs, vitamin C has been on the shortlist of possible interventions due to its beneficial role as an immune booster and inherent antioxidant properties. Within this manuscript, a detailed discussion regarding the intracellular function and inherent properties of vitamin C is conducted. It also provides a comprehensive review of published research pertaining to the differences in expression of the vitamin C transporter under several pathophysiologic conditions. Finally, we review recently published research investigating the efficacy of vitamin C administration in treating viral infection and life-threatening conditions. Overall, this manuscript aims to present existing information regarding the extent to which vitamin C can be an effective treatment for COVID-19 and possible explanations as to why it may work in some individuals but not in others.
Collapse
Affiliation(s)
- Taylor Patterson
- Department of Medicine, Augusta University, Augusta, GA 30912, USA.
| | - Carlos M Isales
- Center for Healthy Aging, Augusta University, Augusta, GA 30912, USA
- Department of Cell biology and anatomy, Augusta University, Augusta, GA 30912, USA
| | - Sadanand Fulzele
- Department of Medicine, Augusta University, Augusta, GA 30912, USA.
- Center for Healthy Aging, Augusta University, Augusta, GA 30912, USA
- Department of Cell biology and anatomy, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
7
|
Potter ML, Hill WD, Isales CM, Hamrick MW, Fulzele S. MicroRNAs are critical regulators of senescence and aging in mesenchymal stem cells. Bone 2021; 142:115679. [PMID: 33022453 PMCID: PMC7901145 DOI: 10.1016/j.bone.2020.115679] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/16/2020] [Accepted: 07/28/2020] [Indexed: 01/10/2023]
Abstract
MicroRNAs (miRNAs) have recently come under scrutiny for their role in various age-related diseases. Similarly, cellular senescence has been linked to disease and aging. MicroRNAs and senescence likely play an intertwined role in driving these pathologic states. In this review, we present the connection between these two drivers of age-related disease concerning mesenchymal stem cells (MSCs). First, we summarize key miRNAs that are differentially expressed in MSCs and other musculoskeletal lineage cells during senescence and aging. Additionally, we also reviewed miRNAs that are regulated via traditional senescence-associated secretory phenotype (SASP) cytokines in MSC. Lastly, we summarize miRNAs that have been found to target components of the cell cycle arrest pathways inherently activated in senescence. This review attempts to highlight potential miRNA targets for regenerative medicine applications in age-related musculoskeletal disease.
Collapse
Affiliation(s)
- Matthew L Potter
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - William D Hill
- Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC, 29403, United States of America
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America; Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America; Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, United States of America.
| |
Collapse
|
8
|
Ye D, Chen C, Wang Q, Zhang Q, Li S, Liu H. Short-wave enhances mesenchymal stem cell recruitment in fracture healing by increasing HIF-1 in callus. Stem Cell Res Ther 2020; 11:382. [PMID: 32894200 PMCID: PMC7487968 DOI: 10.1186/s13287-020-01888-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 01/14/2023] Open
Abstract
Background As a type of high-frequency electrotherapy, a short-wave can promote the fracture healing process; yet, its underlying therapeutic mechanisms remain unclear. Purpose To observe the effect of Short-Wave therapy on mesenchymal stem cell (MSC) homing and relative mechanisms associated with fracture healing. Materials and methods For in vivo study, the effect of Short-Wave therapy to fracture healing was examined in a stabilized femur fracture model of 40 SD rats. Radiography was used to analyze the morphology and microarchitecture of the callus. Additionally, fluorescence assays were used to analyze the GFP-labeled MSC homing after treatment in 20 nude mice with a femoral fracture. For in vitro study, osteoblast from newborn rats simulated fracture site was first irradiated by the Short-Wave; siRNA targeting HIF-1 was used to investigate the role of HIF-1. Osteoblast culture medium was then collected as chemotaxis content of MSC, and the migration of MSC from rats was evaluated using wound healing assay and trans-well chamber test. The expression of HIF-1 and its related factors were quantified by q RT-PCR, ELISA, and Western blot. Results Our in vivo experiment indicated that Short-Wave therapy could promote MSC migration, increase local and serum HIF-1 and SDF-1 levels, induce changes in callus formation, and improve callus microarchitecture and mechanical properties, thus speeding up the healing process of the fracture site. Moreover, the in vitro results further indicated that Short-Wave therapy upregulated HIF-1 and SDF-1 expression in osteoblast and its cultured medium, as well as the expression of CXCR-4, β-catenin, F-actin, and phosphorylation levels of FAK in MSC. On the other hand, the inhibition of HIF-1α was significantly restrained by the inhibition of HIF-1α in osteoblast, and it partially inhibited the migration of MSC. Conclusions These results suggested that Short-Wave therapy could increase HIF-1 in callus, which is one of the crucial mechanisms of chemotaxis MSC homing in fracture healing.
Collapse
Affiliation(s)
- Dongmei Ye
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China.
| | - Chen Chen
- Department of Anatomy, Medical College of Dalian University, Dalian, China
| | - Qiwen Wang
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China.,Department of Rehabilitation, The people's Hospital of Longhua District, Shenzhen, China
| | - Qi Zhang
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Sha Li
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Hongwei Liu
- Department of Rehabilitation, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| |
Collapse
|
9
|
Wang Y, Bian Y, Zhou L, Feng B, Weng X, Liang R. Biological evaluation of bone substitute. Clin Chim Acta 2020; 510:544-555. [PMID: 32798511 DOI: 10.1016/j.cca.2020.08.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 01/02/2023]
Abstract
Critical-sized defects (CSDs) caused by trauma, tumor resection, or skeletal abnormalities create a high demand for bone repair materials (BRMs). Over the years, scientists have been trying to develop BRMs and evaluate their efficacy using numerous developed methods. BRMs are characterized by osteogenesis and angiogenesis promoting properties, the latter of which has rarely been studied in vitro and in vivo. While blood vessels are required to provide nutrients. Bone mass maintains a dynamic balance under the joint action of osteolytic and osteogenic activity in which monocytes differentiate into osteolytic cells, and osteoprogenitor cells differentiate into osteogenic cells. This review would be helpful for inexperienced researchers as well as present a comprehensive overview of methods used to investigate the effect of BRMs on osteogenic cells, osteolytic cells, and blood vessels, as well as their biocompatibility and biological performance. This review is expected to facilitate further research and development of new BRMs.
Collapse
Affiliation(s)
- Yingjie Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yanyan Bian
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Lizhi Zhou
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Bin Feng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Xisheng Weng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
10
|
Fiorani M, Scotti M, Guidarelli A, Burattini S, Falcieri E, Cantoni O. SVCT2-Dependent plasma and mitochondrial membrane transport of ascorbic acid in differentiating myoblasts. Pharmacol Res 2020; 159:105042. [PMID: 32580031 DOI: 10.1016/j.phrs.2020.105042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022]
Abstract
The Na+-dependent Vitamin C transporter 2 (SVCT2) is expressed in the plasma and mitochondrial membranes of various cell types. This notion was also established in proliferating C2C12 myoblasts (Mb), in which the transporter was characterised by a high and low affinity in the plasma and mitochondrial membranes, respectively. In addition, the mitochondrial expression of SVCT2 appeared particularly elevated and, consistently, a brief pre-exposure to low concentrations of Ascorbic Acid (AA) abolished mitochondrial superoxide formation selectively induced by the cocktail arsenite/ATP. Early myotubes (Mt) derived from these cells after 4 days of differentiation presented evidence of slightly increased SVCT2 expression, and were characterised by kinetic parameters for plasma membrane transport of AA in line with those detected in Mb. Confocal microscopy studies indicated that the mitochondrial expression of SVCT2 is well preserved in Mt with one or two nuclei, but progressively reduced in Mt with three or more nuclei. Cellular and mitochondrial expression of SVCT2 was found reduced in day 7 Mt. While the uptake studies were compromised by the poor purity of the mitochondrial preparations obtained from day 4 Mt, we nevertheless obtained evidence of poor transport of the vitamin using the same functional studies successfully employed with Mb. Indeed, even greater concentrations of/longer pre-exposure to AA failed to induce scavenging of mitochondrial superoxide in Mt. These results are therefore indicative of a severely reduced mitochondrial uptake of the vitamin in early Mt, attributable to decreased expression as well as impaired activity of mitochondrial SVCT2.
Collapse
Affiliation(s)
- Mara Fiorani
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, 61029 Urbino, Italy.
| | - Maddalena Scotti
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, 61029 Urbino, Italy.
| | - Andrea Guidarelli
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, 61029 Urbino, Italy.
| | - Sabrina Burattini
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, 61029 Urbino, Italy.
| | - Elisabetta Falcieri
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, 61029 Urbino, Italy.
| | - Orazio Cantoni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, 61029 Urbino, Italy.
| |
Collapse
|
11
|
Weavers H, Wood W, Martin P. Injury Activates a Dynamic Cytoprotective Network to Confer Stress Resilience and Drive Repair. Curr Biol 2019; 29:3851-3862.e4. [PMID: 31668626 PMCID: PMC6868510 DOI: 10.1016/j.cub.2019.09.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 02/07/2023]
Abstract
In healthy individuals, injured tissues rapidly repair themselves following damage. Within a healing skin wound, recruited inflammatory cells release a multitude of bacteriocidal factors, including reactive oxygen species (ROS), to eliminate invading pathogens. Paradoxically, while these highly reactive ROS confer resistance to infection, they are also toxic to host tissues and may ultimately delay repair. Repairing tissues have therefore evolved powerful cytoprotective "resilience" machinery to protect against and tolerate this collateral damage. Here, we use in vivo time-lapse imaging and genetic manipulation in Drosophila to dissect the molecular and cellular mechanisms that drive tissue resilience to wound-induced stress. We identify a dynamic, cross-regulatory network of stress-activated cytoprotective pathways, linking calcium, JNK, Nrf2, and Gadd45, that act to both "shield" tissues from oxidative damage and promote efficient damage repair. Ectopic activation of these pathways confers stress protection to naive tissue, while their inhibition leads to marked delays in wound closure. Strikingly, the induction of cytoprotection is tightly linked to the pathways that initiate the inflammatory response, suggesting evolution of a fail-safe mechanism for tissue protection each time inflammation is triggered. A better understanding of these resilience mechanisms-their identities and precise spatiotemporal regulation-is of major clinical importance for development of therapeutic interventions for all pathologies linked to oxidative stress, including debilitating chronic non-healing wounds.
Collapse
Affiliation(s)
- Helen Weavers
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK.
| | - Will Wood
- School of Cellular and Molecular Medicine, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK; Centre for Inflammation Research, University of Edinburgh, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK; School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK; School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
12
|
Age-Dependent Oxidative Stress Elevates Arginase 1 and Uncoupled Nitric Oxide Synthesis in Skeletal Muscle of Aged Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1704650. [PMID: 31205583 PMCID: PMC6530149 DOI: 10.1155/2019/1704650] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/04/2019] [Indexed: 12/21/2022]
Abstract
Aging is associated with reduced muscle mass (sarcopenia) and poor bone quality (osteoporosis), which together increase the incidence of falls and bone fractures. It is widely appreciated that aging triggers systemic oxidative stress, which can impair myoblast cell survival and differentiation. We previously reported that arginase plays an important role in oxidative stress-dependent bone loss. We hypothesized that arginase activity is dysregulated with aging in muscles and may be involved in muscle pathophysiology. To investigate this, we analyzed arginase activity and its expression in skeletal muscles of young and aged mice. We found that arginase activity and arginase 1 expression were significantly elevated in aged muscles. We also demonstrated that SOD2, GPx1, and NOX2 increased with age in skeletal muscle. Most importantly, we also demonstrated elevated levels of peroxynitrite formation and uncoupling of eNOS in aged muscles. Our in vitro studies using C2C12 myoblasts showed that the oxidative stress treatment increased arginase activity, decreased cell survival, and increased apoptotic markers. These effects were reversed by treatment with an arginase inhibitor, 2(S)-amino-6-boronohexanoic acid (ABH). Our study provides strong evidence that L-arginine metabolism is altered in aged muscle and that arginase inhibition could be used as a novel therapeutic target for age-related muscle complications.
Collapse
|
13
|
Wang C, Tanjaya J, Shen J, Lee S, Bisht B, Pan HC, Pang S, Zhang Y, Berthiaume EA, Chen E, Da Lio AL, Zhang X, Ting K, Guo S, Soo C. Peroxisome Proliferator-Activated Receptor-γ Knockdown Impairs Bone Morphogenetic Protein-2-Induced Critical-Size Bone Defect Repair. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:648-664. [PMID: 30593824 PMCID: PMC6412314 DOI: 10.1016/j.ajpath.2018.11.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/13/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
The Food and Drug Administration-approved clinical dose (1.5 mg/mL) of bone morphogenetic protein-2 (BMP2) has been reported to induce significant adverse effects, including cyst-like adipose-infiltrated abnormal bone formation. These undesirable complications occur because of increased adipogenesis, at the expense of osteogenesis, through BMP2-mediated increases in the master regulatory gene for adipogenesis, peroxisome proliferator-activated receptor-γ (PPARγ). Inhibiting PPARγ during osteogenesis has been suggested to drive the differentiation of bone marrow stromal/stem cells toward an osteogenic, rather than an adipogenic, lineage. We demonstrate that knocking down PPARγ while concurrently administering BMP2 can reduce adipogenesis, but we found that it also impairs BMP2-induced osteogenesis and leads to bone nonunion in a mouse femoral segmental defect model. In addition, in vitro studies using the mouse bone marrow stromal cell line M2-10B4 and mouse primary bone marrow stromal cells confirmed that PPARγ knockdown inhibits BMP2-induced adipogenesis; attenuates BMP2-induced cell proliferation, migration, invasion, and osteogenesis; and escalates BMP2-induced cell apoptosis. More important, BMP receptor 2 and 1B expression was also significantly inhibited by the combined BMP2 and PPARγ knockdown treatment. These findings indicate that PPARγ is critical for BMP2-mediated osteogenesis during bone repair. Thus, uncoupling BMP2-mediated osteogenesis and adipogenesis using PPARγ inhibition to combat BMP2's adverse effects may not be feasible.
Collapse
Affiliation(s)
- Chenchao Wang
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, People's Republic of China; Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Justine Tanjaya
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Jia Shen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Soonchul Lee
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Bharti Bisht
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Hsin Chuan Pan
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Shen Pang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Yulong Zhang
- Departments of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, California
| | - Emily A Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Eric Chen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Andrew L Da Lio
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Shu Guo
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, People's Republic of China.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
14
|
Fariyike B, Singleton Q, Hunter M, Hill WD, Isales CM, Hamrick MW, Fulzele S. Role of MicroRNA-141 in the Aging Musculoskeletal System: A Current Overview. Mech Ageing Dev 2019; 178:9-15. [PMID: 30528652 PMCID: PMC6998035 DOI: 10.1016/j.mad.2018.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/31/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023]
Abstract
MicroRNA's are small non-coding RNAs that regulate the expression of genes by targeting the 3' UTR's of mRNA. Studies reveal that miRNAs play a pivotal role in normal musculoskeletal function such as mesenchymal stem cell differentiation, survivability and apoptosis, osteogenesis, and chondrogenesis. Changes in normal miRNA expression have been linked to a number of pathological disease processes. Additionally, with aging, it is noted that there is dysregulation in the normal function of stem cell differentiation, bone formation/degradation, chondrocyte function, and muscle degeneration. Due to the change in expression of miRNA in degenerative musculoskeletal pathology, it is believed that these molecules may be at least partially responsible for cellular dysfunction. A number of miRNAs have already been identified to play a role in osteoarthritis, osteoporosis and sarcopenia. One miRNA that has become of interest recently is miRNA 141. The purpose of this article is to review the current literature available on miRNA 141 and how it could play a role in osteoporosis, osteoarthritis and musculoskeletal pathology overall.
Collapse
Affiliation(s)
- Babatunde Fariyike
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - Quante Singleton
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - Monte Hunter
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - William D Hill
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Medicine, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States.
| |
Collapse
|
15
|
Differentiation of Promonocytic U937 Cells to Monocytes Is Associated with Reduced Mitochondrial Transport of Ascorbic Acid. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4194502. [PMID: 29576847 PMCID: PMC5822789 DOI: 10.1155/2018/4194502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/30/2017] [Accepted: 12/26/2017] [Indexed: 12/20/2022]
Abstract
Growth of promonocytic U937 cells in the presence of DMSO promotes their differentiation to monocytes. After 4 days of culture in differentiating medium, these cells ceased to proliferate, displayed downregulated ryanodine receptor expression, and responded to specific stimuli with enhanced NADPH-oxidase-derived superoxide formation or cytosolic phospholipase A2-dependent arachidonic acid release. We found that the 4-day differentiation process is also associated with downregulated SVCT2 mRNA expression, in the absence of apparent changes in SVCT2 protein expression and transport rate of ascorbic acid (AA). Interestingly, under the same conditions, these cells accumulated lower amounts of the vitamin in their mitochondria, with an ensuing reduced response to external stimuli sensitive to the mitochondrial fraction of AA. Further analyses demonstrated an unexpected increase in mitochondrial SVCT2 protein expression, however, associated with reduced SVCT2-dependent AA uptake in isolated mitochondria. A decrease in the transporter Vmax, with no change in affinity, was found to account for this response. Differentiation of promonocytic cells to monocytes is therefore characterized by decreased SVCT2 mRNA expression that, even prior to the onset of SVCT2 protein downregulation or apparent changes in plasma membrane transport activity, impacts on the mitochondrial accumulation of the vitamin through a decreased Vmax of the transporter.
Collapse
|
16
|
Burke J, Hunter M, Kolhe R, Isales C, Hamrick M, Fulzele S. Therapeutic potential of mesenchymal stem cell based therapy for osteoarthritis. Clin Transl Med 2016; 5:27. [PMID: 27510262 PMCID: PMC4980326 DOI: 10.1186/s40169-016-0112-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease affecting articular cartilage in joints, and it is a leading cause of disability in the United States. Current pharmacological treatment strategies are ineffective to prevent the OA progression; however, cellular therapies have the potential to regenerate the lost cartilage, combat cartilage degeneration, provide pain relief, and improve patient mobility. One of the most promising sources of cellular regenerative medicine is from mesenchymal stem cells (MSCs). MSCs can be isolated from adipose tissue, bone marrow, synovial tissue, and other sources. The aim of this review is to compile recent advancement in cellular based therapy more specifically in relation to MSCs in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- John Burke
- Department of Orthopedics, Georgia Regents University, Augusta, GA, USA
| | - Monte Hunter
- Department of Orthopedics, Georgia Regents University, Augusta, GA, USA
| | - Ravindra Kolhe
- Department of Pathology, Georgia Regents University, Augusta, GA, USA
| | - Carlos Isales
- Department of Orthopedics, Georgia Regents University, Augusta, GA, USA.,Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA
| | - Mark Hamrick
- Department of Cell Biology and Anatomy, Georgia Regents University, Augusta, GA, USA.,Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Orthopedics, Georgia Regents University, Augusta, GA, USA. .,Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA. .,Department of Orthopedics Surgery, Augusta University, Augusta, GA, 30904, USA.
| |
Collapse
|
17
|
15-Deoxy-Δ(12,14)-Prostaglandin J2 Inhibits Homing of Bone Marrow-Derived Mesenchymal Stem Cells Triggered by Chronic Liver Injury via Redox Pathway. PPAR Res 2015; 2015:876160. [PMID: 26457076 PMCID: PMC4592740 DOI: 10.1155/2015/876160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/27/2015] [Indexed: 12/24/2022] Open
Abstract
It has been reported that bone marrow-derived mesenchymal stem cells (BMSCs) have capacity to migrate to the damaged liver and contribute to fibrogenesis in chronic liver diseases. 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), an endogenous ligand for peroxisome proliferator-activated receptor gamma (PPARγ), is considered a new inhibitor of cell migration. However, the actions of 15d-PGJ2 on BMSC migration remain unknown. In this study, we investigated the effects of 15d-PGJ2 on the migration of BMSCs using a mouse model of chronic liver fibrosis and primary mouse BMSCs. Our results demonstrated that in vivo, 15d-PGJ2 administration inhibited the homing of BMSCs to injured liver by flow cytometric analysis and, in vitro, 15d-PGJ2 suppressed primary BMSC migration in a dose-dependent manner determined by Boyden chamber assay. Furthermore, the repressive effect of 15d-PGJ2 was blocked by reactive oxygen species (ROS) inhibitor, but not PPARγ antagonist, and action of 15d-PGJ2 was not reproduced by PPARγ synthetic ligands. In addition, 15d-PGJ2 triggered a significant ROS production and cytoskeletal remodeling in BMSCs. In conclusion, our results suggest that 15d-PGJ2 plays a crucial role in homing of BMSCs to the injured liver dependent on ROS production, independently of PPARγ, which may represent a new strategy in the treatment of liver fibrosis.
Collapse
|
18
|
Bostancıoğlu RB, Peksen C, Genc H, Gürbüz M, Karel FB, Koparal AS, Dogan A, Kose N, Koparal AT. Analyses of the modulatory effects of antibacterial silver doped calcium phosphate-based ceramic nano-powder on proliferation, survival, and angiogenic capacity of different mammalian cells in vitro. ACTA ACUST UNITED AC 2015; 10:045024. [PMID: 26306474 DOI: 10.1088/1748-6041/10/4/045024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In this study, the antibacterial, cytotoxic, and angiogenic activities of silver doped calcium phosphate-based inorganic powder (ABT or PAG) were systematically investigated. ABT powders containing varying silver content were fabricated using a wet chemical manufacturing method. Antibacterial efficiencies of the ABT powders were investigated using a standard test with indicator bacteria and yeast. The cytotoxic effects of ABT on three different fibroblast cells and human umbilical vein endothelial cells (HUVECs) were assessed using MTT assay. ABT powder exhibits concentration-related cytotoxicity characteristics. Apoptotic activity, attachment capability, and wound healing effects were examined on fibroblasts. The angiogenic activity of ABT was investigated by tube formation assay in HUVECs; 10 μg ml(-1) and 100 μg ml(-1) concentrations of the highest metal ion content of ABT did not disrupt the tube formation of HUVECs. All these tests showed that ABT does not compromise the survival of the cells and might impose regeneration ability to various cell types. These results indicate that silver doped calcium phosphate-based inorganic powder with an optimal silver content has good potential for developing new biomaterials for implant applications.
Collapse
|
19
|
Mohammed BM, Fisher BJ, Kraskauskas D, Ward S, Wayne JS, Brophy DF, Fowler AA, Yager DR, Natarajan R. Vitamin C promotes wound healing through novel pleiotropic mechanisms. Int Wound J 2015; 13:572-84. [PMID: 26290474 DOI: 10.1111/iwj.12484] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/27/2015] [Accepted: 07/14/2015] [Indexed: 01/11/2023] Open
Abstract
Vitamin C (VitC) or ascorbic acid (AscA), a cofactor for collagen synthesis and a primary antioxidant, is rapidly consumed post-wounding. Parenteral VitC administration suppresses pro-inflammatory responses while promoting anti-inflammatory and pro-resolution effects in human/murine sepsis. We hypothesised that VitC could promote wound healing by altering the inflammatory, proliferative and remodelling phases of wound healing. Mice unable to synthesise VitC (Gulo(-/-) ) were used in this study. VitC was provided in the water (sufficient), withheld from another group (deficient) and supplemented by daily intra-peritoneal infusion (200 mg/kg, deficient + AscA) in a third group. Full thickness excisional wounds (6 mm) were created and tissue collected on days 7 and 14 for histology, quantitative polymerase chain reaction (qPCR) and Western blotting. Human neonatal dermal fibroblasts (HnDFs) were used to assess effects of In conclusion, VitC favorably on proliferation. Histological analysis showed improved wound matrix deposition and organisation in sufficient and deficient +AscA mice. Wounds from VitC sufficient and deficient + AscA mice had reduced expression of pro-inflammatory mediators and higher expression of wound healing mediators. Supplementation of HnDF with AscA induced the expression of self-renewal genes and promoted fibroblast proliferation. VitC favourably impacts the spatiotemporal expression of transcripts associated with early resolution of inflammation and tissue remodelling.
Collapse
Affiliation(s)
- Bassem M Mohammed
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA.,Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Bernard J Fisher
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Donatas Kraskauskas
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Susan Ward
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Jennifer S Wayne
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Donald F Brophy
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA
| | - Alpha A Fowler
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Dorne R Yager
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Ramesh Natarajan
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
20
|
Sangani R, Periyasamy-Thandavan S, Kolhe R, Bhattacharyya MH, Chutkan N, Hunter M, Isales C, Hamrick M, Hill WD, Fulzele S. MicroRNAs-141 and 200a regulate the SVCT2 transporter in bone marrow stromal cells. Mol Cell Endocrinol 2015; 410:19-26. [PMID: 25617715 PMCID: PMC4824062 DOI: 10.1016/j.mce.2015.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/07/2015] [Accepted: 01/07/2015] [Indexed: 12/16/2022]
Abstract
Vitamin C is a micro-nutrient which plays an important role in bone marrow stromal cell (BMSCs) differentiation to osteogenesis. This vitamin is transported into the BMSCs through the sodium dependent vitamin C transporter 2 (SVCT2). We previously reported that knockdown of the SVCT2 transporter decreases osteogenic differentiation. However, our understanding of the post-transcriptional regulatory mechanism of the SVCT2 transporter remains poor. MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally regulate the messenger RNAs of protein-coding genes. In this study, we aimed to investigate the impact of miR-141 and miR-200a on SVCT2 expression. We found that mouse BMSCs expressed miR-141 and miR-200a and repressed SVCT2 expression at the functional level by targeting the 3'-untranslated region of mRNA. We also found that miR-141 and miR-200a decreased osteogenic differentiation. Furthermore, miRNA inhibitors increased SVCT2 and osteogenic gene expression in BMSCs. Taken together, these results indicate that both miRNAs are novel regulators of the SVCT2 transporter and play an important role in the osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Rajnikumar Sangani
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Ravindra Kolhe
- Department of Pathology, Georgia Regents University, Augusta, GA 30912, USA
| | - Maryka H Bhattacharyya
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Monte Hunter
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Carlos Isales
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Mark Hamrick
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - William D Hill
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
21
|
Sangani R, Periyasamy-Thandavan S, Pathania R, Ahmad S, Kutiyanawalla A, Kolhe R, Bhattacharyya MH, Chutkan N, Hunter M, Hill WD, Hamrick M, Isales C, Fulzele S. The crucial role of vitamin C and its transporter (SVCT2) in bone marrow stromal cell autophagy and apoptosis. Stem Cell Res 2015. [PMID: 26210298 DOI: 10.1016/j.scr.2015.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Vitamin C is an antioxidant that plays a vital role in various biological processes including bone formation. Previously, we reported that vitamin C is transported into bone marrow stromal cells (BMSCs) through the sodium dependent Vitamin C Transporter 2 (SVCT2) and this transporter plays an important role in osteogenic differentiation. Furthermore, this transporter is regulated by oxidative stress. To date, however, the exact role of vitamin C and its transporter (SVCT2) in ROS regulated autophagy and apoptosis in BMSCs is poorly understood. In the present study, we observed that oxidative stress decreased survival of BMSCs in a dose-dependent manner and induced growth arrest in the G1 phase of the cell cycle. These effects were accompanied by the induction of autophagy, confirmed by P62 and LC3B protein level and punctate GFP-LC3B distribution. The supplementation of vitamin C significantly rescued the BMSCs from oxidative stress by regulating autophagy. Knockdown of the SVCT2 transporter in BMSCs synergistically decreased cell survival even under low oxidative stress conditions. Also, supplementing vitamin C failed to rescue cells from stress. Our results reveal that the SVCT2 transporter plays a vital role in the mechanism of BMSC survival under stress conditions. Altogether, this study has given new insight into the role of the SVCT2 transporter in oxidative stress related autophagy and apoptosis in BMSCs.
Collapse
Affiliation(s)
- Rajnikumar Sangani
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Rajneesh Pathania
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, GA 30912, USA
| | - Saif Ahmad
- Department of Ophthalmology, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Ravindra Kolhe
- Department of Pathology, Georgia Regents University, Augusta, GA 30912, USA
| | - Maryka H Bhattacharyya
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Norman Chutkan
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Monte Hunter
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - William D Hill
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Mark Hamrick
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Carlos Isales
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
22
|
Kao TT, Chu CY, Lee GH, Hsiao TH, Cheng NW, Chang NS, Chen BH, Fu TF. Folate deficiency-induced oxidative stress contributes to neuropathy in young and aged zebrafish--implication in neural tube defects and Alzheimer's diseases. Neurobiol Dis 2014; 71:234-44. [PMID: 25131448 DOI: 10.1016/j.nbd.2014.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/08/2014] [Accepted: 08/02/2014] [Indexed: 12/26/2022] Open
Abstract
Folate is a nutrient essential for the development, function and regeneration of nervous systems. Folate deficiency has been linked to many neurological disorders including neural tube defects in fetus and Alzheimer's diseases in the elderly. However, the etiology underlying these folate deficiency-associated diseases is not completely understood. In this study, zebrafish transgenic lines with timing and duration-controllable folate deficiency were developed by ectopically overexpressing a recombinant EGFP-γ-glutamyl hydrolase (γGH). Impeded neural crest cell migration was observed in the transgenic embryos when folate deficiency was induced in early stages, leading to defective neural tube closure and hematopoiesis. Adding reduced folate or N-acetylcysteine reversed the phenotypic anomalies, supporting the causal link between the increased oxidative stress and the folate deficiency-induced abnormalities. When folate deficiency was induced in aged fish accumulation of beta-amyloid and phosphorylated Tau protein were found in the fish brain cryo-sections. Increased autophagy and accumulation of acidic autolysosome were apparent in folate deficient neuroblastoma cells, which were reversed by reduced folate or N-acetylcysteine supplementation. Decreased expression of cathepsin B, a lysosomal protease, was also observed in cells and tissue with folate deficiency. We concluded that folate deficiency-induced oxidative stress contributed to the folate deficiency-associated neuropathogenesis in both early and late stages of life.
Collapse
Affiliation(s)
- Tseng-Ting Kao
- The Institute of Basic Medical Science, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yi Chu
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Gang-Hui Lee
- The Institute of Basic Medical Science, National Cheng Kung University, Tainan, Taiwan
| | - Tsun-Hsien Hsiao
- The Institute of Basic Medical Science, National Cheng Kung University, Tainan, Taiwan
| | - Nai-Wei Cheng
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Nan-Shan Chang
- The Institute of Basic Medical Science, National Cheng Kung University, Tainan, Taiwan; Institute of Molecular Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Fun Fu
- The Institute of Basic Medical Science, National Cheng Kung University, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|