1
|
Chen PF, Huang GX, Gu WT, Zhuang GD, Chen C, Wang SM, Tang D. Exploration of the Mechanisms of Bu-Yang-Huan-Wu Decoction in the Treatment of Diabetic Peripheral Neuropathy by Integrating of Serum Pharmacochemistry and Network Pharmacology. Chem Biodivers 2024; 21:e202400910. [PMID: 39105318 DOI: 10.1002/cbdv.202400910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Indexed: 08/07/2024]
Abstract
Diabetic peripheral neuropathy (DPN) is a significant and frequent complication of diabetes. Bu-Yang-Huan-Wu Decoction (BHD) is a classic traditional Chinese herbal prescription that is commonly used in modern clinical practice for the effective treatment of DPN, but the underlying mechanism is not yet clearly defined. The chemical constituents of BHD were characterized by UPLC-Q-Orbitrap HR MS/MS, and a total of 101 chemical components were identified, including 30 components absorbed into blood. An interaction network of "compound-target-disease" interactions was constructed based on the compounds detected absorbed in blood and their corresponding targets of diabetic neuropathy acquired from disease gene databases, and the possible biological targets and potential signalling pathways of BHD were predicted via network pharmacology analysis. Subsequently, methylglyoxal-induced (MGO-induced) Schwann cells (SCs) were used to identify the active ingredients in blood components of BHD and verify the molecular mechanisms of BHD. Through network topological analysis, 30 shared targets strongly implicated in the anti-DPN effects of BHD were identifed. Combined network pharmacology and in vitro cellular analysis, we found that the active ingredient of BHD may treat DPN by modulating the AGEs/RAGE pathway. This study provides valuable evidence for future mechanistic studies and potential therapeutic applications for patients with DPN.
Collapse
Affiliation(s)
- Peng-Fei Chen
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Guang-Xiao Huang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wen-Ting Gu
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Guo-Dong Zhuang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Chong Chen
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Shu-Mei Wang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Dan Tang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering & Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| |
Collapse
|
2
|
Mokhtari S, Mahdavi AH, Jafarpour F, Andani MR, Dattilo M, Nasr-Esfahani MH. Taurine, alpha lipoic acid and vitamin B6 ameliorate the reduced developmental competence of immature mouse oocytes exposed to methylglyoxal. Sci Rep 2024; 14:17937. [PMID: 39095405 PMCID: PMC11297043 DOI: 10.1038/s41598-024-66785-5] [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: 12/23/2023] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
Advanced glycation end products (AGEs) are the final products of the Maillard reaction, formed through the interaction of carbohydrates and proteins. Reactive dicarbonyl compounds such as methylglyoxal (MGO) serve as precursors for AGEs formation. Elevated levels of MGO/AGEs are observed in conditions like obesity, polycystic ovarian syndrome (PCOS), and diabetes, negatively impacting oocyte development. Previous studies have shown that hydrogen sulfide, a gasotransmitter with anti-AGEs effects, is produced in a process influenced by vitamin B6. R-α-lipoic acid (ALA) inhibits protein glycation and AGEs formation while stimulating glutathione (GSH) production. Taurine mitigates oxidative stress and acts as an anti-glycation compound, preventing in vitro glycation and AGEs accumulation. This study aimed to explore the ameliorative effects of a micronutrient support (Taurine, ALA and B6: TAB) on mouse oocytes challenged with MGO. Our results indicate that MGO reduces oocyte developmental competence, while TAB supplementation improves maturation, fertilization, and blastocyst formation rates. TAB also restores cell lineage allocation, redox balance and mitigates mitochondrial dysfunction in MGO-challenged oocytes. Furthermore, cumulus cells express key enzymes in the transsulfuration pathway, and TAB enhances their mRNA expression. However, TAB does not rescue MGO-induced damage in denuded oocytes, emphasizing the supportive role of cumulus cells. Overall, these findings suggest that TAB interventions may have significant implications for addressing reproductive dysfunctions associated with elevated MGO/AGEs levels. This study highlights the potential of TAB supplementation in preserving the developmental competence of COCs exposed to MGO stress, providing insights into mitigating the impact of dicarbonyl stress on oocyte quality and reproductive outcomes.
Collapse
Affiliation(s)
- Saba Mokhtari
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Amir Hossein Mahdavi
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, Iran.
| | - Farnoosh Jafarpour
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohsen Rahimi Andani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
3
|
Sun X, Liu N, Sun C, Xu Y, Ding D, Kong J. The inhibitory effect of vitamin D on myocardial homocysteine levels involves activation of Nrf2-mediated methionine synthase. J Steroid Biochem Mol Biol 2023; 231:106303. [PMID: 36990164 DOI: 10.1016/j.jsbmb.2023.106303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/18/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Homocysteine (Hcy) is a synthetic amino acid containing sulfhydryl group, which is an intermediate product of the deep metabolic pathway of methionine and cysteine. The abnormal increase in fasting plasma total Hcy concentration caused by various factors is called hyperhomocysteine (HHcy). HHcy is closely relevant to the occurrence and progression of diverse cardiovascular and cerebrovascular diseases, such as coronary heart disease, hypertension and diabetes, etc. Vitamin D/vitamin D receptor (VDR) pathway is pointed out that prevent cardiovascular disease by reducing serum homocysteine levels. Our research is designed to explore the potential mechanism of vitamin D in the prevention and treatment of HHcy. METHODS AND RESULTS The Hcy and 25(OH)D3 levels in mouse myocardial tissue, serum or myocardial cells were detected using ELISA kits. The expression levels of VDR, Nrf2 and methionine synthase (MTR) were observed using Western blotting, immunohistochemistry and real time polymerase chain reaction (PCR). General information of the mice, including diet, water intake and body weight, was recorded. Vitamin D up-regulated the mRNA and protein expression of Nrf2 and MTR in mouse myocardial tissue and cells. CHIP assay determined that the combination of Nrf2 binding to the S1 site of the MTR promoter in cardiomyocytes using traditional PCR and real time PCR. Dual Luciferase Assay was applied to detect the transcriptional control of Nrf2 on MTR. The up-regulation effect of Nrf2 on MTR was verified by Nrf2 knockout and overexpression in cardiomyocytes. The role of Nrf2 in vitamin D inhibition of Hcy was revealed using Nrf2-knockdown HL-1 cells and Nrf2 heterozygous mice. Western blotting, real time PCR, IHC staining and ELISA showed that Nrf2 deficiency could restrain the increase in MTR expression and the decrease in Hcy level induced by vitamin D. The transcriptional activities of Nrf2/MTR were activated by vitamin D/VDR with a decrease in Hcy. CONCLUSION Vitamin D/VDR upregulates MTR in an Nrf2-dependent manner, thereby reducing the risk of HHcy.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ning Liu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Can Sun
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ding Ding
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Juan Kong
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
4
|
Aschner M, Skalny AV, Ke T, da Rocha JBT, Paoliello MMB, Santamaria A, Bornhorst J, Rongzhu L, Svistunov AA, Djordevic AB, Tinkov AA. Hydrogen Sulfide (H 2S) Signaling as a Protective Mechanism against Endogenous and Exogenous Neurotoxicants. Curr Neuropharmacol 2022; 20:1908-1924. [PMID: 35236265 PMCID: PMC9886801 DOI: 10.2174/1570159x20666220302101854] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/10/2022] [Accepted: 02/27/2022] [Indexed: 11/22/2022] Open
Abstract
In view of the significant role of H2S in brain functioning, it is proposed that H2S may also possess protective effects against adverse effects of neurotoxicants. Therefore, the objective of the present review is to discuss the neuroprotective effects of H2S against toxicity of a wide spectrum of endogenous and exogenous agents involved in the pathogenesis of neurological diseases as etiological factors or key players in disease pathogenesis. Generally, the existing data demonstrate that H2S possesses neuroprotective effects upon exposure to endogenous (amyloid β, glucose, and advanced-glycation end-products, homocysteine, lipopolysaccharide, and ammonia) and exogenous (alcohol, formaldehyde, acrylonitrile, metals, 6-hydroxydopamine, as well as 1-methyl-4-phenyl- 1,2,3,6- tetrahydropyridine (MPTP) and its metabolite 1-methyl-4-phenyl pyridine ion (MPP)) neurotoxicants. On the one hand, neuroprotective effects are mediated by S-sulfhydration of key regulators of antioxidant (Sirt1, Nrf2) and inflammatory response (NF-κB), resulting in the modulation of the downstream signaling, such as SIRT1/TORC1/CREB/BDNF-TrkB, Nrf2/ARE/HO-1, or other pathways. On the other hand, H2S appears to possess a direct detoxicative effect by binding endogenous (ROS, AGEs, Aβ) and exogenous (MeHg) neurotoxicants, thus reducing their toxicity. Moreover, the alteration of H2S metabolism through the inhibition of H2S-synthetizing enzymes in the brain (CBS, 3-MST) may be considered a significant mechanism of neurotoxicity. Taken together, the existing data indicate that the modulation of cerebral H2S metabolism may be used as a neuroprotective strategy to counteract neurotoxicity of a wide spectrum of endogenous and exogenous neurotoxicants associated with neurodegeneration (Alzheimer's and Parkinson's disease), fetal alcohol syndrome, hepatic encephalopathy, environmental neurotoxicant exposure, etc. In this particular case, modulation of H2S-synthetizing enzymes or the use of H2S-releasing drugs should be considered as the potential tools, although the particular efficiency and safety of such interventions are to be addressed in further studies.
Collapse
Affiliation(s)
- Michael Aschner
- Address correspondence to this author at the Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; E-mail
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
6
|
Fehsel K, Christl J. Comorbidity of osteoporosis and Alzheimer's disease: Is `AKT `-ing on cellular glucose uptake the missing link? Ageing Res Rev 2022; 76:101592. [PMID: 35192961 DOI: 10.1016/j.arr.2022.101592] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2023]
Abstract
Osteoporosis and Alzheimer's disease (AD) are both degenerative diseases. Osteoporosis often proceeds cognitive deficits, and multiple studies have revealed common triggers that lead to energy deficits in brain and bone. Risk factors for osteoporosis and AD, such as obesity, type 2 diabetes, aging, chemotherapy, vitamin deficiency, alcohol abuse, and apolipoprotein Eε4 and/or Il-6 gene variants, reduce cellular glucose uptake, and protective factors, such as estrogen, insulin, exercise, mammalian target of rapamycin inhibitors, hydrogen sulfide, and most phytochemicals, increase uptake. Glucose uptake is a fine-tuned process that depends on an abundance of glucose transporters (Gluts) on the cell surface. Gluts are stored in vesicles under the plasma membrane, and protective factors cause these vesicles to fuse with the membrane, resulting in presentation of Gluts on the cell surface. This translocation depends mainly on AKT kinase signaling and can be affected by a range of factors. Reduced AKT kinase signaling results in intracellular glucose deprivation, which causes endoplasmic reticulum stress and iron depletion, leading to activation of HIF-1α, the transcription factor necessary for higher Glut expression. The link between diseases and aging is a topic of growing interest. Here, we show that diseases that affect the same biochemical pathways tend to co-occur, which may explain why osteoporosis and/or diabetes are often associated with AD.
Collapse
|
7
|
Reduced Levels of H2S in Diabetes-Associated Osteoarthritis Are Linked to Hyperglycaemia, Nrf-2/HO-1 Signalling Downregulation and Chondrocyte Dysfunction. Antioxidants (Basel) 2022; 11:antiox11040628. [PMID: 35453313 PMCID: PMC9024787 DOI: 10.3390/antiox11040628] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/26/2022] Open
Abstract
Different findings indicate that type 2 diabetes is an independent risk factor for osteoarthritis (OA). However, the mechanisms underlying the connection between both diseases remain unclear. Changes in the balance of hydrogen sulphide (H2S) are thought to play an important role in the pathogenesis of diabetes and its complications, although its role is still controversial. In this study, we examined the modulation of H2S levels in serum and chondrocytes from OA diabetic (DB) and non-diabetic (non-DB) patients and in cells under glucose stress, in order to elucidate whether impairment in H2S-mediated signalling could participate in the onset of DB-related OA. Here, we identified a reduction in H2S synthesis in the cartilage from OA-DB patients and in cells under glucose stress, which is associated with hyperglycaemia-mediated dysregulation of chondrocyte metabolism. In addition, our results indicate that H2S is an inductor of the Nrf-2/HO-1 signalling pathway in cartilage, but is also a downstream target of Nrf-2 transcriptional activity. Thereby, impairment of the H2S/Nrf-2 axis under glucose stress or DB triggers chondrocyte catabolic responses, favouring the disruption of cartilage homeostasis that characterizes OA pathology. Finally, our findings highlight the benefits of the use of exogeneous sources of H2S in the treatment of DB-OA patients, and warrant future clinical studies.
Collapse
|
8
|
Regulatory Effects of Astragaloside IV on Hyperglycemia-Induced Mitophagy in Schwann Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7864308. [PMID: 35069769 PMCID: PMC8767404 DOI: 10.1155/2022/7864308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/04/2021] [Accepted: 12/16/2021] [Indexed: 01/17/2023]
Abstract
OBJECTIVE This study aimed to observe the regulatory effects of astragaloside IV (AS-IV) on hyperglycemia-induced mitochondrial damage and mitophagy in Schwann cells and to provide references for clinical trials on AS-IV in the treatment of diabetic peripheral neuropathy. METHODS Schwann cells were grown in a high-glucose medium to construct an autophagy model; the cells were then treated with AS-IV and N-acetylcysteine (control) to observe the regulatory effects of AS-IV on oxidative stress and mitophagy. RESULTS AS-IV exhibited antioxidant activity and inhibited the overactivation of autophagy in Schwann cells, significantly reducing the level of reactive oxygen species and downregulating the expression of autophagy-related proteins (LC3, PINK, and Parkin) under hyperglycemic conditions, thereby exerting a protective effect on mitochondrial morphology and membrane potential. CONCLUSION AS-IV can maintain the mitochondrial function of Schwann cells under hyperglycemic conditions by effectively alleviating oxidative stress and overactivation of mitophagy. The evidence from this study supports an AS-IV-based therapeutic strategy against diabetic peripheral neuropathy.
Collapse
|
9
|
Advanced glycation end-products as potential triggering factors of self-reactivity against myelin antigens in Multiple Sclerosis. Med Hypotheses 2021; 157:110702. [PMID: 34666261 DOI: 10.1016/j.mehy.2021.110702] [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: 05/19/2021] [Revised: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 11/20/2022]
Abstract
Multiple Sclerosis (MS) is a demyelinating autoimmune disease in which autoreactive T lymphocytes infiltrate the central nervous system (CNS) and react against antigens derived from proteins of the myelin sheath. The reason why T lymphocytes recognize certain myelin antigens as exogenous, activating the autoimmune response, remains unknown and represents the key to understand the pathogenesis of MS. Neurons are characterized by an elevated glycolytic metabolism. Methylglyoxal (MG) is a highly reactive α-oxoaldehyde spontaneously formed as a by-product of glycolysis, and it reacts with proteins, nucleotides and phospholipids forming stable adducts called advanced glycation end-products (AGEs). Several studies demonstrate that MG-derived AGEs accumulate in the plasma and brain of MS patients. Furthermore, there are evidences that post-myelinated oligodendrocytes, the myelin-forming glial cells, increase their glycolytic metabolism to maintain their survival and functions, likely explaining the progressive accumulation of MG in MS lesions. The hypothesis proposed here is that the MG-derived AGEs, accumulated on the proteins composing the myelin sheath, are responsible for the altered antigen presentation process, mimicking exogenous antigens and triggering the autoimmune response. If this hypothesis will be experimentally confirmed a new pathogenic mechanism of MS will be identified.
Collapse
|
10
|
Chen L, Chen Z, Xu Z, Feng W, Yang X, Qi Z. Polydatin protects Schwann cells from methylglyoxal induced cytotoxicity and promotes crushed sciatic nerves regeneration of diabetic rats. Phytother Res 2021; 35:4592-4604. [PMID: 34089208 DOI: 10.1002/ptr.7177] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/25/2021] [Accepted: 05/07/2021] [Indexed: 01/03/2023]
Abstract
Oxidative stress plays the main role in the pathogenesis of diabetes mellitus and peripheral neuropathy. Polydatin (PD) has been shown to exhibit strong antioxidative and antiinflammatory effects. At present, no research has focused on the possible effects of PD on Schwann cells and impaired peripheral nerves in diabetic models. Here, we used an in vitro Schwann cell damage model induced by methylglyoxal and an in vivo diabetic sciatic nerve crush model to study problems in such an area. In our experiment, we demonstrated that PD potently alleviated the decrease of cellular viability, prevented reactive oxygen species generation, and suppressed mitochondrial depolarization as well as cellular apoptosis in damaged Schwann cells. Moreover, we found that PD could upregulate Nrf2 and Glyoxalase 1 (GLO1) expression and inhibit Keap1 and receptor of AGEs (RAGE) expression of damaged Schwann cells. Finally, our in vivo experiment showed that PD could promote sciatic nerves repair of diabetic rats. Our results revealed that PD exhibited prominent neuroprotective effects on Schwann cells and sciatic nerves in diabetic models. The molecular mechanisms were associated with activating Nfr2 and GLO1 and inhibiting Keap1 and RAGE.
Collapse
Affiliation(s)
- Lulu Chen
- Department No.16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zixiang Chen
- Department No.16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuqiu Xu
- Department No.16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weifeng Feng
- Department No.16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaonan Yang
- Department No.16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zuoliang Qi
- Department No.16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Maternal vitamin D deficiency influences long-chain polyunsaturated fatty acids and pregnancy outcome in association with alterations in one-carbon metabolism. Nutr Res 2021; 86:37-49. [PMID: 33482597 DOI: 10.1016/j.nutres.2020.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 11/10/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023]
Abstract
Preeclampsia is a pregnancy-specific disorder, leading to maternal and infant morbidity and mortality. Abnormal placentation has been reported in preeclampsia. Nutrients like vitamin D and long-chain polyunsaturated fatty acids (LCPUFA) are known to play a role in placental development. In an animal model, we have previously demonstrated that maternal vitamin D deficiency increases the thromboxane/prostacyclin ratio and contributes to inflammation and vasoconstriction. We hypothesize that maternal vitamin D status influences placental LCPUFA metabolism through alterations in one carbon metabolism in women with preeclampsia. To test this hypothesis, we recruited 69 normotensive control (NC) women and 50 women with preeclampsia. Women with preeclampsia had lower placental protein and mRNA levels of cystathionine-β-synthase (CBS), higher plasma malondialdehyde (MDA) levels and higher levels of arachidonic acid (AA) and total omega-6 fatty acids in the placenta. Women with preeclampsia also demonstrated higher placental mRNA levels of cyclooxygenase-2 (COX-2) as compared to NC women. Maternal 25(OH)D levels were negatively associated with maternal plasma MDA levels. Placental vitamin D receptor (VDR) levels were positively associated with CBS while maternal MDA levels were positively associated with serum levels of thromboxane-B2 (TXB2) levels. Our findings indicate that vitamin D deficiency increases oxidative stress through alterations in one carbon metabolism to influence pro-inflammatory omega-6 metabolic pathway in the placenta. This study demonstrates a possible mechanism through which vitamin D deficiency can result in an imbalance in the LCPUFA metabolites and contribute to placental inflammation and endothelial dysfunction in preeclampsia.
Collapse
|
12
|
Yang CT, Devarie-Baez NO, Hamsath A, Fu XD, Xian M. S-Persulfidation: Chemistry, Chemical Biology, and Significance in Health and Disease. Antioxid Redox Signal 2020; 33:1092-1114. [PMID: 31547682 PMCID: PMC7583347 DOI: 10.1089/ars.2019.7889] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: S-Persulfidation generates persulfide adducts (RSSH) on both small molecules and proteins. This process is believed to be critical in the regulation of biological functions of reactive sulfur species such as H2S, as well as in signal transduction. S-Persulfidation also plays regulatory roles in human health and diseases. Recent Advances: Some mechanisms underlying the generation of low-molecular-weight persulfides and protein S-persulfidation in living organisms have been uncovered. Some methods for the specific delivery of persulfides and the detection of persulfides in biological systems have been developed. These advances help to pave the road to better understand the functions of S-persulfidation. Critical Issues: Persulfides are highly reactive and unstable. Currently, their identification relies on trapping them by S-alkylation, but this is not always reliable due to rapid sulfur exchange reactions. Therefore, the presence, identity, and fates of persulfides in biological environments are sometimes difficult to track. Future Directions: Further understanding the fundamental chemistry/biochemistry of persulfides and development of more reliable detection methods are needed. S-Persulfidation in specific protein targets is essential in organismal physiological health and human disease states. Besides cardiovascular and neuronal systems, the roles of persulfidation in other systems need to be further explored. Contradictory results of persulfidation in biology, especially in cancer, need to be clarified.
Collapse
Affiliation(s)
- Chun-Tao Yang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Protein Modification and Degradation Key Lab of Guangzhou and Guangdong, Key Laboratory of Molecular Clinical Pharmacology in School of Pharmaceutics Science, Guangzhou Medical University, Guangzhou, China.,Department of Chemistry, Washington State University, Pullman, Washington, USA
| | - Nelmi O Devarie-Baez
- Department of Chemistry, Washington State University-Tri Cities, Richland, Washington, USA
| | - Akil Hamsath
- Department of Chemistry, Washington State University, Pullman, Washington, USA
| | - Xiao-Dong Fu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Protein Modification and Degradation Key Lab of Guangzhou and Guangdong, Key Laboratory of Molecular Clinical Pharmacology in School of Pharmaceutics Science, Guangzhou Medical University, Guangzhou, China
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, Washington, USA
| |
Collapse
|
13
|
Karhu E, Zukerman R, Eshraghi RS, Mittal J, Deth RC, Castejon AM, Trivedi M, Mittal R, Eshraghi AA. Nutritional interventions for autism spectrum disorder. Nutr Rev 2020; 78:515-531. [PMID: 31876938 DOI: 10.1093/nutrit/nuz092] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Autism spectrum disorder (ASD) is an increasingly prevalent neurodevelopmental disorder with considerable clinical heterogeneity. With no cure for the disorder, treatments commonly center around speech and behavioral therapies to improve the characteristic social, behavioral, and communicative symptoms of ASD. Gastrointestinal disturbances are commonly encountered comorbidities that are thought to be not only another symptom of ASD but to also play an active role in modulating the expression of social and behavioral symptoms. Therefore, nutritional interventions are used by a majority of those with ASD both with and without clinical supervision to alleviate gastrointestinal and behavioral symptoms. Despite a considerable interest in dietary interventions, no consensus exists regarding optimal nutritional therapy. Thus, patients and physicians are left to choose from a myriad of dietary protocols. This review, summarizes the state of the current clinical and experimental literature on nutritional interventions for ASD, including gluten-free and casein-free, ketogenic, and specific carbohydrate diets, as well as probiotics, polyunsaturated fatty acids, and dietary supplements (vitamins A, C, B6, and B12; magnesium and folate).
Collapse
Affiliation(s)
- Elisa Karhu
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Ryan Zukerman
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Rebecca S Eshraghi
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Division of Gastroenterology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Jeenu Mittal
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Richard C Deth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Ana M Castejon
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Malav Trivedi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | | | | |
Collapse
|
14
|
Yan L, Jiaqiong L, Yue G, Xiaoyong L, Xuexian T, Ming L, Yinglan L, Xinxue L, Zena H. Atorvastatin protects against contrast-induced acute kidney injury via upregulation of endogenous hydrogen sulfide. Ren Fail 2020; 42:270-281. [PMID: 33685337 PMCID: PMC7144258 DOI: 10.1080/0886022x.2020.1740098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background Contrast-induced acute kidney injury (CIAKI) is the third leading cause of acute renal failure in hospitalized patients. This study was aimed to investigate whether atorvastatin could upregulate the expression of hydrogen sulfide (H2S) and hence protect against CIAKI. Methods We treated male rats and NRK-52E cells by iopromide to establish in vivo and in vitro models of CIAKI. Pretreatment with atorvastatin was given in CIAKI rats to investigate its effect on CIAKI. We collected serum and urine samples to detect renal function. We obtained kidney tissue for histological analysis and detection of protein concentration. We tested the serum concentration of H2S and renal expression of two H2S synthetases [cystathionine γ-lyase (CSE) and cystathionine-β synthase (CBS)]. NaHS was pretreated in NRK-52E cells to testify its underlying effect on contrast-induced injury. Results Atorvastatin significantly ameliorated renal dysfunction and morphological changes in CIAKI rats, as well as inflammation, apoptosis, and excessive oxidative stress. Atorvastatin also markedly increased the serum concentration of H2S and renal expression of CSE and CBS. Moreover, pretreatment with NaHS in NRK-52E cells considerably attenuated contrast-induced cell death and inflammation. Conclusion Atorvastatin protects against CIAKI via upregulation of endogenous hydrogen sulfide.
Collapse
Affiliation(s)
- Lin Yan
- Department of Nephrology, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lin Jiaqiong
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guo Yue
- Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Xiaoyong
- Department of General Surgery, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tan Xuexian
- Department of Pathology, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Long Ming
- Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Yinglan
- Department of Endocrine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Liao Xinxue
- Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huang Zena
- Department of General Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
15
|
Kim DH, Meza CA, Clarke H, Kim JS, Hickner RC. Vitamin D and Endothelial Function. Nutrients 2020; 12:E575. [PMID: 32098418 PMCID: PMC7071424 DOI: 10.3390/nu12020575] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/21/2022] Open
Abstract
Vitamin D is known to elicit a vasoprotective effect, while vitamin D deficiency is a risk factor for endothelial dysfunction (ED). ED is characterized by reduced bioavailability of a potent endothelium-dependent vasodilator, nitric oxide (NO), and is an early event in the development of atherosclerosis. In endothelial cells, vitamin D regulates NO synthesis by mediating the activity of the endothelial NO synthase (eNOS). Under pathogenic conditions, the oxidative stress caused by excessive production of reactive oxygen species (ROS) facilitates NO degradation and suppresses NO synthesis, consequently reducing NO bioavailability. Vitamin D, however, counteracts the activity of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase which produces ROS, and improves antioxidant capacity by enhancing the activity of antioxidative enzymes such as superoxide dismutase. In addition to ROS, proinflammatory mediators such as TNF-α and IL-6 are risk factors for ED, restraining NO and eNOS bioactivity and upregulating the expression of various atherosclerotic factors through the NF-κB pathway. These proinflammatory activities are inhibited by vitamin D by suppressing NF-κB signaling and production of proinflammatory cytokines. In this review, we discuss the diverse activities of vitamin D in regulating NO bioavailability and endothelial function.
Collapse
Affiliation(s)
- Do-Houn Kim
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
- Center for Advancing Exercise and Nutrition Research on Aging, Florida State University, Tallahassee, FL 32306, USA
| | - Cesar A. Meza
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
| | - Holly Clarke
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
| | - Jeong-Su Kim
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
- Center for Advancing Exercise and Nutrition Research on Aging, Florida State University, Tallahassee, FL 32306, USA
- Institute of Sports Sciences and Medicine, College of Human Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Robert C. Hickner
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA; (D.-H.K.); (C.A.M.); (H.C.); (J.-S.K.)
- Institute of Sports Sciences and Medicine, College of Human Sciences, Florida State University, Tallahassee, FL 32306, USA
- Department of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences, University of KwaZulu-Natal, Westville 4041, South Africa
| |
Collapse
|
16
|
Liu YP, Shao SJ, Guo HD. Schwann cells apoptosis is induced by high glucose in diabetic peripheral neuropathy. Life Sci 2020; 248:117459. [PMID: 32092332 DOI: 10.1016/j.lfs.2020.117459] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
Diabetic peripheral neuropathy (DPN) is a common complication of diabetes mellitus that affects approximately half of patients with diabetes. Current treatment regimens cannot treat DPN effectively. Schwann cells (SCs) are very sensitive to glucose concentration and insulin, and closely associated with the occurrence and development of type 1 diabetic mellitus (T1DM) and DPN. Apoptosis of SCs is induced by hyperglycemia and is involved in the pathogenesis of DPN. This review considers the pathological processes of SCs apoptosis under high glucose, which include the following: oxidative stress, inflammatory reactions, endoplasmic reticulum stress, autophagy, nitrification and signaling pathways (PI3K/AKT, ERK, PERK/Nrf2, and Wnt/β-catenin). The clarification of mechanisms underlying SCs apoptosis induced by high glucose will help us to understand and identify more effective strategies for the treatment of T1DM DPN.
Collapse
Affiliation(s)
- Yu-Pu Liu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shui-Jin Shao
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hai-Dong Guo
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
17
|
Xu S, Li J, Zhai M, Yao X, Liu H, Deng T, Cai H, Zhang W, Zhang W, Lou J, Peng L. 1,25-(OH) 2D 3 protects Schwann cells against advanced glycation end products-induced apoptosis through PKA-NF-κB pathway. Life Sci 2019; 225:107-116. [PMID: 30928409 DOI: 10.1016/j.lfs.2019.03.068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/21/2019] [Accepted: 03/27/2019] [Indexed: 11/28/2022]
Abstract
AIMS To explore the effect and mechanism of 1, 25-(OH)2D3 on Schwann cell apoptosis induced by advanced glycation end products. MAIN METHODS Schwann cells, isolated from rodent sciatic nerve were incubated with AGE-modified bovine serum albumin(AGE) to mimic diabetic conditions and 1,25-(OH)2D3 was used as protector. Cell apoptosis was detected by PI/Annexin-V staining, caspase 3 activity assay and western blotting for caspase 3 and PARP. The activation of protein kinase A (PKA) and nuclear factor kappa-B (NF-κB) was evaluated by western blot. Immunofluorescent staining was used for intercellular location of NF-κB. Cytokine secretion was evaluated by enzyme-linked immunosorbent assay. KEY FINDINGS Schwann cell apoptosis accelerated after incubating with AGE. However, if combining 1,25-(OH)2D3 with AGE, apoptosis decreased significantly. 1,25-(OH)2D3 enhanced PKA activity, but inhibited AGE-induced nuclear translocation of NF-κB. Furthermore, PKA activator (8-bromoadenoside cyclic adenoside monophosphate, 8-Br-cAMP) or NF-κB inhibitor (caffeic acid phenethyl ester, CAPE) could reduce the apoptosis, decreased cleaved caspase 3 and cleaved PARP, suggesting the involvement of PKA and NF-κB pathways in the protection of 1,25-(OH)2D3 on Schwann cells. Moreover, 8-Br-cAMP and CAPE could inhibit AGE-induced secretion of interleukin(IL)-1β, prostaglandin E2(PEG2) and cyclooxygenase 2(COX2). Interestingly, 8-Br-cAMP decreased phospho-NF-κB and inhibited nucleus translocation of NF-κB. It hinted at the regulation of PKA to NF-κB. Finally, a pre-treatment of H-89 (an inhibitor of PKA) could block the protection of 1,25-(OH)2D3 on cell apoptosis. In conclusion, 1,25-(OH)2D3 could protect Schwann cell against AGE-induced apoptosis through PKA/NF-κB pathway. SIGNIFICANCE These findings provide experimental rationales for using vitamin D for diabetic neuropathy.
Collapse
Affiliation(s)
- Shiqing Xu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jing Li
- Department of Stomatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Min Zhai
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiaoqi Yao
- Department of Ultrasonic Diagnosis, China-Japan Friendship Hospital, Beijing 100029, China
| | - Honglin Liu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Tingting Deng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Hanqing Cai
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wan Zhang
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wenjian Zhang
- Suzhou Rely Bio-Tech Co., Ltd., Suzhou 215103, China
| | - Jinning Lou
- Suzhou Rely Bio-Tech Co., Ltd., Suzhou 215103, China
| | - Liang Peng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
18
|
Yang CT, Chen L, Xu S, Day JJ, Li X, Xian M. Recent Development of Hydrogen Sulfide Releasing/Stimulating Reagents and Their Potential Applications in Cancer and Glycometabolic Disorders. Front Pharmacol 2017; 8:664. [PMID: 29018341 PMCID: PMC5623001 DOI: 10.3389/fphar.2017.00664] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/06/2017] [Indexed: 12/24/2022] Open
Abstract
As an important endogenous gaseous signaling molecule, hydrogen sulfide (H2S) exerts various effects in the body. A variety of pathological changes, such as cancer, glycometabolic disorders, and diabetes, are associated with altered endogenous levels of H2S, especially decreased. Therefore, the supplement of H2S is of great significance for the treatment of diseases containing the above pathological changes. At present, many efforts have been made to increase the in vivo levels of H2S by administration of gaseous H2S, simple inorganic sulfide salts, sophisticated synthetic slow-releasing controllable H2S donors or materials, and using H2S stimulating agents. In this article, we reviewed the recent development of H2S releasing/stimulating reagents and their potential applications in two common pathological processes including cancer and glycometabolic disorders.
Collapse
Affiliation(s)
- Chun-Tao Yang
- Affiliated Cancer Hospital and Institute, Key Laboratory of Protein Modification and Degradation in School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Chemistry, Washington State University, Pullman, WA, United States
| | - Li Chen
- Affiliated Cancer Hospital and Institute, Key Laboratory of Protein Modification and Degradation in School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shi Xu
- Department of Chemistry, Washington State University, Pullman, WA, United States
| | - Jacob J Day
- Department of Chemistry, Washington State University, Pullman, WA, United States
| | - Xiang Li
- Affiliated Cancer Hospital and Institute, Key Laboratory of Protein Modification and Degradation in School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, WA, United States
| |
Collapse
|
19
|
Yang CT, Lai ZZ, Zheng ZH, Kang JM, Xian M, Wang RY, Shi K, Meng FH, Li X, Chen L, Zhang H. A novel pH-controlled hydrogen sulfide donor protects gastric mucosa from aspirin-induced injury. J Cell Mol Med 2017; 21:2441-2451. [PMID: 28387464 PMCID: PMC5618702 DOI: 10.1111/jcmm.13166] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/16/2017] [Indexed: 12/19/2022] Open
Abstract
Hydrogen sulphide (H2S) serves as a vital gastric mucosal defence under acid condition. Non‐steroidal anti‐inflammatory drugs (NSAIDs) are among widely prescribed medications with effects of antipyresis, analgesia and anti‐inflammation. However, their inappropriate use causes gastric lesions and endogenous H2S deficiency. In this work, we reported the roles of a novel pH‐controlled H2S donor (JK‐1) in NSAID‐related gastric lesions. We found that JK‐1 could release H2S under mild acidic pH and increase solution pH value. Intragastrical administration of aspirin (ASP), one of NSAIDs, to mice elicited significant gastric lesions, evidenced by mucosal festering and bleeding. It also led to infiltration of inflammatory cells and resultant releases of IL‐6 and TNF‐α, as well as oxidative injury including myeloperoxidase (MPO) induction and GSH depletion. In addition, the ASP administration statistically inhibited H2S generation in gastric mucosa, while up‐regulated cyclooxygenase (COX)‐2 and cystathionine gamma lyase (CSE) expression. Importantly, these adverse effects of ASP were prevented by the intragastrical pre‐administration of JK‐1. However, JK‐1 alone did not markedly alter the property of mouse stomachs. Furthermore, in vitro cellular experiments showed the exposure of gastric mucosal epithelial (GES‐1) cells to HClO, imitating MPO‐driven oxidative injury, decreased cell viability, increased apoptotic rate and damaged mitochondrial membrane potential, which were reversed by pre‐treatment with JK‐1. In conclusion, JK‐1 was proved to be an acid‐sensitive H2S donor and could attenuate ASP‐related gastric lesions through reconstruction of endogenous gastric defence. This work indicates the possible treatment of adverse effects of NSAIDs with pH‐controlled H2S donors in the future.
Collapse
Affiliation(s)
- Chun-Tao Yang
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhen-Zhen Lai
- NanShan School of the First Clinical College, Guangzhou Medical University, Guangzhou, 510120, China
| | - Ze-Hang Zheng
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jian-Ming Kang
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - Rui-Yu Wang
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Kun Shi
- Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Fu-Hui Meng
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiang Li
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Li Chen
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Hui Zhang
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Quality Control Section of Academic Affairs, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
20
|
Garcia‐Gil M, Pierucci F, Vestri A, Meacci E. Crosstalk between sphingolipids and vitamin D3: potential role in the nervous system. Br J Pharmacol 2017; 174:605-627. [PMID: 28127747 PMCID: PMC6398521 DOI: 10.1111/bph.13726] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/16/2016] [Accepted: 01/18/2017] [Indexed: 12/14/2022] Open
Abstract
Sphingolipids are both structural and bioactive compounds. In particular, ceramide and sphingosine 1-phosphate regulate cell fate, inflammation and excitability. 1-α,25-dihydroxyvitamin D3 (1,25(OH)2 D3 ) is known to play an important physiological role in growth and differentiation in a variety of cell types, including neural cells, through genomic actions mediated by its specific receptor, and non-genomic effects that result in the activation of specific signalling pathways. 1,25(OH)2 D3 and sphingolipids, in particular sphingosine 1-phosphate, share many common effectors, including calcium regulation, growth factors and inflammatory cytokines, but it is still not known whether they can act synergistically. Alterations in the signalling and concentrations of sphingolipids and 1,25(OH)2 D3 have been found in neurodegenerative diseases and fingolimod, a structural analogue of sphingosine, has been approved for the treatment of multiple sclerosis. This review, after a brief description of the role of sphingolipids and 1,25(OH)2 D3 , will focus on the potential crosstalk between sphingolipids and 1,25(OH)2 D3 in neural cells.
Collapse
Affiliation(s)
- Mercedes Garcia‐Gil
- Department of BiologyUniversity of PisaPisaItaly
- Interdepartmental Research Center Nutrafood ‘Nutraceuticals and Food for Health’University of PisaPisaItaly
| | - Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, Molecular and Applied Biology Research UnitUniversity of FlorenceFlorenceItaly
- Interuniversitary Miology InstitutesItaly
| | - Ambra Vestri
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, Molecular and Applied Biology Research UnitUniversity of FlorenceFlorenceItaly
- Interuniversitary Miology InstitutesItaly
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, Molecular and Applied Biology Research UnitUniversity of FlorenceFlorenceItaly
- Interuniversitary Miology InstitutesItaly
| |
Collapse
|
21
|
Matafome P, Rodrigues T, Sena C, Seiça R. Methylglyoxal in Metabolic Disorders: Facts, Myths, and Promises. Med Res Rev 2017; 37:368-403. [PMID: 27636890 DOI: 10.1002/med.21410] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/07/2016] [Accepted: 08/12/2016] [Indexed: 08/26/2024]
Abstract
Glucose and fructose metabolism originates the highly reactive byproduct methylglyoxal (MG), which is a strong precursor of advanced glycation end products (AGE). The MG has been implicated in classical diabetic complications such as retinopathy, nephropathy, and neuropathy, but has also been recently associated with cardiovascular diseases and central nervous system disorders such as cerebrovascular diseases and dementia. Recent studies even suggested its involvement in insulin resistance and beta-cell dysfunction, contributing to the early development of type 2 diabetes and creating a vicious circle between glycation and hyperglycemia. Despite several drugs and natural compounds have been identified in the last years in order to scavenge MG and inhibit AGE formation, we are still far from having an effective strategy to prevent MG-induced mechanisms. This review summarizes the endogenous and exogenous sources of MG, also addressing the current controversy about the importance of exogenous MG sources. The mechanisms by which MG changes cell behavior and its involvement in type 2 diabetes development and complications and the pathophysiological implication are also summarized. Particular emphasis will be given to pathophysiological relevance of studies using higher MG doses, which may have produced biased results. Finally, we also overview the current knowledge about detoxification strategies, including modulation of endogenous enzymatic systems and exogenous compounds able to inhibit MG effects on biological systems.
Collapse
Affiliation(s)
- Paulo Matafome
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
- Department of Complementary Sciences, Coimbra Health School (ESTeSC), Instituto Politécnico de Coimbra, 3045-601, Coimbra, Portugal
| | - Tiago Rodrigues
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Cristina Sena
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Raquel Seiça
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| |
Collapse
|
22
|
Ding T, Chen W, Li J, Ding J, Mei X, Hu H. High Glucose Induces Mouse Mesangial Cell Overproliferation via Inhibition of Hydrogen Sulfide Synthesis in a TLR-4-Dependent Manner. Cell Physiol Biochem 2017; 41:1035-1043. [DOI: 10.1159/000461483] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 12/19/2016] [Indexed: 11/19/2022] Open
Abstract
Background/Aims: Overproliferation of mesangial cells was believed to play an important role in the progress of diabetic nephropathy, one of the primary complications of diabetes. Hydrogen sulfide (H2S), a well-known and pungent gas with the distinctive smell of rotten eggs, was discovered to play a protective role in diabetic nephropathy. Methods: MTT assay was used to examine the viability of mesangial cells. Small interfering RNA was used to knock down the expression of TLR4 while specific inhibitor LY294002 to suppress the function of PI3K. H2S generation rate was determined by a H2S micro-respiration sensor. Results: Glucose of 25mM induced significant mesangial cells proliferation, which was accomplished by significantly inhibited endogenous H2S synthesis. And exogenous H2S treatment by NaHS markedly mitigated the overproliferation of mouse mesangial cells. Furthermore, it was found that H2S deficiency could result in TLR4 activation. And H2S supplementation remarkably inhibited TLR4 expression and curbed the mesangial cell overproliferation. Besides, PI3K/Akt pathway inhibition also significantly ameliorated the cell overproliferation. Conclusion: High glucose (HG) induces mouse mesangial cell overproliferation via inhibition of hydrogen sulfide synthesis in a TLR-4-dependent manner. And PI3K/Akt pathway might also play a vital part in the HG-induced mesangial cell overproliferation.
Collapse
|
23
|
Yang CT, Meng FH, Chen L, Li X, Cen LJ, Wen YH, Li CC, Zhang H. Inhibition of Methylglyoxal-Induced AGEs/RAGE Expression Contributes to Dermal Protection by N-Acetyl-L-Cysteine. Cell Physiol Biochem 2017; 41:742-754. [PMID: 28214842 DOI: 10.1159/000458734] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND/AIM Accumulation of advanced glycation end products (AGEs) is a major cause of diabetes mellitus (DM) skin complications. Methylglyoxal (MGO), a reactive dicarbonyl compound, is a crucial intermediate of AGEs generation. N-acetyl-L-cysteine (NAC), an active ingredient of some medicines, can induce endogenous GSH and hydrogen sulfide generation, and set off a condensation reaction with MGO. However, there is rare evidence to show NAC can alleviate DM-induced skin injury through inhibition of AGEs generation or toxicity. The present study aimed to observe the effects of NAC on MGO-induced inflammatory injury and investigate the roles of AGEs and its receptor (RAGE) in NAC's dermal protection in human HaCaT keratinocytes. METHODS The cells were exposed to MGO to simulate a high MGO status in diabetic blood or tissues. The content of AGEs in serum or cell medium was measured with ELISA. The protective effects of NAC against MGO-induce injury were evaluated by administration before MGO one hour, in virtue of cell viability, mitochondrial membrane potential, inflammation reaction, nuclear factor (NF)-κB activation, matrix metalloproteinase (MMP)-9 expression, as well as cellular behavioral function. RESULTS We found the AGEs levels of patients with DM were elevated comparing with healthy volunteers. The in vitro AGEs generation was also able to be enhanced by the exposure of HaCaT cells to MGO, which reduced dose-dependently cellular viability, damaged mitochondrial function, triggered secretion of interleukin (IL)-6 and IL-8, activated NF-κB and upregulated MMP-9 expression. Furthermore, the exposure caused cellular adhesion and migration dysfunction, as well as collagen type I inhibition. Importantly, before the exposure to MGO, the preconditioning with NAC significantly attenuated MGO-induced AGEs generation, improved cellular viability and mitochondrial function, partially reversed the overexpression of proinflammatory factors and MMP-9, as well as the activation of NF-κB. Lastly, NAC blocked MGO-induced RAGE upregulation, and inhibition of RAGE with its neutralizing antibody significantly alleviated MGO-induced NF-κB activation, MMP-9 upregulation and inflammatory injury in HaCaT cells. CONCLUSION The present work indicates the administration of NAC can prevent MGO-induced dermal inflammatory injury through inhibition of AGEs/RAGE signal, which may provide a basal support for the treatment of diabetic skin complications with NAC-containing medicines in the future.
Collapse
Affiliation(s)
- Chun-Tao Yang
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China,Affiliated Cancer Hospital & Institute, Guangzhou, China
| | - Fu-Hui Meng
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China
| | - Li Chen
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China
| | - Xiang Li
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China
| | - Lai-Jian Cen
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China
| | - Yu-Hua Wen
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China
| | - Cai-Chen Li
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China
| | - Hui Zhang
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China,Affiliated Cancer Hospital & Institute, Guangzhou, China,Quality Control Section of Academic Affairs, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|