1
|
Hazer Rosberg DB, Mahlapuu M, Perez R, Dahlin LB. Lactoferrin-derived peptide PXL01 impacts nerve regeneration after sciatic nerve reconstruction in healthy and diabetic rats. Front Cell Dev Biol 2025; 13:1565285. [PMID: 40260416 PMCID: PMC12009942 DOI: 10.3389/fcell.2025.1565285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Introduction Although advanced surgical techniques are available, satisfactory functional outcomes after peripheral nerve injuries are uncommon. Hence, immune-modulating factors such as PXL01, a lactoferrin-derived peptide that improves axonal outgrowth in injured human digital nerves, have gained attention. We previously reported a short-term immunosuppressive effect of PXL01 after the repair of transected rat sciatic nerves, but it had no effect on nerve regeneration. Here, we investigated the potential of PXL01 to improve nerve regeneration in healthy rats and in a rat model of type 2 diabetes (Goto-Kakizaki [GK] rats). Methods A 10-mm sciatic nerve defect was created in healthy (n = 14) and diabetic GK rats (n = 14) and reconstructed using nerve autografts. Immediately after surgery, PXL01 or sodium chloride (control, placebo) (n = 7 for each treatment) was administered around the autograft. On day 8, immunohistochemical staining of the sciatic nerve and dorsal root ganglia (DRGs) was performed to analyze axonal outgrowth (neurofilament staining); inflammation (CD68 and CD206 macrophage staining in nerve); Schwann cell and sensory neuron activation (transcription factor ATF3 staining in nerve and DRGs) and apoptosis (cleaved caspase 3 staining in nerve); and neuroprotection (heat shock protein [HSP27] staining in nerve and DRGs). Results PXL01 had no impact on the macrophage response in the autografts but increased axonal outgrowth and HSP27 expression in the DRGs of healthy and diabetic rats, despite a lower number of activated Schwann cells in the autograft. Diabetes affected axonal outgrowth, Schwann cell and macrophage responses, and HSP27 expression. These effects were observed in the sciatic nerve as well as the DRG. Discussion Application of PXL01, despite having no impact on macrophages, may improve axonal outgrowth and affects Schwann cell activation in autograft-reconstructed sciatic nerves, as well as conveys neuroprotection (HSP27 expression) in the DRGs of healthy and diabetic GK rats. Diabetes influenced nerve regeneration in such autografts. Therefore, PXL01 is a promising candidate to improve nerve regeneration.
Collapse
Affiliation(s)
- Derya Burcu Hazer Rosberg
- Department of Translational Medicine – Hand Surgery, Lund University, Malmö, Sweden
- Department of Hand Surgery, Skåne University Hospital, Malmö, Sweden
| | - Margit Mahlapuu
- Department of Chemistry and Molecular Biology, Göteborg University, Göteborg, Sweden
| | - Raquel Perez
- Department of Translational Medicine – Hand Surgery, Lund University, Malmö, Sweden
- Unit for Social Epidemiology, Department of Clinical Sciences (Malmö), Lund University, Malmö, Sweden
| | - Lars B. Dahlin
- Department of Translational Medicine – Hand Surgery, Lund University, Malmö, Sweden
- Department of Hand Surgery, Skåne University Hospital, Malmö, Sweden
- Department of Biomedical & Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
2
|
Cross K, Vetter SW, Alam Y, Hasan MZ, Nath AD, Leclerc E. Role of the Receptor for Advanced Glycation End Products (RAGE) and Its Ligands in Inflammatory Responses. Biomolecules 2024; 14:1550. [PMID: 39766257 PMCID: PMC11673996 DOI: 10.3390/biom14121550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Since its discovery in 1992, the receptor for advanced glycation end products (RAGE) has emerged as a key receptor in many pathological conditions, especially in inflammatory conditions. RAGE is expressed by most, if not all, immune cells and can be activated by many ligands. One characteristic of RAGE is that its ligands are structurally very diverse and belong to different classes of molecules, making RAGE a promiscuous receptor. Many of RAGE ligands are damaged associated molecular patterns (DAMPs) that are released by cells under inflammatory conditions. Although RAGE has been at the center of a lot of research in the past three decades, a clear understanding of the mechanisms of RAGE activation by its ligands is still missing. In this review, we summarize the current knowledge of the role of RAGE and its ligands in inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (K.C.); (S.W.V.); (Y.A.); (M.Z.H.); (A.D.N.)
| |
Collapse
|
3
|
Zglejc-Waszak K, Jozwik M, Thoene M, Wojtkiewicz J. Role of Receptor for Advanced Glycation End-Products in Endometrial Cancer: A Review. Cancers (Basel) 2024; 16:3192. [PMID: 39335163 PMCID: PMC11430655 DOI: 10.3390/cancers16183192] [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: 08/12/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Endometrial cancer (EC) is the most common gynecological malignancy. EC is associated with metabolic disorders that may promote non-enzymatic glycation and activate the receptor for advanced glycation end-products (RAGE) signaling pathways. Thus, we assumed that RAGE and its ligands may contribute to EC. Of particular interest is the interaction between diaphanous-related formin 1 (Diaph1) and RAGE during the progression of human cancers. Diaph1 is engaged in the proper organization of actin cytoskeletal dynamics, which is crucial in cancer invasion, metastasis, angiogenesis, and axonogenesis. However, the detailed molecular role of RAGE in EC remains uncertain. In this review, we discuss epigenetic factors that may play a key role in the RAGE-dependent endometrial pathology. We propose that DNA methylation may regulate the activity of the RAGE pathway in the uterus. The accumulation of negative external factors, such as hyperglycemia, inflammation, and oxidative stress, may interfere with the DNA methylation process. Therefore, further research should take into account the role of epigenetic mechanisms in EC progression.
Collapse
Affiliation(s)
- Kamila Zglejc-Waszak
- Department of Anatomy, Faculty of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Marcin Jozwik
- Department of Gynecology and Obstetrics, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-045 Olsztyn, Poland
| | - Michael Thoene
- Department of Medical Biology, Faculty of Health Sciences, University of Warmia and Mazury in Olsztyn, Żołnierska 14C Str., 10-561 Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| |
Collapse
|
4
|
Fu M, Zhengran L, Yingli L, Tong W, Liyang C, Xi G, Xiongyi Y, Mingzhe C, Guoguo Y. The contribution of adiponectin to diabetic retinopathy progression: Association with the AGEs-RAGE pathway. Heliyon 2024; 10:e36111. [PMID: 39296166 PMCID: PMC11409038 DOI: 10.1016/j.heliyon.2024.e36111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/09/2024] [Accepted: 08/09/2024] [Indexed: 09/21/2024] Open
Abstract
Diabetic retinopathy (DR) is a chronic complication of diabetes. Given that adiponectin plays a key role in DR progression, this study aims to elucidate the molecular mechanisms of sDR progression related to adiponectin. First, we extracted the microarray dataset GSE60436 from the Gene Expression Omnibus (GEO) database to identify hub genes associated with DR. Pathway enrichment analysis revealed a focus on inflammation, oxidative stress, and metabolic disease pathways. Gene Set Enrichment Analysis (GSEA) identified nine significant pathways related to DR. Immune infiltration analysis indicated increased infiltration of fibroblasts and endothelial cells in DR patients. Second, at the gene level, single-cell RNA sequencing (scRNA-seq) results showed a decrease in ADIPOQ gene expression as the disease progressed in our mouse models. At the protein level, ELISA results from sera of 31 patients and 11 control subjects demonstrated significantly lower adiponectin expression in the proliferative diabetic retinopathy (PDR) group compared to controls. Our findings reveal that adiponectin is involved in the advanced glycation end products (AGEs) and receptor of advanced glycation end products (RAGE) axis, as evidenced by hub gene analysis, scRNA-seq, and ELISA. In conclusion, adiponectin acts as a central molecule in the AGEs-RAGE axis, regulated by ADIPOQ, to influence DR progression.
Collapse
Affiliation(s)
- Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Li Zhengran
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- The Second Clinical School, Southern Medical University, Guangzhou, China
| | - Li Yingli
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wu Tong
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- The First Clinical School, Southern Medical University, Guangzhou, China
| | - Cai Liyang
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guo Xi
- School of Psychological and Cognitive Sciences and Beijing Key Laboratory of Behavior and Mental Health, Peking University, Beijing, China
| | - Yang Xiongyi
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- The Second Clinical School, Southern Medical University, Guangzhou, China
| | - Cao Mingzhe
- Department of Ophthalmology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong Province, China
| | - Yi Guoguo
- Department of Ophthalmology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Mizukami H. Pathological evaluation of the pathogenesis of diabetes mellitus and diabetic peripheral neuropathy. Pathol Int 2024; 74:438-453. [PMID: 38888200 PMCID: PMC11551828 DOI: 10.1111/pin.13458] [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: 04/03/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024]
Abstract
Currently, there are more than 10 million patients with diabetes mellitus in Japan. Therefore, the need to explore the pathogenesis of diabetes and the complications leading to its cure is becoming increasingly urgent. Pathological examination of pancreatic tissues from patients with type 2 diabetes reveals a decrease in the volume of beta cells because of a combination of various stresses. In human type 2 diabetes, islet amyloid deposition is a unique pathological change characterized by proinflammatory macrophage (M1) infiltration into the islets. The pathological changes in the pancreas with islet amyloid were different according to clinical factors, which suggests that type 2 diabetes can be further subclassified based on islet pathology. On the other hand, diabetic peripheral neuropathy is the most frequent diabetic complication. In early diabetic peripheral neuropathy, M1 infiltration in the sciatic nerve evokes oxidative stress or attenuates retrograde axonal transport, as clearly demonstrated by in vitro live imaging. Furthermore, islet parasympathetic nerve density and beta cell volume were inversely correlated in type 2 diabetic Goto-Kakizaki rats, suggesting that diabetic peripheral neuropathy itself may contribute to the decrease in beta cell volume. These findings suggest that the pathogenesis of diabetes mellitus and diabetic peripheral neuropathy may be interrelated.
Collapse
Affiliation(s)
- Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiAomoriJapan
| |
Collapse
|
6
|
Chong ZZ, Menkes DL, Souayah N. Targeting neuroinflammation in distal symmetrical polyneuropathy in diabetes. Drug Discov Today 2024; 29:104087. [PMID: 38969091 DOI: 10.1016/j.drudis.2024.104087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Diabetic distal symmetric polyneuropathy is the most common type of peripheral neuropathy complication of diabetes mellitus. Neuroinflammation is emerging as an important contributor to diabetes-induced neuropathy. Long-term hyperglycemia results in increased production of advanced glycation end products (AGEs). AGEs interact with their receptors to activate intracellular signaling, leading to the release of various inflammatory cytokines. Increased release of inflammatory cytokines is associated with diabetes, diabetic neuropathy, and neuropathic pain. Thus, anti-inflammatory intervention is a potential therapy for diabetic distal symmetric polyneuropathy. Further characterization of inflammatory mechanisms might identify novel therapeutic targets to mitigate diabetic neuropathy.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Department of Neurology, Rutgers University, New Jersey Medical School, Newark, NJ 07103, USA.
| | - Daniel L Menkes
- Department of Neurology, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Nizar Souayah
- Department of Neurology, Rutgers University, New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
7
|
Duval C, Criscuolo F, Bertile F. Glycation resistance and life-history traits: lessons from non-conventional animal models. Biol Lett 2024; 20:20230601. [PMID: 38863347 DOI: 10.1098/rsbl.2023.0601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/12/2024] [Indexed: 06/13/2024] Open
Abstract
Glycation reactions play a key role in the senescence process and are involved in numerous age-related pathologies, such as diabetes complications or Alzheimer's disease. As a result, past studies on glycation have mostly focused on human and laboratory animal models for medical purposes. Very little is known about glycation and its link to senescence in wild animal species. Yet, despite feeding on high-sugar diets, several bat and bird species are long-lived and seem to escape the toxic effects of high glycaemia. The study of these models could open new avenues both for understanding the mechanisms that coevolved with glycation resistance and for treating the damaging effects of glycations in humans. Our understanding of glycaemia's correlation to proxies of animals' pace of life is emerging in few wild species; however, virtually nothing is known about their resistance to glycation, nor on the relationship between glycation, species' life-history traits and individual fitness. Our review summarizes the scarce current knowledge on the links between glycation and life-history traits in non-conventional animal models, highlighting the predominance of avian research. We also investigate some key molecular and physiological parameters involved in glycation regulation, which hold promise for future research on fitness and senescence of individuals.
Collapse
Affiliation(s)
- Cyrielle Duval
- University of Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien, UMR 7178 , Strasbourg 67000, France
- Infrastructure de Protéomique, ProFi , Strasbourg FR2048, France
| | - François Criscuolo
- University of Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien, UMR 7178 , Strasbourg 67000, France
| | - Fabrice Bertile
- University of Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien, UMR 7178 , Strasbourg 67000, France
- Infrastructure de Protéomique, ProFi , Strasbourg FR2048, France
| |
Collapse
|
8
|
Yorek M. Combination therapy is it in the future for successfully treating peripheral diabetic neuropathy? Front Endocrinol (Lausanne) 2024; 15:1357859. [PMID: 38812811 PMCID: PMC11133577 DOI: 10.3389/fendo.2024.1357859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/16/2024] [Indexed: 05/31/2024] Open
Abstract
In 2022, the Center for Disease Control and Prevention reported that 11.3% of the United States population, 37.3 million people, had diabetes and 38% of the population had prediabetes. A large American study conducted in 2021 and supported by many other studies, concluded that about 47% of diabetes patients have peripheral neuropathy and that diabetic neuropathy was present in 7.5% of patients at the time of diabetes diagnosis. In subjects deemed to be pre-diabetes and impaired glucose tolerance there was a wide range of prevalence estimates (interquartile range (IQR): 6%-34%), but most studies (72%) reported a prevalence of peripheral neuropathy ≥10%. There is no recognized treatment for diabetic peripheral neuropathy (DPN) other than good blood glucose control. Good glycemic control slows progression of DPN in patients with type 1 diabetes but for patients with type 2 diabetes it is less effective. With obesity and type 2 diabetes at epidemic levels the need of a treatment for DPN could not be more important. In this article I will first present background information on the "primary" mechanisms shown from pre-clinical studies to contribute to DPN and then discuss mono- and combination therapies that have demonstrated efficacy in animal studies and may have success when translated to human subjects. I like to compare the challenge of finding an effective treatment for DPN to the ongoing work being done to treat hypertension. Combination therapy is the recognized approach used to normalize blood pressure often requiring two, three or more drugs in addition to lifestyle modification to achieve the desired outcome. Hypertension, like DPN, is a progressive disease caused by multiple mechanisms. Therefore, it seems likely as well as logical that combination therapy combined with lifestyle adjustments will be required to successfully treat DPN.
Collapse
Affiliation(s)
- Mark Yorek
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
- Department of Veterans Affairs Iowa City Health Care System, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
9
|
Jarosławska J, Kordas B, Miłowski T, Juranek JK. Mammalian Diaphanous1 signalling in neurovascular complications of diabetes. Eur J Neurosci 2024; 59:2628-2645. [PMID: 38491850 DOI: 10.1111/ejn.16310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/18/2024] [Indexed: 03/18/2024]
Abstract
Over the past few decades, diabetes gradually has become one of the top non-communicable disorders, affecting 476.0 million in 2017 and is predicted to reach 570.9 million people in 2025. It is estimated that 70 to 100% of all diabetic patients will develop some if not all, diabetic complications over the course of the disease. Despite different symptoms, mechanisms underlying the development of diabetic complications are similar, likely stemming from deficits in both neuronal and vascular components supplying hyperglycaemia-susceptible tissues and organs. Diaph1, protein diaphanous homolog 1, although mainly known for its regulatory role in structural modification of actin and related cytoskeleton proteins, in recent years attracted research attention as a cytoplasmic partner of the receptor of advanced glycation end-products (RAGE) a signal transduction receptor, whose activation triggers an increase in proinflammatory molecules, oxidative stressors and cytokines in diabetes and its related complications. Both Diaph1 and RAGE are also a part of the RhoA signalling cascade, playing a significant role in the development of neurovascular disturbances underlying diabetes-related complications. In this review, based on the existing knowledge as well as compelling findings from our past and present studies, we address the role of Diaph1 signalling in metabolic stress and neurovascular degeneration in diabetic complications. In light of the most recent developments in biochemical, genomic and transcriptomic research, we describe current theories on the aetiology of diabetes complications, highlighting the function of the Diaph1 signalling system and its role in diabetes pathophysiology.
Collapse
Affiliation(s)
- Julia Jarosławska
- Department of Biological Functions of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Bernard Kordas
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Tadeusz Miłowski
- Department of Emergency Medicine, School of Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Judyta K Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
10
|
Liu Z, Yang W, Chen J, Wang Q. Circulating HMGB1 in acute ischemic stroke and its association with post-stroke cognitive impairment. PeerJ 2024; 12:e17309. [PMID: 38708343 PMCID: PMC11067911 DOI: 10.7717/peerj.17309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
Background Ischemic stroke frequently leads to a condition known as post-stroke cognitive impairment (PSCI). Timely recognition of individuals susceptible to developing PSCI could facilitate the implementation of personalized strategies to mitigate cognitive deterioration. High mobility group box 1 (HMGB1) is a protein released by ischemic neurons and implicated in inflammation after stroke. Circulating levels of HMGB1 could potentially serve as a prognostic indicator for the onset of cognitive impairment following ischemic stroke. Objective To investigate the predictive value of circulating HMGB1 concentrations in the acute phase of ischemic stroke for the development of cognitive dysfunction at the 3-month follow-up. Methods A total of 192 individuals experiencing their initial episode of acute cerebral infarction were prospectively recruited for this longitudinal investigation. Concentrations of circulating HMGB1 were quantified using an enzyme-linked immunosorbent assay (ELISA) technique within the first 24 hours following hospital admission. Patients underwent neurological evaluation including NIHSS scoring. Neuropsychological evaluation was conducted at the 3-month follow-up after the cerebrovascular event, employing the Montreal Cognitive Assessment (MoCA) as the primary tool for assessing cognitive performance. Multivariable logistic regression models were employed to investigate the relationship between circulating HMGB1 concentrations and cognitive dysfunction following stroke, which was operationalized as a MoCA score below 26, while controlling for potential confounders including demographic characteristics, stroke severity, vascular risk factors, and laboratory parameters. Results Of 192 patients, 84 (44%) developed PSCI. Circulating HMGB1 concentrations were significantly elevated in individuals who developed cognitive dysfunction following stroke compared to those who maintained cognitive integrity (8.4 ± 1.2 ng/mL vs 4.6 ± 0.5 ng/mL, respectively; p < 0.001). The prevalence of PSCI showed a dose-dependent increase with higher HMGB1 quartiles. After controlling for potential confounders such as demographic factors (age, gender, and education), stroke severity, vascular risk factors, and laboratory parameters in a multivariable logistic regression model, circulating HMGB1 concentrations emerged as a significant independent predictor of cognitive dysfunction following stroke (regression coefficient = 0.236, p < 0.001). Conclusion Circulating HMGB1 concentrations quantified within the first 24 hours following acute cerebral infarction are significantly and independently correlated with the likelihood of developing cognitive dysfunction at the 3-month follow-up, even after accounting for potential confounding factors. HMGB1 may be a novel biomarker to identify patients likely to develop post-stroke cognitive impairment for targeted preventive interventions.
Collapse
Affiliation(s)
- Zhenbao Liu
- Department of Neurology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weixia Yang
- Department of Neurology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianxin Chen
- Department of Critical Care Medicine, Jinan First People’s Hospital, Shandong Traditional Chinese Medicine University, Jinan, Shandong, China
| | - Qian Wang
- Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| |
Collapse
|
11
|
Aktas G. Association between the Prognostic Nutritional Index and Chronic Microvascular Complications in Patients with Type 2 Diabetes Mellitus. J Clin Med 2023; 12:5952. [PMID: 37762893 PMCID: PMC10531521 DOI: 10.3390/jcm12185952] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
The prognostic nutritional index (PNI) is associated with inflammatory conditions. Since type 2 diabetes mellitus (T2DM) and its microvascular complications produce a significant inflammatory burden, we aimed to compare the PNI levels of the subjects with T2DM to those of healthy individuals. Furthermore, we aimed to compare the PNI levels of the diabetic subjects, with and without microvascular complications. The study cohort consisted of T2DM patients and healthy volunteers. The general characteristics, laboratory data, and PNI of the T2DM and control groups were compared. We further compared the PNI levels of the diabetic patients, with and without diabetic microvascular complications. The PNI levels of the T2DM patients and the control group were 51.6 (30.1-73.8)% and 64.8 (49.4-76)%, respectively (p < 0.001). Subgroup analyses revealed that the PNI was lower in the diabetic subjects with diabetic microvascular complications than in the diabetic patients without microvascular complications (p < 0.001), in patients with diabetic nephropathy compared to those without nephropathy (p < 0.001), in patients with diabetic retinopathy compared to those without retinopathy (p < 0.001), and in patients with diabetic neuropathy compared to those without neuropathy (p < 0.001). In conclusion, we assert that assessing the PNI may yield additional diagnostic value in regards to the timely determination of diabetic microvascular complications.
Collapse
Affiliation(s)
- Gulali Aktas
- Department of Internal Medicine, Abant Izzet Baysal University Hospital, 14280 Bolu, Turkey
| |
Collapse
|
12
|
Zglejc-Waszak K, Mukherjee K, Korytko A, Lewczuk B, Pomianowski A, Wojtkiewicz J, Banach M, Załęcki M, Nowicka N, Jarosławska J, Kordas B, Wąsowicz K, Juranek JK. Novel insights into the nervous system affected by prolonged hyperglycemia. J Mol Med (Berl) 2023; 101:1015-1028. [PMID: 37462767 PMCID: PMC10400689 DOI: 10.1007/s00109-023-02347-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 06/15/2023] [Accepted: 06/28/2023] [Indexed: 08/05/2023]
Abstract
Multiple molecular pathways including the receptor for advanced glycation end-products-diaphanous related formin 1 (RAGE-Diaph1) signaling are known to play a role in diabetic peripheral neuropathy (DPN). Evidence suggests that neuropathological alterations in type 1 diabetic spinal cord may occur at the same time as or following peripheral nerve abnormalities. We demonstrated that DPN was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. More than 500 differentially expressed genes (DEGs) belonging to multiple functional pathways were identified in diabetic spinal cord and of those the most enriched was RAGE-Diaph1 related PI3K-Akt pathway. Only seven of spinal cord DEGs overlapped with DEGs from type 1 diabetic sciatic nerve and only a single gene cathepsin E (CTSE) was common for both type 1 and type 2 diabetic mice. In silico analysis suggests that molecular changes in spinal cord may act synergistically with RAGE-Diaph1 signaling axis in the peripheral nerve. KEY MESSAGES: Molecular perturbations in spinal cord may be involved in the progression of diabetic peripheral neuropathy. Diabetic peripheral neuropathy was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. In silico analysis revealed that PI3K-Akt signaling axis related to RAGE-Diaph1 was the most enriched biological pathway in diabetic spinal cord. Cathepsin E may be the target molecular hub for intervention against diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland.
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Virginia Tech, VA, 24016, USA
| | - Agnieszka Korytko
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland
| | - Bogdan Lewczuk
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Poland
| | - Andrzej Pomianowski
- Internal Medicine Department, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland
| | - Marta Banach
- Department of Neurology, Collegium Medicum, Jagiellonian University, 31-008, Krakow, Poland
| | - Michał Załęcki
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Poland
| | - Natalia Nowicka
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland
| | - Julia Jarosławska
- Department of Biological Functions of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748, Olsztyn, Poland
| | - Bernard Kordas
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland
| | - Krzysztof Wąsowicz
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719, Olsztyn, Poland
| | - Judyta K Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-085, Olsztyn, Poland.
| |
Collapse
|
13
|
Zglejc-Waszak K, Schmidt AM, Juranek JK. The receptor for advanced glycation end products and its ligands' expression in OVE26 diabetic sciatic nerve during the development of length-dependent neuropathy. Neuropathology 2023; 43:84-94. [PMID: 35915909 DOI: 10.1111/neup.12852] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/15/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023]
Abstract
Type 1 diabetes (T1D) may affect the peripheral nervous system and alter the expression of proteins contributing to inflammation and cellular cytoskeleton dysfunction, in most cases leading to the development of diabetic length-dependent neuropathy (DLDN). In the present study, we performed immunohistochemistry (IHC) to probe the expression of the receptor for advanced glycation end products (RAGE); its key ligands, high-mobility group box 1 (HMGB1), S100 calcium-binding protein B (S100B), and carboxymethyl-lysine (CML - advanced glycation end products (AGE)); and its cytoplasmic tail-binding partner, diaphanous related formin 1 (DIAPH1) and associated molecules, beta-actin (ACTB) and profilin 1 (PFN1) proteins in sciatic nerves harvested from seven-month old FVB/OVE26 mice with genetically-mediated T1D. We found that the amount of RAGE, HMGB1, and S100B proteins was elevated in diabetic vs the non-diabetic groups, while the amount of DIAPH1, ACTB, as well as PFN1 proteins did not differ between these groups. Moreover, our data revealed linear dependence between RAGE and HMGB1 proteins. Interaction criss-cross of selected sets of proteins in the sciatic nerve revealed that there were connected in a singular network. Our results indicate that T1D may alter expression patterns of RAGE axis proteins and thus contribute to DLDN.
Collapse
Affiliation(s)
- Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, University of Warmia and Mazury in Olsztyn, School of Medicine, Collegium Medicum, Olsztyn, Poland
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Judyta K Juranek
- Department of Human Physiology and Pathophysiology, University of Warmia and Mazury in Olsztyn, School of Medicine, Collegium Medicum, Olsztyn, Poland.,Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
14
|
Sen CK, Roy S, Khanna S. Diabetic Peripheral Neuropathy Associated with Foot Ulcer: One of a Kind. Antioxid Redox Signal 2023. [PMID: 35850520 DOI: 10.1089/ars.2022.0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Significance: Diabetic peripheral neuropathy (DPN) associated with a diabetic foot ulcer (DFU) is likely to be complicated with critical factors such as biofilm infection and compromised skin barrier function of the diabetic skin. Repaired skin with a history of biofilm infection is known to be compromised in barrier function. Loss of barrier function is also observed in the oxidative stress affected diabetic and aged skin. Recent Advances: Loss of barrier function makes the skin prone to biofilm infection and cellulitis, which contributes to chronic inflammation and vasculopathy. Hyperglycemia favors biofilm formation as glucose lowering led to reduction in biofilm development. While vasculopathy limits oxygen supply, the O2 cost of inflammation is high increasing hypoxia severity. Critical Issues: The host nervous system can be inhabited by bacteria. Because electrical impulses are a part of microbial physiology, polymicrobial colonization of the host's neural circuit is likely to influence transmission of action potential. The identification of perineural apatite in diabetic patients with peripheral neuropathy suggests bacterial involvement. DPN starts in both feet at the same time. Future Directions: Pair-matched studies of DPN in the foot affected with DFU (i.e., DFU-DPN) compared with DPN in the without ulcer, and intact skin barrier function, are likely to provide critical insight that would help inform effective care strategies. This review characterizes DFU-DPN from a translational science point of view presenting a new paradigm that recognizes the current literature in the context of factors that are unique to DFU-DPN.
Collapse
Affiliation(s)
- Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Savita Khanna
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
15
|
Osonoi S, Mizukami H, Takeuchi Y, Sugawa H, Ogasawara S, Takaku S, Sasaki T, Kudoh K, Ito K, Sango K, Nagai R, Yamamoto Y, Daimon M, Yamamoto H, Yagihashi S. RAGE activation in macrophages and development of experimental diabetic polyneuropathy. JCI Insight 2022; 7:160555. [PMID: 36477360 PMCID: PMC9746912 DOI: 10.1172/jci.insight.160555] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/19/2022] [Indexed: 12/12/2022] Open
Abstract
It is suggested that activation of receptor for advanced glycation end products (RAGE) induces proinflammatory response in diabetic nerve tissues. Macrophage infiltration is invoked in the pathogenesis of diabetic polyneuropathy (DPN), while the association between macrophage and RAGE activation and the downstream effects of macrophages remain to be fully clarified in DPN. This study explored the role of RAGE in the pathogenesis of DPN through the modified macrophages. Infiltrating proinflammatory macrophages impaired insulin sensitivity, atrophied the neurons in dorsal root ganglion, and slowed retrograde axonal transport (RAT) in the sciatic nerve of type 1 diabetic mice. RAGE-null mice showed an increase in the population of antiinflammatory macrophages, accompanied by intact insulin sensitivity, normalized ganglion cells, and RAT. BM transplantation from RAGE-null mice to diabetic mice protected the peripheral nerve deficits, suggesting that RAGE is a major determinant for the polarity of macrophages in DPN. In vitro coculture analyses revealed proinflammatory macrophage-elicited insulin resistance in the primary neuronal cells isolated from dorsal root ganglia. Applying time-lapse recording disclosed a direct impact of proinflammatory macrophage and insulin resistance on the RAT deficits in primary neuronal cultures. These results provide a potentially novel insight into the development of RAGE-related DPN.
Collapse
Affiliation(s)
- Sho Osonoi
- Department of Pathology and Molecular Medicine and,Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | | | - Yuki Takeuchi
- Department of Pathology and Molecular Medicine and,Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hikari Sugawa
- Laboratory of Food and Regulation Biology, Department of Bioscience, School of Agriculture, Tokai University, Higashi-ku, Kumamoto, Japan
| | | | - Shizuka Takaku
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | | | | | - Koichi Ito
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki, Japan
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Ryoji Nagai
- Laboratory of Food and Regulation Biology, Department of Bioscience, School of Agriculture, Tokai University, Higashi-ku, Kumamoto, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Makoto Daimon
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | | | | |
Collapse
|
16
|
Juranek J, Mukherjee K, Kordas B, Załęcki M, Korytko A, Zglejc-Waszak K, Szuszkiewicz J, Banach M. Role of RAGE in the Pathogenesis of Neurological Disorders. Neurosci Bull 2022; 38:1248-1262. [PMID: 35729453 PMCID: PMC9554177 DOI: 10.1007/s12264-022-00878-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/03/2022] [Indexed: 11/30/2022] Open
Abstract
This review reflects upon our own as well as other investigators' studies on the role of receptor for advanced glycation end-products (RAGE), bringing up the latest information on RAGE in physiology and pathology of the nervous system. Over the last ten years, major progress has been made in uncovering many of RAGE-ligand interactions and signaling pathways in nervous tissue; however, the translation of these discoveries into clinical practice has not come to fruition yet. This is likely, in part to be the result of our incomplete understanding of this crucial signaling pathway. Clinical trials examining the therapeutic efficacy of blocking RAGE-external ligand interactions by genetically engineered soluble RAGE or an endogenous RAGE antagonist, has not stood up to its promise; however, other trials with different blocking agents are being considered with hope for therapeutic success in diseases of the nervous system.
Collapse
Affiliation(s)
- Judyta Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-085, Olsztyn, Poland.
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Blacksburg, VA, 24016, USA
| | - Bernard Kordas
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-085, Olsztyn, Poland
| | - Michał Załęcki
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719, Olsztyn, Poland
| | - Agnieszka Korytko
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-085, Olsztyn, Poland
| | - Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-085, Olsztyn, Poland
| | - Jarosław Szuszkiewicz
- Department of Materials and Machines Technology, Faculty of Technical Sciences, University of Warmia and Mazury, 10-719, Olsztyn, Poland
| | - Marta Banach
- Department of Neurology, Collegium Medicum, Jagiellonian University, 31-008, Kraków, Poland.
| |
Collapse
|
17
|
Hyperglycemia-triggered ATF6-CHOP pathway aggravates acute inflammatory liver injury by β-catenin signaling. Cell Death Dis 2022; 8:115. [PMID: 35289326 PMCID: PMC8921205 DOI: 10.1038/s41420-022-00910-z] [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: 10/09/2021] [Revised: 01/08/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022]
Abstract
Although hyperglycemia has been documented as an unfavorable element that can further induce liver ischemia–reperfusion injury (IRI), the related molecular mechanisms remain to be clearly elaborated. This study investigated the effective manner of endoplasmic reticulum (ER) stress signaling in hyperglycemia-exacerbated liver IRI. Here we demonstrated that in the liver tissues and Kupffer cells (KCs) of DM patients and STZ-induced hyperglycemic mice, the ER stress-ATF6-CHOP signaling pathway is activated. TLR4-mediated pro-inflammatory activation was greatly attenuated by the addition of 4-phenylbutyrate (PBA), one common ER stress inhibitor. The liver IRI in hyperglycemic mice was also significantly reduced after PBA treatment. In addition, deficiency of CHOP (CHOP−/−) obviously alleviates the hepatic IRI, and pro-inflammatory effects deteriorated by hyperglycemia. In hyperglycemic mice, β-catenin expression was suppressed while the ATF6-CHOP signal was activated. In the liver tissues of PBA-treated or CHOP−/− hyperglycemic mice, the expression of β-catenin was restored. Furthermore, CHOP deficiency can induce protection against hyperglycemia-related liver IRI, which was disrupted by the knockdown of β-catenin will cause this protection to disappear. High glucose (HG) treatment stimulated ATF6-CHOP signaling, reduced cellular β-catenin accumulation, and promoted the TLR4-related inflammation of BMDMs. But the above effects were partially rescued in BMDMs with CHOP deficiency or by PBA treatment. In BMDMs cultured in HG conditions, the anti-inflammatory functions of CHOP−/− were destroyed by the knockdown of β-catenin. Finally, chimeric mice carrying WT or CHOP−/− BMDMs by bone marrow transplantation were adopted to verify the above conclusion. The current study suggested that hyperglycemia could trigger ER stress-ATF6-CHOP axis, inhibit β-catenin activation, accelerate inflammation, and deteriorate liver IRI, thus providing the treatment potential for management of sterile liver inflammation in DM patients.
Collapse
|
18
|
Kinscherf NA, Pehar M. Role and Therapeutic Potential of RAGE Signaling in Neurodegeneration. Curr Drug Targets 2022; 23:1191-1209. [PMID: 35702767 PMCID: PMC9589927 DOI: 10.2174/1389450123666220610171005] [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: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/29/2022] [Indexed: 01/03/2023]
Abstract
Activation of the receptor for advanced glycation end products (RAGE) has been shown to play an active role in the development of multiple neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis. Although originally identified as a receptor for advanced glycation end products, RAGE is a pattern recognition receptor able to bind multiple ligands. The final outcome of RAGE signaling is defined in a context and cell type specific manner and can exert both neurotoxic and neuroprotective functions. Contributing to the complexity of the RAGE signaling network, different RAGE isoforms with distinctive signaling capabilities have been described. Moreover, multiple RAGE ligands bind other receptors and RAGE antagonism can significantly affect their signaling. Here, we discuss the outcome of celltype specific RAGE signaling in neurodegenerative pathologies. In addition, we will review the different approaches that have been developed to target RAGE signaling and their therapeutic potential. A clear understanding of the outcome of RAGE signaling in a cell type- and disease-specific manner would contribute to advancing the development of new therapies targeting RAGE. The ability to counteract RAGE neurotoxic signaling while preserving its neuroprotective effects would be critical for the success of novel therapies targeting RAGE signaling.
Collapse
Affiliation(s)
- Noah Alexander Kinscherf
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Mariana Pehar
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI, USA
| |
Collapse
|
19
|
Jaroslawska J, Korytko A, Zglejc-Waszak K, Antonowski T, Pomianowski AS, Wasowicz K, Wojtkiewicz J, Juranek JK. Peripheral Neuropathy Presents Similar Symptoms and Pathological Changes in Both High-Fat Diet and Pharmacologically Induced Pre- and Diabetic Mouse Models. Life (Basel) 2021; 11:life11111267. [PMID: 34833143 PMCID: PMC8618965 DOI: 10.3390/life11111267] [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: 10/19/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 12/27/2022] Open
Abstract
The objective of the study was to compare the effects of experimentally induced type 1 or type 2 diabetes (T1D or T2D) on the functional, structural and biochemical properties of mouse peripheral nerves. Eight-week-old C57BL/6 mice were randomly assigned into three groups, including the control (CTRL, chow-fed), STZ (streptozotocin (STZ)-injected), and HFD (high-fat diet (HFD)-fed) group. After 18-weeks of experimental treatment, HFD mice had higher body weights and elevated levels of plasma lipids, while STZ mice developed hyperglycemia. STZ-treated mice, after an extended period of untreated diabetes, developed motor and sensory nerve conduction-velocity deficits. Moreover, relative to control fibers, pre- and diabetic axons were lower in number and irregular in shape. Animals from both treatment groups manifested a pronounced overexpression of nNOS and a reduced expression of SOD1 proteins in the sciatic nerve, indicating oxidative–nitrosative stress and ineffective antioxidant protection in the peripheral nervous system of these mice. Collectively, STZ- and HFD-treated mice revealed similar characteristics of peripheral nerve damage, including a number of morphological and electrophysiological pathologies in the sciatic nerve. While hyperglycemia is a large component of diabetic neuropathy pathogenesis, the non-hyperglycemic effects of diabetes, including dyslipidemia, may also be of importance in the development of this condition.
Collapse
Affiliation(s)
- Julia Jaroslawska
- Department of Biological Functions of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
- Correspondence: (J.J.); (J.K.J.)
| | - Agnieszka Korytko
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.K.); (K.Z.-W.); (T.A.); (J.W.)
| | - Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.K.); (K.Z.-W.); (T.A.); (J.W.)
| | - Tomasz Antonowski
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.K.); (K.Z.-W.); (T.A.); (J.W.)
| | - Andrzej S. Pomianowski
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-718 Olsztyn, Poland;
| | - Krzysztof Wasowicz
- Department of Pathophysiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-718 Olsztyn, Poland;
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.K.); (K.Z.-W.); (T.A.); (J.W.)
| | - Judyta K. Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.K.); (K.Z.-W.); (T.A.); (J.W.)
- Correspondence: (J.J.); (J.K.J.)
| |
Collapse
|
20
|
Zglejc-Waszak K, Mukherjee K, Juranek JK. The cross-talk between RAGE and DIAPH1 in neurological complications of diabetes: A review. Eur J Neurosci 2021; 54:5982-5999. [PMID: 34449932 DOI: 10.1111/ejn.15433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 01/10/2023]
Abstract
Neuropathy, or dysfunction of peripheral nerve, is one of the most common neurological manifestation in patients with diabetes mellitus (DM). DM is typically associated with a hyperglycaemic milieu, which promotes non-enzymatic glycation of proteins. Proteins with advanced glycation are known to engage a cell-surface receptor called the receptor for advanced glycation end products (RAGE). Thus, it is reasonable to assume that RAGE and its associated molecule-mediated cellular signalling may contribute to DM-induced symmetrical axonal (length-dependent) neuropathy. Of particular interest is diaphanous related formin 1 (DIAPH1), a cytoskeletal organizing molecule, which interacts with the cytosolic domain of RAGE and whose dysfunction may precipitate axonopathy/neuropathy. Indeed, it has been demonstrated that both RAGE and DIAPH1 are expressed in the motor and sensory fibres of nerve harvested from DM animal models. Although the detailed molecular role of RAGE and DIAPH1 in diabetic neurological complications remains unclear, here we will discuss available evidence of their involvement in peripheral diabetic neuropathy. Specifically, we will discuss how a hyperglycaemic environment is not only likely to elevate advanced glycation end products (ligands of RAGE) and induce a pro-inflammatory environment but also alter signalling via RAGE and DIAPH1. Further, hyperglycaemia may regulate epigenetic mechanisms that interacts with RAGE signalling. We suggest the cumulative effect of hyperglycaemia on RAGE-DIAPH1-mediated signalling may be disruptive to axonal cytoskeletal organization and transport and is therefore likely to play a key role in pathogenesis of diabetic symmetrical axonal neuropathy.
Collapse
Affiliation(s)
- Kamila Zglejc-Waszak
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Virginia Tech Roanoke, Roanoke, Virginia, USA
| | - Judyta Karolina Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
21
|
Takahashi K, Mizukami H, Osonoi S, Ogasawara S, Hara Y, Kudoh K, Takeuchi Y, Sasaki T, Daimon M, Yagihashi S. Inhibitory effects of xanthine oxidase inhibitor, topiroxostat, on development of neuropathy in db/db mice. Neurobiol Dis 2021; 155:105392. [PMID: 34000416 DOI: 10.1016/j.nbd.2021.105392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation and oxidative stress contribute to the pathophysiology of diabetic neuropathy. According to recent evidence, the modulation of macrophage polarization in peripheral nerves represents a potential therapeutic target for diabetic neuropathy. Xanthine oxidase, which is a form of xanthin oxidoreductase, is the rate-limiting enzyme that catalyzes the degradation of hypoxanthine and xanthine into uric acid. Activation of xanthine oxidase promotes oxidative stress and macrophage activation. A preclinical study reported the beneficial effects of xanthine oxidase inhibitors on peripheral nerve dysfunction in experimental models of diabetes. However, the detailed mechanisms remain unknown. In this study, we examined the effect of the xanthine oxidase inhibitor topiroxostat on macrophage polarization and peripheral neuropathy in an obese diabetic model, db/db mice. First, the effects of xanthine oxidase inhibitors on cultured macrophages and dorsal root ganglion neurons exposed to xanthine oxidase were assessed. Furthermore, five-week-old db/db mice were administered the xanthine oxidase inhibitors topiroxostat [1 mg/kg/day (dbT1) or 2 mg/kg/day (dbT2)] or febuxostat [1 mg/kg (dbF)]. Glucose metabolism and body weight were evaluated during the experimental period. At 4 and 8 weeks of treatment, peripheral nerve functions such as nerve conduction velocities, thermal thresholds and pathology of skin and sciatic nerves were evaluated. The mRNA expression of molecules related to inflammation and oxidative stress was also measured in sciatic nerves. Untreated db/db mice and the nondiabetic db strain (db/m) were studied for comparison. An in vitro study showed that topiroxostat suppressed macrophage activation and proinflammatory but not anti-inflammatory polarization, and prevented the reduction in neurite outgrowth from neurons exposed to xanthine oxidase. Neuropathic changes exemplified by delayed nerve conduction and reduced intraepidermal nerve fiber density developed in db/db mice. These deficits were significantly prevented in the treated group, most potently in dbT2. Protective effects were associated with the suppression of macrophage infiltration, cytokine expression, and oxidative stress in the sciatic nerve and decreased plasma xanthine oxidoreductase activity. Our results revealed the beneficial effects of the xanthine oxidase inhibitor topiroxostat on neuropathy development in a mouse model of type 2 diabetes. The suppression of proinflammatory macrophage activation and oxidative stress-induced damage were suggested to be involved in this process.
Collapse
Affiliation(s)
- Kazuhisa Takahashi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan.
| | - Sho Osonoi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Saori Ogasawara
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Yutaro Hara
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; Department of Gastroenterological Surgery and Pediatric Surgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Kazuhiro Kudoh
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Yuki Takeuchi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Takanori Sasaki
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Makoto Daimon
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Soroku Yagihashi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| |
Collapse
|
22
|
Strom A, Strassburger K, Schmuck M, Shevalye H, Davidson E, Zivehe F, Bönhof G, Reimer R, Belgardt BF, Fleming T, Biermann B, Burkart V, Müssig K, Szendroedi J, Yorek MA, Fritsche E, Nawroth PP, Roden M, Ziegler D. Interaction between magnesium and methylglyoxal in diabetic polyneuropathy and neuronal models. Mol Metab 2020; 43:101114. [PMID: 33166742 PMCID: PMC7704399 DOI: 10.1016/j.molmet.2020.101114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/28/2022] Open
Abstract
Objective The lack of effective treatments against diabetic sensorimotor polyneuropathy demands the search for new strategies to combat or prevent the condition. Because reduced magnesium and increased methylglyoxal levels have been implicated in the development of both type 2 diabetes and neuropathic pain, we aimed to assess the putative interplay of both molecules with diabetic sensorimotor polyneuropathy. Methods In a cross-sectional study, serum magnesium and plasma methylglyoxal levels were measured in recently diagnosed type 2 diabetes patients with (n = 51) and without (n = 184) diabetic sensorimotor polyneuropathy from the German Diabetes Study baseline cohort. Peripheral nerve function was assessed using nerve conduction velocity and quantitative sensory testing. Human neuroblastoma cells (SH-SY5Y) and mouse dorsal root ganglia cells were used to characterize the neurotoxic effect of methylglyoxal and/or neuroprotective effect of magnesium. Results Here, we demonstrate that serum magnesium concentration was reduced in recently diagnosed type 2 diabetes patients with diabetic sensorimotor polyneuropathy and inversely associated with plasma methylglyoxal concentration. Magnesium, methylglyoxal, and, importantly, their interaction were strongly interrelated with methylglyoxal-dependent nerve dysfunction and were predictive of changes in nerve function. Magnesium supplementation prevented methylglyoxal neurotoxicity in differentiated SH-SY5Y neuron-like cells due to reduction of intracellular methylglyoxal formation, while supplementation with the divalent cations zinc and manganese had no effect on methylglyoxal neurotoxicity. Furthermore, the downregulation of mitochondrial activity in mouse dorsal root ganglia cells and consequently the enrichment of triosephosphates, the primary source of methylglyoxal, resulted in neurite degeneration, which was completely prevented through magnesium supplementation. Conclusions These multifaceted findings reveal a novel putative pathophysiological pathway of hypomagnesemia-induced carbonyl stress leading to neuronal damage and merit further investigations not only for diabetic sensorimotor polyneuropathy but also other neurodegenerative diseases associated with magnesium deficiency and impaired energy metabolism. Magnesium and methylglyoxal levels were inversely associated in individuals with type 2 diabetes and distal sensorimotor polyneuropathy. Magnesium, methylglyoxal, and their interaction were associated with methylglyoxal-dependent nerve dysfunction. Under experimental conditions, magnesium supplementation prevented methylglyoxal-mediated neurotoxicity. Magnesium downregulates intracellular methylglyoxal production.
Collapse
Affiliation(s)
- Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| | - Klaus Strassburger
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Martin Schmuck
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Hanna Shevalye
- Department of Internal Medicine, University of Iowa, Iowa City, USA
| | - Eric Davidson
- Department of Internal Medicine, University of Iowa, Iowa City, USA
| | - Fariba Zivehe
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Gidon Bönhof
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Rudolph Reimer
- Microscopy and Image Analysis Technology Platform, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Bengt-Frederik Belgardt
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Institute for Vascular and Islet Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Barbara Biermann
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Mark A Yorek
- Department of Internal Medicine, University of Iowa, Iowa City, USA; Iowa City VA Healthcare System, Iowa City, USA
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Peter P Nawroth
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
| | | |
Collapse
|
23
|
Suzuki A, Yabu A, Nakamura H. Advanced glycation end products in musculoskeletal system and disorders. Methods 2020; 203:179-186. [PMID: 32987130 DOI: 10.1016/j.ymeth.2020.09.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/18/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
The human population is ageing globally, and the number of old people is increasing yearly. Diabetes is common in the elderly, and the number of diabetic patients is also increasing. Elderly and diabetic patients often have musculoskeletal disorder, which are associated with advanced glycation end products (AGEs). AGEs are heterogeneous molecules derived from non-enzymatic products of the reaction of glucose or other sugar derivatives with proteins or lipids, and many different types of AGEs have been identified. AGEs are a biomarker for ageing and for evaluating disease conditions. Fluorescence, spectroscopy, mass spectrometry, chromatography, and immunological methods are commonly used to measure AGEs, but there is no standardized evaluation method because of the heterogeneity of AGEs. The formation of AGEs is irreversible, and they accumulate in tissue, eventually causing damage. AGE accumulation has been confirmed in neuromusculoskeletal tissues, including bones, cartilage, muscles, tendons, ligaments, and nerves, where they adversely affect biomechanical properties by causing charge changes and forming cross-linkages. AGEs also bind to receptors, such as the receptor for AGEs (RAGE), and induce inflammation by intracellular signal transduction. These mechanisms cause many varied aging and diabetes-related pathological conditions, such as osteoporosis, osteoarthritis, sarcopenia, tendinopathy, and neuropathy. Understanding of AGEs related pathomechanism may lead to develop novel methods for the prevention and therapy of such disorders which affect patients' quality of life. Herein, we critically review the current methodology used for detecting AGEs, and present potential mechanisms by which AGEs cause or exacerbate musculoskeletal disorders.
Collapse
Affiliation(s)
- Akinobu Suzuki
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Japan.
| | - Akito Yabu
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Japan
| | - Hiroaki Nakamura
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Japan
| |
Collapse
|
24
|
Paeschke S, Baum P, Toyka KV, Blüher M, Koj S, Klöting N, Bechmann I, Thiery J, Kosacka J, Nowicki M. The Role of Iron and Nerve Inflammation in Diabetes Mellitus Type 2-Induced Peripheral Neuropathy. Neuroscience 2019; 406:496-509. [DOI: 10.1016/j.neuroscience.2019.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 12/11/2022]
|
25
|
Leitão L, Alves CJ, Sousa DM, Neto E, Conceição F, Lamghari M. The alliance between nerve fibers and stem cell populations in bone marrow: life partners in sickness and health. FASEB J 2019; 33:8697-8710. [PMID: 31017803 DOI: 10.1096/fj.201900454r] [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] [Indexed: 12/21/2022]
Abstract
The bone marrow (BM) is the central hematopoietic organ in adult mammals, with great potential to be used as a tool to improve the efficacy of the body's response to a number of malignancies and stressful conditions. The nervous system emerges as a critical regulatory player of the BM both under homeostatic and pathologic settings, with essential roles in cellular anchorage and egress, stem cell differentiation, and endothelial cell permeability. This review collects the current knowledge on the interplay between the nervous system and the BM cell populations, with a focus on how the nervous system modulates hematopoietic stem and progenitor cell, mesenchymal stromal cell, and endothelial progenitor cell activity in BM. We have also highlighted the pathologies that have been associated with disturbances in the neuronal signaling in BM and discussed if targeting the nervous system, either by modulating the activity of specific neuronal circuits or by pharmacologically leveling the activity of sympathetic and sensorial signaling-responsive cells in BM, is a promising therapeutic approach to tackling pathologies from BM origin.-Leitão, L., Alves, C. J., Sousa, D. M., Neto, E., Conceição, F., Lamghari, M. The alliance between nerve fibers and stem cell populations in bone marrow: life partners in sickness and health.
Collapse
Affiliation(s)
- Luís Leitão
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Cecília J Alves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Daniela M Sousa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Estrela Neto
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Francisco Conceição
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
26
|
Zhou J, Zhang Z, Qian G. Neuropathy and inflammation in diabetic bone marrow. Diabetes Metab Res Rev 2019; 35:e3083. [PMID: 30289199 DOI: 10.1002/dmrr.3083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 09/05/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022]
Abstract
Diabetes impairs the bone marrow (BM) architecture and function as well as the mobilization of immature cells into the bloodstream and number of potential regenerative cells. Circadian regulation of bone immature cell migration is regulated by β-adrenergic receptors, which are expressed on haematopoietic stem cells, mesenchymal stem cells, and osteoblasts in the BM. Diabetes is associated with a substantially lower number of sympathetic nerve terminal endings in the BM; thus, diabetic neuropathy plays a critical role in BM dysfunction. Treatment with mesenchymal stem cells, BM mononuclear cells, haematopoietic stem cells, and stromal cells ameliorates the dysfunction of diabetic neuropathy, which occurs, in part, through secreted neurotrophic factors, growth factors, adipokines, and polarizing macrophage M2 cells and inhibiting inflammation. Inflammation may be a therapeutic target for BM stem cells to improve diabetic neuropathy. Given that angiogenic and neurotrophic effects are two major barriers to effective diabetic neuropathy therapy, targeting BM stem cells may provide a novel approach to develop these types of treatments.
Collapse
Affiliation(s)
- Jiyin Zhou
- National Drug Clinical Trial Institution, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zuo Zhang
- National Drug Clinical Trial Institution, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Guisheng Qian
- Institute of Respiratory Diseases, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
27
|
Fishman SL, Sonmez H, Basman C, Singh V, Poretsky L. The role of advanced glycation end-products in the development of coronary artery disease in patients with and without diabetes mellitus: a review. Mol Med 2018; 24:59. [PMID: 30470170 PMCID: PMC6251169 DOI: 10.1186/s10020-018-0060-3] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/04/2018] [Indexed: 12/18/2022] Open
Abstract
Background Traditional risk factors are insufficient to explain all cases of coronary artery disease (CAD) in patients with diabetes mellitus (DM). Advanced glycation end-products (AGEs) and their receptors may play important roles in the development and progression of CAD. Body Hyperglycemia is the hallmark feature of DM. An increase in the incidence of both micro-and macrovascular complications of diabetes has been observed with increased duration of hyperglycemia. This association persists even after glycemic control has been achieved, suggesting an innate mechanism of “metabolic memory.” AGEs are glycated proteins that may serve as mediators of metabolic memory due to their increased production in the setting of hyperglycemia and generally slow turnover. Elevated AGE levels can lead to abnormal cross linking of extracellular and intracellular proteins disrupting their normal structure and function. Furthermore, activation of AGE receptors can induce complex signaling pathways leading to increased inflammation, oxidative stress, enhanced calcium deposition, and increased vascular smooth muscle apoptosis, contributing to the development of atherosclerosis. Through these mechanisms, AGEs may be important mediators of the development of CAD. However, clinical studies regarding the role of AGEs and their receptors in advancing CAD are limited, with contradictory results. Conclusion AGEs and their receptors may be useful biomarkers for the presence and severity of CAD. Further studies are needed to evaluate the utility of circulating and tissue AGE levels in identifying asymptomatic patients at risk for CAD or to identify patients who may benefit from invasive intervention.
Collapse
Affiliation(s)
- Sarah Louise Fishman
- Division of Endocrinology, Department of Medicine, Lenox Hill Hospital, Northwell Health, 110 East 59th St #8B, New York, NY, 10022, USA
| | - Halis Sonmez
- Center for Diabetes and Endocrinology, 111 Salem Tpke, Norwich, CT, 06360, USA
| | - Craig Basman
- Department of Cardiology, Lenox Hill Hospital, Northwell Health, 100 East 77th St, New York, NY, 10065, USA
| | - Varinder Singh
- Department of Cardiology, Lenox Hill Hospital, Northwell Health, 100 East 77th St, New York, NY, 10065, USA
| | - Leonid Poretsky
- Division of Endocrinology, Department of Medicine, Lenox Hill Hospital, Northwell Health, 110 East 59th St #8B, New York, NY, 10022, USA.
| |
Collapse
|
28
|
Kobayashi M, Zochodne DW. Diabetic neuropathy and the sensory neuron: New aspects of pathogenesis and their treatment implications. J Diabetes Investig 2018; 9:1239-1254. [PMID: 29533535 PMCID: PMC6215951 DOI: 10.1111/jdi.12833] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/20/2018] [Accepted: 03/03/2018] [Indexed: 12/17/2022] Open
Abstract
Diabetic polyneuropathy (DPN) continues to be generally considered as a "microvascular" complication of diabetes mellitus alongside nephropathy and retinopathy. The microvascular hypothesis, however, might be tempered by the concept that diabetes directly targets dorsal root ganglion sensory neurons. This neuron-specific concept, supported by accumulating evidence, might account for important features of DPN, such as its early sensory neuron degeneration. Diabetic sensory neurons develop neuronal atrophy alongside a series of messenger ribonucleic acid (RNA) changes related to declines in structural proteins, increases in heat shock protein, increases in the receptor for advanced glycation end-products, declines in growth factor signaling and other changes. Insulin is recognized as a potent neurotrophic factor, and insulin ligation enhances neurite outgrowth through activation of the phosphoinositide 3-kinase-protein kinase B pathway within sensory neurons and attenuates phenotypic features of experimental DPN. Several interventions, including glucagon-like peptide-1 agonism, and phosphatase and tensin homolog inhibition to activate growth signals in sensory neurons, or heat shock protein overexpression, prevent or reverse neuropathic abnormalities in experimental DPN. Diabetic sensory neurons show a unique pattern of microRNA alterations, a key element of messenger RNA silencing. For example, let-7i is widely expressed in sensory neurons, supports their growth and is depleted in experimental DPN; its replenishment improves features of DPN models. Finally, impairment of pre-messenger RNA splicing in diabetic sensory neurons including abnormal nuclear RNA metabolism and structure with loss of survival motor neuron protein, a neuron survival molecule, and overexpression of CWC22, a splicing factor, offer further novel insights. The present review addresses these new aspects of DPN sensory neurodegeneration.
Collapse
Affiliation(s)
- Masaki Kobayashi
- Department of Neurology and Neurological ScienceGraduate School of MedicineTokyo Medical and Dental UniversityTokyoJapan
- Department of NeurologyYokufukai Geriatric HospitalTokyoJapan
| | - Douglas W Zochodne
- Division of Neurology and Department of MedicineNeuroscience and Mental Health InstituteFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
29
|
Yuan C, Hu J, Parathath S, Grauer L, Cassella CB, Bagdasarov S, Goldberg IJ, Ramasamy R, Fisher EA. Human Aldose Reductase Expression Prevents Atherosclerosis Regression in Diabetic Mice. Diabetes 2018; 67:1880-1891. [PMID: 29891593 PMCID: PMC6110315 DOI: 10.2337/db18-0156] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 05/25/2018] [Indexed: 12/19/2022]
Abstract
Guidelines to reduce cardiovascular risk in diabetes include aggressive LDL lowering, but benefits are attenuated compared with those in patients without diabetes. Consistent with this, we have reported in mice that hyperglycemia impaired atherosclerosis regression. Aldose reductase (AR) is thought to contribute to clinical complications of diabetes by directing glucose into pathways producing inflammatory metabolites. Mice have low levels of AR, thus raising them to human levels would be a more clinically relevant model to study changes in diabetes under atherosclerosis regression conditions. Donor aortae from Western diet-fed Ldlr-/- mice were transplanted into normolipidemic wild-type, Ins2Akita (Akita+/- , insulin deficient), human AR (hAR) transgenic, or Akita+/- /hAR mice. Akita+/- mice had impaired plaque regression as measured by changes in plaque size and the contents of CD68+ cells (macrophages), lipids, and collagen. Supporting synergy between hyperglycemia and hAR were the even more pronounced changes in these parameters in Akita+/- /hAR mice, which had atherosclerosis progression in spite of normolipidemia. Plaque CD68+ cells from the Akita+/- /hAR mice had increased oxidant stress and expression of inflammation-associated genes but decreased expression of anti-inflammatory genes. In summary, hAR expression amplifies impaired atherosclerosis regression in diabetic mice, likely by interfering with the expected reduction in plaque macrophage inflammation.
Collapse
MESH Headings
- Aldehyde Reductase/genetics
- Aldehyde Reductase/metabolism
- Animals
- Aorta/physiopathology
- Aorta/transplantation
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/physiopathology
- Biomarkers/blood
- Biomarkers/metabolism
- Crosses, Genetic
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/physiopathology
- Diet, Western/adverse effects
- Disease Models, Animal
- Disease Progression
- Gene Expression Regulation
- Humans
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Oxidative Stress
- Plaque, Atherosclerotic/immunology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/physiopathology
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Species Specificity
Collapse
Affiliation(s)
- Chujun Yuan
- Marc and Ruti Bell Vascular Biology Program, Leon Charney Division of Cardiology, New York University School of Medicine, New York, NY
- Department of Medicine, New York University School of Medicine, New York, NY
| | - Jiyuan Hu
- Division of Biostatistics, Department of Population Health, New York University School of Medicine, New York, NY
| | - Saj Parathath
- Marc and Ruti Bell Vascular Biology Program, Leon Charney Division of Cardiology, New York University School of Medicine, New York, NY
- Department of Medicine, New York University School of Medicine, New York, NY
| | - Lisa Grauer
- Marc and Ruti Bell Vascular Biology Program, Leon Charney Division of Cardiology, New York University School of Medicine, New York, NY
- Department of Medicine, New York University School of Medicine, New York, NY
| | - Courtney Blachford Cassella
- Marc and Ruti Bell Vascular Biology Program, Leon Charney Division of Cardiology, New York University School of Medicine, New York, NY
- Department of Medicine, New York University School of Medicine, New York, NY
| | - Svetlana Bagdasarov
- Department of Medicine, New York University School of Medicine, New York, NY
- Diabetes Research Center, Division of Endocrinology, New York University School of Medicine, New York, NY
| | - Ira J Goldberg
- Department of Medicine, New York University School of Medicine, New York, NY
- Diabetes Research Center, Division of Endocrinology, New York University School of Medicine, New York, NY
| | - Ravichandran Ramasamy
- Department of Medicine, New York University School of Medicine, New York, NY
- Diabetes Research Center, Division of Endocrinology, New York University School of Medicine, New York, NY
| | - Edward A Fisher
- Marc and Ruti Bell Vascular Biology Program, Leon Charney Division of Cardiology, New York University School of Medicine, New York, NY
- Department of Medicine, New York University School of Medicine, New York, NY
| |
Collapse
|
30
|
Chen J, Chopp M. Exosome Therapy for Stroke. Stroke 2018; 49:1083-1090. [PMID: 29669873 PMCID: PMC6028936 DOI: 10.1161/strokeaha.117.018292] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Jieli Chen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Geriatrics, Tianjin Medical University General Hospital, China (J.C.)
- Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, China (J.C.)
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Physics, Oakland University, Rochester, MI (M.C.)
| |
Collapse
|
31
|
Hyperglycemia in Stroke Impairs Polarization of Monocytes/Macrophages to a Protective Noninflammatory Cell Type. J Neurosci 2017; 36:9313-25. [PMID: 27605608 DOI: 10.1523/jneurosci.0473-16.2016] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 07/14/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Hyperglycemia is common in patients with acute stroke, even in those without preexisting diabetes, and denotes a bad outcome. However, the mechanisms underlying the detrimental effects of hyperglycemia are largely unclear. In a mouse model of ischemic stroke, we found that hyperglycemia increased the infarct volume and decreased the number of protective noninflammatory monocytes/macrophages in the ischemic brain. Ablation of peripheral monocytes blocked the detrimental effect of hyperglycemia, suggesting that monocytes are required. In hyperglycemic mice, α-dicarbonyl glucose metabolites, the precursors for advanced glycation end products, were significantly elevated in plasma and ischemic brain tissue. The receptor of advanced glycation end products, AGER (previously known as RAGE), interfered with polarization of macrophages to a noninflammatory phenotype. When Ager was deleted, hyperglycemia did not aggravate ischemic brain damage any longer. Independently of AGER, methylglyoxal reduced the release of endothelial CSF-1 (M-CSF), which stimulates polarization of macrophages to a noninflammatory phenotype in the microenvironment of the ischemic brain. In summary, our study identified α-dicarbonyls and AGER as mediators by which hyperglycemia lowers the number of protective noninflammatory macrophages and consequently increases ischemic brain damage. Modulating the metabolism of α-dicarbonyls or blocking AGER may improve the treatment of stroke patients with hyperglycemia. SIGNIFICANCE STATEMENT Although glucose is the main energy substrate of the brain, hyperglycemia aggravates ischemic brain damage in acute stroke. So far, clinical trials have indicated that insulin treatment provides no solution to this common clinical problem. This study shows, in an experimental stroke model, that hyperglycemia interferes with the polarization of monocytes/macrophages to a protective cell type. Key players are α-dicarbonyls and the receptor for advanced glycation end products (AGER). Deletion of AGER normalized monocyte/macrophage polarization and reversed the detrimental effects of hyperglycemia, suggesting new avenues to treat stroke patients.
Collapse
|
32
|
Brown BN, Haschak MJ, Lopresti ST, Stahl EC. Effects of age-related shifts in cellular function and local microenvironment upon the innate immune response to implants. Semin Immunol 2017; 29:24-32. [PMID: 28539184 DOI: 10.1016/j.smim.2017.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/18/2017] [Accepted: 05/11/2017] [Indexed: 12/14/2022]
Abstract
The host macrophage response is now well recognized as a predictor of the success or failure of biomaterial implants following placement. More specifically, shifts from an "M1" pro-inflammatory towards a more "M2-like" anti-inflammatory macrophage polarization profile have been shown to result in enhanced material integration and/or tissue regeneration downstream. As a result, a number of biomaterials-based approaches to controlling macrophage polarization have been developed. However, the ability to promote such activity is predicated upon an in-depth, context-dependent understanding of the host response to biomaterials. Recent work has shown the impacts of both tissue location and tissue status (i.e. underlying pathology) upon the host innate immune response to implants, representing a departure from a focus upon implant material composition and form. Thus, the ideas of "biocompatibility," the host macrophage reaction, and ideal material requirements and modification strategies may need to be revisited on a patient, tissue, and disease basis. Immunosenescence, dysregulation of macrophage function, and delayed resolution of immune responses in aged individuals have all been demonstrated, suggesting that the host response to biomaterials in aged individuals should differ from that in younger individuals. However, despite the increasing usage of implantable medical devices in aged patients, few studies examining the effects of aging upon the host response to biomaterials and the implications of this response for long-term integration and function have been performed. The objective of the present manuscript is to review the putative effects of aging upon the host response to implanted materials and to advance the hypothesis that age-related changes in the local microenvrionement, with emphasis on the extracellular matrix, play a previously unrecognized role in determining the host response to implants.
Collapse
Affiliation(s)
- Bryan N Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, United States; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA 15213, United States.
| | - Martin J Haschak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, United States
| | - Samuel T Lopresti
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, United States
| | - Elizabeth C Stahl
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States; Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, 200 Lothrop St., Pittsburgh, PA 15261, United States
| |
Collapse
|
33
|
de la Hoz CL, Cheng C, Fernyhough P, Zochodne DW. A model of chronic diabetic polyneuropathy: benefits from intranasal insulin are modified by sex and RAGE deletion. Am J Physiol Endocrinol Metab 2017; 312:E407-E419. [PMID: 28223295 PMCID: PMC5451527 DOI: 10.1152/ajpendo.00444.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/06/2017] [Accepted: 02/18/2017] [Indexed: 01/11/2023]
Abstract
Human diabetic polyneuropathy (DPN) is a progressive complication of chronic diabetes mellitus. Preliminary evidence has suggested that intranasal insulin, in doses insufficient to alter hyperglycemia, suppresses the development of DPN. In this work we confirm this finding, but demonstrate that its impact is modified by sex and deletion of RAGE, the receptor for advanced glycosylation end products. We serially evaluated experimental DPN in male and female wild-type mice and male RAGE null (RN) mice, each with nondiabetic controls, during 16 wk of diabetes, the final 8 wk including groups given intranasal insulin. Age-matched nondiabetic female mice had higher motor and sensory conduction velocities than their male counterparts and had lesser conduction slowing from chronic diabetes. Intranasal insulin improved slowing in both sexes. In male RN mice, there was less conduction slowing with chronic diabetes, and intranasal insulin provided limited benefits. Rotarod testing and hindpaw grip power offered less consistent impacts. Mechanical sensitivity and thermal sensitivity were respectively but disparately changed and improved with insulin in wild-type female and male mice but not RN male mice. These studies confirm that intranasal insulin improves indexes of experimental DPN but indicates that females with DPN may differ in their underlying phenotype. RN mice had partial but incomplete protection from underlying DPN and lesser impacts from insulin. We also identify an important role for sex in the development of DPN and report evidence that insulin and AGE-RAGE pathways in its pathogenesis may overlap.
Collapse
Affiliation(s)
- Cristiane L de la Hoz
- Department of Clinical Neurosciences, the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; and
| | - Chu Cheng
- Department of Clinical Neurosciences, the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; and
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St Boniface Hospital Research Centre and Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, Alberta Diabetes Institute, University of Alberta, Alberta, Canada;
- Department of Clinical Neurosciences, the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; and
| |
Collapse
|
34
|
Hidmark AS, Nawroth PP, Fleming T. STZ causes depletion of immune cells in sciatic nerve and dorsal root ganglion in experimental diabetes. J Neuroimmunol 2017; 306:76-82. [DOI: 10.1016/j.jneuroim.2017.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 02/06/2017] [Accepted: 03/10/2017] [Indexed: 12/20/2022]
|
35
|
Sango K, Mizukami H, Horie H, Yagihashi S. Impaired Axonal Regeneration in Diabetes. Perspective on the Underlying Mechanism from In Vivo and In Vitro Experimental Studies. Front Endocrinol (Lausanne) 2017; 8:12. [PMID: 28203223 PMCID: PMC5285379 DOI: 10.3389/fendo.2017.00012] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/16/2017] [Indexed: 12/21/2022] Open
Abstract
Axonal regeneration after peripheral nerve injury is impaired in diabetes, but its precise mechanisms have not been elucidated. In this paper, we summarize the progress of research on altered axonal regeneration in animal models of diabetes and cultured nerve tissues exposed to hyperglycemia. Impaired nerve regeneration in animal diabetes can be attributed to dysfunction of neurons and Schwann cells, unfavorable stromal environment supportive of regenerating axons, and alterations of target tissues receptive to reinnervation. In particular, there are a number of factors such as enhanced activity of the negative regulators of axonal regeneration (e.g., phosphatase and tensin homolog deleted on chromosome 10 and Rho/Rho kinase), delayed Wallerian degeneration, alterations of the extracellular matrix components, enhanced binding of advanced glycation endproducts (AGEs) with the receptor for AGE, and delayed muscle reinnervation that can be obstacles to functional recovery after an axonal injury. It is also noteworthy that we and others have observed excessive neurite outgrowth from peripheral sensory ganglion explants from streptozotocin (STZ)-diabetic mice in culture and enhanced regeneration of small nerve fibers after sciatic nerve injury in STZ-induced diabetic rats. The excess of abortive neurite outgrowth may lead to misconnections of axons and target organs, which may interfere with appropriate target reinnervation and functional repair. Amelioration of perturbed nerve regeneration may be crucial for the future management of diabetic neuropathy.
Collapse
Affiliation(s)
- Kazunori Sango
- Diabetic Neuropathy Project, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- *Correspondence: Kazunori Sango,
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | | | - Soroku Yagihashi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
36
|
Shekhtman A, Ramasamy R, Schmidt AM. Glycation & the RAGE axis: targeting signal transduction through DIAPH1. Expert Rev Proteomics 2016; 14:147-156. [PMID: 27967251 DOI: 10.1080/14789450.2017.1271719] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION The consequences of chronic disease are vast and unremitting; hence, understanding the pathogenic mechanisms mediating such disorders holds promise to identify therapeutics and diminish the consequences. The ligands of the receptor for advanced glycation end products (RAGE) accumulate in chronic diseases, particularly those characterized by inflammation and metabolic dysfunction. Although first discovered and reported as a receptor for advanced glycation end products (AGEs), the expansion of the repertoire of RAGE ligands implicates the receptor in diverse milieus, such as autoimmunity, chronic inflammation, obesity, diabetes, and neurodegeneration. Areas covered: This review summarizes current knowledge regarding the ligand families of RAGE and data from human subjects and animal models on the role of the RAGE axis in chronic diseases. The recent discovery that the cytoplasmic domain of RAGE binds to the formin homology 1 (FH1) domain, DIAPH1, and that this interaction is essential for RAGE ligand-stimulated signal transduction, is discussed. Finally, we review therapeutic opportunities targeting the RAGE axis as a means to mitigate chronic diseases. Expert commentary: With the aging of the population and the epidemic of cardiometabolic disease, therapeutic strategies to target molecular pathways that contribute to the sequelae of these chronic diseases are urgently needed. In this review, we propose that the ligand/RAGE axis and its signaling nexus is a key factor in the pathogenesis of chronic disease and that therapeutic interruption of this pathway may improve quality and duration of life.
Collapse
Affiliation(s)
- Alexander Shekhtman
- a Department of Chemistry , University at Albany, State University of New York , Albany , NY , 12222 , USA
| | - Ravichandran Ramasamy
- b Diabetes Research Program, Division of Endocrinology, Department of Medicine , NYU Langone Medical Center , New York , NY , 10016 , USA
| | - Ann Marie Schmidt
- b Diabetes Research Program, Division of Endocrinology, Department of Medicine , NYU Langone Medical Center , New York , NY , 10016 , USA
| |
Collapse
|
37
|
Aikawa E, Fujita R, Asai M, Kaneda Y, Tamai K. Receptor for Advanced Glycation End Products-Mediated Signaling Impairs the Maintenance of Bone Marrow Mesenchymal Stromal Cells in Diabetic Model Mice. Stem Cells Dev 2016; 25:1721-1732. [PMID: 27539289 DOI: 10.1089/scd.2016.0067] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Bone marrow mesenchymal stromal cells (BM-MSCs) have been demonstrated to contribute to tissue regeneration. However, chronic pathological conditions, such as diabetes and aging, can result in a decreased number and/or quality of BM-MSCs. We therefore investigated the maintenance mechanism of BM-MSCs by studying signaling through the receptor for advanced glycation end products (RAGE), which is thought to be activated under various pathological conditions. The abundance of endogenous BM-MSCs decreased in a type 2 diabetes mellitus (DM2) model, as determined by performing colony-forming unit (CFU) assays. Flow cytometric analysis revealed that the prevalence of the Lin-/ckit-/CD106+/CD44- BM population, which was previously identified as a slow-cycling BM-MSC population, also decreased. Furthermore, in a streptozotocin-induced type 1 DM model (DM1), the CFUs of fibroblasts and the prevalence of the Lin-/ckit-/CD106+/CD44- BM population also significantly decreased. BM-MSCs in RAGE knockout (KO) mice were resistant to such reduction induced by streptozotocin treatment, suggesting that chronic RAGE signaling worsened the maintenance mechanism of BM-MSCs. Using an in vitro culture condition, BM-MSCs from RAGE-KO mice showed less proliferation and expressed significantly more Nanog and Oct-4, which are key factors in multipotency, than did wild-type BM-MSCs. Furthermore, RAGE-KO BM-MSCs showed a greater capacity for differentiation into mesenchymal lineages, such as adipocytes and osteocytes. These data suggested that RAGE signaling inhibition is useful for maintaining BM-MSCs in vitro. Together, our findings indicated that perturbation of BM-MSCs in DM could be partially explained by chronic RAGE signaling and that targeting the RAGE signaling pathway is a viable approach for maintaining BM-MSCs under chronic pathological conditions.
Collapse
Affiliation(s)
- Eriko Aikawa
- 1 Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University , Suita, Japan
| | - Ryo Fujita
- 1 Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University , Suita, Japan .,2 Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences , Nagasaki, Japan .,3 Division of Gene Therapy Science, Graduate School of Medicine, Osaka University , Suita, Japan
| | - Maiko Asai
- 4 Faculty of Medicine, Hiroshima University , Higashihiroshima, Japan
| | - Yasufumi Kaneda
- 3 Division of Gene Therapy Science, Graduate School of Medicine, Osaka University , Suita, Japan
| | - Katsuto Tamai
- 1 Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University , Suita, Japan
| |
Collapse
|
38
|
Bang OY, Kim EH, Cha JM, Moon GJ. Adult Stem Cell Therapy for Stroke: Challenges and Progress. J Stroke 2016; 18:256-266. [PMID: 27733032 PMCID: PMC5066440 DOI: 10.5853/jos.2016.01263] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/15/2016] [Accepted: 09/18/2016] [Indexed: 02/06/2023] Open
Abstract
Stroke is one of the leading causes of death and physical disability among adults. It has been 15 years since clinical trials of stem cell therapy in patients with stroke have been conducted using adult stem cells like mesenchymal stem cells and bone marrow mononuclear cells. Results of randomized controlled trials showed that adult stem cell therapy was safe but its efficacy was modest, underscoring the need for new stem cell therapy strategies. The primary limitations of current stem cell therapies include (a) the limited source of engraftable stem cells, (b) the presence of optimal time window for stem cell therapies, (c) inherited limitation of stem cells in terms of growth, trophic support, and differentiation potential, and (d) possible transplanted cell-mediated adverse effects, such as tumor formation. Here, we discuss recent advances that overcome these hurdles in adult stem cell therapy for stroke.
Collapse
Affiliation(s)
- Oh Young Bang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea
| | - Eun Hee Kim
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea
| | - Jae Min Cha
- Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology, Samsung Electronics Co., Ltd., Seoul, Korea.,Medical Device Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Gyeong Joon Moon
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea.,Stem cell and Regenerative Medicine Institute, Samsung Biomedical Research Institute, Seoul, Korea
| |
Collapse
|
39
|
Kato J, Agalave NM, Svensson CI. Pattern recognition receptors in chronic pain: Mechanisms and therapeutic implications. Eur J Pharmacol 2016; 788:261-273. [PMID: 27343378 DOI: 10.1016/j.ejphar.2016.06.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/13/2022]
Abstract
For the individual, it is vital to promptly detect and recognize a danger that threatens the integrity of the body. Pattern recognition receptors (PRRs) are several classes of protein families originally classified as receptors detecting exogenous pathogens. PRRs are also capable of recognizing molecules released from damaged tissues (damage-associated molecular pattern molecules; DAMPs) and thereby contribute to danger recognition. Importantly, it is now evident that PRRs, such as toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE), are not only expressed in peripheral immune cells but also present in neurons and glial cells in the nervous system. These PRR-expressing cells work in concert, enabling highly sensitive danger recognition. However, this sensitiveness can act as a double-edged sword. Accumulated evidence has led to the hypothesis that aberrant activation of PRRs may play a crucial role in the pathogenesis of pathological pain. Indeed, numerous studies employing gene deletion or pharmacological inhibition of PRRs successfully reversed or prevented pathological pain in experimental animal models. Furthermore, a number of preclinical studies have shown the therapeutic potential of targeting PRRs for chronic pain. Here, we review the current knowledge regarding the role of PRRs in chronic pain and discuss the promise and challenges of targeting PRRs as a novel therapeutic approach for chronic pain.
Collapse
Affiliation(s)
- Jungo Kato
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Nilesh M Agalave
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Juranek JK, Daffu GK, Geddis MS, Li H, Rosario R, Kaplan BJ, Kelly L, Schmidt AM. Soluble RAGE Treatment Delays Progression of Amyotrophic Lateral Sclerosis in SOD1 Mice. Front Cell Neurosci 2016; 10:117. [PMID: 27242430 PMCID: PMC4860390 DOI: 10.3389/fncel.2016.00117] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/22/2016] [Indexed: 12/12/2022] Open
Abstract
The etiology of amyotrophic lateral sclerosis (ALS), a fatal motor neuron disorder characterized by progressive muscle weakness and spasticity, remains largely unknown. Approximately 5-10% of cases are familial, and of those, 15-20% are associated with mutations in the gene encoding Cu/Zn superoxide dismutase (SOD1). Mutations of the SOD1 gene interrupt cellular homeostasis and contribute to cellular toxicity evoked by the presence of altered SOD1, along with other toxic species, such as advanced glycation end products (AGEs). AGEs trigger activation of their chief cell surface receptor, RAGE (receptor for advanced glycation end products), and induce RAGE-dependent cellular stress and inflammation in neurons, thereby affecting their function and leading to apoptosis. Here, we show for the first time that the expression of RAGE is higher in the SOD1 transgenic mouse model of ALS vs. wild-type mouse spinal cord. We tested whether pharmacological blockade of RAGE may delay the onset and progression of disease in this mouse model. Our findings reveal that treatment of SOD1 transgenic mice with soluble RAGE (sRAGE), a natural competitor of RAGE that sequesters RAGE ligands and blocks their interaction with cell surface RAGE, significantly delays the progression of ALS and prolongs life span compared to vehicle treatment. We demonstrate that in sRAGE-treated SOD1 transgenic animals at the final stage of the disease, a significantly higher number of neurons and lower number of astrocytes is detectable in the spinal cord. We conclude that RAGE antagonism may provide a novel therapeutic strategy for ALS intervention.
Collapse
Affiliation(s)
- Judyta K Juranek
- Division of Endocrinology, Department of Medicine, New York University Langone Medical CenterNew York, NY, USA; Department of Surgery, Columbia University Medical CenterNew York, NY, USA
| | - Gurdip K Daffu
- Division of Endocrinology, Department of Medicine, New York University Langone Medical Center New York, NY, USA
| | - Matthew S Geddis
- Department of Surgery, Columbia University Medical CenterNew York, NY, USA; Department of Science, Borough of Manhattan Community College-City University of New YorkNew York, NY, USA
| | - Huilin Li
- Division of Biostatistics, Department of Population Health, New York University Langone Medical Center New York, NY, USA
| | - Rosa Rosario
- Division of Endocrinology, Department of Medicine, New York University Langone Medical CenterNew York, NY, USA; Department of Surgery, Columbia University Medical CenterNew York, NY, USA
| | - Benjamin J Kaplan
- Department of Surgery, Columbia University Medical Center New York, NY, USA
| | - Lauren Kelly
- Department of Surgery, Columbia University Medical Center New York, NY, USA
| | - Ann Marie Schmidt
- Division of Endocrinology, Department of Medicine, New York University Langone Medical CenterNew York, NY, USA; Department of Surgery, Columbia University Medical CenterNew York, NY, USA
| |
Collapse
|
41
|
López-Díez R, Shekhtman A, Ramasamy R, Schmidt AM. Cellular mechanisms and consequences of glycation in atherosclerosis and obesity. Biochim Biophys Acta Mol Basis Dis 2016; 1862:2244-2252. [PMID: 27166197 DOI: 10.1016/j.bbadis.2016.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/28/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023]
Abstract
Post-translational modification of proteins imparts diversity to protein functions. The process of glycation represents a complex set of pathways that mediates advanced glycation endproduct (AGE) formation, detoxification, intracellular disposition, extracellular release, and induction of signal transduction. These processes modulate the response to hyperglycemia, obesity, aging, inflammation, and renal failure, in which AGE formation and accumulation is facilitated. It has been shown that endogenous anti-AGE protective mechanisms are thwarted in chronic disease, thereby amplifying accumulation and detrimental cellular actions of these species. Atop these considerations, receptor for advanced glycation endproducts (RAGE)-mediated pathways downregulate expression and activity of the key anti-AGE detoxification enzyme, glyoxalase-1 (GLO1), thereby setting in motion an interminable feed-forward loop in which AGE-mediated cellular perturbation is not readily extinguished. In this review, we consider recent work in the field highlighting roles for glycation in obesity and atherosclerosis and discuss emerging strategies to block the adverse consequences of AGEs. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.
Collapse
Affiliation(s)
- Raquel López-Díez
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, New York, NY 10016, United States
| | - Alexander Shekhtman
- Department of Chemistry, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, New York, NY 10016, United States
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, NYU Langone Medical Center, New York, NY 10016, United States.
| |
Collapse
|
42
|
Chen H, Virk MS, Chen F. Phenolic acids inhibit the formation of advanced glycation end products in food simulation systems depending on their reducing powers and structures. Int J Food Sci Nutr 2016; 67:400-11. [DOI: 10.3109/09637486.2016.1166187] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hengye Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei Province, PR China
| | - Muhammad Safiullah Virk
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei Province, PR China
| | - Fusheng Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei Province, PR China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Wuhan, Hubei Province, PR China
| |
Collapse
|
43
|
Yagihashi S. Glucotoxic Mechanisms and Related Therapeutic Approaches. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 127:121-49. [PMID: 27133148 DOI: 10.1016/bs.irn.2016.03.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropathy is the earliest and commonest complication of diabetes. With increasing duration of diabetes, frequency and severity of neuropathy are worsened. Long-term hyperglycemia is therefore implicated in the development of this disorder. Nerve tissues require glucose energy to function and survive. Upon excessive glucose entry into the peripheral nerve, the glycolytic pathway and collateral glucose-utilizing pathways are overactivated and initiate adverse effects on nerve tissues. During hyperglycemia, flux through the polyol pathway, formation of advanced glycation end-products, production of free radicals, flux into the glucosamine pathway, and protein kinase C activity are all enhanced to negatively influence nerve function and structure. Suppression of these aberrant metabolic pathways has succeeded in prevention and inhibition of the development of neuropathy in animal models with diabetes. Satisfactory results were not attained, however, in patients with diabetes and further clinical trials are required. In this review, the author summarizes the hitherto proposed theories on the pathogenesis of diabetic neuropathy related to glucose metabolism and future prospects for the effective treatment of neuropathy.
Collapse
Affiliation(s)
- S Yagihashi
- Hirosaki University Graduate School of Medicine, Hirosaki, Japan.
| |
Collapse
|
44
|
Ray R, Juranek JK, Rai V. RAGE axis in neuroinflammation, neurodegeneration and its emerging role in the pathogenesis of amyotrophic lateral sclerosis. Neurosci Biobehav Rev 2015; 62:48-55. [PMID: 26724598 DOI: 10.1016/j.neubiorev.2015.12.006] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 10/19/2015] [Accepted: 12/10/2015] [Indexed: 12/13/2022]
Abstract
RAGE, the receptor of advanced glycation end-products, is thought to be one of the potential contributors to the neurodegeneration. It has been shown that RAGE activation triggers an increase in proinflammatory molecules, oxidative stressors and cytokines. RAGE involvement has been documented in the pathogenesis of a number of neurodegenerative diseases such amyotrophic lateral sclerosis (ALS), Alzheimer's, Parkinson's, Huntington's, Creutzfeld-Jakob' diseases and various neurodegenerative conditions such as diabetic neuropathy, familial amyloid polyneuropathy, Charcot neuroarthropathy and vasculitic neuropathy. Although the detailed mechanisms of RAGE contribution to the neurodegeneration remains unclear, studies indicate that RAGE detrimental actions are exerted via its binding to the pro-inflammatory ligands such as advanced glycation end-products, S100/calgranulin and amphoterin and subsequent activation of downstream regulatory pathways such as NF-κB, STAT and JKN pathways. Here, in this review we attempt to shed light onto molecular events and pathological pathways involved in neuroinflammation, neurodegeneration and its emerging role in the pathogenesis of amyotrophic lateral sclerosis (ALS)--a progressive and fatal neurodegenerative disorder, summarizing current knowledge and the prospect of RAGE in the pathogenesis of this disastrous disease.
Collapse
Affiliation(s)
- Rashmi Ray
- Institute of Life Sciences, Bhubaneswar 751023, India; Manipal University, Karnataka 576104, India
| | - Judyta K Juranek
- Department of Medicine, New York University Medical Center, New York, USA; Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury, 10-557 Olsztyn, Poland.
| | - Vivek Rai
- Institute of Life Sciences, Bhubaneswar 751023, India.
| |
Collapse
|
45
|
Ramasamy R, Shekhtman A, Schmidt AM. The multiple faces of RAGE--opportunities for therapeutic intervention in aging and chronic disease. Expert Opin Ther Targets 2015; 20:431-46. [PMID: 26558318 DOI: 10.1517/14728222.2016.1111873] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION This review focuses on the multi-ligand receptor of the immunoglobulin superfamily--receptor for advanced glycation endproducts (RAGE). The accumulation of the multiple ligands of RAGE in cellular stress milieux links RAGE to the pathobiology of chronic disease and natural aging. AREAS COVERED In this review, we present a discussion on the ligands of RAGE and the implications of these ligand families in disease. We review the recent literature on the role of ligand-RAGE interaction in the consequences of natural aging; the macro- and microvascular complications of diabetes; obesity and insulin resistance; autoimmune disorders and chronic inflammation; and tumors and Alzheimer's disease. We discuss the mechanisms of RAGE signaling through its intracellular binding effector molecule--the formin DIAPH1. Physicochemical evidence of how the RAGE cytoplasmic domain binds to the FH1 (formin homology 1) domain of DIAPH1, and the consequences thereof, are also reviewed. EXPERT OPINION We discuss the modalities of RAGE antagonism currently in preclinical and clinical studies. Finally, we present the rationale behind potentially targeting the RAGE cytoplasmic domain-DIAPH1 interaction as a logical strategy for therapeutic intervention in the pathological settings of chronic diseases and aging wherein RAGE ligands accumulate and signal.
Collapse
Affiliation(s)
- Ravichandran Ramasamy
- a Diabetes Research Program, Division of Endocrinology, Department of Medicine , New York University Langone Medical Center , New York , NY 10016 , USA
| | - Alexander Shekhtman
- b Department of Chemistry , University at Albany, State University of New York , Albany , NY 12222 , USA
| | - Ann Marie Schmidt
- a Diabetes Research Program, Division of Endocrinology, Department of Medicine , New York University Langone Medical Center , New York , NY 10016 , USA
| |
Collapse
|
46
|
Yue S, Zhou H, Zhu J, Rao J, Busuttil RW, Kupiec-Weglinski JW, Lu L, Zhai Y. Hyperglycemia and liver ischemia reperfusion injury: a role for the advanced glycation endproduct and its receptor pathway. Am J Transplant 2015; 15:2877-87. [PMID: 26112980 PMCID: PMC9438741 DOI: 10.1111/ajt.13360] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 04/17/2015] [Accepted: 04/19/2015] [Indexed: 01/25/2023]
Abstract
Although pretransplant diabetes is a risk factor for mortality post-liver transplant, the underlying mechanism has not been fully defined. In a murine liver partial warm ischemia model, we addressed the question of how diabetes/hyperglycemia impacted tissue inflammatory injuries against ischemia reperfusion (IR), focusing on the advanced glycation endproduct (AGE) and its receptor (RAGE) pathway. Our results showed that hepatocellular injury was exacerbated in streptozotocin-induced diabetic mice against IR, in association with hyper-inflammatory immune activation in livers. Serum levels of AGEs, but not HMGB1, were increased in diabetic mice in response to liver IR. Both RAGE antagonist peptides and small interfering RNA alleviated liver injuries and inhibited inflammatory immune activation against IR in diabetic, but not normal, mice. Kupffer cells (KCs)/macrophages, but not hepatocytes, from diabetic mice expressed significantly higher levels of RAGE, leading to their hyper-inflammatory responsiveness to both TLR ligands and AGEs. In vitro, hyperglycemia increased macrophage RAGE expression and enhanced their TLR responses. Our results demonstrated that activation of the AGE-RAGE signaling pathway in KCs was responsible for hyper-inflammatory immune responses and exacerbated hepatocellular injuries in diabetic/hyperglycemic hosts against liver IR.
Collapse
Affiliation(s)
- Shi Yue
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA
| | - Haoming Zhou
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA,Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jianjun Zhu
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA,Liver Surgery, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jianhua Rao
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA,Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ronald W. Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA
| | - Ling Lu
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
47
|
Litwinoff E, Hurtado Del Pozo C, Ramasamy R, Schmidt AM. Emerging Targets for Therapeutic Development in Diabetes and Its Complications: The RAGE Signaling Pathway. Clin Pharmacol Ther 2015; 98:135-44. [PMID: 25974754 DOI: 10.1002/cpt.148] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 05/08/2015] [Accepted: 05/08/2015] [Indexed: 12/16/2022]
Abstract
Types 1 and 2 diabetes are on the rise worldwide. Although the treatment of hyperglycemia has benefited from recent advances, aggressive efforts to maintain euglycemia may be fraught with risk, especially in older subjects or in subjects vulnerable to hypoglycemic unawareness. Hence, strategies to prevent and treat the complications of hyperglycemia are essential. In this review we summarize recent updates on the biology of the receptor for advanced glycation endproducts (RAGE) in the pathogenesis of both micro- and macrovascular complications of diabetes, insights from the study of mouse models of obesity and diabetic complications, and from associative studies in human subjects. The study of the mechanisms and consequences of the interaction of the RAGE cytoplasmic domain with the formin, mDia1, in RAGE signal transduction, will be discussed. Lastly, we review the "state-of-the-art" on RAGE-directed therapeutics. Tackling RAGE/mDia1 may identify a novel class of therapeutics preventing diabetes and its complications.
Collapse
Affiliation(s)
- Ems Litwinoff
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - C Hurtado Del Pozo
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - R Ramasamy
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - A M Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
48
|
Mietto BS, Mostacada K, Martinez AMB. Neurotrauma and inflammation: CNS and PNS responses. Mediators Inflamm 2015; 2015:251204. [PMID: 25918475 PMCID: PMC4397002 DOI: 10.1155/2015/251204] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/24/2015] [Accepted: 03/09/2015] [Indexed: 01/09/2023] Open
Abstract
Traumatic injury to the central nervous system (CNS) or the peripheral nervous system (PNS) triggers a cascade of events which culminate in a robust inflammatory reaction. The role played by inflammation in the course of degeneration and regeneration is not completely elucidated. While, in peripheral nerves, the inflammatory response is assumed to be essential for normal progression of Wallerian degeneration and regeneration, CNS trauma inflammation is often associated with poor recovery. In this review, we discuss key mechanisms that trigger the inflammatory reaction after nervous system trauma, emphasizing how inflammations in both CNS and PNS differ from each other, in terms of magnitude, cell types involved, and effector molecules. Knowledge of the precise mechanisms that elicit and maintain inflammation after CNS and PNS tissue trauma and their effect on axon degeneration and regeneration is crucial for the identification of possible pharmacological drugs that can positively affect the tissue regenerative capacity.
Collapse
Affiliation(s)
- Bruno Siqueira Mietto
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, 21941-550 Rio de Janeiro, RJ, Brazil
| | - Klauss Mostacada
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, 21941-550 Rio de Janeiro, RJ, Brazil
| | - Ana Maria Blanco Martinez
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, 21941-550 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
49
|
Tsukamoto M, Sango K, Niimi N, Yanagisawa H, Watabe K, Utsunomiya K. Upregulation of galectin-3 in immortalized Schwann cells IFRS1 under diabetic conditions. Neurosci Res 2015; 92:80-5. [DOI: 10.1016/j.neures.2014.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/23/2014] [Accepted: 11/27/2014] [Indexed: 01/08/2023]
|
50
|
Ma K, Xu Y, Wang C, Li N, Li K, Zhang Y, Li X, Yang Q, Zhang H, Zhu X, Bai H, Ben J, Ding Q, Li K, Jiang Q, Xu Y, Chen Q. A cross talk between class A scavenger receptor and receptor for advanced glycation end-products contributes to diabetic retinopathy. Am J Physiol Endocrinol Metab 2014; 307:E1153-65. [PMID: 25352436 DOI: 10.1152/ajpendo.00378.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In response to hyperglycemia in patients with diabetes, many signaling pathways contribute to the pathogenesis of diabetic complications, including diabetic retinopathy (DR). Excessive production of inflammatory mediators plays an important role in this process. Amadori-glycated albumin, one of the major forms of advanced glycated end-products, has been implicated in DR by inducing inflammatory responses in microglia/macrophages. Our goal was to delineate the potential cross talk between class A scavenger receptor (SR-A) and the receptor for advanced glycated end-product (RAGE) in the context of DR. We show here that SR-A ablation caused an exacerbated form of DR in streptozotocin-injected C57BL/6J mice as evidenced by fundus imaging and electroretinography. Immunohistochemical staining and RT-PCR assay indicated that there was augmented activation of proinflammatory macrophages with upregulated synthesis of proinflammatory mediators in the retina in Sr-a(-/-) mice. Overexpression of SR-A suppressed RAGE-induced mitogen-activated protein kinase (MAPK) signaling, whereas RAGE activation in macrophages favored a proinflammatory (M1) phenotype in the absence of SR-A. Mechanistic analysis on bone marrow-derived macrophages and HEK293 cell line revealed that SR-A interacted with and inhibited the phosphorylation of mitogen-activated protein kinase kinase 7, the major kinase in the RAGE-MAPK-NF-κB signaling, thereby leading to diminished secretion of proinflammatory cytokines. Our findings suggest that the antagonism between SR-A and RAGE contributes to the pathogenesis of DR by nurturing a disease-prone macrophage phenotype. Therefore, specific agonist that boosts SR-A signaling could potentially provide benefits in the prevention and/or intervention of DR.
Collapse
Affiliation(s)
- Ke Ma
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Yiming Xu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Chenchen Wang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Nan Li
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Kexue Li
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Yan Zhang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Xiaoyu Li
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Qing Yang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Hanwen Zhang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Xudong Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Hui Bai
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Jingjing Ben
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Qingqing Ding
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Keran Li
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Xu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| | - Qi Chen
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Center, Nanjing Medical University, Nanjing, China; and
| |
Collapse
|