1
|
Eid SA, Elzinga SE, Kim B, Rumora AE, Hayes JM, Carter A, Pacut C, Allouch AM, Koubek EJ, Feldman EL. High-Intensity Interval Training, Caloric Restriction, or Their Combination Have Beneficial Effects on Metabolically Acquired Peripheral Neuropathy. Diabetes 2024; 73:1895-1907. [PMID: 39163551 PMCID: PMC11493763 DOI: 10.2337/db23-0997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Peripheral neuropathy (PN) is a prevalent and debilitating complication of obesity, prediabetes, and type 2 diabetes, which remains poorly understood and lacks disease-modifying therapies. Fortunately, diet and/or exercise have emerged as effective treatment strategies for PN. Here, we examined the impact of caloric restriction (CR) and high-intensity interval training (HIIT) interventions, alone or combined (HIIT-CR), on metabolic and PN outcomes in high-fat diet (HFD) mice. HFD feeding alone resulted in obesity, impaired glucose tolerance, and PN. Peripheral nerves isolated from these mice also developed insulin resistance (IR). CR and HIIT-CR, but not HIIT alone, improved HFD-induced metabolic dysfunction. However, all interventions improved PN to similar extents. When examining the underlying neuroprotective mechanisms in whole nerves, we found that CR and HIIT-CR activate the fuel-sensing enzyme AMPK. We then performed complimentary in vitro work in Schwann cells, the glia of peripheral nerves. Treating primary Schwann cells with the saturated fatty acid palmitate to mimic prediabetic conditions caused IR, which was reversed by the AMPK activator, AICAR. Together, these results enhance our understanding of PN pathogenesis, the differential mechanisms by which diet and exercise may improve PN, and Schwann cell-specific contributions to nerve insulin signaling and PN progression. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | | | - Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Amy E. Rumora
- Department of Neurology, University of Michigan, Ann Arbor, MI
- Department of Neurology, Columbia University, New York, NY
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Andrew Carter
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Crystal Pacut
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Adam M. Allouch
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Emily J. Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
2
|
Cao B, Li K, Ke J, Zhao D. Trajectories of Remnant Cholesterol are Associated with Diabetic Foot Ulcer in Adult Patients with Type 2 Diabetes: A Retrospective Cohort Study. Diabetes Metab Syndr Obes 2024; 17:3043-3051. [PMID: 39166153 PMCID: PMC11334913 DOI: 10.2147/dmso.s461330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/03/2024] [Indexed: 08/22/2024] Open
Abstract
Objective This study aimed to investigate the potential association between long-term variations in remnant cholesterol (RC) levels and the development of diabetic foot ulcers (DFU) in participants with type 2 diabetes (T2D). Methods This was a retrospective cohort study. Variation in RC was assessed by the following metrics: mean, standard deviation (SD), coefficient of variation (CV) and trajectories pattern of RC. To identify RC trajectories, we employed the latent class mixture model. The primary endpoint was the development of DFU, and the time-to-event data were analyzed using Cox regression. Results A total of 1874 patients with T2D were included, with a median follow-up duration of 4.7 years. Among them, 129 individuals (6.9%) developed DFU. The proportion of DFU was significantly higher in the U-shaped group compared to the median group (P for trend < 0.001). Upon adjustment for confounding variables, the U-shaped trajectory correlated with a higher risk of DFU, demonstrating a hazard ratio (HR) of 2.57 (95% CI, 1.54-4.27). Subgroup analysis showed the U-shaped trajectory had a higher DFU risk regardless of gender (HR=2.40 and 2.81, respectively), glycemic control (HR=1.89 and 7.41, respectively), smoking (HR=2.36 and 2.93, respectively), or hypertension (HR=2.30 and 2.97, respectively). No association was found between mean, SD and CV of RC and DFU. Conclusion A U-shape trajectory of RC was independently associated with an elevated risk of DFU among patients with T2D.
Collapse
Affiliation(s)
- Bin Cao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| | - Kun Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People’s Republic of China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 101149, People’s Republic of China
| |
Collapse
|
3
|
Eid SA, Elzinga SE, Guo K, Hinder LM, Hayes JM, Pacut CM, Koubek EJ, Hur J, Feldman EL. Transcriptomic profiling of sciatic nerves and dorsal root ganglia reveals site-specific effects of prediabetic neuropathy. Transl Res 2024; 270:24-41. [PMID: 38556110 PMCID: PMC11166517 DOI: 10.1016/j.trsl.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/01/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Peripheral neuropathy (PN) is a severe and frequent complication of obesity, prediabetes, and type 2 diabetes characterized by progressive distal-to-proximal peripheral nerve degeneration. However, a comprehensive understanding of the mechanisms underlying PN, and whether these mechanisms change during PN progression, is currently lacking. Here, gene expression data were obtained from distal (sciatic nerve; SCN) and proximal (dorsal root ganglia; DRG) injury sites of a high-fat diet (HFD)-induced mouse model of obesity/prediabetes at early and late disease stages. Self-organizing map and differentially expressed gene analyses followed by pathway enrichment analysis identified genes and pathways altered across disease stage and injury site. Pathways related to immune response, inflammation, and glucose and lipid metabolism were consistently dysregulated with HFD-induced PN, irrespective of injury site. However, regulation of oxidative stress was unique to the SCN while dysregulated Hippo and Notch signaling were only observed in the DRG. The role of the immune system and inflammation in disease progression was supported by an increase in the percentage of immune cells in the SCN with PN progression. Finally, when comparing these data to transcriptomic signatures from human patients with PN, we observed conserved pathways related to metabolic dysregulation across species, highlighting the translational relevance of our mouse data. Our findings demonstrate that PN is associated with distinct site-specific molecular re-programming in the peripheral nervous system, identifying novel, clinically relevant therapeutic targets.
Collapse
Affiliation(s)
- Stéphanie A. Eid
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sarah E. Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Guo
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lucy M. Hinder
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Crystal M. Pacut
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily J. Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
4
|
Dobrowolska-Zrałka K, Janek Ł, Pawlik-Sobecka L, Smereka J, Regulska-Ilow B. Association of the Length of Service in the 24/48 Shift of Firefighters of the State Fire Service in Wroclaw on Selected Serum Biochemical Parameters of Nutritional Status. Nutrients 2024; 16:2467. [PMID: 39125346 PMCID: PMC11314308 DOI: 10.3390/nu16152467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The aim of the study was to evaluate the association of the quality of diet as calculated by the Nutrient Rich Food index (NRF9.3), and length of service (LS) (≤10 years vs. >10 years) with selected serum biochemical parameters, the proportions of different lipid profile fractions and advanced glycation endproduct (AGE) values of 108 firefighters from the State Fire Service in Wroclaw. The LS officers > 10 years had significantly higher total cholesterol (211.50 (184.00-254.00) vs. 184.00 (166.00-194.00)), LDL (123.75 (108.20-167.90) vs. 105.18 (90.24-119.00)) non-HDL (151.70 (132.00-196.70) vs. 122.00 (106.00-140.00)), triglycerides (118.50 (96.00-158.00) vs. 78.00 (67.00-103.00)) and lower HDL concentrations (51.30 (45.60-56.70) vs. 58.00 (51.70-66.10)) compared to firefighters in the LS ≤ 10 years group. Significant differences between the seniority groups were also noted for all lipid profile ratios. Regardless of the officers' seniority, systolic blood pressure was observed at the highest normal level of 134.4 ± 14.4 in the LS ≤ 10 years group and 139.5 ± 14.3 in the LS > 10 years group. Advanced glycation endproduct values were significantly dependent on diet quality, as expressed by the NRF9.3 index and on the TG/HDL ratio, but not on seniority. Diet quality, as expressed by the NRF9.3 index, had a significant association with GLU and FI levels, and components of the lipid profile between seniority groups. As NRF9.3 increased, TG/HDL, LDL/HDL, TC/HDL, and non-HDL/HDL ratios decreased. AGEs were significantly affected by NRF9.3 and significantly associated with TG/HDL. Firefighters' diets, as assessed by the NRF9.3 index, had a significant association with predictors of insulin resistance, diabetes, and cardiometabolic predictors between seniority groups. The nutritional education of firefighters (and other professional groups working irregularly), especially those with longer tenure (e.g., >10 years), is necessary to prevent the development of, e.g., CVD, MetS, and T2DM, which contribute towards a reduced ability to perform professional duties.
Collapse
Affiliation(s)
- Karolina Dobrowolska-Zrałka
- Department of Dietetics and Bromatology, Wroclaw Medical University, ul. Borowska 211, 50-556 Wroclaw, Poland;
| | - Łucja Janek
- Statistical Analysis Center, Wroclaw Medical University, ul. K. Marcinkowskiego 2-6, 50-368 Wroclaw, Poland;
| | - Lilla Pawlik-Sobecka
- Division of Basic Medical Sciences, Wroclaw Medical University, ul. Borowska 211, 50-556 Wroclaw, Poland;
| | - Jacek Smereka
- Department of Emergency Medical Service, Wroclaw Medical University, ul. Parkowa 34, 51-616 Wroclaw, Poland;
| | - Bożena Regulska-Ilow
- Department of Dietetics and Bromatology, Wroclaw Medical University, ul. Borowska 211, 50-556 Wroclaw, Poland;
| |
Collapse
|
5
|
Ratan Y, Rajput A, Pareek A, Pareek A, Kaur R, Sonia S, Kumar R, Singh G. Recent Advances in Biomolecular Patho-Mechanistic Pathways behind the Development and Progression of Diabetic Neuropathy. Biomedicines 2024; 12:1390. [PMID: 39061964 PMCID: PMC11273858 DOI: 10.3390/biomedicines12071390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetic neuropathy (DN) is a neurodegenerative disorder that is primarily characterized by distal sensory loss, reduced mobility, and foot ulcers that may potentially lead to amputation. The multifaceted etiology of DN is linked to a range of inflammatory, vascular, metabolic, and other neurodegenerative factors. Chronic inflammation, endothelial dysfunction, and oxidative stress are the three basic biological changes that contribute to the development of DN. Although our understanding of the intricacies of DN has advanced significantly over the past decade, the distinctive mechanisms underlying the condition are still poorly understood, which may be the reason behind the lack of an effective treatment and cure for DN. The present study delivers a comprehensive understanding and highlights the potential role of the several pathways and molecular mechanisms underlying the etiopathogenesis of DN. Moreover, Schwann cells and satellite glial cells, as integral factors in the pathogenesis of DN, have been enlightened. This work will motivate allied research disciplines to gain a better understanding and analysis of the current state of the biomolecular mechanisms behind the pathogenesis of DN, which will be essential to effectively address every facet of DN, from prevention to treatment.
Collapse
Affiliation(s)
- Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Aishwarya Rajput
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Ranjeet Kaur
- Adesh Institute of Dental Sciences and Research, Bathinda 151101, Punjab, India;
| | - Sonia Sonia
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India;
| | - Rahul Kumar
- Baba Ragav Das Government Medical College, Gorakhpur 273013, Uttar Pradesh, India;
| | - Gurjit Singh
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| |
Collapse
|
6
|
Cheng Y, Chen Y, Li K, Liu S, Pang C, Gao L, Xie J, Wenjing LV, Yu H, Deng B. How inflammation dictates diabetic peripheral neuropathy: An enlightening review. CNS Neurosci Ther 2024; 30:e14477. [PMID: 37795833 PMCID: PMC11017439 DOI: 10.1111/cns.14477] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/28/2023] [Accepted: 09/08/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) constitutes a debilitating complication associated with diabetes. Although, the past decade has seen rapid developments in understanding the complex etiology of DPN, there are no approved therapies that can halt the development of DPN, or target the damaged nerve. Therefore, clarifying the pathogenesis of DPN and finding effective treatment are the crucial issues for the clinical management of DPN. AIMS This review is aiming to summary the current knowledge on the pathogenesis of DPN, especially the mechanism and application of inflammatory response. METHODS We systematically summarized the latest studies on the pathogenesis and therapeutic strategies of diabetic neuropathy in PubMed. RESULTS In this seminal review, the underappreciated role of immune activation in the progression of DPN is scrutinized. Novel insights into the inflammatory regulatory mechanisms of DPN have been unearthed, illuminating potential therapeutic strategies of notable clinical significance. Additionally, a nuanced examination of DPN's complex etiology, including aberrations in glycemic control and insulin signaling pathways, is presented. Crucially, an emphasis has been placed on translating these novel understandings into tangible clinical interventions to ameliorate patient outcomes. CONCLUSIONS This review is distinguished by synthesizing cutting-edge mechanisms linking inflammation to DPN and identifying innovative, inflammation-targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yifan Cheng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouChina
| | - Yinuo Chen
- Department of NeurologyFirst Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
- First School of Clinical MedicineWenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Kezheng Li
- Department of NeurologyFirst Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
- First School of Clinical MedicineWenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Shuwei Liu
- First School of Clinical MedicineWenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Chunyang Pang
- Department of NeurologyFirst Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Lingfei Gao
- Department of NeurologyFirst Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Jiali Xie
- Department of Neurology, Shanghai East HospitalTongji UniversityShanghaiP.R. China
| | - L. V. Wenjing
- Department of GeriatricsThe Affiliated Hospital of Qingdao UniversityQingdaoShandong ProvinceChina
| | - Huan Yu
- Department of PediatricsSecond Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Binbin Deng
- Department of NeurologyFirst Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
- First School of Clinical MedicineWenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| |
Collapse
|
7
|
Cai M, Li S, Cai K, Du X, Han J, Hu J. Empowering mitochondrial metabolism: Exploring L-lactate supplementation as a promising therapeutic approach for metabolic syndrome. Metabolism 2024; 152:155787. [PMID: 38215964 DOI: 10.1016/j.metabol.2024.155787] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Mitochondrial dysfunction plays a critical role in the pathogenesis of metabolic syndrome (MetS), affecting various cell types and organs. In MetS animal models, mitochondria exhibit decreased quality control, characterized by abnormal morphological structure, impaired metabolic activity, reduced energy production, disrupted signaling cascades, and oxidative stress. The aberrant changes in mitochondrial function exacerbate the progression of metabolic syndrome, setting in motion a pernicious cycle. From this perspective, reversing mitochondrial dysfunction is likely to become a novel and powerful approach for treating MetS. Unfortunately, there are currently no effective drugs available in clinical practice to improve mitochondrial function. Recently, L-lactate has garnered significant attention as a valuable metabolite due to its ability to regulate mitochondrial metabolic processes and function. It is highly likely that treating MetS and its related complications can be achieved by correcting mitochondrial homeostasis disorders. In this review, we comprehensively discuss the complex relationship between mitochondrial function and MetS and the involvement of L-lactate in regulating mitochondrial metabolism and associated signaling pathways. Furthermore, it highlights recent findings on the involvement of L-lactate in common pathologies of MetS and explores its potential clinical application and further prospects, thus providing new insights into treatment possibilities for MetS.
Collapse
Affiliation(s)
- Ming Cai
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China; Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuyao Li
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China
| | - Keren Cai
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China
| | - Xinlin Du
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China
| | - Jia Han
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China.
| | - Jingyun Hu
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai 201299, PR China.
| |
Collapse
|
8
|
Karimi MA, Vaezi A, Ansari A, Archin I, Dadgar K, Rasouli A, Ghannadikhosh P, Alishiri G, Tizro N, Gharei F, Imanparvar S, Salehi S, Mazhari SA, Etemadi MH, Alipour M, Deravi N, Naziri M. Lipid variability and risk of microvascular complications in patients with diabetes: a systematic review and meta-analysis. BMC Endocr Disord 2024; 24:4. [PMID: 38167035 PMCID: PMC10759662 DOI: 10.1186/s12902-023-01526-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND AND AIMS The current systematic review aimed to elucidate the effects of lipid variability on microvascular complication risk in diabetic patients. The lipid components studied were as follows: High-density lipoprotein (HDL), High-density lipoprotein (LDL), Triglyceride (TG), Total Cholesterol (TC), and Remnant Cholesterol (RC). METHOD We carried out a systematic search in multiple databases, including PubMed, Web of Science, and SCOPUS, up to October 2nd, 2023. After omitting the duplicates, we screened the title and abstract of the studies. Next, we retrieved and reviewed the full text of the remaining articles and included the ones that met our inclusion criteria in the study. RESULT In this research, we examined seven studies, comprising six cohort studies and one cross-sectional study. This research was conducted in Hong Kong, China, Japan, Taiwan, Finland, and Italy. The publication years of these articles ranged from 2012 to 2022, and the duration of each study ranged from 5 to 14.3 years. The study group consisted of patients with type 2 diabetes aged between 45 and 84 years, with a diabetes history of 7 to 12 years. These studies have demonstrated that higher levels of LDL, HDL, and TG variability can have adverse effects on microvascular complications, especially nephropathy and neuropathic complications. TG and LDL variability were associated with the development of albuminuria and GFR decline. Additionally, reducing HDL levels showed a protective effect against microalbuminuria. However, other studies did not reveal an apparent relationship between lipid variations and microvascular complications, such as retinopathy. Current research lacks geographic and demographic diversity. Increased HDL, TG, and RC variability have been associated with several microvascular difficulties. Still, the pathogenic mechanism is not entirely known, and understanding how lipid variability affects microvascular disorders may lead to novel treatments. Furthermore, the current body of this research is restricted in its coverage. This field's lack of thorough investigations required a more extensive study and comprehensive effort. CONCLUSION The relationship between lipid variation (LDL, HDL, and TG) (adverse effects) on microvascular complications, especially nephropathy and neuropathic (and maybe not retinopathy), is proven. Physicians and health policymakers should be highly vigilant to lipid variation in a general population.
Collapse
Affiliation(s)
- Mohammad Amin Karimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Vaezi
- Student Research Committee, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Ansari
- Medical Student, Shantou University Medical College, Shantou, Guangdong, China
| | - Iman Archin
- Kazan (Volga Region) Federal University, Kazan, Russia
| | - Kiarash Dadgar
- Young Researchers Elite Club, Islamic Azad University Tehran Medical Branch, Tehran, Iran
| | - Asma Rasouli
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Parna Ghannadikhosh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Goharsharieh Alishiri
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Neda Tizro
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Gharei
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saba Imanparvar
- School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sakineh Salehi
- Department of Medicine, Ardabil Medical Sciences Branch, Islamic Azad University, Ardabil, Iran
| | | | | | - Milad Alipour
- Medical Student, Department of Medicine, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | - Niloofar Deravi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mahdyieh Naziri
- Students Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Hashim M, Badruddeen, Akhtar J, Khan MI, Ahmad M, Islam A, Ahmad A. Diabetic Neuropathy: An Overview of Molecular Pathways and Protective Mechanisms of Phytobioactives. Endocr Metab Immune Disord Drug Targets 2024; 24:758-776. [PMID: 37867264 DOI: 10.2174/0118715303266444231008143430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/31/2023] [Accepted: 08/25/2023] [Indexed: 10/24/2023]
Abstract
Diabetic neuropathy (DN) is a common and debilitating complication of diabetes mellitus that affects the peripheral nerves and causes pain, numbness, and impaired function. The pathogenesis of DN involves multiple molecular mechanisms, such as oxidative stress, inflammation, and pathways of advanced glycation end products, polyol, hexosamine, and protein kinase C. Phytochemicals are natural compounds derived from plants that have various biological activities and therapeutic potential. Flavonoids, terpenes, alkaloids, stilbenes, and tannins are some of the phytochemicals that have been identified as having protective potential for diabetic neuropathy. These compounds can modulate various cellular pathways involved in the development and progression of neuropathy, including reducing oxidative stress and inflammation and promoting nerve growth and repair. In this review, the current evidence on the effects of phytochemicals on DN by focusing on five major classes, flavonoids, terpenes, alkaloids, stilbenes, and tannins, are summarized. This compilation also discusses the possible molecular targets of numerous pathways of DN that these phytochemicals modulate. These phytochemicals may offer a promising alternative or complementary approach to conventional drugs for DN management by modulating multiple pathological pathways and restoring nerve function.
Collapse
Affiliation(s)
- Mohd Hashim
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Badruddeen
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | | | - Mohammad Ahmad
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Asad Ahmad
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
10
|
Silsby M, Feldman EL, Dortch RD, Roth A, Haroutounian S, Rajabally YA, Vucic S, Shy ME, Oaklander AL, Simon NG. Advances in diagnosis and management of distal sensory polyneuropathies. J Neurol Neurosurg Psychiatry 2023; 94:1025-1039. [PMID: 36997315 PMCID: PMC10544692 DOI: 10.1136/jnnp-2021-328489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/23/2023] [Indexed: 04/01/2023]
Abstract
Distal sensory polyneuropathy (DSP) is characterised by length-dependent, sensory-predominant symptoms and signs, including potentially disabling symmetric chronic pain, tingling and poor balance. Some patients also have or develop dysautonomia or motor involvement depending on whether large myelinated or small fibres are predominantly affected. Although highly prevalent, diagnosis and management can be challenging. While classic diabetes and toxic causes are well-recognised, there are increasingly diverse associations, including with dysimmune, rheumatological and neurodegenerative conditions. Approximately half of cases are initially considered idiopathic despite thorough evaluation, but often, the causes emerge later as new symptoms develop or testing advances, for instance with genetic approaches. Improving and standardising DSP metrics, as already accomplished for motor neuropathies, would permit in-clinic longitudinal tracking of natural history and treatment responses. Standardising phenotyping could advance research and facilitate trials of potential therapies, which lag so far. This review updates on recent advances and summarises current evidence for specific treatments.
Collapse
Affiliation(s)
- Matthew Silsby
- Neurology, Westmead Hospital, Westmead, New South Wales, Australia
- Brain and Nerve Research Centre, Sydney Medical School, The University of Sydney, New South Wales, Australia
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard D Dortch
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Institute of Imaging Science, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University Institute of Imaging Science, Nashville, Tennessee, USA
| | - Alison Roth
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University School of Medicine in Saint Louis, St Louis, Missouri, USA
| | - Yusuf A Rajabally
- Inflammatory Neuropathy Clinic, Department of Neurology, University Hospitals Birmingham, Aston Medical School, Aston University, Birmingham, UK
| | - Steve Vucic
- Brain and Nerve Research Centre, Sydney Medical School, The University of Sydney, New South Wales, Australia
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Anne Louise Oaklander
- Nerve Unit, Departments of Neurology and Pathology (Neuropathology), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Neil G Simon
- Northern Beaches Clinical School, Macquarie University, Frenchs Forest, New South Wales, Australia
| |
Collapse
|
11
|
Kim B, Kang Y, Mendelson FE, Hayes JM, Savelieff MG, Nagrath S, Feldman EL. Palmitate and glucose increase amyloid precursor protein in extracellular vesicles: Missing link between metabolic syndrome and Alzheimer's disease. J Extracell Vesicles 2023; 12:e12340. [PMID: 37898562 PMCID: PMC10613125 DOI: 10.1002/jev2.12340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 06/05/2023] [Accepted: 06/11/2023] [Indexed: 10/30/2023] Open
Abstract
The metabolic syndrome (MetS) and Alzheimer's disease share several pathological features, including insulin resistance, abnormal protein processing, mitochondrial dysfunction and elevated inflammation and oxidative stress. The MetS constitutes elevated fasting glucose, obesity, dyslipidaemia and hypertension and increases the risk of developing Alzheimer's disease, but the precise mechanism remains elusive. Insulin resistance, which develops from a diet rich in sugars and saturated fatty acids, such as palmitate, is shared by the MetS and Alzheimer's disease. Extracellular vesicles (EVs) are also a point of convergence, with altered dynamics in both the MetS and Alzheimer's disease. However, the role of palmitate- and glucose-induced insulin resistance in the brain and its potential link through EVs to Alzheimer's disease is unknown. We demonstrate that palmitate and high glucose induce insulin resistance and amyloid precursor protein phosphorylation in primary rat embryonic cortical neurons and human cortical stem cells. Palmitate also triggers insulin resistance in oligodendrocytes, the supportive glia of the brain. Palmitate and glucose enhance amyloid precursor protein secretion from cortical neurons via EVs, which induce tau phosphorylation when added to naïve neurons. Additionally, EVs from palmitate-treated oligodendrocytes enhance insulin resistance in recipient neurons. Overall, our findings suggest a novel theory underlying the increased risk of Alzheimer's disease in MetS mediated by EVs, which spread Alzheimer's pathology and insulin resistance.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| | - Yoon‐Tae Kang
- Department of Chemical Engineering and Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Faye E. Mendelson
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| | - John M. Hayes
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| | - Masha G. Savelieff
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| | - Sunitha Nagrath
- Department of Chemical Engineering and Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
12
|
Eid SA, Rumora AE, Beirowski B, Bennett DL, Hur J, Savelieff MG, Feldman EL. New perspectives in diabetic neuropathy. Neuron 2023; 111:2623-2641. [PMID: 37263266 PMCID: PMC10525009 DOI: 10.1016/j.neuron.2023.05.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/20/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
Diabetes prevalence continues to climb with the aging population. Type 2 diabetes (T2D), which constitutes most cases, is metabolically acquired. Diabetic peripheral neuropathy (DPN), the most common microvascular complication, is length-dependent damage to peripheral nerves. DPN pathogenesis is complex, but, at its core, it can be viewed as a state of impaired metabolism and bioenergetics failure operating against the backdrop of long peripheral nerve axons supported by glia. This unique peripheral nerve anatomy and the injury consequent to T2D underpins the distal-to-proximal symptomatology of DPN. Earlier work focused on the impact of hyperglycemia on nerve damage and bioenergetics failure, but recent evidence additionally implicates contributions from obesity and dyslipidemia. This review will cover peripheral nerve anatomy, bioenergetics, and glia-axon interactions, building the framework for understanding how hyperglycemia and dyslipidemia induce bioenergetics failure in DPN. DPN and painful DPN still lack disease-modifying therapies, and research on novel mechanism-based approaches is also covered.
Collapse
Affiliation(s)
- Stephanie A Eid
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amy E Rumora
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Bogdan Beirowski
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Neuroscience Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Masha G Savelieff
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
13
|
Hu Y, Chen C, Liang Z, Liu T, Hu X, Wang G, Hu J, Xie X, Liu Z. Compound Qiying Granules alleviates diabetic peripheral neuropathy by inhibiting endoplasmic reticulum stress and apoptosis. Mol Med 2023; 29:98. [PMID: 37464341 PMCID: PMC10354983 DOI: 10.1186/s10020-023-00698-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/11/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) is a major complication of diabetes. This study aimed to investigate the therapeutic effects and molecular mechanisms of Compound Qiying Granules (CQYG) for DPN. METHODS Rats and RSC96 cells of DPN models were established to evaluate the therapeutic effects of CQYG. Then the morphology and apoptotic changes of sciatic nerves were detected. Further, tandem mass tag based quantitative proteomics technology was used to identify differentially expressed proteins (DEPs) and the underlying molecular mechanisms. Protein expression of key signaling pathways was also detected. RESULTS CQYG treatment significantly improved blood glucose and oxidative stress levels, and further reduced nerve fiber myelination lesions, denervation, and apoptosis in DPN rats. Further, 2176 DEPs were found in CQYG treated DPN rats. Enrichment analysis showed that protein processing in the endoplasmic reticulum (ER), and apoptosis were all inhibited after CQYG treatment. Next, CQYG treatment reduced inflammatory factor expression, mitochondrial damage, and apoptosis in RSC96 cells which induced by high glucose. Transmission electron microscopy results found that CQYG treatment improved the morphology of nerve myelin, mitochondria, and ER. CQYG treatment decreased ER stress and apoptosis pathway proteins that were highly expressed in DPN models. In addition, we also predicted the potential targets of CQYG in DEPs. CONCLUSIONS CQYG exerts neuroprotective effects in experimental diabetic neuropathy through anti-ER stress and anti-apoptosis.
Collapse
Affiliation(s)
- Yan Hu
- Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Chen Chen
- Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Zhengting Liang
- Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Tao Liu
- Xinjiang Medical University, Urumqi, 830011, Xinjiang, China.
- Traditional Chinese Medicine Hospital Affiliated With Xinjiang Medical University, Urumqi, 830000, Xinjiang, China.
| | - Xiaoling Hu
- Traditional Chinese Medicine Hospital Affiliated With Xinjiang Medical University, Urumqi, 830000, Xinjiang, China
| | - Guanying Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Jinxia Hu
- Traditional Chinese Medicine Hospital Affiliated With Xinjiang Medical University, Urumqi, 830000, Xinjiang, China.
| | - Xiaolin Xie
- Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Zhiyan Liu
- Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| |
Collapse
|
14
|
Eid SA, Noureldein M, Kim B, Hinder LM, Mendelson FE, Hayes JM, Hur J, Feldman EL. Single-cell RNA-seq uncovers novel metabolic functions of Schwann cells beyond myelination. J Neurochem 2023; 166:367-388. [PMID: 37328915 PMCID: PMC11141588 DOI: 10.1111/jnc.15877] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/04/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023]
Abstract
Schwann cells (SCs) support peripheral nerves under homeostatic conditions, independent of myelination, and contribute to damage in prediabetic peripheral neuropathy (PN). Here, we used single-cell RNA sequencing to characterize the transcriptional profiles and intercellular communication of SCs in the nerve microenvironment using the high-fat diet-fed mouse, which mimics human prediabetes and neuropathy. We identified four major SC clusters, myelinating, nonmyelinating, immature, and repair in healthy and neuropathic nerves, in addition to a distinct cluster of nerve macrophages. Myelinating SCs acquired a unique transcriptional profile, beyond myelination, in response to metabolic stress. Mapping SC intercellular communication identified a shift in communication, centered on immune response and trophic support pathways, which primarily impacted nonmyelinating SCs. Validation analyses revealed that neuropathic SCs become pro-inflammatory and insulin resistant under prediabetic conditions. Overall, our study offers a unique resource for interrogating SC function, communication, and signaling in nerve pathophysiology to help inform SC-specific therapies.
Collapse
Affiliation(s)
- Stéphanie A. Eid
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mohamed Noureldein
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lucy M. Hinder
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Faye E. Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
15
|
Enders J, Elliott D, Wright DE. Emerging Nonpharmacologic Interventions to Treat Diabetic Peripheral Neuropathy. Antioxid Redox Signal 2023; 38:989-1000. [PMID: 36503268 PMCID: PMC10402707 DOI: 10.1089/ars.2022.0158] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 12/14/2022]
Abstract
Significance: Diabetic peripheral neuropathy (DPN), a complication of metabolic syndrome, type I and type II diabetes, leads to sensory changes that include slow nerve conduction, nerve degeneration, loss of sensation, pain, and gate disturbances. These complications remain largely untreatable, although tight glycemic control can prevent neuropathy progression. Nonpharmacologic approaches remain the most impactful to date, but additional advances in treatment approaches are needed. Recent Advances: This review highlights several emerging interventions, including a focus on dietary interventions and physical activity, that continue to show promise for treating DPN. We provide an overview of our current understanding of how exercise can improve aspects of DPN. We also highlight new studies in which a ketogenic diet has been used as an intervention to prevent and reverse DPN. Critical Issues: Both exercise and consuming a ketogenic diet induce systemic and cellular changes that collectively improve complications associated with DPN. Both interventions may involve similar signaling pathways and benefits but also impact DPN through unique mechanisms. Future Directions: These lifestyle interventions are critically important as personalized medicine approaches will likely be needed to identify specific subsets of neuropathy symptoms and deficits in patients, and determine the most impactful treatment. Overall, these two interventions have the potential to provide meaningful relief for patients with DPN and provide new avenues to identify new therapeutic targets.
Collapse
Affiliation(s)
- Jonathan Enders
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Daniel Elliott
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Douglas E. Wright
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
16
|
Thomas S, Enders J, Kaiser A, Rovenstine L, Heslop L, Hauser W, Chadwick A, Wright D. Abnormal intraepidermal nerve fiber density in disease: A scoping review. Front Neurol 2023; 14:1161077. [PMID: 37153658 PMCID: PMC10157176 DOI: 10.3389/fneur.2023.1161077] [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: 02/07/2023] [Accepted: 03/30/2023] [Indexed: 05/10/2023] Open
Abstract
Background Intraepidermal nerve fiber density (IENFD) has become an important biomarker for neuropathy diagnosis and research. The consequences of reduced IENFD can include sensory dysfunction, pain, and a significant decrease in quality of life. We examined the extent to which IENFD is being used as a tool in human and mouse models and compared the degree of fiber loss between diseases to gain a broader understanding of the existing data collected using this common technique. Methods We conducted a scoping review of publications that used IENFD as a biomarker in human and non-human research. PubMed was used to identify 1,004 initial articles that were then screened to select articles that met the criteria for inclusion. Criteria were chosen to standardize publications so they could be compared rigorously and included having a control group, measuring IENFD in a distal limb, and using protein gene product 9.5 (PGP9.5). Results We analyzed 397 articles and collected information related to publication year, the condition studied, and the percent IENFD loss. The analysis revealed that the use of IENFD as a tool has been increasing in both human and non-human research. We found that IENFD loss is prevalent in many diseases, and metabolic or diabetes-related diseases were the most studied conditions in humans and rodents. Our analysis identified 73 human diseases in which IENFD was affected, with 71 reporting IENFD loss and an overall average IENFD change of -47%. We identified 28 mouse and 21 rat conditions, with average IENFD changes of -31.6% and -34.7%, respectively. Additionally, we present data describing sub-analyses of IENFD loss according to disease characteristics in diabetes and chemotherapy treatments in humans and rodents. Interpretation Reduced IENFD occurs in a surprising number of human disease conditions. Abnormal IENFD contributes to important complications, including poor cutaneous vascularization, sensory dysfunction, and pain. Our analysis informs future rodent studies so they may better mirror human diseases impacted by reduced IENFD, highlights the breadth of diseases impacted by IENFD loss, and urges exploration of common mechanisms that lead to substantial IENFD loss as a complication in disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Douglas Wright
- Sensory Nerve Disorder Lab, Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
17
|
Rayens NT, Cook KJ, McKinley SA, Payne CK. Palmitate-mediated disruption of the endoplasmic reticulum decreases intracellular vesicle motility. Biophys J 2023; 122:1355-1363. [PMID: 36869590 PMCID: PMC10111363 DOI: 10.1016/j.bpj.2023.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/02/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Essential cellular processes such as metabolism, protein synthesis, and autophagy require the intracellular transport of membrane-bound vesicles. The importance of the cytoskeleton and associated molecular motors for transport is well documented. Recent research has suggested that the endoplasmic reticulum (ER) may also play a role in vesicle transport through a tethering of vesicles to the ER. We use single-particle tracking fluorescence microscopy and a Bayesian change-point algorithm to characterize vesicle motility in response to the disruption of the ER, actin, and microtubules. This high-throughput change-point algorithm allows us to efficiently analyze thousands of trajectory segments. We find that palmitate-mediated disruption of the ER leads to a significant decrease in vesicle motility. A comparison with the disruption of actin and microtubules shows that disruption of the ER has a significant impact on vesicle motility, greater than the disruption of actin. Vesicle motility was dependent on cellular region, with greater motility in the cell periphery than the perinuclear region, possibly due to regional differences in actin and the ER. Overall, these results suggest that the ER is an important factor in vesicle transport.
Collapse
Affiliation(s)
- Nathan T Rayens
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina
| | - Keisha J Cook
- School of Mathematical and Statistical Sciences, Clemson University, Clemson, South Carolina
| | - Scott A McKinley
- Department of Mathematics, Tulane University, New Orleans, Louisiana
| | - Christine K Payne
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina.
| |
Collapse
|
18
|
Thomas SJ, Enders J, Kaiser A, Rovenstine L, Heslop L, Hauser W, Chadwick A, Wright DE. Abnormal Intraepidermal Nerve Fiber Density in Disease: A Scoping Review. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.08.23285644. [PMID: 36798392 PMCID: PMC9934806 DOI: 10.1101/2023.02.08.23285644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Background Intraepidermal nerve fiber density (IENFD) has become an important biomarker for neuropathy diagnosis and research. The consequences of reduced IENFD can include sensory dysfunction, pain, and a significant decrease in quality of life. We examined the extent to which IENFD is being used as a tool in human and mouse models and compared the degree of fiber loss between diseases to gain a broader understanding of the existing data collected using this common technique. Methods We conducted a scoping review of publications that used IENFD as a biomarker in human and non-human research. PubMed was used to identify 1,004 initial articles that were then screened to select articles that met the criteria for inclusion. Criteria were chosen to standardize publications so they could be compared rigorously and included having a control group, measuring IENFD in a distal limb, and using protein gene product 9.5 (PGP9.5). Results We analyzed 397 articles and collected information related to publication year, the condition studied, and the percent IENFD loss. The analysis revealed that the use of IENFD as a tool has been increasing in both human and non-human research. We found that IENFD loss is prevalent in many diseases, and metabolic or diabetes-related diseases were the most studied conditions in humans and rodents. Our analysis identified 74 human diseases in which IENFD was affected, with 71 reporting IENFD loss and an overall average IENFD change of -47%. We identified 28 mouse and 21 rat conditions, with average IENFD changes of -31.6 % and - 34.7% respectively. Additionally, we present data describing sub-analyses of IENFD loss according to disease characteristics in diabetes and chemotherapy treatments in humans and rodents. Interpretation Reduced IENFD occurs in a surprising number of human disease conditions. Abnormal IENFD contributes to important complications, including poor cutaneous vascularization, sensory dysfunction, and pain. Our analysis informs future rodent studies so they may better mirror human diseases impacted by reduced IENFD, highlights the breadth of diseases impacted by IENFD loss, and urges exploration of common mechanisms that lead to substantial IENFD loss as a complication in disease.
Collapse
Affiliation(s)
- SJ Thomas
- Sensory Nerve Disorder Lab, University of Kansas Medical Center, Anesthesiology Department, Kansas City, KS, USA
| | - J Enders
- Sensory Nerve Disorder Lab, University of Kansas Medical Center, Anesthesiology Department, Kansas City, KS, USA
| | - A Kaiser
- Sensory Nerve Disorder Lab, University of Kansas Medical Center, Anesthesiology Department, Kansas City, KS, USA
| | - L Rovenstine
- Sensory Nerve Disorder Lab, University of Kansas Medical Center, Anesthesiology Department, Kansas City, KS, USA
| | - L Heslop
- Sensory Nerve Disorder Lab, University of Kansas Medical Center, Anesthesiology Department, Kansas City, KS, USA
| | - W Hauser
- Sensory Nerve Disorder Lab, University of Kansas Medical Center, Anesthesiology Department, Kansas City, KS, USA
| | - A Chadwick
- Sensory Nerve Disorder Lab, University of Kansas Medical Center, Anesthesiology Department, Kansas City, KS, USA
| | - DE Wright
- Sensory Nerve Disorder Lab, University of Kansas Medical Center, Anesthesiology Department, Kansas City, KS, USA
| |
Collapse
|
19
|
Ras J, Kengne AP, Smith DL, Soteriades ES, Leach L. Association between Cardiovascular Disease Risk Factors and Cardiorespiratory Fitness in Firefighters: A Systematic Review and Meta-Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2816. [PMID: 36833514 PMCID: PMC9957465 DOI: 10.3390/ijerph20042816] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 06/12/2023]
Abstract
Approximately 45% of on-duty related mortalities were due to sudden cardiac death, with many of these fatalities related to cardiovascular disease and overexertion, while performing emergency duties. Therefore, the aim of this systematic review was to determine the association between cardiovascular disease risk factors and cardiorespiratory fitness in firefighters. A literature search of PubMed, SCOPUS, Web of Science, Embase, EBSCOHost, and ScienceDirect was conducted; the Rayyan® intelligent systematic review tool was used to screen and select studies for inclusion. The appraisal tool for cross-sectional studies and the Critical Appraisal Skills Programme toolkit were used for methodological assessment of included studies. Data were analyzed using the Review Manager 5.3 and MedCalc® statistical softwares to determine the effects of obesity (Z = 10.29, p < 0.001) and aging (Z = 4.72, p < 0.001) on cardiorespiratory fitness. Furthermore, there was a significant effect for cardiorespiratory fitness level on systolic blood pressure (Z = 5.94, p < 0.001), diastolic blood pressure (Z = 2.45, p < 0.001), total cholesterol levels (Z = 3.80, p < 0.001), low-density lipoprotein cholesterol (Z = 4.44, p < 0.001), triglycerides (Z = 3.76, p < 0.001) and blood glucose concentration (Z = 4.78, p < 0.001). Cardiovascular disease risk factors and cardiorespiratory fitness were significantly and inversely associated in firefighters. Fire service departments should adopt behavioral intervention strategies to maintain optimum cardiovascular disease risk factor profiles and cardiorespiratory fitness among firefighters to ensure their occupational well-being.
Collapse
Affiliation(s)
- Jaron Ras
- Department of Sport, Recreation and Exercise Science, Faculty of Community and Health Sciences, University of the Western Cape, Cape Town 7535, South Africa
| | - Andre P. Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town 7505, South Africa
| | - Denise L. Smith
- Health and Human Physiological Sciences, Skidmore College, Saratoga Springs, NY 12866, USA
| | - Elpidoforos S. Soteriades
- Healthcare Management Program, School of Economics and Management, Open University of Cyprus, Nicosia 2220, Cyprus
- Department of Environmental Health, Environmental and Occupational Medicine and Epidemiology (EOME), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lloyd Leach
- Department of Sport, Recreation and Exercise Science, Faculty of Community and Health Sciences, University of the Western Cape, Cape Town 7535, South Africa
| |
Collapse
|
20
|
Akowuah PK, Lema C, Rumbaut RE, Burns AR. A Low-Fat/Sucrose Diet Rich in Complex Carbohydrates Reverses High-Fat/Sucrose Diet-Induced Corneal Dysregulation. Int J Mol Sci 2023; 24:931. [PMID: 36674448 PMCID: PMC9865780 DOI: 10.3390/ijms24020931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
High-fat/sucrose diet feeding in mice causes loss of corneal nerve function and impairs corneal wound healing. While changing to a diet with a low fat/sugar composition and enrichments in complex carbohydrates mitigates the reduction in nerve function, it remains to be determined if it has an effect on corneal wound healing. In this study, 6-week-old C57BL/6 male mice were fed either a normal diet or a high-fat/sucrose diet for 20 weeks. A third group (diet reversal) was placed on a high-fat/sucrose diet for 10 weeks followed by a normal diet for an additional 10 weeks. A central corneal epithelial abrasion wound was created, and wound closure was monitored. Neutrophil and platelet recruitment was assessed by immunofluorescence microscopy. Mice fed the high-fat/sucrose diet-only had greater adiposity (p < 0.005) than normal diet-only fed mice; diet reversal markedly reduced adiposity. Following corneal abrasion, wound closure was delayed by ~6 h (p ≤ 0.01) and, at 30 h post-wounding, fewer neutrophils reached the wound center and fewer extravascular platelets were present at the limbus (p < 0.05). Diet restored normal wound closure and neutrophil and platelet influx in the injured cornea. These data suggest compositional changes to the diet may be an effective diet-based therapeutic strategy for maintaining or restoring corneal health.
Collapse
Affiliation(s)
| | - Carolina Lema
- College of Optometry, University of Houston, Houston, TX 77204, USA
| | - Rolando E. Rumbaut
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical, Houston, TX 77030, USA
| | - Alan R. Burns
- College of Optometry, University of Houston, Houston, TX 77204, USA
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
21
|
Quiroz-Aldave J, Durand-Vásquez M, Gamarra-Osorio E, Suarez-Rojas J, Jantine Roseboom P, Alcalá-Mendoza R, Coronado-Arroyo J, Zavaleta-Gutiérrez F, Concepción-Urteaga L, Concepción-Zavaleta M. Diabetic neuropathy: Past, present, and future. CASPIAN JOURNAL OF INTERNAL MEDICINE 2023; 14:153-169. [PMID: 37223297 PMCID: PMC10201131 DOI: 10.22088/cjim.14.2.153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/03/2022] [Accepted: 09/06/2022] [Indexed: 05/25/2023]
Abstract
Background A sedentary lifestyle and an unhealthy diet have considerably increased the incidence of diabetes mellitus worldwide in recent decades, which has generated a high rate of associated chronic complications. Methods A narrative review was performed in MEDLINE, EMBASES and SciELO databases, including 162 articles. Results Diabetic neuropathy (DN) is the most common of these complications, mainly producing two types of involvement: sensorimotor neuropathy, whose most common form is symmetric distal polyneuropathy, and autonomic neuropathies, affecting the cardiovascular, gastrointestinal, and urogenital system. Although hyperglycemia is the main metabolic alteration involved in its genesis, the presents of obesity, dyslipidemia, arterial hypertension, and smoking, play an additional role in its appearance. In the pathophysiology, three main phenomena stand out: oxidative stress, the formation of advanced glycosylation end-products, and microvasculature damage. Diagnosis is clinical, and it is recommended to use a 10 g monofilament and a 128 Hz tuning fork as screening tools. Glycemic control and non-pharmacological interventions constitute the mainstay of DN treatment, although there are currently investigations in antioxidant therapies, in addition to pain management. Conclusions Diabetes mellitus causes damage to peripheral nerves, being the most common form of this, distal symmetric polyneuropathy. Control of glycemia and comorbidities contribute to prevent, postpone, and reduce its severity. Pharmacological interventions are intended to relieve pain.
Collapse
Affiliation(s)
| | | | | | | | - Pela Jantine Roseboom
- Division of Emergency Medicine, Hospital Regional Docente de Trujillo, Trujillo, Peru
| | - Rosa Alcalá-Mendoza
- Division of Physical Medicine and Rehabilitation, Hospital Víctor Lazarte Echegaray, Trujillo, Peru
| | - Julia Coronado-Arroyo
- Division of Obstetrics and Gynecology, Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
| | | | | | | |
Collapse
|
22
|
Mázala-de-Oliveira T, Jannini de Sá YAP, Carvalho VDF. Impact of gut-peripheral nervous system axis on the development of diabetic neuropathy. Mem Inst Oswaldo Cruz 2023; 118:e220197. [PMID: 36946851 PMCID: PMC10027071 DOI: 10.1590/0074-02760220197] [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: 08/26/2022] [Accepted: 02/14/2023] [Indexed: 03/22/2023] Open
Abstract
Diabetes is a chronic metabolic disease caused by a reduction in the production and/or action of insulin, with consequent development of hyperglycemia. Diabetic patients, especially those who develop neuropathy, presented dysbiosis, with an increase in the proportion of pathogenic bacteria and a decrease in the butyrate-producing bacteria. Due to this dysbiosis, diabetic patients presented a weakness of the intestinal permeability barrier and high bacterial product translocation to the bloodstream, in parallel to a high circulating levels of pro-inflammatory cytokines such as TNF-α. In this context, we propose here that dysbiosis-induced increased systemic levels of bacterial products, like lipopolysaccharide (LPS), leads to an increase in the production of pro-inflammatory cytokines, including TNF-α, by Schwann cells and spinal cord of diabetics, being crucial for the development of neuropathy.
Collapse
Affiliation(s)
| | | | - Vinicius de Frias Carvalho
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brasil
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação, Rio de Janeiro, RJ, Brasil
- + Corresponding author:
| |
Collapse
|
23
|
Yang C, Zhao X, An X, Zhang Y, Sun W, Zhang Y, Duan Y, Kang X, Sun Y, Jiang L, Lian F. Axonal transport deficits in the pathogenesis of diabetic peripheral neuropathy. Front Endocrinol (Lausanne) 2023; 14:1136796. [PMID: 37056668 PMCID: PMC10086245 DOI: 10.3389/fendo.2023.1136796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a chronic and prevalent metabolic disease that gravely endangers human health and seriously affects the quality of life of hyperglycemic patients. More seriously, it can lead to amputation and neuropathic pain, imposing a severe financial burden on patients and the healthcare system. Even with strict glycemic control or pancreas transplantation, peripheral nerve damage is difficult to reverse. Most current treatment options for DPN can only treat the symptoms but not the underlying mechanism. Patients with long-term diabetes mellitus (DM) develop axonal transport dysfunction, which could be an important factor in causing or exacerbating DPN. This review explores the underlying mechanisms that may be related to axonal transport impairment and cytoskeletal changes caused by DM, and the relevance of the latter with the occurrence and progression of DPN, including nerve fiber loss, diminished nerve conduction velocity, and impaired nerve regeneration, and also predicts possible therapeutic strategies. Understanding the mechanisms of diabetic neuronal injury is essential to prevent the deterioration of DPN and to develop new therapeutic strategies. Timely and effective improvement of axonal transport impairment is particularly critical for the treatment of peripheral neuropathies.
Collapse
|
24
|
Yang HM, Kim J, Shin D, Kim JY, You J, Lee HC, Jang HD, Kim HS. Resistin impairs mitochondrial homeostasis via cyclase-associated protein 1-mediated fission, leading to obesity-induced metabolic diseases. Metabolism 2023; 138:155343. [PMID: 36356648 DOI: 10.1016/j.metabol.2022.155343] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE One of the suggested mechanisms of obesity-induced insulin resistance is mitochondrial dysfunction in target tissues such as skeletal muscle. In our study, we examined whether resistin, an adipokine associated with obesity-mediated insulin resistance, induced metabolic disorders by impairing mitochondrial homeostasis. METHODS The morphology and function of mitochondria of skeletal muscle were examined in resistin-knockout and humanized resistin mice that were subjected to high-fat diet for 3 months. Morphology was examined by transmission electron microscopy. Mitochondria bioenergetics of skeletal muscle were evaluated using a Seahorse XF96 analyzer. Human skeletal myoblasts were used for in vitro studies on signaling mechanisms in responses to resistin. RESULTS A high-fat diet in humanized resistin mice increased fragmented and shorter mitochondria in the skeletal muscle, whereas resistin-knockout mice had healthy normal mitochondria. In vitro studies showed that human resistin treatment impaired mitochondrial homeostasis by inducing mitochondrial fission, leading to a decrease in ATP production and mitochondrial dysfunction. Induction of mitochondrial fission by resistin was accompanied by increased formation of mitochondria-associated ER membranes (MAM). At the same time, resistin induced up-regulation of the protein kinase A (PKA) pathway. This activation of PKA induced phosphorylation of Drp1 at serine 616, leading to Drp1 activation and subsequent induction of mitochondrial fission. The key molecule that mediated human resistin-induced mitochondrial fission was adenylyl cyclase-associated protein 1 (CAP1), which was reported as a bona fide receptor for human resistin. Moreover, our newly developed biomimetic selective blocking peptide could repress human resistin-mediated mitochondrial dysfunction. High-fat diet-fed mice showed lower exercise capacity and higher insulin resistance, which was prevented by a novel peptide to block the binding of resistin to CAP1 or in the CAP1-knockdown mice. CONCLUSIONS Our study demonstrated that human resistin induces mitochondrial dysfunction by inducing abnormal mitochondrial fission. This result suggests that the resistin-CAP1 complex could be a potential therapeutic target for the treatment of obesity-related metabolic diseases such as diabetes and cardiometabolic diseases.
Collapse
Affiliation(s)
- Han-Mo Yang
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Joonoh Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dasom Shin
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ju-Young Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jihye You
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyun-Chae Lee
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyun-Duk Jang
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyo-Soo Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Department of Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Sánchez-Alegría K, Arias C. Functional consequences of brain exposure to saturated fatty acids: From energy metabolism and insulin resistance to neuronal damage. Endocrinol Diabetes Metab 2023; 6:e386. [PMID: 36321333 PMCID: PMC9836261 DOI: 10.1002/edm2.386] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Saturated fatty acids (FAs) are the main component of high-fat diets (HFDs), and high consumption has been associated with the development of insulin resistance, endoplasmic reticulum stress and mitochondrial dysfunction in neuronal cells. In particular, the reduction in neuronal insulin signaling seems to underlie the development of cognitive impairments and has been considered a risk factor for Alzheimer's disease (AD). METHODS This review summarized and critically analyzed the research that has impacted the field of saturated FA metabolism in neurons. RESULTS We reviewed the mechanisms for free FA transport from the systemic circulation to the brain and how they impact neuronal metabolism. Finally, we focused on the molecular and the physiopathological consequences of brain exposure to the most abundant FA in the HFD, palmitic acid (PA). CONCLUSION Understanding the mechanisms that lead to metabolic alterations in neurons induced by saturated FAs could help to develop several strategies for the prevention and treatment of cognitive impairment associated with insulin resistance, metabolic syndrome, or type II diabetes.
Collapse
Affiliation(s)
- Karina Sánchez-Alegría
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
26
|
Canta A, Carozzi VA, Chiorazzi A, Meregalli C, Oggioni N, Rodriguez-Menendez V, Sala B, Melcangi RC, Giatti S, Lombardi R, Bianchi R, Marmiroli P, Cavaletti G. Multimodal Comparison of Diabetic Neuropathy in Aged Streptozotocin-Treated Sprague-Dawley and Zucker Diabetic Fatty Rats. Biomedicines 2022; 11:20. [PMID: 36672528 PMCID: PMC9855818 DOI: 10.3390/biomedicines11010020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/02/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
The development and progression of diabetic polyneuropathy (DPN) are due to multiple mechanisms. The creation of reliable animal models of DPN has been challenging and this issue has not yet been solved. However, despite some recognized differences from humans, most of the current knowledge on the pathogenesis of DPN relies on results achieved using rodent animal models. The simplest experimental DPN model reproduces type 1 diabetes, induced by massive chemical destruction of pancreatic beta cells with streptozotocin (STZ). Spontaneous/transgenic models of diabetes are less frequently used, mostly because they are less predictable in clinical course, more expensive, and require a variable time to achieve homogeneous metabolic conditions. Among them, Zucker diabetic fatty (ZDF) rats represent a typical type 2 diabetes model. Both STZ-induced and ZDF rats have been extensively used, but only very few studies have compared the long-term similarities and differences existing between these two models. Moreover, inconsistencies have been reported regarding several aspects of short-term in vivo studies using these models. In this study, we compared the long-term course of DPN in STZ-treated Sprague-Dawley and ZDF rats with a multimodal set of readout measures.
Collapse
Affiliation(s)
- Annalisa Canta
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Valentina A. Carozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Alessia Chiorazzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Cristina Meregalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Norberto Oggioni
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Virginia Rodriguez-Menendez
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Barbara Sala
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20100 Milano, Italy
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20100 Milano, Italy
| | - Raffaella Lombardi
- Neuroalgology Unit, IRCCS Fondazione Istituto Neurologico “Carlo Besta”, 20100 Milano, Italy
| | - Roberto Bianchi
- Neuroalgology Unit, IRCCS Fondazione Istituto Neurologico “Carlo Besta”, 20100 Milano, Italy
| | - Paola Marmiroli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
27
|
cGAS-STING activation contributes to podocyte injury in diabetic kidney disease. iScience 2022; 25:105145. [PMID: 36176590 PMCID: PMC9513272 DOI: 10.1016/j.isci.2022.105145] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/25/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal diseases. DKD does not have efficacious treatment. The cGAS-STING pathway is activated in podocytes at the early stage of kidney dysfunction, which is associated with the activation of STING downstream effectors TBK1 and NF-κB but not IRF3. Lipotoxicity induces mitochondrial damage and mtDNA leakage to the cytosol through Bcl-2 associated X protein (BAX) in podocytes. BAX-mediated mtDNA cytosolic leakage can activate the cGAS-STING pathway in the absence of lipotoxicity and is sufficient to cause podocyte injury. Depletion of cytosolic mtDNA, genetic STING knockdown, or pharmacological inhibition of STING or TBK1 alleviates podocyte injury and improves renal functions in cultured podocytes or mouse models of diabetes and obesity. These results suggest that the mtDNA-cGAS-STING pathway promotes podocyte injury and is a potential therapeutic target for DKD or other obesity-related kidney diseases.
Collapse
|
28
|
Cardiovascular Disease Risk Factors, Musculoskeletal Health, Physical Fitness, and Occupational Performance in Firefighters: A Narrative Review. JOURNAL OF ENVIRONMENTAL AND PUBLIC HEALTH 2022; 2022:7346408. [PMID: 36193397 PMCID: PMC9526561 DOI: 10.1155/2022/7346408] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/20/2022] [Indexed: 11/18/2022]
Abstract
Introduction Firefighting is a strenuous occupation that requires firefighters to be in peak physical condition. However, many firefighters have risk factors for cardiovascular disease, impaired musculoskeletal health, and are not physically fit for duty, which all negatively impact their occupational performance. Therefore, the aim of this review is to determine the relationship between cardiovascular disease risk factors, musculoskeletal health, physical fitness, and occupational performance in firefighters. Methods The electronic databases PubMed, SCOPUS, and Web of Science were searched online via the library portal of the University of the Western Cape. Publications and grey literature between the years 2000 to present were used. In total, 2607 articles were identified; after the removal of duplicates 1188 articles were then screened, and were excluded for not meeting initial screening criteria. The remaining 209 full-text articles were screened based on the inclusion and exclusion criteria, where 163 articles were excluded. Only studies that were quantitative were included. This left 46 articles that were then finally included in the current narrative review. Results The current literature indicated that significant relationships existed between cardiovascular risk factors, musculoskeletal health, physical fitness, and occupational performance. The results indicated firefighters who were aged, obese, physically inactive, cigarette smokers, and unfit were at the highest risk for cardiovascular and musculoskeletal health complications, and unsatisfactory occupational performance. Musculoskeletal health complications significantly affected occupational performance and work ability and were related to physical fitness of firefighters. Most cardiovascular risk factors were related to physical fitness, and all physical fitness parameters were related to occupational performance in firefighters. Conclusion The overwhelming evidence in the current review established that physical fitness is related to occupational performance. However, the relationship between cardiovascular risk factors and musculoskeletal health in relation to occupational performance is less clear and still understudied. Significant gaps remain in the literature.
Collapse
|
29
|
Rumora AE, Kim B, Feldman EL. A Role for Fatty Acids in Peripheral Neuropathy Associated with Type 2 Diabetes and Prediabetes. Antioxid Redox Signal 2022; 37:560-577. [PMID: 35152728 PMCID: PMC9499450 DOI: 10.1089/ars.2021.0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 11/12/2022]
Abstract
Significance: As the global prevalence of diabetes rises, diabetic complications are also increasing at an alarming rate. Peripheral neuropathy (PN) is the most prevalent complication of diabetes and prediabetes, and is characterized by progressive sensory loss resulting from nerve damage. While hyperglycemia is the major risk factor for PN in type 1 diabetes (T1D), the metabolic syndrome (MetS) underlies the onset and progression of PN in type 2 diabetes (T2D) and prediabetes. Recent Advances: Recent reports show that dyslipidemia, a MetS component, is strongly associated with PN in T2D and prediabetes. Dyslipidemia is characterized by an abnormal plasma lipid profile with uncontrolled lipid levels, and both clinical and preclinical studies implicate a role for dietary fatty acids (FAs) in PN pathogenesis. Molecular studies further show that saturated and unsaturated FAs differentially regulate the nerve lipid profile and nerve function. Critical Issues: We first review the properties of FAs and the neuroanatomy of the peripheral nervous system (PNS). Second, we discuss clinical and preclinical studies that implicate the involvement of FAs in PN. Third, we summarize the potential effects of FAs on nerve function and lipid metabolism within the peripheral nerves, sensory neurons, and Schwann cells. Future Directions: Future directions will focus on identifying molecular pathways in T2D and prediabetes that are modulated by FAs in PN. Determining pathophysiological mechanisms that underlie the injurious effects of saturated FAs and beneficial properties of unsaturated FAs will provide mechanistic targets for developing new targeted therapies to treat PN associated with T2D and prediabetes. Antioxid. Redox Signal. 37, 560-577.
Collapse
Affiliation(s)
- Amy E. Rumora
- Department of Neurology, Columbia University, New York, New York, USA
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Bhumsoo Kim
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
30
|
Rumora AE, Guo K, Hinder LM, O’Brien PD, Hayes JM, Hur J, Feldman EL. A High-Fat Diet Disrupts Nerve Lipids and Mitochondrial Function in Murine Models of Neuropathy. Front Physiol 2022; 13:921942. [PMID: 36072849 PMCID: PMC9441493 DOI: 10.3389/fphys.2022.921942] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
As the prevalence of prediabetes and type 2 diabetes (T2D) continues to increase worldwide, accompanying complications are also on the rise. The most prevalent complication, peripheral neuropathy (PN), is a complex process which remains incompletely understood. Dyslipidemia is an emerging risk factor for PN in both prediabetes and T2D, suggesting that excess lipids damage peripheral nerves; however, the precise lipid changes that contribute to PN are unknown. To identify specific lipid changes associated with PN, we conducted an untargeted lipidomics analysis comparing the effect of high-fat diet (HFD) feeding on lipids in the plasma, liver, and peripheral nerve from three strains of mice (BL6, BTBR, and BKS). HFD feeding triggered distinct strain- and tissue-specific lipid changes, which correlated with PN in BL6 mice versus less robust murine models of metabolic dysfunction and PN (BTBR and BKS mice). The BL6 mice showed significant changes in neutral lipids, phospholipids, lysophospholipids, and plasmalogens within the nerve. Sphingomyelin (SM) and lysophosphatidylethanolamine (LPE) were two lipid species that were unique to HFD BL6 sciatic nerve compared to other strains (BTBR and BKS). Plasma and liver lipids were significantly altered in all murine strains fed a HFD independent of PN status, suggesting that nerve-specific lipid changes contribute to PN pathogenesis. Many of the identified lipids affect mitochondrial function and mitochondrial bioenergetics, which were significantly impaired in ex vivo sural nerve and dorsal root ganglion sensory neurons. Collectively, our data show that consuming a HFD dysregulates the nerve lipidome and mitochondrial function, which may contribute to PN in prediabetes.
Collapse
Affiliation(s)
- Amy E. Rumora
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Department of Neurology, Columbia University, New York, NY, United States
| | - Kai Guo
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Lucy M. Hinder
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Phillipe D. O’Brien
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Junguk Hur
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
31
|
Haddad M, Eid S, Harb F, Massry MEL, Azar S, Sauleau EA, Eid AA. Activation of 20-HETE Synthase Triggers Oxidative Injury and Peripheral Nerve Damage in Type 2 Diabetic Mice. THE JOURNAL OF PAIN 2022; 23:1371-1388. [PMID: 35339661 DOI: 10.1016/j.jpain.2022.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/26/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022]
Abstract
Diabetic Peripheral Neuropathy (DPN), highly prevalent among patients with diabetes, is characterized by peripheral nerve dysfunction. Reactive Oxygen Species (ROS) overproduction has been suggested to orchestrate diabetic complications including DPN. Untargeted antioxidant therapy has exhibited limited efficacy, highlighting a critical need to explore ROS sources altered in a cell-specific manner in DPN. Cytochromes P450 (CYP) enzymes are prominent sources of ROS. Particularly, the 20-HETE synthase, CYP4A, is reported to mediate diabetes-induced renal, retinal, and cardiovascular injuries. This work investigates the role of CYP4A/20-HETE in DPN and their mechanisms of action. Non-obese type 2 Diabetic mice (MKR) were used and treated with a CYP4A-inhibitor (HET0016) or AMPK-activator (Metformin). Peripheral nerves of MKR mice reflect increased CYP4A and 20-HETE levels, concurrent with altered myelin proteins and sensorimotor deficits. This was associated with increased ROS production and altered Beclin-1 and LC3 protein levels, indicative of disrupted autophagic responses in tandem with AMPK inactivation. AMPK activation via Metformin restored nerve integrity, reduced ROS production, and regulated autophagy. Interestingly, similar outcomes were revealed upon HET0016 treatment whereby ROS production, autophagic responses, and AMPK signaling were normalized in diabetic mice. Altogether, the results highlight hyperglycemia-mediated oxidative injury in DPN through a novel CYP4A/20-HETE/AMPK pathological axis. PERSPECTIVE: To our knowledge, this is the first study to highlight the role of CYPs/20-HETE-induced oxidative injury in the pathogenesis of diabetic peripheral neuropathy. Targeting the identified pathological axis CYP4A/20-HETE/AMPK may be of clinical potential in predicting and alleviating peripheral nerve injury in patients with Type 2 Diabetes Mellitus.
Collapse
Affiliation(s)
- Mary Haddad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; Department of Biostatistics, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7357 ICube, University of Strasbourg, Strasbourg, France
| | - Stéphanie Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Frederic Harb
- Department of Life and Earth Sciences, Faculty of Sciences, Lebanese University, Fanar, Lebanon
| | - Mohamed E L Massry
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Sami Azar
- Department of Internal Medicine, Division of Diabetes and Endocrinology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut, Beirut, Lebanon
| | - Erik-Andre Sauleau
- Department of Biostatistics, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7357 ICube, University of Strasbourg, Strasbourg, France
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
32
|
Ding Y, Wang L, Sun J, Shi Y, Li G, Luan X, Zheng G, Zhang G. Remnant Cholesterol and Dyslipidemia Are Risk Factors for Guillain–Barré Syndrome and Severe Guillain–Barré Syndrome by Promoting Monocyte Activation. Front Immunol 2022; 13:946825. [PMID: 35911688 PMCID: PMC9326451 DOI: 10.3389/fimmu.2022.946825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundGuillain–Barré syndrome (GBS) is the most common severe acute paralytic neuropathy, with a mortality rate of 5% and permanent sequelae rate of 10%. Currently, the cause of GBS remains unclear. Therefore, we sought to determine potential predictors for GBS and its severity.MethodsA case–control study was performed at Tiantan Hospital in Beijing from January 2017 to December 2021. Laboratory and clinical characteristics were assessed in recruited GBS patients and healthy control individuals (matched by sex and age). The potential risk factors for GBS and severe GBS were assessed using a logistic regression analysis. The mRNA levels of toll-like receptor 4 (TLR4), toll-like receptor 2 (TLR2) and nuclear factor κB (NF-κB) in GBS patients and control PBMCs were detected by fluorescence quantitative PCR. THP-1 cells were costimulated with LPS and free cholesterol to demonstrate the effect of free cholesterol on monocyte activation.ResultsA total of 147 GBS patients and 153 healthy individuals were included in the study. Logistic regression analyses showed that preceding infection, alcohol consumption, remnant cholesterol, homocysteine and the dyslipidemia index were correlated with a higher risk of GBS. In contrast, increased HDL cholesterol was correlated with a lower risk of GBS. Moreover, remnant cholesterol and the dyslipidemia index were significantly correlated with severe GBS. The mRNA levels of TLR4, TLR2 and NF-κB in the PBMCs of GBS patients were significantly higher than those of healthy individuals. LPS activated THP-1 cells, and free cholesterol treatment increased the expression of TLR4, TLR2, NF-κB and IL-1β mRNA in LPS-activated THP-1 cells.ConclusionDyslipidemia was correlated with the risk of GBS and severe GBS. Remnant cholesterol may promote the activation of monocytes in GBS patients. It may be valuable to control lipid levels in the prevention of GBS and severe GBS.
Collapse
Affiliation(s)
- Yaowei Ding
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lijuan Wang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jialu Sun
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yijun Shi
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guoge Li
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xin Luan
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guanghui Zheng
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Medical Products Administration (NMPA) Key Laboratory for Quality Control of In Vitro Diagnostics, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guojun Zhang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Medical Products Administration (NMPA) Key Laboratory for Quality Control of In Vitro Diagnostics, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Guojun Zhang,
| |
Collapse
|
33
|
Elshareif N, Gavini CK, Mansuy-Aubert V. LXR agonist modifies neuronal lipid homeostasis and decreases PGD2 in the dorsal root ganglia in western diet-fed mice. Sci Rep 2022; 12:10754. [PMID: 35750708 PMCID: PMC9232502 DOI: 10.1038/s41598-022-14604-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/09/2022] [Indexed: 11/09/2022] Open
Abstract
The prevalence of peripheral neuropathy is high in diabetic and overweight populations. Chronic neuropathic pain, a symptom of peripheral neuropathy, is a major disabling symptom that leads to a poor quality of life. Glucose management for diabetic and prediabetic individuals often fail to reduce or improve pain symptoms, therefore, exploring other mechanisms is necessary to identify effective treatments. A large body of evidence suggest that lipid signaling may be a viable target for management of peripheral neuropathy in obese individuals. The nuclear transcription factors, Liver X Receptors (LXR), are known regulators of lipid homeostasis, phospholipid remodeling, and inflammation. Notably, the activation of LXR using the synthetic agonist GW3965, delayed western diet (WD)-induced allodynia in rodents. To further understand the neurobiology underlying the effect of LXR, we used translating ribosome affinity purification and evaluated translatomic changes in the sensory neurons of WD-fed mice treated with the LXR agonist GW3965. We also observed that GW3965 decreased prostaglandin levels and decreased free fatty acid content, while increasing lysophosphatidylcholine, phosphatidylcholine, and cholesterol ester species in the sensory neurons of the dorsal root ganglia (DRG). These data suggest novel downstream interplaying mechanisms that modifies DRG neuronal lipid following GW3965 treatment.
Collapse
Affiliation(s)
- Nadia Elshareif
- Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Chaitanya K Gavini
- Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Virginie Mansuy-Aubert
- Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, 60153, USA.
| |
Collapse
|
34
|
Hou L, Hu C, Ji L, Wang Q, Liang M. The Mitochondrial tRNA Phe 625G>A Mutation in Three Han Chinese Families With Cholecystolithiasis. Front Genet 2022; 13:814729. [PMID: 35719381 PMCID: PMC9198646 DOI: 10.3389/fgene.2022.814729] [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: 11/18/2021] [Accepted: 04/19/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we assessed three Chinese families with inherited cholecystolithiasis and conducted the clinical, genetic, and molecular characterization of these subjects. Eight of eighteen matrilineal relatives had a clinical phenotype in these three families. Sequence analysis of complete mitochondrial genomes in these probands identified the homoplasmic tRNAPhe 625 G > A mutation and distinct sets of mtDNA polymorphisms belonging to haplogroups H2, F4b, and M10a. The 625G > A mutation disturbed the classic G-C base-pairings at a highly conserved position 49 in the T-stem of mitochondrial tRNAs. Molecular dynamics simulation showed that the structure of tRNAphe with 625 G > A mutation was noticeably remodeled while compared with the isoform of the wild type. The occurrence of tRNAPhe 625 G > A mutation in these various genetically unrelated subjects strongly indicates that this mutation is involved in the pathogenesis of cholecystolithiasis. This is the first evidence that tRNA mutations are associated with cholecystolithiasis, and it provided more insights into the genetic mechanism of cholecystolithiasis.
Collapse
Affiliation(s)
- Lingling Hou
- Department of Medical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cuifang Hu
- Attardi Institute of Mitochondrial Biomedicine, Wenzhou Medical University, Wenzhou, China
| | - Lili Ji
- Department of Medical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiongdan Wang
- Department of Medical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Min Liang
- Department of Medical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Attardi Institute of Mitochondrial Biomedicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Hukportie DN, Li F, Zhou R, Zheng J, Wu X, Zou M, Wu X. Lipid variability and risk of microvascular complications in Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial: A post hoc analysis. J Diabetes 2022; 14:365-376. [PMID: 35668633 PMCID: PMC9366577 DOI: 10.1111/1753-0407.13273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Greater lipid variability may cause adverse health events among diabetic patients. We aimed to examine the effect of lipid variability on the risk of diabetic microvascular outcomes among type 2 diabetes mellitus patients. METHODS We assessed the association between visit-to-visit variability (measured by variability independent of mean) in high-density lipoprotein (HDL) cholesterol, low-density lipoprotein-cholesterol (LDL), triglyceride, and remnant cholesterol (RC) measurements among participants involved in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study and the risk of incident microvascular outcomes, including nephropathy, neuropathy, and retinopathy. Cox proportional hazards models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs), adjusted for potential confounders. RESULTS There were 2400, 2470, and 2468 cases of nephropathy, neuropathy, and retinopathy during a follow-up period of 22 600, 21 542, and 26 701 person-years, respectively. Higher levels of HDL, triglyceride, and RC variability were associated with an increased risk of incident nephropathy and neuropathy. Compared with the lowest quartile, the fully adjusted HRs (95% CI) for the highest quartile of HDL, triglyceride, and RC variability for nephropathy risk were 1.57 (1.22, 2.01), 1.50 (1.18, 1.92), and 1.40 (1.09, 1.80), respectively; and for neuropathy, the corresponding risks were 1.36 (1.05, 1.75), 1.47 (1.14, 1.91), and 1.35 (1.04, 1.74), respectively. Null association was observed between LDL variability and all microvascular complications. Additionally, all associations of variability in the other lipids with retinopathy risk were null. CONCLUSION Among individuals with type 2 diabetes mellitus, HDL, triglyceride, and RC variability were associated with increased risks of nephropathy and neuropathy but not retinopathy. TRIAL REGISTRATION ClinicalTrials.gov., no. NCT00000620.
Collapse
Affiliation(s)
- Daniel Nyarko Hukportie
- Department of Epidemiology, School of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Fu‐Rong Li
- Department of Epidemiology, School of Public HealthSouthern Medical UniversityGuangzhouChina
- School of Public Health and Emergency ManagementSouthern University of Science and TechnologyShenzhenChina
| | - Rui Zhou
- Department of Epidemiology, School of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Jia‐Zhen Zheng
- Department of Epidemiology, School of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Xiao‐Xiang Wu
- Department of General Surgery157 Hospital, General Hospital of Guangzhou Military CommandGuangzhouChina
| | - Meng‐Chen Zou
- Department of Endocrinology and Metabolism, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xian‐Bo Wu
- Department of Epidemiology, School of Public HealthSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
36
|
Babetto E, Beirowski B. Of axons that struggle to make ends meet: Linking axonal bioenergetic failure to programmed axon degeneration. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148545. [PMID: 35339437 DOI: 10.1016/j.bbabio.2022.148545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 02/07/2023]
Abstract
Axons are the long, fragile, and energy-hungry projections of neurons that are challenging to sustain. Together with their associated glia, they form the bulk of the neuronal network. Pathological axon degeneration (pAxD) is a driver of irreversible neurological disability in a host of neurodegenerative conditions. Halting pAxD is therefore an attractive therapeutic strategy. Here we review recent work demonstrating that pAxD is regulated by an auto-destruction program that revolves around axonal bioenergetics. We then focus on the emerging concept that axonal and glial energy metabolism are intertwined. We anticipate that these discoveries will encourage the pursuit of new treatment strategies for neurodegeneration.
Collapse
Affiliation(s)
- Elisabetta Babetto
- Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA.
| | - Bogdan Beirowski
- Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
37
|
Chandrasekaran K, Najimi N, Sagi AR, Yarlagadda S, Salimian M, Arvas MI, Hedayat AF, Kevas Y, Kadakia A, Russell JW. NAD + Precursors Repair Mitochondrial Function in Diabetes and Prevent Experimental Diabetic Neuropathy. Int J Mol Sci 2022; 23:4887. [PMID: 35563288 PMCID: PMC9102948 DOI: 10.3390/ijms23094887] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/18/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
Axon degeneration in diabetic peripheral neuropathy (DPN) is associated with impaired NAD+ metabolism. We tested whether the administration of NAD+ precursors, nicotinamide mononucleotide (NMN) or nicotinamide riboside (NR), prevents DPN in models of Type 1 and Type 2 diabetes. NMN was administered to streptozotocin (STZ)-induced diabetic rats and STZ-induced diabetic mice by intraperitoneal injection at 50 or 100 mg/kg on alternate days for 2 months. mice The were fed with a high fat diet (HFD) for 2 months with or without added NR at 150 or 300 mg/kg for 2 months. The administration of NMN to STZ-induced diabetic rats or mice or dietary addition of NR to HFD-fed mice improved sensory function, normalized sciatic and tail nerve conduction velocities, and prevented loss of intraepidermal nerve fibers in skin samples from the hind-paw. In adult dorsal root ganglion (DRG) neurons isolated from HFD-fed mice, there was a decrease in NAD+ levels and mitochondrial maximum reserve capacity. These impairments were normalized in isolated DRG neurons from NR-treated mice. The results indicate that the correction of NAD+ depletion in DRG may be sufficient to prevent DPN but does not significantly affect glucose tolerance, insulin levels, or insulin resistance.
Collapse
Affiliation(s)
- Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Neda Najimi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Avinash R. Sagi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Sushuma Yarlagadda
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Mohammad Salimian
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Muhammed Ikbal Arvas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Ahmad F. Hedayat
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Anand Kadakia
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - James W. Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (A.R.S.); (S.Y.); (M.S.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA
- CAMC Institute for Academic Medicine, 415 Morris Street Suite 300, Charleston, WV 25301, USA
| |
Collapse
|
38
|
Guo K, Savelieff MG, Rumora AE, Alakwaa FM, Callaghan BC, Hur J, Feldman EL. Plasma Metabolomics and Lipidomics Differentiate Obese Individuals by Peripheral Neuropathy Status. J Clin Endocrinol Metab 2022; 107:1091-1109. [PMID: 34878536 PMCID: PMC8947234 DOI: 10.1210/clinem/dgab844] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Indexed: 12/19/2022]
Abstract
CONTEXT Peripheral neuropathy (PN) is a frequent prediabetes and type 2 diabetes (T2D) complication. Multiple clinical studies reveal that obesity and dyslipidemia can also drive PN progression, independent of glycemia, suggesting a complex interplay of specific metabolite and/or lipid species may underlie PN. OBJECTIVE This work aimed to identify the plasma metabolomics and lipidomics signature that underlies PN in an observational study of a sample of individuals with average class 3 obesity. METHODS We performed plasma global metabolomics and targeted lipidomics on obese participants with (n = 44) and without PN (n = 44), matched for glycemic status, vs lean nonneuropathic controls (n = 43). We analyzed data by Wilcoxon, logistic regression, partial least squares-discriminant analysis, and group-lasso to identify differential metabolites and lipids by obesity and PN status. We also conducted subanalysis by prediabetes and T2D status. RESULTS Lean vs obese comparisons, regardless of PN status, identified the most significant differences in gamma-glutamyl and branched-chain amino acid metabolism from metabolomics analysis and triacylglycerols from lipidomics. Stratification by PN status within obese individuals identified differences in polyamine, purine biosynthesis, and benzoate metabolism. Lipidomics found diacylglycerols as the most significant subpathway distinguishing obese individuals by PN status, with additional contributions from phosphatidylcholines, sphingomyelins, ceramides, and dihydroceramides. Stratifying the obese group by glycemic status did not affect discrimination by PN status. CONCLUSION Obesity may be as strong a PN driver as prediabetes or T2D in a sample of individuals with average class 3 obesity, at least by plasma metabolomics and lipidomics profile. Metabolic and complex lipid pathways can differentiate obese individuals with and without PN, independent of glycemic status.
Collapse
Affiliation(s)
- Kai Guo
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Masha G Savelieff
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy E Rumora
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Fadhl M Alakwaa
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Brian C Callaghan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
- Correspondence: Eva L. Feldman, MD, PhD, Department of Neurology, University of Michigan 5017 AAT-BSRB, 109 Zina Pitcher Pl, Ann Arbor, MI 48109-0588, USA.
| |
Collapse
|
39
|
Sivakumar PM, Prabhakar PK, Cetinel S, R N, Prabhawathi V. Molecular Insights on the Therapeutic Effect of Selected Flavonoids on Diabetic Neuropathy. Mini Rev Med Chem 2022; 22:1828-1846. [PMID: 35264089 DOI: 10.2174/1389557522666220309140855] [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: 08/26/2021] [Revised: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 11/22/2022]
Abstract
One of the common clinical complications of diabetes is diabetic neuropathy affecting the nervous system. Painful diabetic neuropathy is widespread and highly prevalent. At least 50% of diabetes patients develop diabetic neuropathy eventually. The four main types of diabetic neuropathy are peripheral neuropathy, autonomic neuropathy, proximal neuropathy (diabetic polyradiculopathy), and mononeuropathy (Focal neuropathy). Glucose control remains the common therapy for diabetic neuropathy due to limited knowledge on early biomarkers that are expressed during nerve damage, thereby limiting the cure through pharmacotherapy. Glucose control dramatically reduces the onset of neuropathy in type 1 diabetes but proves less effective in type 2 diabetes. Therefore, the focus is on various herbal remedies for prevention and treatment. There is numerous research on the use of anticonvulsants and antidepressants for the management of pain in diabetic neuropathy. Extensive research is being done on natural products including the isolation of pure compounds like flavonoids from plants and their effect on diabetic neuropathy. This review focuses on the use of an important of flavonoids such as flavanols (e.g., quercetin, rutin, kaempferol, and isorhamnetin), flavanones (e.g., hesperidin, naringenin and c,lass eriodictyol), and flavones (e.g., apigenin, luteolin, tangeretin, chrysin, and diosmin) for the prevention and treatment of diabetic neuropathy. The mechanisms of action of flavonoids against diabetic neuropathy by their antioxidant, anti-inflammation, anti-glycation properties, etc. are also covered in this review article.
Collapse
Affiliation(s)
- Ponnurengam Malliappan Sivakumar
- Center for Molecular Biology, Institute of Research and Development, Duy Tan University, 03 Quang Trung, Da Nang, Vietnam.
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
| | | | - Sibel Cetinel
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey.
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul 34956, Turkey
| | - Neelakandan R
- Department of Textile Technology, Anna University, Chennai, Tamil Nadu, India
| | - Veluchamy Prabhawathi
- Multidisciplinary Research Unit, Coimbatore Medical College, Coimbatore - 641014, Tamil Nadu, India
| |
Collapse
|
40
|
Yuan P, Song F, Zhu P, Fan K, Liao Q, Huang L, Liu Z. Poly (ADP-ribose) polymerase 1-mediated defective mitophagy contributes to painful diabetic neuropathy in the db/db model. J Neurochem 2022; 162:276-289. [PMID: 35263449 DOI: 10.1111/jnc.15606] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/29/2022]
Abstract
Studies have shown that poly (ADP-ribose) polymerase 1 (PARP1) was involved in the pathological process of diabetes. Mitophagy is widely acknowledged to be a key regulatory process in maintaining reactive oxygen species homeostasis via lysosome degradation of damaged mitochondria. However, the regulatory role of PARP1 in mitophagy-related mitochondrial oxidative injury and progression of painful diabetic neuropathy (PDN) is unclear. In this study, we studied the in vitro and in vivo mechanisms of PARP1-mediated mitophagy blockade in a leptin gene-mutation (db/db) mouse model of PDN. Db/db mice models of PDN were established by assessing the sciatic nerve conduction velocity (SNCV), mechanical withdrawal threshold (MWT), and thermal withdrawal latency (TWL). The results showed that PARP1 activity and mitochondrial injury of dorsal root ganglion (DRG) neurons were increased, and mitophagy was impaired in PDN mice. PARP1 was found to mediate the impairment of mitophagy in DRG neurons isolated from PDN mice. PARP1 inhibitors (PJ34 or AG14361) attenuated diabetes-induced peripheral nerve hyperalgesia, restored DRG neuron mitophagy function and decreased mitochondrial oxidative injury. Mitophagy impairment induced by lysosome deacidificant (DC661) aggravated diabetes-induced DRG neuron mitochondrial oxidative stress and injury. Taken together, our data revealed that PARP1 induced defective mitophagy of DRG neurons is a key mechanism in diabetes-induced peripheral neuropathic injury. Inhibition of PARP1 and restoration of mitophagy function are potential therapeutic targets for PDN.
Collapse
Affiliation(s)
- Pengfei Yuan
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China
| | - Fuhu Song
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Pian Zhu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China
| | - Keke Fan
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China
| | - Qinming Liao
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Lijin Huang
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Zhongjie Liu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Industrial Avenue Central 253, Guangzhou, 510282, Guangdong Province, China
| |
Collapse
|
41
|
Thyroid Hormones, Peripheral White Blood Count, and Dose of Basal Insulin Are Associated with Changes in Nerve Conduction Studies in Adolescents with Type 1 Diabetes. Metabolites 2021; 11:metabo11110795. [PMID: 34822453 PMCID: PMC8619894 DOI: 10.3390/metabo11110795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes (T1D) in the child population is the third most common chronic disease. Diabetic peripheral neuropathy (DPN) is a very disabling and silently developing complication. This prospective, observational study enrolled 182 (93 girls) patients with T1D, aged 16.5-18 years. The aim of the study was to assess the correlation between factors of diabetes metabolic control, blood count, thyroid hormones, thyroid-stimulating hormone (TSH), level of cortisol, vitamin D3, metabolic factors, demographic data, and nerve conduction study (NCS) parameters. We revealed that in multivariate regression models for almost all NCS parameters, beside height and diabetes duration, significant factors were basal insulin dose per kilogram of weight (BID/kg), body mass index (BMI), and thyroid hormones. For conduction velocities of the motor nerves, mean HbA1c exists in models. In all models for all NCS parameters there exists at least one parameter of peripheral white blood cell counts (predominantly monocytes). There is a significant influence of thyroid hormones, peripheral blood white cells count, and BID per weight on parameters of NCS. It is essential to take care of the proper insulin dose per weight of patients and the adequate proportion of basal to prandial insulin.
Collapse
|
42
|
Mou Y, Dein J, Chen Z, Jagdale M, Li XJ. MFN2 Deficiency Impairs Mitochondrial Transport and Downregulates Motor Protein Expression in Human Spinal Motor Neurons. Front Mol Neurosci 2021; 14:727552. [PMID: 34602978 PMCID: PMC8482798 DOI: 10.3389/fnmol.2021.727552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/26/2021] [Indexed: 11/13/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is one of the most common genetically inherited neurological disorders and CMT type 2A (CMT 2A) is caused by dominant mutations in the mitofusin-2 (MFN2) gene. MFN2 is located in the outer mitochondrial membrane and is a mediator of mitochondrial fusion, with an essential role in maintaining normal neuronal functions. Although loss of MFN2 induces axonal neuropathy, the detailed mechanism by which MFN2 deficiency results in axonal degeneration of human spinal motor neurons remains largely unknown. In this study, we generated MFN2-knockdown human embryonic stem cell (hESC) lines using lentivirus expressing MFN2 short hairpin RNA (shRNA). Using these hESC lines, we found that MFN2 loss did not affect spinal motor neuron differentiation from hESCs but resulted in mitochondrial fragmentation and dysfunction as determined by live-cell imaging. Notably, MFN2-knockodwn spinal motor neurons exhibited CMT2A disease-related phenotypes, including extensive perikaryal inclusions of phosphorylated neurofilament heavy chain (pNfH), frequent axonal swellings, and increased pNfH levels in long-term cultures. Importantly, MFN2 deficit impaired anterograde and retrograde mitochondrial transport within axons, and reduced the mRNA and protein levels of kinesin and dynein, indicating the interfered motor protein expression induced by MFN2 deficiency. Our results reveal that MFN2 knockdown induced axonal degeneration of spinal motor neurons and defects in mitochondrial morphology and function. The impaired mitochondrial transport in MFN2-knockdown spinal motor neurons is mediated, at least partially, by the altered motor proteins, providing potential therapeutic targets for rescuing axonal degeneration of spinal motor neurons in CMT2A disease.
Collapse
Affiliation(s)
- Yongchao Mou
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, United States.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Joshua Dein
- MD Program, University of Illinois College of Medicine Rockford, Rockford, IL, United States
| | - Zhenyu Chen
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, United States.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Mrunali Jagdale
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, United States.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Xue-Jun Li
- Department of Biomedical Sciences, University of Illinois College of Medicine Rockford, Rockford, IL, United States.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
43
|
Poitras TM, Munchrath E, Zochodne DW. Neurobiological Opportunities in Diabetic Polyneuropathy. Neurotherapeutics 2021; 18:2303-2323. [PMID: 34935118 PMCID: PMC8804062 DOI: 10.1007/s13311-021-01138-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 12/29/2022] Open
Abstract
This review highlights a selection of potential translational directions for the treatment of diabetic polyneuropathy (DPN) currently irreversible and without approved interventions beyond pain management. The list does not include all diabetic targets that have been generated over several decades of research but focuses on newer work. The emphasis is firstly on approaches that support the viability and growth of peripheral neurons and their ability to withstand a barrage of diabetic alterations. We include a section describing Schwann cell targets and finally how mitochondrial damage has been a common element in discussing neuropathic damage. Most of the molecules and pathways described here have not yet reached clinical trials, but many trials have been negative to date. Nonetheless, these failures clear the pathway for new thoughts over reversing DPN.
Collapse
Affiliation(s)
- Trevor M Poitras
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Easton Munchrath
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Douglas W Zochodne
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada.
| |
Collapse
|
44
|
Baghbani E, Noorolyai S, Shanehbandi D, Mokhtarzadeh A, Aghebati-Maleki L, Shahgoli VK, Brunetti O, Rahmani S, Shadbad MA, Baghbanzadeh A, Silvestris N, Baradaran B. Regulation of immune responses through CD39 and CD73 in cancer: Novel checkpoints. Life Sci 2021; 282:119826. [PMID: 34265363 DOI: 10.1016/j.lfs.2021.119826] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/22/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
The immunosuppressive tumor microenvironment has been implicated in attenuating anti-tumoral immune responses and tumor growth in various cancers. Inhibitory immune checkpoints have been introduced as the primary culprits for developing the immunosuppressive tumor microenvironment. Therefore, a better understanding of the cross-talk between inhibitory immune checkpoints in the tumor microenvironment can pave the way for introducing novel approaches for treating affected patients. Growing evidence indicates that CD39 and CD73, as novel checkpoints, can transform adenosine triphosphate (ATP)-mediated pro-inflammatory tumor microenvironment into an adenosine-mediated immunosuppressive one via the purinergic signaling pathway. Indeed, enzymatic processes of CD39 and CD73 have crucial roles in adjusting the extent, intensity, and chemical properties of purinergic signals. This study aims to review the biological function of CD39 and CD73 and shed light on their significance in regulating anti-tumoral immune responses in various cancers.
Collapse
Affiliation(s)
- Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vahid Khaze Shahgoli
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Cancer and Inflammation Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Shima Rahmani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nicola Silvestris
- IRCCS Bari, Italy, Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO, University of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
45
|
Sloan G, Selvarajah D, Tesfaye S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat Rev Endocrinol 2021; 17:400-420. [PMID: 34050323 DOI: 10.1038/s41574-021-00496-z] [Citation(s) in RCA: 188] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 02/08/2023]
Abstract
Diabetic sensorimotor peripheral neuropathy (DSPN) is a serious complication of diabetes mellitus and is associated with increased mortality, lower-limb amputations and distressing painful neuropathic symptoms (painful DSPN). Our understanding of the pathophysiology of the disease has largely been derived from animal models, which have identified key potential mechanisms. However, effective therapies in preclinical models have not translated into clinical trials and we have no universally accepted disease-modifying treatments. Moreover, the condition is generally diagnosed late when irreversible nerve damage has already taken place. Innovative point-of-care devices have great potential to enable the early diagnosis of DSPN when the condition might be more amenable to treatment. The management of painful DSPN remains less than optimal; however, studies suggest that a mechanism-based approach might offer an enhanced benefit in certain pain phenotypes. The management of patients with DSPN involves the control of individualized cardiometabolic targets, a multidisciplinary approach aimed at the prevention and management of foot complications, and the timely diagnosis and management of neuropathic pain. Here, we discuss the latest advances in the mechanisms of DSPN and painful DSPN, originating both from the periphery and the central nervous system, as well as the emerging diagnostics and treatments.
Collapse
Affiliation(s)
- Gordon Sloan
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Dinesh Selvarajah
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Department of Oncology and Human Metabolism, University of Sheffield, Sheffield, UK
| | - Solomon Tesfaye
- Diabetes Research Unit, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| |
Collapse
|
46
|
Rumora AE, Guo K, Alakwaa FM, Andersen ST, Reynolds EL, Jørgensen ME, Witte DR, Tankisi H, Charles M, Savelieff MG, Callaghan BC, Jensen TS, Feldman EL. Plasma lipid metabolites associate with diabetic polyneuropathy in a cohort with type 2 diabetes. Ann Clin Transl Neurol 2021; 8:1292-1307. [PMID: 33955722 PMCID: PMC8164865 DOI: 10.1002/acn3.51367] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The global rise in type 2 diabetes is associated with a concomitant increase in diabetic complications. Diabetic polyneuropathy is the most frequent type 2 diabetes complication and is associated with poor outcomes. The metabolic syndrome has emerged as a major risk factor for diabetic polyneuropathy; however, the metabolites associated with the metabolic syndrome that correlate with diabetic polyneuropathy are unknown. METHODS We conducted a global metabolomics analysis on plasma samples from a subcohort of participants from the Danish arm of Anglo-Danish-Dutch study of Intensive Treatment of Diabetes in Primary Care (ADDITION-Denmark) with and without diabetic polyneuropathy versus lean control participants. RESULTS Compared to lean controls, type 2 diabetes participants had significantly higher HbA1c (p = 0.0028), BMI (p = 0.0004), and waist circumference (p = 0.0001), but lower total cholesterol (p = 0.0001). Out of 991 total metabolites, we identified 15 plasma metabolites that differed in type 2 diabetes participants by diabetic polyneuropathy status, including metabolites belonging to energy, lipid, and xenobiotic pathways, among others. Additionally, these metabolites correlated with alterations in plasma lipid metabolites in type 2 diabetes participants based on neuropathy status. Further evaluating all plasma lipid metabolites identified a shift in abundance, chain length, and saturation of free fatty acids in type 2 diabetes participants. Importantly, the presence of diabetic polyneuropathy impacted the abundance of plasma complex lipids, including acylcarnitines and sphingolipids. INTERPRETATION Our explorative study suggests that diabetic polyneuropathy in type 2 diabetes is associated with novel alterations in plasma metabolites related to lipid metabolism.
Collapse
Affiliation(s)
- Amy E. Rumora
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | - Kai Guo
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
- Department of Biomedical SciencesUniversity of North DakotaGrand ForksNorth Dakota
| | - Fadhl M. Alakwaa
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | | | - Evan L. Reynolds
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | - Marit E. Jørgensen
- Steno Diabetes Center CopenhagenGentofteDenmark
- University of Southern DenmarkOdenseDenmark
| | - Daniel R. Witte
- Department of Public HealthAarhus UniversityAarhusDenmark
- Danish Diabetes AcademyOdenseDenmark
| | - Hatice Tankisi
- Department of Clinical NeurophysiologyAarhus UniversityAarhusDenmark
| | - Morten Charles
- Department of Public HealthAarhus UniversityAarhusDenmark
| | - Masha G. Savelieff
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | - Brian C. Callaghan
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| | - Troels S. Jensen
- Danish Pain Research CenterDepartment of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichigan
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichigan
| |
Collapse
|
47
|
Sajic M, Rumora AE, Kanhai AA, Dentoni G, Varatharajah S, Casey C, Brown RDR, Peters F, Hinder LM, Savelieff MG, Feldman EL, Smith KJ. High Dietary Fat Consumption Impairs Axonal Mitochondrial Function In Vivo. J Neurosci 2021; 41:4321-4334. [PMID: 33785643 PMCID: PMC8143198 DOI: 10.1523/jneurosci.1852-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 02/11/2021] [Accepted: 03/15/2021] [Indexed: 01/25/2023] Open
Abstract
Peripheral neuropathy (PN) is the most common complication of prediabetes and diabetes. PN causes severe morbidity for Type 2 diabetes (T2D) and prediabetes patients, including limb pain followed by numbness resulting from peripheral nerve damage. PN in T2D and prediabetes is associated with dyslipidemia and elevated circulating lipids; however, the molecular mechanisms underlying PN development in prediabetes and T2D are unknown. Peripheral nerve sensory neurons rely on axonal mitochondria to provide energy for nerve impulse conduction under homeostatic conditions. Models of dyslipidemia in vitro demonstrate mitochondrial dysfunction in sensory neurons exposed to elevated levels of exogenous fatty acids. Herein, we evaluated the effect of dyslipidemia on mitochondrial function and dynamics in sensory axons of the saphenous nerve of a male high-fat diet (HFD)-fed murine model of prediabetes to identify mitochondrial alterations that correlate with PN pathogenesis in vivo We found that the HFD decreased mitochondrial membrane potential (MMP) in axonal mitochondria and reduced the ability of sensory neurons to conduct at physiological frequencies. Unlike mitochondria in control axons, which dissipated their MMP in response to increased impulse frequency (from 1 to 50 Hz), HFD mitochondria dissipated less MMP in response to axonal energy demand, suggesting a lack of reserve capacity. The HFD also decreased sensory axonal Ca2+ levels and increased mitochondrial lengthening and expression of PGC1α, a master regulator of mitochondrial biogenesis. Together, these results suggest that mitochondrial dysfunction underlies an imbalance of axonal energy and Ca2+ levels and impairs impulse conduction within the saphenous nerve in prediabetic PN.SIGNIFICANCE STATEMENT Diabetes and prediabetes are leading causes of peripheral neuropathy (PN) worldwide. PN has no cure, but development in diabetes and prediabetes is associated with dyslipidemia, including elevated levels of saturated fatty acids. Saturated fatty acids impair mitochondrial dynamics and function in cultured neurons, indicating a role for mitochondrial dysfunction in PN progression; however, the effect of elevated circulating fatty acids on the peripheral nervous system in vivo is unknown. In this study, we identify early pathogenic events in sensory nerve axons of mice with high-fat diet-induced PN, including alterations in mitochondrial function, axonal conduction, and intra-axonal calcium, that provide important insight into potential PN mechanisms associated with prediabetes and dyslipidemia in vivo.
Collapse
Affiliation(s)
- Marija Sajic
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Amy E Rumora
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Anish A Kanhai
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Giacomo Dentoni
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Sharlini Varatharajah
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Caroline Casey
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Ryan D R Brown
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Fabian Peters
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| | - Lucy M Hinder
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Masha G Savelieff
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan 48109
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, WC1N 3BG, United Kingdom
| |
Collapse
|
48
|
Diaz A, Muñoz-Arenas G, Venegas B, Vázquez-Roque R, Flores G, Guevara J, Gonzalez-Vergara E, Treviño S. Metforminium Decavanadate (MetfDeca) Treatment Ameliorates Hippocampal Neurodegeneration and Recognition Memory in a Metabolic Syndrome Model. Neurochem Res 2021; 46:1151-1165. [PMID: 33559829 DOI: 10.1007/s11064-021-03250-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/02/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
The consumption of foods rich in carbohydrates, saturated fat, and sodium, accompanied by a sedentary routine, are factors that contribute to the progress of metabolic syndrome (MS). In this way, they cause the accumulation of body fat, hypertension, dyslipidemia, and hyperglycemia. Additionally, MS has been shown to cause oxidative stress, inflammation, and death of neurons in the hippocampus. Consequently, spatial and recognition memory is affected. It has recently been proposed that metformin decavanadate (MetfDeca) exerts insulin mimetic effects that enhance metabolism in MS animals; however, what effects it can cause on the hippocampal neurons of rats with MS are unknown. The objective of the work was to evaluate the effect of MetfDeca on hippocampal neurodegeneration and recognition memory in rats with MS. Administration of MetfDeca for 60 days in MS rats improved object recognition memory (NORt). In addition, MetfDeca reduced markers of oxidative stress and hippocampal neuroinflammation. Accompanied by an increase in the density and length of the dendritic spines of the hippocampus of rats with MS. We conclude that MetfDeca represents an important therapeutic agent to treat MS and induce neuronal and cognitive restoration mechanisms.
Collapse
Affiliation(s)
- Alfonso Diaz
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Guadalupe Muñoz-Arenas
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Berenice Venegas
- Faculty of Biological Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Rubén Vázquez-Roque
- Laboratory of Neuropsychiatry, Institute of Physiology, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Gonzalo Flores
- Laboratory of Neuropsychiatry, Institute of Physiology, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Jorge Guevara
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Samuel Treviño
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico.
| |
Collapse
|
49
|
Palavicini JP, Chen J, Wang C, Wang J, Qin C, Baeuerle E, Wang X, Woo JA, Kang DE, Musi N, Dupree JL, Han X. Early disruption of nerve mitochondrial and myelin lipid homeostasis in obesity-induced diabetes. JCI Insight 2020; 5:137286. [PMID: 33148881 PMCID: PMC7710310 DOI: 10.1172/jci.insight.137286] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 09/30/2020] [Indexed: 12/17/2022] Open
Abstract
Diabetic neuropathy is a major complication of diabetes. Current treatment options alleviate pain but do not stop the progression of the disease. At present, there are no approved disease-modifying therapies. Thus, developing more effective therapies remains a major unmet medical need. Seeking to better understand the molecular mechanisms driving peripheral neuropathy, as well as other neurological complications associated with diabetes, we performed spatiotemporal lipidomics, biochemical, ultrastructural, and physiological studies on PNS and CNS tissue from multiple diabetic preclinical models. We unraveled potentially novel molecular fingerprints underlying nerve damage in obesity-induced diabetes, including an early loss of nerve mitochondrial (cardiolipin) and myelin signature (galactosylceramide, sulfatide, and plasmalogen phosphatidylethanolamine) lipids that preceded mitochondrial, myelin, and axonal structural/functional defects; started in the PNS; and progressed to the CNS at advanced diabetic stages. Mechanistically, we provided substantial evidence indicating that these nerve mitochondrial/myelin lipid abnormalities are (surprisingly) not driven by hyperglycemia, dysinsulinemia, or insulin resistance, but rather associate with obesity/hyperlipidemia. Importantly, our findings have major clinical implications as they open the door to novel lipid-based biomarkers to diagnose and distinguish different subtypes of diabetic neuropathy (obese vs. nonobese diabetics), as well as to lipid-lowering therapeutic strategies for treatment of obesity/diabetes-associated neurological complications and for glycemic control.
Collapse
Affiliation(s)
- Juan P. Palavicini
- Barshop Institute for Longevity and Aging Studies and
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Juan Chen
- Barshop Institute for Longevity and Aging Studies and
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunyan Wang
- Barshop Institute for Longevity and Aging Studies and
| | - Jianing Wang
- Barshop Institute for Longevity and Aging Studies and
| | - Chao Qin
- Barshop Institute for Longevity and Aging Studies and
| | - Eric Baeuerle
- Barshop Institute for Longevity and Aging Studies and
| | - Xinming Wang
- Byrd Alzheimer’s Center and Research Institute, USF Health Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jung A. Woo
- Byrd Alzheimer’s Center and Research Institute, USF Health Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - David E. Kang
- Byrd Alzheimer’s Center and Research Institute, USF Health Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies and
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jeffrey L. Dupree
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Research Service, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, Virginia, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies and
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
50
|
Hossain MJ, Kendig MD, Wild BM, Issar T, Krishnan AV, Morris MJ, Arnold R. Evidence of Altered Peripheral Nerve Function in a Rodent Model of Diet-Induced Prediabetes. Biomedicines 2020; 8:biomedicines8090313. [PMID: 32872256 PMCID: PMC7555926 DOI: 10.3390/biomedicines8090313] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Peripheral neuropathy (PN) is a debilitating complication of diabetes that affects >50% of patients. Recent evidence suggests that obesity and metabolic disease, which often precede diabetes diagnosis, may influence PN onset and severity. We examined this in a translationally relevant model of prediabetes induced by a cafeteria (CAF) diet in Sprague–Dawley rats (n = 15 CAF versus n = 15 control). Neuropathy phenotyping included nerve conduction, tactile sensitivity, intraepidermal nerve fiber density (IENFD) and nerve excitability testing, an in vivo measure of ion channel function and membrane potential. Metabolic phenotyping included body composition, blood glucose and lipids, plasma hormones and inflammatory cytokines. After 13 weeks diet, CAF-fed rats demonstrated prediabetes with significantly elevated fasting blood glucose, insulin and impaired glucose tolerance as well as obesity and dyslipidemia. Nerve conduction, tactile sensitivity and IENFD did not differ; however, superexcitability was significantly increased in CAF-fed rats. Mathematical modeling demonstrated this was consistent with a reduction in sodium–potassium pump current. Moreover, superexcitability correlated positively with insulin resistance and adiposity, and negatively with fasting high-density lipoprotein cholesterol. In conclusion, prediabetic rats over-consuming processed, palatable foods demonstrated altered nerve function that preceded overt PN. This work provides a relevant model for pathophysiological investigation of diabetic complications.
Collapse
Affiliation(s)
- Md Jakir Hossain
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia; (M.J.H.); (M.D.K.); (B.M.W.); (M.J.M.)
| | - Michael D. Kendig
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia; (M.J.H.); (M.D.K.); (B.M.W.); (M.J.M.)
| | - Brandon M. Wild
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia; (M.J.H.); (M.D.K.); (B.M.W.); (M.J.M.)
| | - Tushar Issar
- Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia; (T.I.); (A.V.K.)
| | - Arun V. Krishnan
- Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia; (T.I.); (A.V.K.)
| | - Margaret J. Morris
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia; (M.J.H.); (M.D.K.); (B.M.W.); (M.J.M.)
| | - Ria Arnold
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia; (M.J.H.); (M.D.K.); (B.M.W.); (M.J.M.)
- Correspondence: ; Tel.: +61-293858709
| |
Collapse
|