1
|
Aljumaah MR, Roach J, Hu Y, Gunstad J, Azcarate-Peril MA. Microbial dipeptidyl peptidases of the S9B family as host-microbe isozymes. SCIENCE ADVANCES 2025; 11:eads5721. [PMID: 40173242 PMCID: PMC11964003 DOI: 10.1126/sciadv.ads5721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 02/26/2025] [Indexed: 04/04/2025]
Abstract
Human dipeptidyl peptidase 4 (hDPP-4) has been a pharmacological target for metabolic diseases, particularly diabetes, since the early 2000s. As a ubiquitous enzyme found in both prokaryotic and eukaryotic organisms, hDPP-4 plays crucial roles in host homeostasis and disease progression. While many studies have explored hDPP-4's properties, research on gut microbially derived DPP-4 (mDPP-4) remains limited. This review discusses the significance of mDPP-4 and its health implications, analyzing crystal structures of mDPP-4 in comparison to human counterparts. We examine how hDPP-4 inhibitors could influence gut microbiome composition and mDPP-4 activity. Additionally, this review connects ongoing discussions regarding DPP-4 substrate specificity and potential access routes for mDPP-4, emphasizing the urgent need for further research on mDPP-4's role in health and improve the precision of DPP-4 inhibitor therapies.
Collapse
Affiliation(s)
- Mashael R. Aljumaah
- Center for Gastrointestinal Biology and Disease (CGIBD), Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, UNC Microbiome Core, University of North Carolina, Chapel Hill, NC, USA
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Jeffery Roach
- Center for Gastrointestinal Biology and Disease (CGIBD), Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, UNC Microbiome Core, University of North Carolina, Chapel Hill, NC, USA
| | - Yunan Hu
- Center for Gastrointestinal Biology and Disease (CGIBD), Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, UNC Microbiome Core, University of North Carolina, Chapel Hill, NC, USA
| | - John Gunstad
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
| | - M. Andrea Azcarate-Peril
- Center for Gastrointestinal Biology and Disease (CGIBD), Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, UNC Microbiome Core, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Gong B, Wang T, Sun L. Evolution and therapeutic potential of glucagon-like peptide 2 analogs. Biochem Pharmacol 2025; 233:116758. [PMID: 39842552 DOI: 10.1016/j.bcp.2025.116758] [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: 08/22/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Glucagon-like peptide 2 (GLP-2) is a proglucagon-derived peptide released by intestinal endocrine cells. However, its therapeutic potential is limited by rapid inactivation via dipeptidyl peptidase-IV. The elucidation of three-dimensional structures of G-protein-coupled receptors, including GLP-2 receptor, has facilitated the rational design of novel peptide therapeutics. Recent studies have explored various structural modifications based on the structure of GLP-2, such as amino acid substitution, lipidation, and fusion with proteins, to extend the half-life of GLP-2 and enhance its biological activity. One promising avenue involves the development of multifunctional molecules targeting multiple pharmacological systems to boost therapeutic efficacy. This paper reviews the recent advancements in understanding GLP-2, including its physiological roles and structure-activity relationships, and evaluates the development prospects of GLP-2 analogs.
Collapse
Affiliation(s)
- Binbin Gong
- College of Medicine, Jiaxing University, Jiaxing 314001, PR China; College of Pharmacy, Zhejiang University of Technology, Hangzhou 310000, PR China
| | - Ting Wang
- College of Medicine, Jiaxing University, Jiaxing 314001, PR China
| | - Lidan Sun
- College of Medicine, Jiaxing University, Jiaxing 314001, PR China; Taizhou Hospital, Zhejiang University, Taizhou 317000, PR China.
| |
Collapse
|
3
|
Mukherjee K, Xiao C. GLP-2 regulation of intestinal lipid handling. Front Physiol 2024; 15:1358625. [PMID: 38426205 PMCID: PMC10902918 DOI: 10.3389/fphys.2024.1358625] [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: 12/20/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Lipid handling in the intestine is important for maintaining energy homeostasis and overall health. Mishandling of lipids in the intestine contributes to dyslipidemia and atherosclerotic cardiovascular diseases. Despite advances in this field over the past few decades, significant gaps remain. The gut hormone glucagon-like peptide-2 (GLP-2) has been shown to play pleotropic roles in the regulation of lipid handling in the intestine. Of note, GLP-2 exhibits unique actions on post-prandial lipid absorption and post-absorptive release of intestinally stored lipids. This review aims to summarize current knowledge in how GLP-2 regulates lipid processing in the intestine. Elucidating the mechanisms of GLP-2 regulation of intestinal lipid handling not only improves our understanding of GLP-2 biology, but also provides insights into how lipids are processed in the intestine, which offers opportunities for developing novel strategies towards prevention and treatment of dyslipidemia and atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
| | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
4
|
Masai K, Nakayama Y, Shin K, Sugahara C, Miyazaki I, Yasuhara T, Date I, Asanuma M. Neurogenesis impairment with glial activation in the hippocampus-connected regions of intracerebroventricular streptozotocin-injected mice. Neurosci Lett 2024; 820:137598. [PMID: 38110145 DOI: 10.1016/j.neulet.2023.137598] [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: 06/14/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023]
Abstract
Adult neurogenesis in the hippocampus and subventricular zone (SVZ) is impaired by intracerebroventricular administration of streptozotocin (icv-STZ) to rodents. Although neural cells in the several brain regions which connect with the hippocampus or SVZ is thought to be involved in the adult neurogenesis, few studies have investigated morphological alterations of glial cells in these areas. The present study revealed that icv-STZ induces reduction of neural progenitor cells and a dramatic increase in reactive astrocytes and microglia especially in the hippocampus and various hippocampus-connected brain areas. In contrast, there was no significant neuronal damage excluding demyelination of the stria medullaris. The results indicate the hippocampal neurogenesis impairment of this model might be occurred by activated glial cells in the hippocampus, or hippocampus-connected regions.
Collapse
Affiliation(s)
- Kaori Masai
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yuta Nakayama
- Department of Medical Neurobiology, Okayama University Medical School, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kotaro Shin
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| |
Collapse
|
5
|
Pal B, Chattopadhyay M. Recent clinical and pharmacological advancements of incretin-based therapy and the effects of incretin on physiology. JOURNAL OF DIABETOLOGY 2024; 15:24-37. [DOI: 10.4103/jod.jod_117_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/03/2024] [Indexed: 12/11/2024] Open
Abstract
Abstract
A novel therapeutic target for diabetes mellitus is incretin-based therapies, glucagon-like peptide-1, and glucose-dependent insulinotropic polypeptides are released from the gastrointestinal (GI) tract and act on beta cells of pancreatic islets by increasing the secretion of insulin. The management and prevention of diabetes require habitual and pharmacological therapies along with quality and healthy lifestyle. This includes maintaining the body weight, blood glucose level, cardiovascular risk, complexity, and co-morbidities. The utilization of glucagon-like peptide-1 (GLP-1) agonists is an object of research with favorable hemoglobin A1C levels and weight loss in type 1 diabetic patients. However, cost-effectiveness and tolerability, remain significant barriers for patients to using these medications. The risk of suicidal tendencies and thoughts of self-harm have been increased in patients receiving GLP-1 receptor agonists. Tirzepatide treatment showed a potent glucose-lowering effect and promoted weight loss with minimum GI adverse effects in animal studies as well as phase I and II human trials, in comparison with established GLP-1 receptor agonists. The glucose-dependent insulinotropic polypeptide receptor (GIPR) peptide-antagonist effectively blocks the action of gastric-inhibitory-polypeptide (GIP) in vitro and ex vivo in human pancreas and in vivo in rodent models. However, incretin-based therapies have received enormous attention in the last few decades for the treatment of diabetes, obesity, and other repurposing including central nervous system disorders. Therefore, in this article, we demonstrate the overview, physiological, and pharmacological advances of incretin-based pharmacotherapies and their physiological roles. Furthermore, the recent updates of glucagon-like peptide-1 receptor agonist, Glucagon-like peptide-2 receptor agonist, GLP-1/GIP co-agonists, GIP/GLP-1/glucagon triple agonist and GIP-antagonist are also discussed.
Collapse
Affiliation(s)
- Bhaskar Pal
- Department of Pharmacology, Charaktala College of Pharmacy, Charaktala, Debipur, West Bengal, India
| | - Moitreyee Chattopadhyay
- Department of Pharmaceutical Technology, Maulana Abul Kalam Azad University of Technology, Nadia, West Bengal, India
| |
Collapse
|
6
|
Bathini P, Dupanloup I, Zenaro E, Terrabuio E, Fischer A, Ballabani E, Doucey MA, Alberi L. Systemic Inflammation Causes Microglial Dysfunction With a Vascular AD phenotype. Brain Behav Immun Health 2022; 28:100568. [PMID: 36704658 PMCID: PMC9871075 DOI: 10.1016/j.bbih.2022.100568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/12/2022] [Accepted: 11/26/2022] [Indexed: 12/24/2022] Open
Abstract
Background Studies in rodents and humans have indicated that inflammation outside CNS (systemic inflammation) affects brain homeostasis contributing to neurodevelopmental disorders. Itis becoming increasingly evident that such early insults may also belinked to neurodegenerative diseases like late-onset Alzheimer's disease (AD). Importantly, lifestyle and stress, such as viral or bacterial infection causing chronic inflammation, may contribute to neurodegenerative dementia. Systemic inflammatory response triggers a cascade of neuroinflammatory responses, altering brain transcriptome, cell death characteristic of AD, and vascular dementia. Our study aimed to assess the temporal evolution of the pathological impact of systemic inflammation evoked by prenatal and early postnatal peripheral exposure of viral mimetic Polyinosinic:polycytidylic acid (PolyI:C) and compare the hippocampal transcriptomic changes with the profiles of human post-mortem AD and vascular dementia brain specimens. Methods We have engineered the PolyI:C sterile infection model in wildtype C57BL6 mice to achieve chronic low-grade systemic inflammation. We have conducted a cross-sectional analysis of aging PolyI:C and Saline control mice (3 months, 6 months, 9 months, and 16 months), taking the hippocampus as a reference brain region, and compared the brain aging phenotype to AD progression in humans with mild AD, severe AD, and Controls (CTL), in parallel to Vascular dementia (VaD) patients' specimens. Results We found that PolyI:C mice display both peripheral and central inflammation with a peak at 6 months, associated with memory deficits. The hippocampus is characterized by a pronounced and progressive tauopathy. In PolyI:C brains, microglia undergo aging-dependent morphological shifts progressively adopting a phagocytic phenotype. Transcriptomic analysis reveals a profound change in gene expression throughout aging, with a peak in differential expression at 9 months. We show that the proinflammatory marker Lcn2 is one of the genes with the strongest upregulation in PolyI:C mice upon aging. Validation in brains from patients with increasing severity of AD and VaD shows the reproducibility of some gene targets in vascular dementia specimens as compared to AD ones. Conclusions The PolyI:C model of sterile infection demonstrates that peripheral chronic inflammation causes progressive tau hyperphosphorylation, changes in microglia morphology, astrogliosis and gene reprogramming reflecting increased neuroinflammation, vascular remodeling, and the loss of neuronal functionality seen to some extent in human AD and Vascular dementia suggesting early immune insults could be crucial in neurodegenerative diseases.
Collapse
Affiliation(s)
- Praveen Bathini
- Department of Medicine, University of Fribourg, Fribourg, Switzerland,Corresponding author.
| | | | - Elena Zenaro
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Eleonora Terrabuio
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Amrei Fischer
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Edona Ballabani
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | | | - Lavinia Alberi
- Department of Medicine, University of Fribourg, Fribourg, Switzerland,Swiss Integrative Center for Human Health, Fribourg, Switzerland,Corresponding author. Swiss Integrative Centre of Human Health, Passage du Cardinal 13B, CH-1700, Fribourg, Switzerland.
| |
Collapse
|
7
|
Kurowska-Rucińska E, Ruciński J, Myślińska D, Grembecka B, Wrona D, Majkutewicz I. Dimethyl Fumarate Alleviates Adult Neurogenesis Disruption in Hippocampus and Olfactory Bulb and Spatial Cognitive Deficits Induced by Intracerebroventricular Streptozotocin Injection in Young and Aged Rats. Int J Mol Sci 2022; 23:ijms232415449. [PMID: 36555093 PMCID: PMC9779626 DOI: 10.3390/ijms232415449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
The disorder of adult neurogenesis is considered an important mechanism underlying the learning and memory impairment observed in Alzheimer's disease (AD). The sporadic nonhereditary form of AD (sAD) affects over 95% of AD patients and is related to interactions between genetic and environmental factors. An intracerebroventricular injection of streptozotocin (STZ-ICV) is a representative and well-established method to induce sAD-like pathology. Dimethyl fumarate (DMF) has antioxidant and anti-inflammatory properties and is used for multiple sclerosis treatment. The present study determines whether a 26-day DMF therapy ameliorates the disruption of adult neurogenesis and BDNF-related neuroprotection in the hippocampus and olfactory bulb (OB) in an STZ-ICV rat model of sAD. Considering age as an important risk factor for developing AD, this study was performed using 3-month-old (the young group) and 22-month-old (the aged group) male Wistar rats. Spatial cognitive functions were evaluated with the Morris water maze task. Immunofluorescent labelling was used to assess the parameters of adult neurogenesis and BDNF-related neuroprotection in the hippocampus and OB. Our results showed that the STZ-ICV evoked spatial learning and memory impairment and disturbances in adult neurogenesis and BDNF expression in both examined brain structures. In the aged animals, the deficits were more severe. We found that the DMF treatment significantly alleviated STZ-ICV-induced behavioural and neuronal disorders in both age groups of the rats. Our findings suggest that DMF, due to its beneficial effect on the formation of new neurons and BDNF-related neuroprotection, may be considered as a promising new therapeutic agent in human sAD.
Collapse
|
8
|
Taghadosi Z, Zarifkar A, Razban V, Aligholi H. The effect of chronic stress and its preconditioning on spatial memory as well as hippocampal LRP1 and RAGE expression in a streptozotocin-induced rat model of Alzheimer's disease. Metab Brain Dis 2022; 37:2699-2710. [PMID: 35930096 DOI: 10.1007/s11011-022-01044-y] [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: 01/13/2022] [Accepted: 06/26/2022] [Indexed: 10/16/2022]
Abstract
According to available evidence, prolonged or chronic exposure to stress is detrimental to various brain structures, including the hippocampus. The current study examined the expression of two critical blood-brain barrier receptors required for amyloid-beta clearance to understand better the mechanism by which chronic stress impairs learning and memory in patients with Alzheimer's disease (AD). Rats were randomly assigned to one of two groups in this study: experiment 1 and experiment 2. Each main group was then divided into four subgroups. Rats were bilaterally injected with streptozotocin (STZ, 3 mg/kg, twice) using the intracerebroventricular (ICV) technique to induce the Alzheimer's model. Additionally, they were subjected to foot shock (1 mA, 1 Hz) for 10 s every 60 s (1 h/day) for ten consecutive days prior to and following STZ injection. The Morris Water Maze (MWM) test was used to assess spatial learning and memory. Real-time PCR was used to determine Low-density lipoprotein receptor-related protein-1 (LRP1) and receptor for advanced glycation end-products (RAGE) mRNA levels in the hippocampus. Moreover, the animals' body weights were determined as physiological parameters in all groups. The results indicated that 10-day chronic electric foot shock stress reduced body weight, impaired spatial learning and memory, decreased hippocampal LRP1 mRNA expression, and increased hippocampal RAGE mRNA expression in a rat AD model. It can be concluded that chronic stress in conjunction with AD alters the expression of LRP1 and RAGE in the hippocampus. The findings pave the way for scientists to develop novel treatment strategies for AD.
Collapse
Affiliation(s)
- Zohreh Taghadosi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asadollah Zarifkar
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Aligholi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Activation of TGR5 Ameliorates Streptozotocin-Induced Cognitive Impairment by Modulating Apoptosis, Neurogenesis, and Neuronal Firing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3716609. [PMID: 35464765 PMCID: PMC9033389 DOI: 10.1155/2022/3716609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 01/03/2022] [Accepted: 03/08/2022] [Indexed: 12/15/2022]
Abstract
Takeda G protein-coupled receptor 5 (TGR5) is the first known G protein-coupled receptor specific for bile acids and is recognized as a new and critical target for type 2 diabetes and metabolic syndrome. It is expressed in many brain regions associated with memory such as the hippocampus and frontal cortex. Here, we hypothesize that activation of TGR5 may ameliorate streptozotocin- (STZ-) induced cognitive impairment. The mouse model of cognitive impairment was established by a single intracerebroventricular (ICV) injection of STZ (3.0 mg/kg), and we found that TGR5 activation by its agonist INT-777 (1.5 or 3.0 μg/mouse, ICV injection) ameliorated spatial memory impairment in the Morris water maze and Y-maze tests. Importantly, INT-777 reversed STZ-induced downregulation of TGR5 and glucose usage deficits. Our results further showed that INT-777 suppressed neuronal apoptosis and improved neurogenesis which were involved in tau phosphorylation and CREB-BDNF signaling. Moreover, INT-777 increased action potential firing of excitatory pyramidal neurons in the hippocampal CA3 and medial prefrontal cortex of ICV-STZ groups. Taken together, these findings reveal that activation of TGR5 has a neuroprotective effect against STZ-induced cognitive impairment by modulating apoptosis, neurogenesis, and neuronal firing in the brain and TGR5 might be a novel and potential target for Alzheimer's disease.
Collapse
|
10
|
Desmarais F, Hervé V, Bergeron KF, Ravaut G, Perrotte M, Fyfe-Desmarais G, Rassart E, Ramassamy C, Mounier C. Cerebral Apolipoprotein D Exits the Brain and Accumulates in Peripheral Tissues. Int J Mol Sci 2021; 22:ijms22084118. [PMID: 33923459 PMCID: PMC8073497 DOI: 10.3390/ijms22084118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Apolipoprotein D (ApoD) is a secreted lipocalin associated with neuroprotection and lipid metabolism. In rodent, the bulk of its expression occurs in the central nervous system. Despite this, ApoD has profound effects in peripheral tissues, indicating that neural ApoD may reach peripheral organs. We endeavor to determine if cerebral ApoD can reach the circulation and accumulate in peripheral tissues. Three hours was necessary for over 40% of all the radiolabeled human ApoD (hApoD), injected bilaterally, to exit the central nervous system (CNS). Once in circulation, hApoD accumulates mostly in the kidneys/urine, liver, and muscles. Accumulation specificity of hApoD in these tissues was strongly correlated with the expression of lowly glycosylated basigin (BSG, CD147). hApoD was observed to pass through bEnd.3 blood brain barrier endothelial cells monolayers. However, cyclophilin A did not impact hApoD internalization rates in bEnd.3, indicating that ApoD exit from the brain is either independent of BSG or relies on additional cell types. Overall, our data showed that ApoD can quickly and efficiently exit the CNS and reach the liver and kidneys/urine, organs linked to the recycling and excretion of lipids and toxins. This indicated that cerebral overexpression during neurodegenerative episodes may serve to evacuate neurotoxic ApoD ligands from the CNS.
Collapse
Affiliation(s)
- Frederik Desmarais
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Vincent Hervé
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
| | - Karl F. Bergeron
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
| | - Gaétan Ravaut
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
| | - Morgane Perrotte
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
| | - Guillaume Fyfe-Desmarais
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Eric Rassart
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Charles Ramassamy
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
- Correspondence: (C.R.); (C.M.)
| | - Catherine Mounier
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Correspondence: (C.R.); (C.M.)
| |
Collapse
|
11
|
Sasaki-Hamada S, Fujiwara A, Satoh S, Iwai T, Oka JI. GLP-2 restores impairments in spatial working memory and hippocampal LTD via the MEK/ERK pathway in juvenile-onset diabetes rats. Behav Brain Res 2021; 406:113235. [PMID: 33716118 DOI: 10.1016/j.bbr.2021.113235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 11/18/2022]
Abstract
Type 1 diabetic animal models, generated by injecting streptozotocin (STZ), have been widely used in research. We previously reported that juvenile-onset diabetes mellitus (JDM) rats, which were prepared by administering STZ to 17-day-old rats, developed cognitive impairments and hippocampal synaptic plasticity deficiencies, which were restored by glucagon-like peptide-1 (GLP-1). GLP-1 and GLP-2 are simultaneously derived from proglucagon and act through their own specific receptors. The present study was performed to investigate the potential of GLP-2 in JDM rats. The results obtained demonstrated that GLP-2 restored impairments in spatial working memory and hippocampal long-term depression (LTD) in JDM rats, and that the MEK1/2 inhibitor, U0126, inhibited this recovery. Therefore, GLP-2 has potential in the treatment of cognitive deficits in childhood-onset diabetes.
Collapse
Affiliation(s)
- Sachie Sasaki-Hamada
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan; Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara, 252-0373, Japan
| | - Ayumu Fujiwara
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Show Satoh
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Takashi Iwai
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Kitasato University, Tokyo, 108-8641, Japan
| | - Jun-Ichiro Oka
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
12
|
Zhang Z, Hao L, Shi M, Yu Z, Shao S, Yuan Y, Zhang Z, Hölscher C. Neuroprotective Effects of a GLP-2 Analogue in the MPTP Parkinson's Disease Mouse Model. JOURNAL OF PARKINSONS DISEASE 2021; 11:529-543. [PMID: 33523018 DOI: 10.3233/jpd-202318] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Glucagon-like peptide 2 (GLP-2) is a peptide hormone derived from the proglucagon gene expressed in the intestines, pancreas and brain. Some previous studies showed that GLP-2 improved aging and Alzheimer's disease related memory impairments. Parkinson's disease (PD) is a progressive neurodegenerative disorder, and to date, there is no particular medicine reversed PD symptoms effectively. OBJECTIVE The aim of this study was to evaluate neuroprotective effects of a GLP-2 analogue in the 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) PD mouse model. METHODS In the present study, the protease resistant Gly(2)-GLP-2 (50 nmol/kg ip.) analogue has been tested for 14 days by behavioral assessment, transmission electron microscope, immunofluorescence histochemistry, enzyme-linked immunosorbent assay and western blot in an acute PD mouse model induced by MPTP. For comparison, the incretin receptor dual agonist DA5-CH was tested in a separate group. RESULTS The GLP-2 analogue treatment improved the locomotor and exploratory activity of mice, and improved bradykinesia and movement imbalance of mice. Gly(2)-GLP-2 treatment also protected dopaminergic neurons and restored tyrosine hydroxylase expression levels in the substantia nigra. Gly(2)-GLP-2 furthermore reduced the inflammation response as seen in lower microglia activation, and decreased NLRP3 and interleukin-1β pro-inflammatory cytokine expression levels. In addition, the GLP-2 analogue improved MPTP-induced mitochondrial dysfunction in the substantia nigra. The protective effects were comparable to those of the dual agonist DA5-CH. CONCLUSION The present results demonstrate that Gly(2)-GLP-2 can attenuate NLRP3 inflammasome-mediated inflammation and mitochondrial damage in the substantia nigra induced by MPTP, and Gly(2)-GLP-2 shows neuroprotective effects in this PD animal model.
Collapse
Affiliation(s)
- Zijuan Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China.,Basic Medical Collenge, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Li Hao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China.,Basic Medical Collenge, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ming Shi
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ziyang Yu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Simai Shao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Ye Yuan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| | - Christian Hölscher
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan province, China
| |
Collapse
|
13
|
Su Y, Zhang Z, Li H, Ma J, Sun L, Shao S, Zhang Z, Hölscher C. A GLP-2 Analogue Protects SH-SY5Y and Neuro-2a Cells Against Mitochondrial Damage, Autophagy Impairments and Apoptosis in a Parkinson Model. Drug Res (Stuttg) 2020; 71:43-50. [PMID: 33022720 DOI: 10.1055/a-1266-3263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glucagon-like peptide-2 (GLP-2) is a peptide hormone that belongs to the glucagon-derived peptide family. We have previously shown that analogues of the sister hormone Glucagon-like peptide-1 (GLP-1) showed neuroprotective effects. Here we investigated the effect of a GLP-2 agonist in a cell model of Parkinson's disease (PD) created by treating SH-SY5Y or Neuro-2a cells with 1-Methyl-4-phenyl-pyridine ion (MPP+). Cell viability and cell cytotoxicity was detected by MTT and LDH assays, respectively. The protein expression levels of mitochondrial, autophagy and apoptotic biomarkers including PGC-1α, Mfn2, IRE1, ATG7, LC3B, Beclin1 and Bcl-2 were detected by western blot. Mitochondrial superoxide was detected by MitoSOX Red. In addition, mitochondrial morphology, autophagosome and apoptotic corpuscles were observed by transmission electron microscope (TEM). We found that the GLP-1 and the GLP-2 agonists both protect cells against mitochondrial damage, autophagy impairments and apoptosis induced by MPP+both in SH-SY5Y and Neuro-2a cells. Cell signaling for mitogenesis was enhanced, and oxidative stress levels much reduced by the drugs. This demonstrates for the first time the neuroprotective effects of a GLP-2 analogue in PD cellular models, in which oxidative stress, autophagy and apoptosis play crucial roles. The protective effects were comparable to those seen with the GLP-1 analogue liraglutide. The results suggest that not only GLP-1, but also GLP-2 has neuroprotective properties and may be useful as a novel treatment of PD.
Collapse
Affiliation(s)
| | - Zijuan Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Hao Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jinlian Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Limin Sun
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Simai Shao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Christian Hölscher
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China.,Neurology Department of the Second Associated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
14
|
Can dipeptidyl peptidase-4 inhibitors treat cognitive disorders? Pharmacol Ther 2020; 212:107559. [PMID: 32380197 DOI: 10.1016/j.pharmthera.2020.107559] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022]
Abstract
The linkage of neurodegenerative diseases with insulin resistance (IR) and type 2 diabetes mellitus (T2DM), including oxidative stress, mitochondrial dysfunction, excessive inflammatory responses and abnormal protein processing, and the correlation between cerebrovascular diseases and hyperglycemia has opened a new window for novel therapeutics for these cognitive disorders. Various antidiabetic agents have been studied for their potential treatment of cognitive disorders, among which the dipeptidyl peptidase-4 (DPP-4) inhibitors have been investigated more recently. So far, DPP-4 inhibitors have demonstrated neuroprotection and cognitive improvements in animal models, and cognitive benefits in diabetic patients with or without cognitive impairments. This review aims to summarize the potential mechanisms, advantages and limitations, and currently available evidence for developing DPP-4 inhibitors as a treatment of cognitive disorders.
Collapse
|
15
|
Song L, Piao Z, Yao L, Zhang L, Lu Y. Schisandrin ameliorates cognitive deficits, endoplasmic reticulum stress and neuroinflammation in streptozotocin (STZ)-induced Alzheimer's disease rats. Exp Anim 2020; 69:363-373. [PMID: 32336744 PMCID: PMC7445059 DOI: 10.1538/expanim.19-0146] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Schisandrin, an active component extracted from Schisandra chinensis (Turcz.) Baill has
been reported to alleviate the cognitive impairment in neurodegenerative disorder like
Alzheimer’s disease (AD). However, the mechanism by which schisandrin regulates the
cognitive decline is still unclear. In our study, intracerebroventricular injection of
streptozotocin (STZ) was employed to establish AD model in male Wistar rats, and indicated
dose of schisandrin was further administered. The Morris water maze test was performed to
evaluate the ability of learning and memory in rats with schisandrin treatment. The
results indicated that schisandrin improved the capacity of cognition in STZ-induced rats.
The contents of pro-inflammatory cytokines in brain tissue were determined by ELISA, and
the expressions of these cytokines were assessed by western-blot and immunohistochemistry.
The results showed that treatment of schisandrin significantly reduced the production of
inflammation mediators including tumor necrosis factor-α, interleukin-1β and
interleukin-6. Further study suggested a remarkable decrease in the expressions of ER
stress maker proteins like C/EBP-homologous protein, glucose-regulated protein 78 and
cleaved caspase-12 in the presence of schisandrin, meanwhile the up-regulation of sirtuin
1 (SIRT1) was also observed in the same group. Additionally, the results of western-blot
and EMSA demonstrated that schisandrin inhibited NF-κB signaling in the brain of
STZ-induced rats. In conclusion, schisandrin ameliorated STZ-induced cognitive
dysfunction, ER stress and neuroinflammation which may be associated with up-regulation of
SIRT1. Our study provides novel mechanisms for the neuroprotective effect of schisandrin
in AD treatment.
Collapse
Affiliation(s)
- Lin Song
- School of Life Sciences, Huizhou University, 46 Yanda Avenue, Huizhou, Guangdong 516007, P.R. China
| | - Zhongyuan Piao
- Department of Neurology, Huizhou Third People's Hospital, Huizhou Hospital of Guangzhou Medical University, 1 Xuebei Street, Huizhou, Guangdong 516002, P.R. China
| | - Lifen Yao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang 150001, P.R. China
| | - Limei Zhang
- Department of Obstetrics and Gynecology, Huizhou Third People's Hospital, Huizhou Hospital of Guangzhou Medical University, 1 Xuebei Street, Huizhou, Guangdong 516002, P.R. China
| | - Yichan Lu
- Department of Chinese Medicine, Dalian Maternity and Child Health Care Hospital, 321 Jiefang Road, Dalian, Liaoning 116033, People's Republic of China
| |
Collapse
|