1
|
Elemary T, Nicola M, Abdelrahim MEA, Zaafar D. The impact of DPP-4 inhibitors on cardiovascular disease treatment: a comprehensive review of current therapeutic strategies and future directions. Mol Biol Rep 2025; 52:400. [PMID: 40244362 DOI: 10.1007/s11033-025-10458-7] [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: 10/01/2024] [Accepted: 03/20/2025] [Indexed: 04/18/2025]
Abstract
Patients diagnosed with type 2 diabetes have an increased risk of developing cardiovascular complications. The researchers are currently working on understanding how to prevent these progressions from occurring. Since 2006, dipeptidyl-peptidase-4 inhibitors have been made available to patients as a relatively new treatment for diabetes. These substances inhibit the enzyme known as dipeptidyl peptidase-4 (DPP-4), which in turn increases the levels of the glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1). This results in an increase in the effectiveness of insulin release after meals, which in turn has a positive impact on glycemic control. Regarding the safety of this category of medication in the treatment of cardiovascular disorders, there have been a great deal of debates. Emerging research suggests that DPP-4 inhibitors could be useful in the treatment of a variety of cardiovascular conditions, including coronary atherosclerosis, heart failure, and hypertension, among others. In order to investigate the possibility of using dipeptidyl-peptidase-4 inhibitors as a treatment option for cardiovascular disease, the molecular pathways that are thought to be responsible for their cardioprotective effect will be clarifies throughout the course of this review.
Collapse
Affiliation(s)
- Toka Elemary
- Clinical Pharmacy Department, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| | - Mina Nicola
- Clinical Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed E A Abdelrahim
- Clinical Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Dalia Zaafar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| |
Collapse
|
2
|
Xu S, Wang Q, Qin Y, Yang Q, Xu Y, Zhou Z. Dl-3-n-butylphthalein inhibits neuronal apoptosis and ferroptosis after cerebral ischemia-reperfusion injury in rats by regulating CXCR4. Neurotoxicol Teratol 2025; 108:107434. [PMID: 39956404 DOI: 10.1016/j.ntt.2025.107434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
OBJECTIVE To investigate the anti-apoptosis and anti-ferroptosis effects of dl-3-n-butylphthalide (dl-NBP) on cerebral ischemia-reperfusion injury (CIRI) in rats, and the potential involvement of cysteine-X-cysteine chemokine receptor 4 (CXCR4). METHODS The differentially expressed genes between healthy people and stroke patients were screened by GEO database. A transient middle cerebral artery occlusion rat model was used to induce CIRI in vivo. Rats were randomly divided into sham group, tMCAO group, and dl-NBP + tMCAO group. The therapeutic effect of dl-NBP in vivo and its effect on apoptosis and ferroptosis in brain tissues were evaluated. An in vitro oxygen-glucose deprivation/reperfusion (OGD/R) model was established to simulate CIRI in cultured PC12 cells, and the effects of dl-NBP on apoptosis and ferroptosis were examined. In this model, CXCR4 expression was assessed by western blotting and its involvement in dl-NBP-mediated protection assessed by inhibition with AMD3100. RESULTS In the stroke-related GSE22255 and GSE66724 datasets, a total of six genes with increased co-expression were found, including CXCR4. Dl-NBP treatment significantly reduced both the volume of cerebral infarction and the degree of cerebral edema, and improved neurological function in rats. dl-NBP reduced the degree of apoptosis and ferroptosis and alleviated CIRI both in vivo and in vitro. The pro-survival effects of dl-NBP were significantly reversed after CXCR4 inhibition with AMD3100. CONCLUSION Dl-NBP has anti-apoptotic and anti-ferroptotic effects on CIRI both in vivo and in vitro, and this effect is mediated by CXCR4.
Collapse
Affiliation(s)
- Sifan Xu
- Department of Neurology, First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu City, Anhui Province, PR China
| | - Qi Wang
- Department of Neurology, First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu City, Anhui Province, PR China
| | - Yu Qin
- Department of Neurology, First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu City, Anhui Province, PR China
| | - Qian Yang
- Department of Neurology, First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu City, Anhui Province, PR China
| | - Yang Xu
- Department of Neurology, First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu City, Anhui Province, PR China..
| | - Zhiming Zhou
- Department of Neurology, First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu City, Anhui Province, PR China..
| |
Collapse
|
3
|
Shi S, Li X, Chen Y, Li J, Dai Y. Cardiovascular Therapy Benefits of Novel Antidiabetic Drugs in Patients With Type 2 Diabetes Mellitus Complicated With Cardiovascular Disease: A Network Meta-Analysis. J Diabetes 2025; 17:e70044. [PMID: 39789833 PMCID: PMC11717902 DOI: 10.1111/1753-0407.70044] [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: 09/25/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVE Provide an evidence-based basis for the selection of cardiovascular benefit drugs in Type 2 diabetes mellitus (T2DM) patients with cardiovascular disease (CVD). METHODS Conduct a comprehensive search of all relevant literature from PubMed, Embase, Web of Science, Cochrane Library, and Clinical Trials.gov from their establishment until December 13, 2023, and select randomized controlled trials (RCTs) that meet the pre-established inclusion and exclusion criteria. Use the Cochrane bias risk assessment tool to evaluate the quality of the included literature. Use R 4.3.2 software to conduct network meta-analysis for drug category comparison. RESULTS A total of 24 large-scale randomized controlled trials (RCTs) were included, including 19 intervention measures, and 172 803 patients participated in the study. The results of the network meta-analysis show that: GLP1RA (OR 0.89, 95% CI 0.81-0.97) and SGLT2i (OR 0.91, 95% CI 0.83-0.99) can reduce the occurrence of major adverse cardiovascular events (MACE), GLP1RA (OR 0.88, 95% CI 0.79-0.97) and SGLT2i (OR 0.89, 95% CI 0.81-0.99) reduced the risk of cardiovascular death. SGLT2i (OR 0.68, 95% CI 0.62-0.75) reduced the occurrence of hospitalization for heart failure, GLP1RA (OR 0.88, 95% CI 0.81-0.97) and SGLT2i (OR 0.89, 95% CI 0.80-0.97) reduced the occurrence of all-cause death. CONCLUSION In the comparison of new hypoglycemic drug classes, GLP1RA and SGLT2i reduced MACE, cardiovascular mortality and all-cause mortality in T2DM patients with CVD, with no significant difference in efficacy, and DPP4i was noninferior to placebo. Only GLP1RA reduced the risk of nonfatal stroke, and only SGLT2i reduced the risk of HHF.
Collapse
Affiliation(s)
- Saixian Shi
- School of PharmacySouthwest Medical UniversityLuzhouSichuan ProvinceChina
- Pangang Xichang HospitalXichangSichuan ProvinceChina
| | - Xiaofeng Li
- School of PharmacySouthwest Medical UniversityLuzhouSichuan ProvinceChina
| | - Ye Chen
- School of PharmacySouthwest Medical UniversityLuzhouSichuan ProvinceChina
| | - Jiahao Li
- School of PharmacySouthwest Medical UniversityLuzhouSichuan ProvinceChina
| | - Yan Dai
- Department of PharmacyAffiliated Hospital of Southwest Medical UniversityLuzhouSichuan ProvinceChina
| |
Collapse
|
4
|
Wang X, Chen L, Li W, He Z, Jiang H. Association of dipeptidyl peptidase-4 with Alzheimer's disease: A new therapeutic prospect. J Alzheimers Dis 2025; 103:319-332. [PMID: 39773090 DOI: 10.1177/13872877241304673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Alzheimer's disease (AD) is the most common disease associated with cognitive dysfunction, which is closely associated with type 2 diabetes mellitus (T2DM) in clinical manifestations, pathological changes and prevention. Inhibition of dipeptidyl peptidase 4 (DPP-4) can lower blood glucose levels by stimulating insulin secretion. Besides, it can affect cognitive function through the neuroprotective effect of DPP-4 substrates, such as glucose-dependent insulin peptide and glucagon-like peptide-1, the proteolytic effect on amyloid-β and the protective effect on neuronal structure. This review discusses the relationship between cognitive impairment in T2DM and in AD, summarizes the effect of DPP-4 inhibitor (DPP-4i) on improving cognitive impairment in these two diseases based on the current studies. Given the lack of clinical randomized trials that evaluate the effect of DPP-4i on AD, this review is expected to provide preclinical evidence for DPP-4i as a potential therapy for the treatment and prevention of AD.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Physiology and Pathophysiology, Jiaxing University Medical College, Jiaxing, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Li Chen
- Department of Pathology, Northeast Yunnan Regional Central Hospital, Zhaotong, China
| | - Weijian Li
- Department of Physiology and Pathophysiology, Jiaxing University Medical College, Jiaxing, China
| | - Zhi He
- Department of Physiology and Pathophysiology, Jiaxing University Medical College, Jiaxing, China
| | - Haiying Jiang
- Department of Physiology and Pathophysiology, Jiaxing University Medical College, Jiaxing, China
| |
Collapse
|
5
|
Höfling C, Donkersloot P, Ulrich L, Burghardt S, Opitz M, Geissler S, Schilling S, Cynis H, Michalski D, Roßner S. Dipeptidyl peptidase 4 deficiency improves survival after focal cerebral ischemia in mice and ameliorates microglia activation and specific inflammatory markers. Neurobiol Dis 2024; 201:106671. [PMID: 39293688 DOI: 10.1016/j.nbd.2024.106671] [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: 06/17/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024] Open
Abstract
Dipeptidyl peptidase 4 (DPP4; CD26) is involved in the regulation of various metabolic, immunological, and neurobiological processes in healthy individuals. Observations based on epidemiological data indicate that DPP4 inhibition by gliptins, typically used in patients with diabetes, may reduce the risk for cerebral ischemia and may also improve related outcomes. However, as DPP4 inhibitor application is neither complete nor specific for suppression of DPP4 enzymatic activity and DPP4 has non-enzymatic functions as well, the variety of consequences is a matter of debate. Therefore, we here used DPP4 knock-out (KO) mice to analyze the specific contribution of DPP4 to cellular, immunological, and functional consequences of experimental focal cerebral ischemia. We observed a significantly higher survival rate of DPP4 KO mice after ischemia, which was accompanied by a lower abundance of the pro-inflammatory chemokine CCL2 and reduced activation of Iba1-positive microglia cells in brain tissue of DPP4 KO mice. In addition, after ischemia for 24 h to 72 h, decreased concentrations of CCL5 and CCL12 in plasma and of CCL17 in brain tissue of DPP4 KO mice were observed when compared to wild type mice. Other aspects analyzed, such as the functional Menzies score, astrocyte activation and chemokine levels in plasma and brain tissue were affected by ischemia but appeared to be unaffected by the DPP4 KO genotype. Taken together, experimental ablation of DPP4 functions in mice improves survival and ameliorates aspects of cellular and molecular inflammation after focal cerebral ischemia.
Collapse
Affiliation(s)
- Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; Department of Neurology, University of Leipzig, 04103 Leipzig, Germany
| | - Philippa Donkersloot
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Luise Ulrich
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Sina Burghardt
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Michael Opitz
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Stefanie Geissler
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, 06120 Halle (Saale), Germany
| | - Stephan Schilling
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, 06120 Halle (Saale), Germany; Anhalt University of Applied Sciences, Faculty of Applied Biosciences and Process Engineering, 06366 Köthen, Germany
| | - Holger Cynis
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, 06120 Halle (Saale), Germany; Junior Research Group "Immunomodulation in Pathophysiological Processes" Faculty of Medicine, Martin Luther University Halle-Wittenberg, Germany
| | - Dominik Michalski
- Department of Neurology, University of Leipzig, 04103 Leipzig, Germany
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
6
|
Chen B, Yu X, Horvath-Diano C, Ortuño MJ, Tschöp MH, Jastreboff AM, Schneeberger M. GLP-1 programs the neurovascular landscape. Cell Metab 2024; 36:2173-2189. [PMID: 39357509 DOI: 10.1016/j.cmet.2024.09.003] [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: 04/26/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Readily available nutrient-rich foods exploit our inherent drive to overconsume, creating an environment of overnutrition. This transformative setting has led to persistent health issues, such as obesity and metabolic syndrome. The development of glucagon-like peptide-1 receptor (GLP-1R) agonists reveals our ability to pharmacologically manage weight and address metabolic conditions. Obesity is directly linked to chronic low-grade inflammation, connecting our metabolic environment to neurodegenerative diseases. GLP-1R agonism in curbing obesity, achieved by impacting appetite and addressing associated metabolic defects, is revealing additional benefits extending beyond weight loss. Whether GLP-1R agonism directly impacts brain health or does so indirectly through improved metabolic health remains to be elucidated. In exploring the intricate connection between obesity and neurological conditions, recent literature suggests that GLP-1R agonism may have the capacity to shape the neurovascular landscape. Thus, GLP-1R agonism emerges as a promising strategy for addressing the complex interplay between metabolic health and cognitive well-being.
Collapse
Affiliation(s)
- Bandy Chen
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Xiaofei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Claudia Horvath-Diano
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - María José Ortuño
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Matthias H Tschöp
- Helmholtz Zentrum München, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany
| | - Ania M Jastreboff
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - Marc Schneeberger
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA; Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
7
|
Zhang X, Zheng Y, Wang Z, Zhang G, Yang L, Gan J, Jiang X. Calpain: The regulatory point of cardiovascular and cerebrovascular diseases. Biomed Pharmacother 2024; 179:117272. [PMID: 39153432 DOI: 10.1016/j.biopha.2024.117272] [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: 05/12/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024] Open
Abstract
Calpain, a key member of the Calpain cysteine protease superfamily, performs limited protein hydrolysis in a calcium-dependent manner. Its activity is tightly regulated due to the potential for non-specific cleavage of various intracellular proteins upon aberrant activation. A thorough review of the literature from 2010 to 2023 reveals 121 references discussing cardiovascular and cerebrovascular diseases. Dysregulation of the Calpain system is associated with various pathological phenomena, including lipid metabolism disorders, inflammation, apoptosis, and excitotoxicity. Although recent studies have revealed the significant role of Calpain in cardiovascular and cerebrovascular diseases, the precise mechanisms remain incompletely understood. Exploring the potential of Calpain inhibition as a therapeutic approach for the treatment of cardiovascular and cerebrovascular diseases may emerge as a compelling area of interest for future calpain research.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Yujia Zheng
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Ziyu Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Guangming Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Lin Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Jiali Gan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xijuan Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
8
|
Zhuge F, Zheng L, Pan Y, Ni L, Fu Z, Shi J, Ni Y. DPP-4 inhibition by linagliptin ameliorates age-related mild cognitive impairment by regulating microglia polarization in mice. Exp Neurol 2024; 373:114689. [PMID: 38199510 DOI: 10.1016/j.expneurol.2024.114689] [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: 10/23/2023] [Revised: 12/29/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Extensive preclinical evidence demonstrates a causative link between insulin signaling dysfunction and the pathogenesis of Alzheimer's disease (AD), and diabetic drugs may represent a promising approach to fighting AD. However, it remains to be determined which antidiabetic drugs are more effective in preventing cognitive impairment. Thus, the present study investigated the effect of dipeptidyl peptidase-4 (DPP-4) inhibitor linagliptin on cognitive impairment in middle-aged mice by comparing it with the effect of metformin. We found that DPP-4 activity increased in the hippocampus of middle-aged mice, and DPP-4 was mainly expressed by microglia rather than astrocytes and oligodendrocytes. DPP-4 directly regulated M1/M2 microglia polarization following LPS or IL-4 stimulation, while DPP-4 inhibitor, linagliptin, suppressed M1-polarized activation and induced M2-polarized activation. Both linagliptin and metformin enhanced cognitive ability, increased hippocampal synaptic plasticity and neurogenesis, and decreased age-related oxidative stress and inflammation by regulating microglia polarization in the hippocampus of middle-aged mice. The combination of linagliptin and metformin showed a maximum protective effect compared to the individual drugs alone. Loss of macrophage inflammatory protein-1α (MIP-1α), a DPP-4 substrate, abrogated the cognitive protection and anti-inflammation effects of linagliptin. Therefore, the current investigation exhibits a potential utility for DPP-4 inhibition in attenuating microglia-mediated inflammation and preventing mild cognitive impairment (MCI) in middle-aged mice, and the effect was partly mediated by MIP-1α.
Collapse
Affiliation(s)
- Fen Zhuge
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Liujie Zheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Yuxiang Pan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Liyang Ni
- Food Biochemistry Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Junping Shi
- Department of Infectious Disease, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
9
|
Bertocchi I, Turroni S, Chiazza F. Editorial: Nutritional modulation of central nervous system development, maintenance, plasticity, and recovery. Front Neurosci 2023; 17:1332191. [PMID: 38046658 PMCID: PMC10691758 DOI: 10.3389/fnins.2023.1332191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 12/05/2023] Open
Affiliation(s)
- Ilaria Bertocchi
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute - Cavalieri-Ottolenghi Foundation (NICO), Laboratory of Neuropsychopharmacology, Turin, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Fausto Chiazza
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Novara, Italy
| |
Collapse
|
10
|
Zhang Y, Liu Y, Zhang X, Yong VW, Xue M. Omarigliptin Protects the Integrity of the Blood-Brain Barrier After Intracerebral Hemorrhage in Mice. J Inflamm Res 2023; 16:2535-2548. [PMID: 37342770 PMCID: PMC10278948 DOI: 10.2147/jir.s411017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/10/2023] [Indexed: 06/23/2023] Open
Abstract
Purpose Intracerebral hemorrhage (ICH) is a fatal disease without effective treatment. The damage of the blood-brain barrier (BBB) is a key cause of brain edema and herniation after ICH. Omarigliptin (also known as MK3102) is a potent antidiabetic that inhibits dipeptidyl peptidase (DPP4); the latter has the ability to bind and degrade matrix metalloproteinases (MMPs). The present study aims to investigate the protective effects of omarigliptin against the destruction of BBB following ICH in mice. Methods and Materials Collagenase VII was used to induce ICH in C57BL/6 mice. MK3102 (7 mg/kg/day) was administered after ICH. The modified neurological severity scores (mNSS) were carried out to assess neurological functions. Nissl staining was applied to evaluate neuronal loss. Brain water content, Evans blue extravasation, Western blots, immunohistochemistry and immunofluorescence were used to study the protective effects of BBB with MK3102 at 3 days after ICH. Results MK3102 reduced DPP4 expression and decreased hematoma formation and neurobehavioral deficits of ICH mice. This was correspondent with lowered activation of microglia/macrophages and infiltration of neutrophils after ICH. Importantly, MK3102 protected the integrity of the BBB after ICH, associated with decreased expression of MMP-9, and preservation of the tight junction proteins ZO-1 and Occludin on endothelial cells through putative degradation of MMP-9, and inhibition of the expression of CX43 on astrocytes. Conclusion Omarigliptin protects the integrity of the BBB in mice after ICH injury.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Xiangyu Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - V Wee Yong
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| |
Collapse
|
11
|
Maglinger B, Harp JP, Frank JA, Rupareliya C, McLouth CJ, Pahwa S, Sheikhi L, Dornbos D, Trout AL, Stowe AM, Fraser JF, Pennypacker KR. Inflammatory-associated proteomic predictors of cognitive outcome in subjects with ELVO treated by mechanical thrombectomy. BMC Neurol 2023; 23:214. [PMID: 37280551 PMCID: PMC10243077 DOI: 10.1186/s12883-023-03253-z] [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: 09/10/2022] [Accepted: 05/18/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Emergent Large Vessel Occlusion (ELVO) stroke causes devastating vascular events which can lead to significant cognitive decline and dementia. In the subset of ELVO subjects treated with mechanical thrombectomy (MT) at our institution, we aimed to identify systemic and intracranial proteins predictive of cognitive function at time of discharge and at 90-days. These proteomic biomarkers may serve as prognostic indicators of recovery, as well as potential targets for novel/existing therapeutics to be delivered during the subacute stage of stroke recovery. METHODS At the University of Kentucky Center for Advanced Translational Stroke Sciences, the BACTRAC tissue registry (clinicaltrials.gov; NCT03153683) of human biospecimens acquired during ELVO stroke by MT is utilized for research. Clinical data are collected on each enrolled subject who meets inclusion criteria. Blood samples obtained during thrombectomy were sent to Olink Proteomics for proteomic expression values. Montreal Cognitive Assessments (MoCA) were evaluated with categorical variables using ANOVA and t-tests, and continuous variables using Pearson correlations. RESULTS There were n = 52 subjects with discharge MoCA scores and n = 28 subjects with 90-day MoCA scores. Several systemic and intracranial proteins were identified as having significant correlations to discharge MoCA scores as well as 90-day MoCA scores. Highlighted proteins included s-DPP4, CCL11, IGFBP3, DNER, NRP1, MCP1, and COMP. CONCLUSION We set out to identify proteomic predictors and potential therapeutic targets related to cognitive outcomes in ELVO subjects undergoing MT. Here, we identify several proteins which predicted MoCA after MT, which may serve as therapeutic targets to lessen post-stroke cognitive decline.
Collapse
Affiliation(s)
- Benton Maglinger
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jordan P Harp
- Department of Neurology, University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
| | - Jacqueline A Frank
- Department of Neurology, University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
| | | | | | - Shivani Pahwa
- Department of Neurosurgery, University of Kentucky, Lexington, KY, USA
- Department of Radiology, University of Kentucky, Lexington, KY, USA
| | - Lila Sheikhi
- Department of Neurology, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, University of Kentucky, Lexington, KY, USA
- Department of Radiology, University of Kentucky, Lexington, KY, USA
| | - David Dornbos
- Department of Neurosurgery, University of Kentucky, Lexington, KY, USA
- Department of Radiology, University of Kentucky, Lexington, KY, USA
| | - Amanda L Trout
- Department of Neurology, University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
| | - Ann M Stowe
- Department of Neurology, University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Justin F Fraser
- Department of Neurology, University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, University of Kentucky, Lexington, KY, USA
- Department of Radiology, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Keith R Pennypacker
- Department of Neurology, University of Kentucky, Lexington, KY, USA.
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA.
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
- Department of Neurology and Neuroscience, Center for Advanced Translational Stroke Science, University of Kentucky, Building BBSRB, Office B383, Lexington, KY, 40536, USA.
| |
Collapse
|
12
|
Jiang G, Li X, Liu M, Li H, Shen H, liao J, You W, Fang Q, Chen G. Remote ischemic postconditioning ameliorates stroke injury via the SDF-1α/CXCR4 signaling axis in rats. Brain Res Bull 2023; 197:31-41. [PMID: 36990325 DOI: 10.1016/j.brainresbull.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/14/2023] [Accepted: 03/26/2023] [Indexed: 03/29/2023]
Abstract
Remote Ischemic Postconditioning (RIPostC) has become a research hotspot due to its protective effect on the brain in clinical studies related to ischemic stroke. The purpose of this study is to investigate the protective effect of RIPostC after ischemic stroke in rats. The middle cerebral artery occlusion/reperfusion (MCAO/R) model was established by the wire embolization method. RIPostC was obtained by inducing temporary ischemia in the hind limbs of rats. First, based on the results of short-term behavioral measures and long-term neurological function experiments, RIPostC was found to have a protective effect on the MCAO/R model and to improve neurological recovery in rats. Compared to the sham group, RIPostC upregulated the expression levels of C-X-C motif chemokine receptor 4(CXCR4) in the brain and stromal cell-derived factor-1(SDF-1α) in peripheral blood. In addition, RIPostC upregulated CXCR4 expression on CD34+ stem cells in peripheral blood in flow cytometric assays. Meanwhile, according to the results of EdU/DCX co-staining and CD31 staining, it was found that the effect of RIPostC on ameliorating brain injury via SDF-1α/CXCR4 signaling axis may be associated with vascular neogenesis. Finally, after inhibiting the SDF-1α/CXCR4 signaling axis using AMD3100(Plerixafor), we found that the neuroprotective effect of RIPostC was diminished. Taken together, RIPostC can improve neurobehavioral damage induced by MCAO/R in rats, and its mechanism may be related to SDF-1α/CXCR4 signaling axis. Therefore, RIPostC can be used as an intervention strategy for stroke. SDF-1α/CXCR4 signaling axis can also be a potential target for intervention.
Collapse
|
13
|
Bernstein HG, Keilhoff G, Dobrowolny H, Steiner J. The many facets of CD26/dipeptidyl peptidase 4 and its inhibitors in disorders of the CNS - a critical overview. Rev Neurosci 2023; 34:1-24. [PMID: 35771831 DOI: 10.1515/revneuro-2022-0026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/10/2022] [Indexed: 01/11/2023]
Abstract
Dipeptidyl peptidase 4 is a serine protease that cleaves X-proline or X-alanine in the penultimate position. Natural substrates of the enzyme are glucagon-like peptide-1, glucagon inhibiting peptide, glucagon, neuropeptide Y, secretin, substance P, pituitary adenylate cyclase-activating polypeptide, endorphins, endomorphins, brain natriuretic peptide, beta-melanocyte stimulating hormone and amyloid peptides as well as some cytokines and chemokines. The enzyme is involved in the maintenance of blood glucose homeostasis and regulation of the immune system. It is expressed in many organs including the brain. DPP4 activity may be effectively depressed by DPP4 inhibitors. Apart from enzyme activity, DPP4 acts as a cell surface (co)receptor, associates with adeosine deaminase, interacts with extracellular matrix, and controls cell migration and differentiation. This review aims at revealing the impact of DPP4 and DPP4 inhibitors for several brain diseases (virus infections affecting the brain, tumours of the CNS, neurological and psychiatric disorders). Special emphasis is given to a possible involvement of DPP4 expressed in the brain.While prominent contributions of extracerebral DPP4 are evident for a majority of diseases discussed herein; a possible role of "brain" DPP4 is restricted to brain cancers and Alzheimer disease. For a number of diseases (Covid-19 infection, type 2 diabetes, Alzheimer disease, vascular dementia, Parkinson disease, Huntington disease, multiple sclerosis, stroke, and epilepsy), use of DPP4 inhibitors has been shown to have a disease-mitigating effect. However, these beneficial effects should mostly be attributed to the depression of "peripheral" DPP4, since currently used DPP4 inhibitors are not able to pass through the intact blood-brain barrier.
Collapse
Affiliation(s)
- Hans-Gert Bernstein
- Department of Psychiatry and Psychotherapy, Otto v. Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto v. Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry and Psychotherapy, Otto v. Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Johann Steiner
- Department of Psychiatry and Psychotherapy, Otto v. Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| |
Collapse
|
14
|
Li J, Ji C, Zhang W, Lan L, Ge W. Effect of new glucose-lowering drugs on stroke in patients with type 2 diabetes: A systematic review and Meta-analysis. J Diabetes Complications 2023; 37:108362. [PMID: 36462459 DOI: 10.1016/j.jdiacomp.2022.108362] [Citation(s) in RCA: 4] [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: 08/23/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
AIMS People with diabetes tend to face a higher risk of stroke. Randomized controlled trials (RCTs) have demonstrated the different outcomes of new glucose-lowering drugs marketed in recent years on cardiovascular outcome events. The effects of glucagon-like peptide-1 (GLP-1) agonists, sodium-glucose cotransporter-2 (SGLT-2) inhibitors, and dipeptidyl peptidase-4 (DPP-4) inhibitors on stroke risk were evaluated in published RCTs. METHODS A search of Embase, Cochrane Library, and PubMed databases identified studies with stroke as an outcome event up to 3 December 2021. Risk ratios for stroke outcomes were analyzed using a fixed-effects model. I2 was used to assess the heterogeneity of the study. RESULTS 19 RCTs with 155,027 participants with type 2 diabetes were identified. Pooled analysis showed that compared to placebo, GLP-1 agonists reduced non-fatal stroke by 15 % (RR = 0.85, 95%CI 0.77-0.94, P = 0.002, I2 = 0 %) and total stroke (RR = 0.84, 95%CI 0.77-0.93, P = 0.000, I2 = 0 %) by 16 %. SGLT-2 inhibitors and DPP-4 inhibitors were not significantly associated with lower stroke risk. CONCLUSIONS This meta-analysis indicates that GLP-1 agonists have potential benefits for stroke. However, further studies are needed if GLP-1 agonists are to be used to reduce the risk of stroke in patients with type 2 diabetes. More research is also needed to investigate the effects of new glucose-lowering drugs on different stroke subtypes. SYSTEMATIC REVIEW REGISTRATION This protocol was registered on the International Prospective Register of Systematic Reviews (https://www.crd.york.ac.uk/PROSPERO/; registration number: CRD42022326382).
Collapse
Affiliation(s)
- Jiaxi Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China; Department of Pharmacy, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Cheng Ji
- Department of Pharmacy, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wen Zhang
- Department of Radiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Linyan Lan
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China; Department of Pharmacy, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weihong Ge
- Department of Pharmacy, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
15
|
Zakaria EM, Tawfeek WM, Hassanin MH, Hassaballah MY. Cardiovascular protection by DPP-4 inhibitors in preclinical studies: an updated review of molecular mechanisms. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:1357-1372. [PMID: 35945358 PMCID: PMC9568460 DOI: 10.1007/s00210-022-02279-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022]
Abstract
Dipeptidyl peptidase 4 (DPP4) inhibitors are a class of antidiabetic medications that cause glucose-dependent increase in incretins in diabetic patients. One of the two incretins, glucagon-like peptide-1 (GLP-1), beside its insulinotropic activity, has been studied for extra pancreatic effects. Most of DPP4 inhibitors (DPP4i) have been investigated in in vivo and in vitro models of diabetic and nondiabetic cardiovascular diseases including heart failure, hypertension, myocardial ischemia or infarction, atherosclerosis, and stroke. Results of preclinical studies proved prominent therapeutic potential of DPP4i in cardiovascular diseases, regardless the presence of diabetes. This review aims to present an updated summary of the cardiovascular protective and therapeutic effects of DPP4 inhibitors through the past 5 years focusing on the molecular mechanisms beneath these effects. Additionally, based on the results summary presented here, future studies may be conducted to elucidate or illustrate some of these findings which can add clinical benefits towards management of diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Esraa M Zakaria
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Walaa M Tawfeek
- Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | | | | |
Collapse
|
16
|
You S, Miao M, Lu Z, Bao A, Du J, Che B, Xu T, Zhong C, Cao Y, Liu CF, Zhang Y, He J. Plasma-Soluble Dipeptidyl Peptidase 4 and Risk of Major Cardiovascular Events After Ischemic Stroke: Secondary Analysis of China Antihypertensive Trial in Acute Ischemic Stroke (CATIS). Neurology 2022; 99:e925-e934. [PMID: 35654589 DOI: 10.1212/wnl.0000000000200784] [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: 11/04/2021] [Accepted: 04/11/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Recent studies have suggested that plasma soluble dipeptidyl peptidase-4 (sDPP4) have important physiological effects, which may influence the prognosis of ischemic stroke. Our study aimed to examine the relationship between plasma sDDP4 levels and long-term clinical outcomes among acute ischemic stroke patients. METHODS Secondary analysis was conducted among 3,564 participants (2,270 men and 1,294 women) from the China Antihypertensive Trial in Acute Ischemic Stroke with baseline measurement of plasma sDPP4 levels. We evaluated the associations between plasma sDPP4 levels and 2-year clinical outcomes using logistic regression and Cox regression models. We further investigated the predictive utility of sDPP4 by calculating net reclassification index (NRI) and integrated discrimination improvement (IDI). RESULTS The highest plasma sDPP4 quartile was associated with lower risk of cardiovascular events (HR 0.62, 95% CI 0.45-0.87), recurrent stroke (HR 0.70, 95% CI 0.49-0.99), all-cause mortality (HR 0.62, 95% CI 0.44-0.87), stroke-specific mortality (HR 0.65, 95% CI 0.44-0.94) and poor functional outcomes (OR 0.66, 95% CI 0.53-0.82) at 2 years compared with the lowest sDPP4 category in multivariable models. The addition of plasma sDPP4 to conventional risk factors model significantly improved risk prediction of all outcomes. DISCUSSION In this study, we found that higher plasma sDPP4 levels in acute ischemic stroke patients were associated with decreased risks of cardiovascular events, recurrent stroke, all-cause mortality, and poor functional outcomes after ischemic stroke. These findings suggest that plasma sDPP4 may be a potential prognostic marker for initial risk stratification in patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Shoujiang You
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Mengyuan Miao
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Zian Lu
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Anran Bao
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Jigang Du
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Bizhong Che
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Yongjun Cao
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.,Institutes of Neuroscience, Soochow University, Suzhou 215123, China
| | - Chun-Feng Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.,Institutes of Neuroscience, Soochow University, Suzhou 215123, China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou 215123, China
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| |
Collapse
|
17
|
Kang SM, Park JH. Pleiotropic Benefits of DPP-4 Inhibitors Beyond Glycemic Control. Clin Med Insights Endocrinol Diabetes 2021; 14:11795514211051698. [PMID: 34733107 PMCID: PMC8558587 DOI: 10.1177/11795514211051698] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
Dipeptidyl peptidase (DPP)-4 inhibitors are oral anti-diabetic medications that block the activity of the ubiquitous enzyme DPP-4. Inhibition of this enzyme increases the level of circulating active glucagon-like peptide (GLP)-1 secreted from L-cells in the small intestine. GLP-1 increases the glucose level, dependent on insulin secretion from pancreatic β-cells; it also decreases the abnormally increased level of glucagon, eventually decreasing the blood glucose level in patients with type 2 diabetes. DPP-4 is involved in many physiological processes other than the degradation of GLP-1. Therefore, the inhibition of DPP-4 may have numerous effects beyond glucose control. In this article, we review the pleiotropic effects of DPP-4 inhibitors beyond glucose control, including their strong beneficial effects on the stress induced accelerated senescence of vascular cells, and the possible clinical implications of these effects.
Collapse
Affiliation(s)
- Seon Mee Kang
- Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Jeong Hyun Park
- Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| |
Collapse
|
18
|
Roy A, Sahoo J, Narayanan N, Merugu C, Kamalanathan S, Naik D. Dipeptidyl peptidase-4 inhibitor-induced autoimmune diseases: Current evidence. World J Diabetes 2021; 12:1426-1441. [PMID: 34630898 PMCID: PMC8472501 DOI: 10.4239/wjd.v12.i9.1426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/12/2021] [Accepted: 07/19/2021] [Indexed: 02/06/2023] Open
Abstract
Dipeptidyl peptidase-4 inhibitors (DPP-4i) have an important place in the management of type 2 diabetes. The DPP-4 enzyme is ubiquitously distributed throughout the human body and has multiple substrates through which it regulates several important physiological functions. DPP-4 regulates several immune functions, including T-cell activation, macrophage function, and secretion of cytokines. Studies have reported an increase in autoimmune diseases like bullous pemphigoid, inflammatory bowel disease, and arthritis with DPP-4i use. The relationship of DPP-4i and autoimmune diseases is a complex one and warrants further research into the effect of DPP-4 inhibition on the immune system to understand the pathogenesis more clearly. Whether a particular cluster of autoimmune diseases is associated with DPP-4i use remains an important contentious issue. Nevertheless, a heightened awareness from the clinicians is required to identify and treat any such diseases. Through this review, we explore the clinical and pathophysiological characteristics of this association in light of recent evidence.
Collapse
Affiliation(s)
- Ayan Roy
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, Jodhpur 342005, India
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Jayaprakash Sahoo
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Niya Narayanan
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Chandhana Merugu
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Sadishkumar Kamalanathan
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Dukhabandhu Naik
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| |
Collapse
|
19
|
Deng Y, Guo F, Han X, Huang X. Repetitive transcranial magnetic stimulation increases neurological function and endogenous neural stem cell migration via the SDF-1α/CXCR4 axis after cerebral infarction in rats. Exp Ther Med 2021; 22:1037. [PMID: 34373723 PMCID: PMC8343462 DOI: 10.3892/etm.2021.10469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 06/09/2021] [Indexed: 12/21/2022] Open
Abstract
Neural stem cell (NSC) migration is closely associated with brain development and is reportedly involved during recovery from ischaemic stroke. Chemokine signalling mediated by stromal cell-derived factor 1α (SDF-1α) and its receptor CXC chemokine receptor 4 (CXCR4) has been previously documented to guide the migration of NSCs. Although repetitive transcranial magnetic stimulation (rTMS) can increase neurological function in a rat stroke model, its effects on the migration of NSCs and associated underlying mechanism remain unclear. Therefore, the present study investigated the effects of rTMS on ischaemic stroke following middle cerebral artery occlusion (MCAO). All rats underwent rTMS treatment 24 h after MCAO. Neurological function, using modified Neurological Severity Scores and grip strength test and NSC migration, which were measured using immunofluorescence staining, were analysed at 7 and 14 days after MCAO, before the protein expression levels of the SDF-1α/CXCR4 axis was evaluated using western blot analysis. AMD3100, a CXCR4 inhibitor, was used to assess the effects of SDF-1α/CXCR4 signalling. In addition, neuronal survival was investigated using Nissl staining at 14 days after MCAO. It was revealed that rTMS increased the neurological recovery of rats with MCAO, facilitated the migration of NSC, augmented the expression levels of the SDF-1α/CXCR4 axis and decreased neuronal loss. Furthermore, the rTMS-induced positive responses were significantly abolished by AMD3100. Overall, these results indicated that rTMS conferred therapeutic neuroprotective properties, which can restore neurological function after ischaemic stroke, in a manner that may be associated with the activation of the SDF-1α/CXCR4 axis.
Collapse
Affiliation(s)
- Yuguo Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Feng Guo
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaohua Han
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaolin Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
20
|
Augestad IL, Dekens D, Karampatsi D, Elabi O, Zabala A, Pintana H, Larsson M, Nyström T, Paul G, Darsalia V, Patrone C. Normalisation of glucose metabolism by exendin-4 in the chronic phase after stroke promotes functional recovery in male diabetic mice. Br J Pharmacol 2021; 179:677-694. [PMID: 33973246 PMCID: PMC8820185 DOI: 10.1111/bph.15524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/30/2021] [Accepted: 04/27/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND PURPOSE Glucagon-like peptide-1 (GLP-1) receptor activation decreases stroke risk in people with Type 2 diabetes (T2D), while animal studies have shown the efficacy of this strategy to counteract stroke-induced acute brain damage. However, whether GLP-1 receptor activation also improves recovery in the chronic phase after stroke is unknown. We investigated whether post-acute, chronic administration of the GLP-1 receptor agonist, exendin-4, improves post-stroke recovery and examined possible underlying mechanisms in T2D and non-T2D mice. EXPERIMENTAL APPROACH We induced stroke via transient middle cerebral artery occlusion (tMCAO) in T2D/obese mice (8 months of high-fat diet) and age-matched controls. Exendin-4 was administered for 8 weeks from Day 3 post-tMCAO. We assessed functional recovery by weekly upper-limb grip strength tests. Insulin sensitivity and glycaemia were evaluated at 4 and 8 weeks post-tMCAO. Neuronal survival, stroke-induced neurogenesis, neuroinflammation, atrophy of GABAergic parvalbumin+ interneurons, post-stroke vascular remodelling and fibrotic scar formation were investigated by immunohistochemistry. KEY RESULTS Exendin-4 normalised T2D-induced impairment of forepaw grip strength recovery in correlation with normalised glycaemia and insulin sensitivity. Moreover, exendin-4 counteracted T2D-induced atrophy of parvalbumin+ interneurons and decreased microglia activation. Finally, exendin-4 normalised density and pericyte coverage of micro-vessels and restored fibrotic scar formation in T2D mice. In non-T2D mice, the exendin-4-mediated recovery was minor. CONCLUSION AND IMPLICATIONS Chronic GLP-1 receptor activation mediates post-stroke functional recovery in T2D mice by normalising glucose metabolism and improving neuroplasticity and vascular remodelling in the recovery phase. The results warrant clinical trial of GLP-1 receptor agonists for rehabilitation after stroke in T2D.
Collapse
Affiliation(s)
- Ingrid Lovise Augestad
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Doortje Dekens
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dimitra Karampatsi
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Osama Elabi
- Translational Neurology Group, Department of Clinical Sciences, Wallenberg Neuroscience Center, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Alexander Zabala
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hiranya Pintana
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Martin Larsson
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Nyström
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Sciences, Wallenberg Neuroscience Center, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Vladimer Darsalia
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cesare Patrone
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Chiazza F, Pintana H, Lietzau G, Nyström T, Patrone C, Darsalia V. The Stroke-Induced Increase of Somatostatin-Expressing Neurons is Inhibited by Diabetes: A Potential Mechanism at the Basis of Impaired Stroke Recovery. Cell Mol Neurobiol 2021; 41:591-603. [PMID: 32447613 PMCID: PMC7921043 DOI: 10.1007/s10571-020-00874-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022]
Abstract
Type 2 diabetes (T2D) hampers recovery after stroke, but the underling mechanisms are mostly unknown. In a recently published study (Pintana et al. in Clin Sci (Lond) 133(13):1367-1386, 2019), we showed that impaired recovery in T2D was associated with persistent atrophy of parvalbumin+ interneurons in the damaged striatum. In the current work, which is an extension of the abovementioned study, we investigated whether somatostatin (SOM)+ interneurons are also affected by T2D during the stroke recovery phase. C57Bl/6j mice were fed with high-fat diet or standard diet (SD) for 12 months and subjected to 30-min transient middle cerebral artery occlusion (tMCAO). SOM+ cell number/density in the striatum was assessed by immunohistochemistry 2 and 6 weeks after tMCAO in peri-infarct and infarct areas. This was possible by establishing a computer-based quantification method that compensates the post-stroke tissue deformation and the irregular cell distribution. SOM+ interneurons largely survived the stroke as seen at 2 weeks. Remarkably, 6 weeks after stroke, the number of SOM+ interneurons increased (vs. contralateral striatum) in SD-fed mice in both peri-infarct and infarct areas. However, this increase did not result from neurogenesis. T2D completely abolished this effect specifically in the in the infarct area. The results suggest that the up-regulation of SOM expression in the post-stroke phase could be related to neurological recovery and T2D could inhibit this process. We also present a new and precise method for cell counting in the stroke-damaged striatum that allows to reveal accurate, area-related effects of stroke on cell number.
Collapse
Affiliation(s)
- Fausto Chiazza
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Pharmaceutical Sciences, Università Degli Studi del Piemonte Orientale, Novara, Italy
| | - Hiranya Pintana
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Grazyna Lietzau
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cesare Patrone
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vladimer Darsalia
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Friedman-Levi Y, Liraz-Zaltsman S, Shemesh C, Rosenblatt K, Kesner EL, Gincberg G, Carmichael ST, Silva AJ, Shohami E. Pharmacological blockers of CCR5 and CXCR4 improve recovery after traumatic brain injury. Exp Neurol 2021; 338:113604. [PMID: 33453212 DOI: 10.1016/j.expneurol.2021.113604] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/27/2020] [Accepted: 01/09/2021] [Indexed: 11/16/2022]
Abstract
CCR5 and CXCR4 are structurally related chemokine receptors that belong to the superfamily of G-protein coupled receptors through which the HIV virus enters and infects cells. Both receptors are also related to HIV-associated neurocognitive disorders that include difficulties in concentration and memory, impaired executive functions, psychomotor slowing, depression and irritability, which are also hallmarks of the long-term sequelae of TBI. Moreover, A growing body of evidence attributes negative influences to CCR5 activation on cognition, particularly after stroke and traumatic brain injury (TBI). Here we investigated the effect of their blockage on motor and cognitive functions, on brain tissue loss and preservation and on some of the biochemical pathways involved. We examined the effect of maraviroc, a CCR5 antagonist used in HIV patients as a viral entry inhibitor, and of plerixafor (AMD3100), a CXCR4 antagonist used in cancer patients as an immune-modulator, on mice subjected to closed head injury (CHI). Mice were treated with maraviroc or plerixafor after CHI for the following 4 or 5 days, respectively. Neurobehavior was assessed according to the Neurological Severity Score; cognitive tests were performed by using the Y-maze, Barnes maze and the novel object recognition test; anxiety was evaluated with the open field test. The mice were sacrificed and brain tissues were collected for Western blot, pathological and immunohistochemical analyses. Both drugs enhanced tissue preservation in the cortex, hippocampus, periventricular areas, corpus callosum and striatum, and reduced astrogliosis)GFAP expression). They also increased the levels of synaptic cognition-related signaling molecules such as phosphorylated NR1 and CREB, and the synaptic plasticity protein PSD95. Both treatments also enhanced the expression of CCR5 and CXCR4 on different brain cell types. In summary, the beneficial effects of blocking CCR5 and CXCR4 after CHI suggest that the drugs used in this study, both FDA approved and in clinical use, should be considered for translational research in TBI patients.
Collapse
Affiliation(s)
- Yael Friedman-Levi
- Department of Pharmacology, the Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Sigal Liraz-Zaltsman
- Department of Pharmacology, the Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel; The Joseph Sagol Neuroscience Center, Sheba Medical Center, Israel; Institute for Health and Medical Professions, Department of Sports Therapy, Ono Academic College, Kiryat Ono, Israel.
| | - Chen Shemesh
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Israel.
| | | | - Efrat L Kesner
- Department of Pharmacology, the Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Galit Gincberg
- Department of Pharmacology, the Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, UCLA, LA, CA, USA.
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, LA, CA, USA.
| | - Esther Shohami
- Department of Pharmacology, the Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
23
|
Cuijpers I, Papageorgiou A, Carai P, Herwig M, Mügge A, Klein T, Hamdani N, Jones EAV, Heymans S. Linagliptin prevents left ventricular stiffening by reducing titin cleavage and hypophosphorylation. J Cell Mol Med 2021; 25:729-741. [PMID: 33295687 PMCID: PMC7812306 DOI: 10.1111/jcmm.16122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/12/2020] [Accepted: 11/08/2020] [Indexed: 12/25/2022] Open
Abstract
The metabolic syndrome (MetS) is an escalating problem worldwide, causing left ventricular stiffening, an early characteristic of diastolic dysfunction for which no treatment exists. As diastolic dysfunction and stiffening in MetS patients are associated with increased circulating dipeptidyl peptidase-4 (DPP-4) levels, we investigated whether the clinically approved DPP-4 inhibitor linagliptin reduces left ventricular stiffness in MetS-induced cardiac disease. Sixteen-week-old obese ZSF1 rats, displaying the MetS and left ventricular stiffness, received linagliptin-supplemented or placebo diet for four weeks. Linagliptin significantly reduced obesity, hyperlipidaemia, and hyperglycaemia and improved left ventricular relaxation. This improved relaxation was related to decreased cardiac fibrosis and cardiomyocyte passive stiffness (Fpassive ). The reduced Fpassive was the result of titin isoform switching from the stiff N2B to the more flexible N2BA and increased phosphorylation of total titin and specifically its N2Bus region (S4080 and S3391). Importantly, DPP-4 directly cleaved titin in vitro, resulting in an increased Fpassive , which was prevented by simultaneous administration of linagliptin. In conclusion, linagliptin improves left ventricular stiffness in obese ZSF1 rats by preventing direct DPP4-mediated titin cleavage, as well as by modulating both titin isoform levels and phosphorylation. Reducing left ventricular stiffness by administering linagliptin might prevent MetS-induced early diastolic dysfunction in human.
Collapse
Affiliation(s)
- Ilona Cuijpers
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyCARIM School for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Anna‐Pia Papageorgiou
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyCARIM School for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Paolo Carai
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
| | - Melissa Herwig
- Molecular Cardiology and Experimental CardiologyRuhr University BochumBochumGermany
- Department of CardiologySt. Josef‐HospitalRuhr University BochumBochumGermany
- Institute of PhysiologyRuhr University BochumBochumGermany
| | - Andreas Mügge
- Molecular Cardiology and Experimental CardiologyRuhr University BochumBochumGermany
- Department of CardiologySt. Josef‐HospitalRuhr University BochumBochumGermany
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Nazha Hamdani
- Molecular Cardiology and Experimental CardiologyRuhr University BochumBochumGermany
- Department of CardiologySt. Josef‐HospitalRuhr University BochumBochumGermany
- Institute of PhysiologyRuhr University BochumBochumGermany
- Department of Clinical PharmacologyRuhr University BochumBochumGermany
| | - Elizabeth A. V. Jones
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyCARIM School for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Stephane Heymans
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyCARIM School for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
- Holland Heart HouseICIN‐Netherlands Heart InstituteUtrechtThe Netherlands
| |
Collapse
|
24
|
Elrashidy RA, Hasan RA. Stromal cell-derived factor-1α predominantly mediates the ameliorative effect of linagliptin against cisplatin-induced testicular injury in adult male rats. Cytokine 2020; 136:155260. [DOI: 10.1016/j.cyto.2020.155260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 08/09/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022]
|
25
|
Affiliation(s)
- Lucy Ginn
- Transcriptional Networks in Lung Cancer Group Cancer Research UK Manchester Institute University of Manchester Manchester UK
- Cancer Research UK Lung Cancer Centre of Excellence At Manchester and University College London England UK
| | - Manuela La Montagna
- Transcriptional Networks in Lung Cancer Group Cancer Research UK Manchester Institute University of Manchester Manchester UK
- Cancer Research UK Lung Cancer Centre of Excellence At Manchester and University College London England UK
| | - Qinghua Wu
- College of Life Science Yangtze University Jingzhou Hubei China
- Department of Chemistry Faculty of Science University of Hradec Kralove Hradec Kralove East Bohemia Czech Republic
| | - Lei Shi
- Transcriptional Networks in Lung Cancer Group Cancer Research UK Manchester Institute University of Manchester Manchester UK
- Cancer Research UK Lung Cancer Centre of Excellence At Manchester and University College London England UK
| |
Collapse
|
26
|
El-Deeb OS, Soliman GM, Elesawy RO. Linagliptin, the dipeptidyl peptidase-4 enzyme inhibitor, lessens CHOP and GRP78 biomarkers levels in cisplatin-induced neurobehavioral deficits: A possible restorative gateway. J Biochem Mol Toxicol 2020; 34:e22541. [PMID: 32567747 DOI: 10.1002/jbt.22541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/15/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
Cisplatin (CP) is a cornerstone chemotherapeutic agent, however, its neurotoxicity is a chief cause of its limited usage. Linagliptin, which is a dipeptidyl peptidase-4 enzyme inhibitor, has exhibited considerable neuroprotective potential. We aimed to evaluate the linagliptin modulatory effects on endoplasmic reticulum (ER) stress, redox status, and apoptosis in CP-induced neurotoxicity. Thirty mice were allocated equally into the control group, Group II: CP group, and Group III: linagliptin treated CP group. All groups were subjected to the measurement of hippocampal messenger RNA gene expression of glucose-regulated protein-78 and C/EBP homologous protein (CHOP). Peroxisome proliferator-activated receptor γ coactivator 1α and cleaved caspase-3 levels were assessed by the enzyme-linked immunosorbent assay technique while malondialdehyde, reduced glutathione levels and superoxide dismutase activity were detected spectrophotometrically. Linagliptin ameliorated ER stress and enhanced antioxidant status with cognitive function improvement. Linagliptin may be considered a promising neuroprotective agent owing to its ability to reduce ER/oxidative stress.
Collapse
Affiliation(s)
- Omnia S El-Deeb
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Gehan M Soliman
- Histology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rasha O Elesawy
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
27
|
Augestad IL, Pintana H, Larsson M, Krizhanovskii C, Nyström T, Klein T, Darsalia V, Patrone C. Regulation of Glycemia in the Recovery Phase After Stroke Counteracts the Detrimental Effect of Obesity-Induced Type 2 Diabetes on Neurological Recovery. Diabetes 2020; 69:1961-1973. [PMID: 32540876 DOI: 10.2337/db20-0095] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/09/2020] [Indexed: 11/13/2022]
Abstract
The interplay between obesity and type 2 diabetes (T2D) in poststroke recovery is unclear. Moreover, the impact of glucose control during the chronic phase after stroke is undetermined. We investigated whether obesity-induced T2D impairs neurological recovery after stroke by using a clinically relevant experimental design. We also investigated the potential efficacy of two clinically used T2D drugs: the dipeptidyl peptidase 4 inhibitor linagliptin and the sulfonylurea glimepiride. We induced transient middle cerebral artery occlusion (tMCAO) in T2D/obese mice (after 7 months of high-fat diet [HFD]) and age-matched controls. After stroke, we replaced HFD with standard diet for 8 weeks to mimic the poststroke clinical situation. Linagliptin or glimepiride were administered daily from 3 days after tMCAO for 8 weeks. We assessed neurological recovery weekly by upper-limb grip strength. Brain damage, neuroinflammation, stroke-induced neurogenesis, and atrophy of parvalbumin-positive (PV+) interneurons were quantified by immunohistochemistry. T2D/obesity impaired poststroke neurological recovery in association with hyperglycemia, neuroinflammation, and atrophy of PV+ interneurons. Both drugs counteracted these effects. In nondiabetic mice, only linagliptin accelerated recovery. These findings shed light on the interplay between obesity and T2D in stroke recovery. Moreover, they promote the use of rehabilitative strategies that are based on efficacious glycemia regulation, even if initiated days after stroke.
Collapse
Affiliation(s)
- Ingrid Lovise Augestad
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hiranya Pintana
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Martin Larsson
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Camilla Krizhanovskii
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Research, Södertälje Hospital, Karolinska Institutet, Södertälje, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Vladimer Darsalia
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cesare Patrone
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
Abstract
Previous studies have demonstrated that individuals with type 2 diabetes mellitus (T2DM) have a two- to fourfold propensity to develop cardiovascular disease (CVD) than nondiabetic population, making CVD a major cause of death and disability among people with T2DM. The present treatment options for management of diabetes propose the earlier and more frequent use of new antidiabetic drugs that could control hyperglycaemia and reduce the risk of cardiovascular events. Findings from basic and clinical studies pointed out DPP-4 inhibitors as potentially novel pharmacological tools for cardioprotection. There is a growing body of evidence suggesting that these drugs have ability to protect the heart against acute ischaemia-reperfusion injury as well as reduce the size of infarction. Consequently, the prevention of degradation of the incretin hormones by the use of DPP-4 inhibitors represents a new strategy in the treatment of patients with T2DM and reduction of CV events in these patients. Here, we discuss the cardioprotective effects of DPP-4 inhibitors as well as proposed pathways that these hypoglycaemic agents target in the cardiovascular system.
Collapse
|
29
|
Ferrari F, Moretti A, Villa RF. The treament of hyperglycemia in acute ischemic stroke with incretin-based drugs. Pharmacol Res 2020; 160:105018. [PMID: 32574826 DOI: 10.1016/j.phrs.2020.105018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/21/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Stroke is a major cause of mortality and morbidity worldwide. Considerable experimental and clinical evidence suggests that both diabetes mellitus (DM) and post-stroke hyperglycemia are associated with increased mortality rate and worsened clinical conditions in acute ischemic stroke (AIS) patients. Insulin treatment does not seem to provide convincing benefits for these patients, therefore prompting a change of strategy. The selective agonists of Glucagon-Like Peptide-1 Receptors (GLP-1Ras) and the Inhibitors of Dipeptidyl Peptidase-IV (DPP-IVIs, gliptins) are two newer classes of glucose-lowering drugs used for the treatment of DM. This review examines in detail the rationale for their development and the physicochemical, pharmacokinetic and pharmacodynamic properties and clinical activities. Emphasis will be placed on their neuroprotective effects at cellular and molecular levels in experimental models of acute cerebral ischemia. In perspective, an adequate basis does exist for a novel therapeutic approach to hyperglycemia in AIS patients through the additive treatment with GLP-1Ras plus DPP-IVIs.
Collapse
Affiliation(s)
- Federica Ferrari
- Department of Advanced Diagnostic and Therapeutic Technologies, Section of Neuroradiology, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore 3, 20162 Milano, Italy; Departments of Biology-Biotechnology and Chemistry, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Antonio Moretti
- Departments of Biology-Biotechnology and Chemistry, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Roberto Federico Villa
- Departments of Biology-Biotechnology and Chemistry, Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy.
| |
Collapse
|
30
|
Poupon-Bejuit L, Rocha-Ferreira E, Thornton C, Hagberg H, Rahim AA. Neuroprotective Effects of Diabetes Drugs for the Treatment of Neonatal Hypoxia-Ischemia Encephalopathy. Front Cell Neurosci 2020; 14:112. [PMID: 32435185 PMCID: PMC7218053 DOI: 10.3389/fncel.2020.00112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/08/2020] [Indexed: 12/15/2022] Open
Abstract
The perinatal period represents a time of great vulnerability for the developing brain. A variety of injuries can result in death or devastating injury causing profound neurocognitive deficits. Hypoxic-ischemic neonatal encephalopathy (HIE) remains the leading cause of brain injury in term infants during the perinatal period with limited options available to aid in recovery. It can result in long-term devastating consequences with neurologic complications varying from mild behavioral deficits to severe seizure, intellectual disability, and/or cerebral palsy in the newborn. Despite medical advances, the only viable option is therapeutic hypothermia which is classified as the gold standard but is not used, or may not be as effective in preterm cases, infection-associated cases or low resource settings. Therefore, alternatives or adjunct therapies are urgently needed. Ongoing research continues to advance our understanding of the mechanisms contributing to perinatal brain injury and identify new targets and treatments. Drugs used for the treatment of patients with type 2 diabetes mellitus (T2DM) have demonstrated neuroprotective properties and therapeutic efficacy from neurological sequelae following HIE insults in preclinical models, both alone, or in combination with induced hypothermia. In this short review, we have focused on recent findings on the use of diabetes drugs that provide a neuroprotective effect using in vitro and in vivo models of HIE that could be considered for clinical translation as a promising treatment.
Collapse
Affiliation(s)
| | - Eridan Rocha-Ferreira
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claire Thornton
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Henrik Hagberg
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ahad A. Rahim
- UCL School of Pharmacy, University College London, London, United Kingdom
| |
Collapse
|
31
|
Shao S, Xu Q, Yu X, Pan R, Chen Y. Dipeptidyl peptidase 4 inhibitors and their potential immune modulatory functions. Pharmacol Ther 2020; 209:107503. [PMID: 32061923 PMCID: PMC7102585 DOI: 10.1016/j.pharmthera.2020.107503] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/30/2020] [Indexed: 12/25/2022]
Abstract
Dipeptidyl peptidase 4 (DPP4) inhibitors (DPP4is) are oral anti-diabetic drugs (OADs) for the treatment of type 2 diabetes mellitus (T2DM) through inhibiting the degradation of incretin peptides. Numerous investigations have been focused on the effects of DPP4is on glucose homeostasis. However, there are limited evidences demonstrating their Potential modulatory functions in the immune system. DPP4, originally known as the lymphocyte cell surface protein CD26, is widely expressed in many types of immune cells including CD4(+) and CD8(+) T cells, B cells, NK cells, dendritic cells, and macrophages; and regulate the functions of these cells. In addition, DPP4 is capable of modulating plenty of cytokines, chemokines and peptide hormones. Accordingly, DPP4/CD26 is speculated to be involved in various immune/inflammatory diseases and DPP4is may become a new drug class applied in these diseases. This review focuses on the regulatory effects of DPP4is on immune functions and their possible underlying mechanisms. Further clinical studies will be necessitated to fully evaluate the administration of DPP4is in diabetic patients with or without immune diseases.
Collapse
Affiliation(s)
- Shiying Shao
- Division of Endocrinology, Department of Internal Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - QinQin Xu
- Division of Endocrinology, Department of Internal Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Xuefeng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Ruping Pan
- Department of Nuclear Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Yong Chen
- Division of Endocrinology, Department of Internal Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China.
| |
Collapse
|
32
|
Zanozina OV, Sorokina YA, Lovtsova LV. Glucose-lowering medication selection in patients with diabetes and acute cerebrovascular accident. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2020. [DOI: 10.15829/1728-8800-2020-1-2378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- O. V. Zanozina
- Privolzhsky Research Medical University; N.A. Semashko Nizhny Novgorod Regional Clinical Hospital
| | | | | |
Collapse
|
33
|
Lietzau G, Magni G, Kehr J, Yoshitake T, Candeias E, Duarte AI, Pettersson H, Skogsberg J, Abbracchio MP, Klein T, Nyström T, Ceruti S, Darsalia V, Patrone C. Dipeptidyl peptidase-4 inhibitors and sulfonylureas prevent the progressive impairment of the nigrostriatal dopaminergic system induced by diabetes during aging. Neurobiol Aging 2020; 89:12-23. [PMID: 32143981 DOI: 10.1016/j.neurobiolaging.2020.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 02/08/2023]
Abstract
The nigrostriatal dopaminergic system (NDS) controls motor activity, and its impairment during type 2 diabetes (T2D) progression could increase Parkinson's disease risk in diabetics. If so, whether glycemia regulation prevents this impairment needs to be addressed. We investigated whether T2D impairs the NDS and whether dipeptidyl peptidase-4 inhibition (DPP-4i; a clinical strategy against T2D but also neuroprotective in animal models) prevents this effect, in middle-aged mice. Neither T2D (induced by 12 months of high-fat diet) nor aging (14 months) changed striatal dopamine content assessed by high-performance liquid chromatography. However, T2D reduced basal and amphetamine-stimulated striatal extracellular dopamine, assessed by microdialysis. Both the DPP-4i linagliptin and the sulfonylurea glimepiride (an antidiabetic comparator unrelated to DPP-4i) counteracted these effects. The functional T2D-induced effects did not correlate with NDS neuronal/glial alterations. However, aging itself affected striatal neurons/glia, and the glia effects were counteracted mainly by DPP-4i. These findings show NDS functional pathophysiology in T2D and suggest the preventive use of two unrelated anti-T2D drugs. Moreover, DPP-4i counteracted striatal age-related glial alterations suggesting striatal rejuvenation properties.
Collapse
Affiliation(s)
- Grazyna Lietzau
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Giulia Magni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Jan Kehr
- Pronexus Analytical AB, Bromma, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Takashi Yoshitake
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Emanuel Candeias
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana I Duarte
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Hans Pettersson
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Maria P Abbracchio
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Thomas Nyström
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stefania Ceruti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Vladimer Darsalia
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Cesare Patrone
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
34
|
DPP-4 Inhibitor Linagliptin is Neuroprotective in Hyperglycemic Mice with Stroke via the AKT/mTOR Pathway and Anti-apoptotic Effects. Neurosci Bull 2019; 36:407-418. [PMID: 31808042 DOI: 10.1007/s12264-019-00446-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/10/2019] [Indexed: 12/11/2022] Open
Abstract
Dipeptidyl peptidase 4 (DPP-4) inhibitors have been shown to have neuroprotective effects in diabetic patients suffering from stroke, but less research has focused on patients with mild hyperglycemia below the threshold for a diagnosis of diabetes. In this investigation, a hyperglycemic mouse model was generated by intraperitoneal injection of streptozotocin and then subjected to focal cerebral ischemia. We demonstrated that the DPP-4 inhibitor linagliptin significantly decreased the infarct volume, reduced neuronal cell death, decreased inflammation, and improved neurological deficit compared with control mice. Linagliptin up-regulated the expression of p-Akt and p-mTOR and regulated the apoptosis factors Bcl-2, Bax, and caspase 9. Taken together, these results suggest that linagliptin exerts a neuroprotective action likely through activation of the Akt/mTOR pathway along with anti-apoptotic and anti-inflammatory mechanisms. Therefore, linagliptin may be considered as a therapeutic treatment for stroke patients with mild hyperglycemia.
Collapse
|
35
|
Kim TH, Lee K, Park IB, Choi CS, Ahn TH, Lee DH. The effects of DPP4 inhibitors on the levels of plasma catecholamines and their metabolites in patients with type 2 diabetes. Diabetes Res Clin Pract 2019; 156:107832. [PMID: 31513823 DOI: 10.1016/j.diabres.2019.107832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/11/2019] [Accepted: 08/29/2019] [Indexed: 01/21/2023]
Abstract
AIMS Dipeptidyl peptidase 4 inhibitors (DPP4Is) can increase sympathetic activity. We aimed to evaluate the direct association between serum DPP4 activity and sympathetic activity in humans. METHODS Fasting serum DPP4 activity and plasma levels of catecholamines and their metabolites were measured in 211 patients with type 2 diabetes mellitus (T2DM) treated with DPP4I (n = 146) or non-DPP4I therapy (n = 65) and in healthy control subjects (n = 30). RESULTS Although there were no differences in plasma levels of catecholamines and their metabolites between the DPP4I and non-DPP4I groups, the levels in both of these groups were lower than those in the healthy control group. In DPP4I-treated patients, serum DPP4 activity showed an inverse correlation with plasma levels of norepinephrine (NE) (r = -0.339, p < 0.01), metanephrine (MET) (r = -0.251, p < 0.01) and normetanephrine (r = -0.312, p < 0.001). In addition, plasma MET level showed a weak inverse correlation with serum DPP4 activity in the combined T2DM group. In DPP4I-treated patients, the inverse correlation between DPP4 activity and plasma NE remained significant even after multiple adjustments. CONCLUSIONS Our results suggest that although sympathetic activity is lower in patients with T2DM, the greater the suppression of DPP4 activity by DPP4I therapy, the greater the increase in sympathetic activity is, which may have clinical implications in high risk T2DM patients.
Collapse
Affiliation(s)
- Tae Hun Kim
- Gachon University, School of Medicine, Incheon, Republic of Korea
| | - Kiyoung Lee
- Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Ie Byung Park
- Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Cheol Soo Choi
- Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Tae Hoon Ahn
- Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea.
| | - Dae Ho Lee
- Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea.
| |
Collapse
|
36
|
Farr OM, Pilitsi E, Mantzoros CS. Of mice and men: incretin actions in the central nervous system. Metabolism 2019; 98:121-135. [PMID: 31173757 DOI: 10.1016/j.metabol.2019.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023]
Abstract
Incretins have risen to the forefront of therapies for obesity and related metabolic complications, primarily because of their efficacy and relatively few side effects. Importantly, their efficacy in altering energy balance and decreasing body weight is apparently through actions in the central nervous system (CNS); the latter may have implications beyond obesity per se, i.e. in other disease states associated with obesity including CNS-related disorders. Here, we first describe the role of the CNS in energy homeostasis and then the current state of knowledge in terms of incretin physiology, pathophysiology and efficacy in preclinical and clinical studies. In the future, more clinical studies are needed to fully map mechanistic pathways underlying incretin actions and outcomes in the human CNS. Additionally, future research will likely lead to the discovery of additional novel incretins and/or more efficacious medications with less side effects through the improvement of current compounds with properties that would allow them to have more favorable pharmacokinetic and pharmacodynamic profiles and/or by combining known and novel incretins into safe and more efficacious combination therapies leading ultimately to more tangible benefits for our patients.
Collapse
Affiliation(s)
- Olivia M Farr
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, United States of America.
| | - Eleni Pilitsi
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, United States of America
| | - Christos S Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215, United States of America; Section of Endocrinology, VA Boston Healthcare System, Boston, MA 02130, United States of America
| |
Collapse
|
37
|
Tsivgoulis G, Katsanos AH, Mavridis D, Lambadiari V, Roffe C, Macleod MJ, Sevcik P, Cappellari M, Nevšímalová M, Toni D, Ahmed N. Association of Baseline Hyperglycemia With Outcomes of Patients With and Without Diabetes With Acute Ischemic Stroke Treated With Intravenous Thrombolysis: A Propensity Score-Matched Analysis From the SITS-ISTR Registry. Diabetes 2019; 68:1861-1869. [PMID: 31217175 DOI: 10.2337/db19-0440] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/17/2019] [Indexed: 11/13/2022]
Abstract
Available data from observational studies on the association of admission hyperglycemia (aHG) with outcomes of patients with acute ischemic stroke (AIS) treated with intravenous thrombolysis (IVT) are contradictory, especially when stratified by diabetes mellitus (DM) history. We assessed the association of aHG (≥144 mg/dL) with outcomes stratified by DM history using propensity score-matched (PSM) data from the SITS-ISTR. The primary safety outcome was symptomatic intracranial hemorrhage (SICH); 3-month functional independence (FI) (modified Rankin Scale [mRS] scores 0-2) represented the primary efficacy outcome. Patients with and without aHG did not differ in baseline characteristics both in the non-DM (n = 12,318) and DM (n = 6,572) PSM subgroups. In the non-DM group, patients with aHG had lower 3-month FI rates (53.3% vs. 57.9%, P < 0.001), higher 3-month mortality rates (19.2% vs. 16.0%, P < 0.001), and similar symptomatic intracerebral hemorrhage (SICH) rates (1.7% vs. 1.8%, P = 0.563) compared with patients without aHG. Similarly, in the DM group, patients with aHG had lower rates of 3-month favorable functional outcome (mRS scores 0-1, 34.1% vs. 39.3%, P < 0.001) and FI (48.2% vs. 52.5%, P < 0.001), higher 3-month mortality rates (23.7% vs. 19.9%, P < 0.001), and similar SICH rates (2.2% vs. 2.7%, P = 0.224) compared with patients without aHG. In conclusion, aHG was associated with unfavorable 3-month clinical outcomes in patients with and without DM and AIS treated with IVT.
Collapse
Affiliation(s)
- Georgios Tsivgoulis
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN
| | - Aristeidis H Katsanos
- Second Department of Neurology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurology, St. Josef-Hospital, Ruhr University, Bochum, Germany
| | - Dimitris Mavridis
- Department of Primary Education, University of Ioannina, Ioannina, Greece
- Faculté de Médecine, Université Paris Descartes, Paris, France
| | - Vaia Lambadiari
- Research Unit and Diabetes Center, Second Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christine Roffe
- University Hospital of North Midlands, Stoke-on-Trent, U.K., and Keele University, Keele, U.K
| | - Mary Joan Macleod
- Division of Applied Medicine, University of Aberdeen, Foresterhill, U.K
| | - Petr Sevcik
- Department of Neurology, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Manuel Cappellari
- Stroke Unit, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | | | - Danilo Toni
- Neurovascular Unit, Policlinico Umberto I, Department of Human Neurosciences, University of Rome, "La Sapienza," Rome, Italy
| | - Niaz Ahmed
- Department of Neurology, Karolinska University Hospital Solna, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
38
|
Neuroprotective Properties of Linagliptin: Focus on Biochemical Mechanisms in Cerebral Ischemia, Vascular Dysfunction and Certain Neurodegenerative Diseases. Int J Mol Sci 2019; 20:ijms20164052. [PMID: 31434198 PMCID: PMC6719127 DOI: 10.3390/ijms20164052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 02/08/2023] Open
Abstract
Linagliptin is a representative of dipeptidyl peptidase 4 (DPP-4) inhibitors which are registered and used effectively in a treatment of diabetes mellitus type 2. They increase the levels of active forms of endogenous incretins such as GLP-1 and GIP by inhibiting their enzymatic decomposition. Scientific reports suggest beneficial effects of linagliptin administration via immunological and biochemical pathways involved in neuroprotective processes of CNS. Linagliptin’s administration leads to a decrease in the concentration of proinflammatory factors such as: TNF-α, IL-6 and increases the number of anti-inflammatory patrolling monocytes CX3CR1bright. Significant reduction in Aβ42 level has been associated with the use of linagliptin implying potential application in Alzheimer’s disease. Linagliptin improved vascular functions by increasing production of nitric oxide (NO) and limiting concentration of apolipoprotein B. Linagliptin-induced decrease in macrophages infiltration may provide improvement in atheromatous plaque stabilization. Premedication with linagliptin increases neuron’s survival after stroke and augments neuronal stem cells proliferation. It seems to be connected with SDF-1α/CXCR4 signaling pathway. Linagliptin prevented abnormal proliferation and migration of rat brain microvascular endothelial cells in a state of hypoperfusion via SIRT1/HIF-1α/VEGF pathway. The article presents a summary of the studies assessing neuroprotective properties of linagliptin with special emphasis on cerebral ischemia, vascular dysfunction and neurodegenerative diseases.
Collapse
|
39
|
Trettel F, Di Castro MA, Limatola C. Chemokines: Key Molecules that Orchestrate Communication among Neurons, Microglia and Astrocytes to Preserve Brain Function. Neuroscience 2019; 439:230-240. [PMID: 31376422 DOI: 10.1016/j.neuroscience.2019.07.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/01/2019] [Accepted: 07/19/2019] [Indexed: 12/19/2022]
Abstract
In the CNS, chemokines and chemokine receptors are involved in pleiotropic physiological and pathological activities. Several evidences demonstrated that chemokine signaling in the CNS plays key homeostatic roles and, being expressed on neurons, glia and endothelial cells, chemokines mediate the bidirectional cross-talk among parenchymal cells. An efficient communication between neurons and glia is crucial to establish and maintain a healthy brain environment which ensures normal functionality. Glial cells behave as active sensors of environmental changes induced by neuronal activity or detrimental insults, supporting and exerting neuroprotective activities. In this review we summarize the evidence that chemokines (CXCL12, CX3CL1, CXCL16 and CCL2) modulate neuroprotective processes upon different noxious stimuli and participate to orchestrate neurons-microglia-astrocytes action to preserve and limit brain damage. This article is part of a Special Issue entitled: Honoring Ricardo Miledi - outstanding neuroscientist of XX-XXI centuries.
Collapse
Affiliation(s)
- Flavia Trettel
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Maria Amalia Di Castro
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy; IRCCS Neuromed, Via Atinense 19, 86077, Pozzilli, Italy
| |
Collapse
|
40
|
Nash B, Tarn K, Irollo E, Luchetta J, Festa L, Halcrow P, Datta G, Geiger JD, Meucci O. Morphine-Induced Modulation of Endolysosomal Iron Mediates Upregulation of Ferritin Heavy Chain in Cortical Neurons. eNeuro 2019; 6:ENEURO.0237-19.2019. [PMID: 31300544 PMCID: PMC6675873 DOI: 10.1523/eneuro.0237-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 01/01/2023] Open
Abstract
HIV-associated neurocognitive disorders (HAND) remain prevalent and are aggravated by µ-opioid use. We have previously shown that morphine and other µ-opioids may contribute to HAND by inhibiting the homeostatic and neuroprotective chemokine receptor CXCR4 in cortical neurons, and this novel mechanism depends on upregulation of the protein ferritin heavy chain (FHC). Here, we examined the cellular events and potential mechanisms involved in morphine-mediated FHC upregulation using rat cortical neurons of either sex in vitro and in vivo. Morphine dose dependently increased FHC protein levels in primary neurons through µ-opioid receptor (µOR) and Gαi-protein signaling. Cytoplasmic FHC levels were significantly elevated, but nuclear FHC levels and FHC gene expression were unchanged. Morphine-treated rats also displayed increased FHC levels in layer 2/3 neurons of the prefrontal cortex. Importantly, both in vitro and in vivo FHC upregulation was accompanied by loss of mature dendritic spines, which was also dependent on µOR and Gαi-protein signaling. Moreover, morphine upregulated ferritin light chain (FLC), a component of the ferritin iron storage complex, suggesting that morphine altered neuronal iron metabolism. Indeed, prior to FHC upregulation, morphine increased cytoplasmic labile iron levels as a function of decreased endolysosomal iron. In line with this, chelation of endolysosomal iron (but not extracellular iron) blocked morphine-induced FHC upregulation and dendritic spine reduction, whereas iron overloading mimicked the effect of morphine on FHC and dendritic spines. Overall, these data demonstrate that iron mediates morphine-induced FHC upregulation and consequent dendritic spine deficits and implicate endolysosomal iron efflux to the cytoplasm in these effects.
Collapse
Affiliation(s)
- Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Kevin Tarn
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Jared Luchetta
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Lindsay Festa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Peter Halcrow
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102
| |
Collapse
|
41
|
Hu J, Yang C, Wang H, Li J, Tan X, Wang J, Zhang B, Zhao Y. An up-to-date evaluation of alogliptin benzoate for the treatment of type 2 diabetes. Expert Opin Pharmacother 2019; 20:1679-1687. [PMID: 31335214 DOI: 10.1080/14656566.2019.1645124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: A growth in the market for anti-diabetic drugs, along with an ever-increasing population suffering from type 2 diabetes mellitus (T2DM), requires a critical re-evaluation of anti-diabetic drugs used for a long time, in order to provide up-to-date practical prescribing information for clinicians. Alogliptin benzoate was firstly approved in 2010 in Japan for T2DM, both as a monotherapy or in combination with other anti-diabetic drugs. Areas covered: This article provides a comprehensive review of the latest data on alogliptin benzoate, including hypoglycemic activity and safety. Expert opinion: The cumulative evidence for alogliptin benzoate is robust with regards to glycemic efficacy and safety. Low hypoglycemia risks and weight changes support its consideration as a first-line medication for T2DM, either as a monotherapy or in combination therapy with other anti-diabetic drugs such as metformin. Ongoing trials will look to better analyze and address its safety and efficacy in pediatric patients and expand our clinical knowledge of this medication.
Collapse
Affiliation(s)
- Jingbo Hu
- Institute of Drug Discovery Technology, Ningbo University , Ningbo , China
| | - Chunlin Yang
- Department of pharmacy, Ningbo University affiliated Yangming Hospital , Yuyao , China
| | - Hongbo Wang
- Department of pharmacy, Ningbo University affiliated Yangming Hospital , Yuyao , China
| | - Jing Li
- Department of pharmacy, Ningbo University affiliated Yangming Hospital , Yuyao , China
| | - Xueying Tan
- College of pharmacy, Zhejiang Pharmaceutical College , Ningbo , China
| | - Jinhui Wang
- Institute of Drug Discovery Technology, Ningbo University , Ningbo , China
| | - Bin Zhang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, College of Food and Pharmaceutical Sciences, Ningbo University , Ningbo , China
| | - Yufen Zhao
- Institute of Drug Discovery Technology, Ningbo University , Ningbo , China
| |
Collapse
|
42
|
Larsson M, Patrone C, von Euler M, Holst JJ, Nathanson D. GLP-1 secretion in acute ischemic stroke: association with functional outcome and comparison with healthy individuals. Cardiovasc Diabetol 2019; 18:91. [PMID: 31307484 PMCID: PMC6628501 DOI: 10.1186/s12933-019-0896-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/09/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Glucagon-like peptide-1 (GLP-1) treatment has been shown to reduce stroke incidence in diabetes and also to be neuroprotective in experimental stroke models. The prognostic value of endogenous levels of GLP-1 in the recovery phase after stroke remains to be elucidated. The aim of the study was to investigate the potential association between GLP-1 levels and functional outcome after stroke and to determine whether GLP-1 is altered in the acute phase of stroke compared to 3 months post stroke and to healthy controls. METHODS Fasting GLP-1 was measured on hospital day 2-4 in patients without previously known diabetes (n = 59) that received recombinant tissue plasminogen activator (rtPA) for ischemic stroke. Fasting GLP-1 was measured again after 3 months and neurologic outcome was measured as modified Rankin Scale (mRS). mRS ≥ 2 was considered as unfavorable outcome. A control group of healthy individuals (n = 27) was recruited and their fasting GLP-1 was measured. RESULTS Fasting GLP-1 was higher in the patients that suffered a stroke compared to healthy controls (25.1 vs. 18.0 pmol/L; p = 0.004). The GLP-1 levels did not change significantly at the 3-month follow up OGTT (25.8 vs. 25.6; p = 0.80). There was no significant association between GLP-1 levels and unfavorable mRS (OR 1.03, 95% CI 0.95-1.12, p = 0.50). CONCLUSIONS Endogenous GLP-1 levels in patients that recently suffered an ischemic stroke are higher than in healthy controls and remained unchanged at the 3 months follow-up, possibly indicating an elevation of the levels of GLP-1 already pre-stroke. However, no association between endogenous GLP-1 and functional outcome of stroke 3 months post stroke was found.
Collapse
Affiliation(s)
- Martin Larsson
- Department of Clinical Science and Education at Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden.
| | - Cesare Patrone
- Department of Clinical Science and Education at Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden
| | - Mia von Euler
- Department of Clinical Science and Education at Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden
| | - Jens J Holst
- Department of Biomedical Sciences and NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David Nathanson
- Department of Clinical Science and Education at Södersjukhuset, Karolinska Institutet, Sjukhusbacken 10, 118 83, Stockholm, Sweden
| |
Collapse
|
43
|
Obesity-induced type 2 diabetes impairs neurological recovery after stroke in correlation with decreased neurogenesis and persistent atrophy of parvalbumin-positive interneurons. Clin Sci (Lond) 2019; 133:1367-1386. [PMID: 31235555 DOI: 10.1042/cs20190180] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/08/2019] [Accepted: 06/24/2019] [Indexed: 01/11/2023]
Abstract
Type 2 diabetes (T2D) hampers stroke recovery though largely undetermined mechanisms. Few preclinical studies have investigated the effect of genetic/toxin-induced diabetes on long-term stroke recovery. However, the effects of obesity-induced T2D are mostly unknown. We aimed to investigate whether obesity-induced T2D worsens long-term stroke recovery through the impairment of brain's self-repair mechanisms - stroke-induced neurogenesis and parvalbumin (PV)+ interneurons-mediated neuroplasticity. To mimic obesity-induced T2D in the middle-age, C57bl/6j mice were fed 12 months with high-fat diet (HFD) and subjected to transient middle cerebral artery occlusion (tMCAO). We evaluated neurological recovery by upper-limb grip strength at 1 and 6 weeks after tMCAO. Gray and white matter damage, stroke-induced neurogenesis, and survival and potential atrophy of PV-interneurons were quantitated by immunohistochemistry (IHC) at 2 and 6 weeks after tMCAO. Obesity/T2D impaired neurological function without exacerbating brain damage. Moreover, obesity/T2D diminished stroke-induced neural stem cell (NSC) proliferation and neuroblast formation in striatum and hippocampus at 2 weeks after tMCAO and abolished stroke-induced neurogenesis in hippocampus at 6 weeks. Finally, stroke resulted in the atrophy of surviving PV-interneurons 2 weeks after stroke in both non-diabetic and obese/T2D mice. However, after 6 weeks, this effect selectively persisted in obese/T2D mice. We show in a preclinical setting of clinical relevance that obesity/T2D impairs neurological functions in the stroke recovery phase in correlation with reduced neurogenesis and persistent atrophy of PV-interneurons, suggesting impaired neuroplasticity. These findings shed light on the mechanisms behind impaired stroke recovery in T2D and could facilitate the development of new stroke rehabilitative strategies for obese/T2D patients.
Collapse
|
44
|
Ahn CH, Lim S. Effects of Thiazolidinedione and New Antidiabetic Agents on Stroke. J Stroke 2019; 21:139-150. [PMID: 31161759 PMCID: PMC6549069 DOI: 10.5853/jos.2019.00038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with hyperglycemia are at a high risk of cardio- and cerebrovascular diseases. Diabetes patients also have poor outcomes after cerebrovascular disease development. Several classes of drugs are used for diabetes management in clinical practice. Thiazolidinedione (TZD) was introduced in the late 1990s, and new antidiabetic agents have been introduced since 2000. After issues with rosiglitazone in 2007, the U.S. Food and Drug Administration strongly recommended that trials investigating cardiovascular risk associated with new antidiabetic medications should be conducted before drug approval in the United States, to prove the safety of these new drugs and to determine their superiority to previous medications. Currently, results are available from two studies with TZD focusing on cardiovascular diseases, including stroke, and from 12 cardiovascular outcome trials focusing on major adverse cardiovascular events associated with new antidiabetic agents (four with dipeptidyl peptidase-4 inhibitors, three with sodium-glucose cotransporter-2 inhibitors, and five with glucagon-like peptide-1 analogues). These studies showed different results for primary cardiovascular outcomes and stroke prevention. It is important to determine whether prescription of TZD or new antidiabetic medications compared to conventional treatment, such as sulfonylurea or insulin, is better for stroke management. Furthermore, it is unclear whether drugs in the same class show greater safety and efficacy than other drugs for stroke management.
Collapse
Affiliation(s)
- Chang Ho Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
45
|
Cai X, Zhu Z, Zhang Y, Tian X. SDF-1α promotes repair of myocardial ischemic necrosis zones in rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1956-1967. [PMID: 31934018 PMCID: PMC6949651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/19/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To explore the repair effect of stromal cell-derived factor-1α (SDF-1α) on myocardial ischemic necrosis zones. METHODS Lentivirus (LV-SDF-1α-GFP) containing SDF-1α target gene was established, the separated and cultured neonatal rat cardiac fibroblasts were transfected, and caudal intravenous injection of isoproterenol was conducted to prepare a rat model of myocardial ischemia. Small animal ultrasound was used to evaluate the effect on cardiac functions. Morphology and immunofluorescence were used to observe the change of ischemic necrosis zones and expressions of stem cellular markers c-kit, CD34, nkx2.5, and nanog, and a quantitative analysis was performed. RESULTS The established LV-SDF-1α-GFP was used to transfect myocardial fibroblasts which presented GFP green fluorescent expression and could secrete SDF-1α. The small animal ultrasound system showed that rat cardiac functions of the lentivirus group and cell group were improved to different degrees, myocardial ischemic necrosis zones of lentivirus group and cell group were reduced, and differences had statistical significances (P<0.05). Immunofluorescence showed that expressions of stem cellular markers c-kit, CD34, nkx2.5 and nanog in myocardial tissue ischemic zones in both the lentivirus group and cell group increased, and differences through inter-group comparison had statistical significances (P<0.05). CONCLUSION SDF-1α can promote migration and proliferation of stem cells into the myocardial ischemic necrosis zone, participate in repair of the myocardial necrosis zone, and improve cardiac function.
Collapse
Affiliation(s)
- Xinhua Cai
- Department of Histology and Embryology, Xinxiang Medical UniversityXinxiang 453003, Henan Province, China
| | - Zhanzhan Zhu
- The 7th People’s Hospital of ZhengzhouZhengzhou 450006, Henan Province, China
| | - Yongchun Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical UniversityWeihui 453100, Henan Province, China
| | - Xiangqin Tian
- Department of Histology and Embryology, Xinxiang Medical UniversityXinxiang 453003, Henan Province, China
| |
Collapse
|
46
|
Darsalia V, Johansen OE, Lietzau G, Nyström T, Klein T, Patrone C. Dipeptidyl Peptidase-4 Inhibitors for the Potential Treatment of Brain Disorders; A Mini-Review With Special Focus on Linagliptin and Stroke. Front Neurol 2019; 10:493. [PMID: 31139140 PMCID: PMC6518970 DOI: 10.3389/fneur.2019.00493] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/23/2019] [Indexed: 12/21/2022] Open
Abstract
Cerebral stroke is a leading cause of death and persistent disability of elderly in the world. Although stroke prevention by targeting several risk factors such as diabetes and hypertension has decreased the stroke incidence, the total number of strokes is increasing due to the population aging and new preventive therapies are needed. Moreover, post-stroke acute pharmacological strategies aimed to reduce stroke-induced brain injury have failed in clinical trials despite being effective in animal models. Finally, approximately 30% of surviving stroke patients do not recover from stroke and remain permanently dependent on supportive care in activities of daily living. Therefore, strategies to improve stroke recovery in the post-acute phase are highly needed. Linagliptin is a dipeptidyl peptidase-4 inhibitor which is clinically approved to reduce hyperglycemia in type 2 diabetes. The regulation of glycemia by dipeptidyl peptidase-4 inhibition is mainly achieved by preventing endogenous glucagon-like peptide-1 (GLP-1) degradation. Interestingly, linagliptin has also shown glycaemia-independent beneficial effects in animal models of stroke, Parkinson's disease and Alzheimer's disease. In some case the preclinical data have been supported with some clinical data. Although potentially very interesting for the development of new strategies against stroke and neurodegenerative disorders, the mode of action of linagliptin in the brain is still largely unknown and seems to occur in a GLP-1R-independent manner. The purpose of this mini-review is to summarize and discuss the recent experimental and clinical work regarding the effects of linagliptin in the central nervous system, with special emphasis on acute neuroprotection, stroke prevention and post-stroke recovery. We also highlight the main questions in this research field that need to be addressed in clinical perspective.
Collapse
Affiliation(s)
- Vladimer Darsalia
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Grazyna Lietzau
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Cesare Patrone
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
47
|
Erbil D, Eren CY, Demirel C, Küçüker MU, Solaroğlu I, Eser HY. GLP-1's role in neuroprotection: a systematic review. Brain Inj 2019; 33:734-819. [PMID: 30938196 DOI: 10.1080/02699052.2019.1587000] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) is a target for treatment of diabetes; however, its function in the brain is not well studied. In this systematic review, we aimed to analyze the neuroprotective role of GLP-1 and its defined mechanisms. Methods: We searched 'Web of Science' and 'Pubmed' to identify relevant studies using GLP-1 as the keyword. Two hundred and eighty-nine clinical and preclinical studies have been included. Data have been presented by grouping neurodegenerative, neurovascular and specific cell culture models. Results: Recent literature shows that GLP-1 and its agonists, DPP-4 inhibitors and combined GLP-1/GIP molecules are effective in partially or fully reversing the effects of neurotoxic compounds, neurovascular complications of diabetes, neuropathological changes related with Alzheimer's disease, Parkinson's disease or vascular occlusion. Possible mechanisms that provide neuroprotection are enhancing the viability of the neurons and restoring neurite outgrowth by increased neurotrophic factors, increasing subventricular zone progenitor cells, decreasing apoptosis, decreasing the level of pro-inflammatory factors, and strengthening blood-brain barrier. Conclusion: Based on the preclinical studies, GLP-1 modifying agents are promising targets for neuroprotection. On the other hand, the number of clinical studies that investigate GLP-1 as a treatment is low and further clinical trials are needed for a benchside to bedside translation of recent findings.
Collapse
Affiliation(s)
- Damla Erbil
- a School of Medicine , Koç University , Istanbul , Turkey
| | - Candan Yasemin Eren
- b Research Center for Translational Medicine , Koç University , Istanbul , Turkey
| | - Cağrı Demirel
- a School of Medicine , Koç University , Istanbul , Turkey
| | | | - Ihsan Solaroğlu
- a School of Medicine , Koç University , Istanbul , Turkey.,b Research Center for Translational Medicine , Koç University , Istanbul , Turkey
| | - Hale Yapıcı Eser
- a School of Medicine , Koç University , Istanbul , Turkey.,b Research Center for Translational Medicine , Koç University , Istanbul , Turkey
| |
Collapse
|
48
|
Milonas D, Didangelos T, Hatzitolios AI, Tziomalos K. Incretin-Based Antihyperglycemic Agents for the Management of Acute Ischemic Stroke in Patients with Diabetes Mellitus: A Review. Diabetes Ther 2019; 10:429-435. [PMID: 30725400 PMCID: PMC6437305 DOI: 10.1007/s13300-019-0580-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Indexed: 12/18/2022] Open
Abstract
Diabetes mellitus (DM) is a major risk factor for ischemic stroke. Moreover, patients with DM suffer more severe strokes and have worse functional outcome following an acute stroke than patients without DM. In this context, data from animal studies and emerging evidence from clinical studies suggest that incretin-based antihyperglycemic agents might exert beneficial effects in patients with DM who suffer ischemic stroke. It appears that these agents exert neuroprotective actions that might both reduce infarct size and promote recovery. The present review summarizes the evidence on the potential role of incretin-based antihyperglycemic agents in the management of acute ischemic stroke.
Collapse
Affiliation(s)
- Dimitrios Milonas
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece.
| | - Triantafyllos Didangelos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Apostolos I Hatzitolios
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| | - Konstantinos Tziomalos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, Thessaloniki, Greece
| |
Collapse
|
49
|
Huang X, Wan M, Yang Q, Ding X, Zhou Z. The stromal cell-derived factor-1 α (SDF-1α)/cysteine-X-cysteine chemokine receptor 4 (CXCR4) axis: a possible prognostic indicator of acute ischemic stroke. J Int Med Res 2019; 47:1897-1907. [PMID: 30760134 PMCID: PMC6567759 DOI: 10.1177/0300060519827173] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Objective The stromal cell-derived factor-1α/cysteine-X-cysteine chemokine receptor 4 (SDF-1α/CXCR4) axis promotes neuroprotection and angiogenesis in animal studies. Few studies have investigated the potential clinical implications of the SDF-1α/CXCR4 axis in patients with acute ischemic stroke (AIS). We evaluated the prognostic values of the SDF-1α/CXCR4 axis in patients with proximal middle cerebral artery occlusion. Methods Fifty-five patients and 18 age- and sex-matched volunteers were enrolled. Baseline clinical characteristics and risk factors of stroke were recorded. Peripheral whole blood cells were double stained with anti-CD34 and anti-CXCR4 (CD184). CD34+CXCR4+ cells were analyzed by flow cytometry. Plasma SDF-1α levels were measured by enzyme-linked immunosorbent assay. Results In the AIS group, plasma SDF-1α levels and the number of circulating CD34+CXCR4+ cells were significantly higher than those in controls. Day 1 SDF-1α levels were negatively correlated with infarct volume (r = −0.521) and the initial National Institutes of Health Stroke Scale score (r = −0.489). SDF-1α levels (day 1: r = −0.514; day 3: r = −0.275; day 7: r = −0.375) and circulating CD34+CXCR4+ cells (day 7: r = −0.282) were inversely associated with the 90-day modified Rankin Scale score. Conclusion The SDF-1α/CXCR4 axis has potential applications for predicting the clinical outcome of AIS.
Collapse
Affiliation(s)
- Xianjun Huang
- 1 Department of Neurology, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui Province, China
| | - Mei Wan
- 2 Department of Neurology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei Province, China
| | - Qian Yang
- 1 Department of Neurology, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui Province, China
| | - Xianhui Ding
- 1 Department of Neurology, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui Province, China
| | - Zhiming Zhou
- 1 Department of Neurology, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui Province, China
| |
Collapse
|
50
|
Hao FL, Han XF, Wang XL, Zhao ZR, Guo AH, Lu XJ, Zhao XF. The neurovascular protective effect of alogliptin in murine MCAO model and brain endothelial cells. Biomed Pharmacother 2018; 109:181-187. [PMID: 30396075 DOI: 10.1016/j.biopha.2018.10.064] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/12/2022] Open
Abstract
Endothelial damage and blood brain barrier disruption contribute to ischemic stroke and brain injury. Gliptins are a novel class of treatment agents for diabetes, and recent studies have linked the use of gliptins to neuroprotection. Alogliptin is a type of orally available gliptin that was approved for clinical use by the FDA in 2013. In this study, we investigated the neurovascular protective effects of alogliptin both in vivo and in vitro. In a murine middle cerebral artery occlusion (MCAO) stroke model, administration of alogliptin ameliorated cerebral infarction and disruption of brain vascular permeability, and restored expression of the endothelial tight junction proteins occludin and zona occludens 1 (ZO-1). In brain vascular endothelial cells exposed to oxygen and glucose deprivation/reperfusion (OGD/R), alogliptin prevented OGD/R-induced high permeability of the endothelial monolayer. Alogliptin treatment recovered the reduction in occludin and ZO-1 induced by OGD/R. Moreover, alogliptin treatment prevented OGD/R-induced induction of metalloproteinase (MMP)-2 and MMP-9, and restored expression of tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2. Collectively, our data indicate that alogliptin can improve neurovascular integrity and exerts neuroprotective effects.
Collapse
Affiliation(s)
- Feng-Li Hao
- Department of Neurology, Xianyang Hospital of Yan'an University, Xianyang, 712000, China
| | - Xiao-Fang Han
- Internal Medicine, Xianyang Hospital of Yan'an University, Xianyang, China
| | - Xiao-Li Wang
- Department of Neurology, Air Force Military Medical University, Xi'an, China
| | - Zhi-Ru Zhao
- Department of Neurology, Xianyang Hospital of Yan'an University, Xianyang, 712000, China.
| | - Ai-Hong Guo
- Department of Neurology, Xianyang Hospital of Yan'an University, Xianyang, 712000, China
| | - Xin-Jian Lu
- Department of Neurology, Xianyang Hospital of Yan'an University, Xianyang, 712000, China
| | - Xiong-Fei Zhao
- Department of Neurology, Xianyang Hospital of Yan'an University, Xianyang, 712000, China
| |
Collapse
|