1
|
Pardridge WM. Treatment of Parkinson's disease with biologics that penetrate the blood-brain barrier via receptor-mediated transport. Front Aging Neurosci 2023; 15:1276376. [PMID: 38035276 PMCID: PMC10682952 DOI: 10.3389/fnagi.2023.1276376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
Parkinson's disease (PD) is characterized by neurodegeneration of nigral-striatal neurons in parallel with the formation of intra-neuronal α-synuclein aggregates, and these processes are exacerbated by neuro-inflammation. All 3 components of PD pathology are potentially treatable with biologics. Neurotrophins, such as glial derived neurotrophic factor or erythropoietin, can promote neural repair. Therapeutic antibodies can lead to disaggregation of α-synuclein neuronal inclusions. Decoy receptors can block the activity of pro-inflammatory cytokines in brain. However, these biologic drugs do not cross the blood-brain barrier (BBB). Biologics can be made transportable through the BBB following the re-engineering of the biologic as an IgG fusion protein, where the IgG domain targets an endogenous receptor-mediated transcytosis (RMT) system within the BBB, such as the insulin receptor or transferrin receptor. The receptor-specific antibody domain of the fusion protein acts as a molecular Trojan horse to ferry the biologic into brain via the BBB RMT pathway. This review describes the re-engineering of all 3 classes of biologics (neurotrophins, decoy receptor, therapeutic antibodies) for BBB delivery and treatment of PD. Targeting the RMT pathway at the BBB also enables non-viral gene therapy of PD using lipid nanoparticles (LNP) encapsulated with plasmid DNA encoding therapeutic genes. The surface of the lipid nanoparticle is conjugated with a receptor-specific IgG that triggers RMT of the LNP across the BBB in vivo.
Collapse
|
2
|
Pardridge WM. Receptor-mediated drug delivery of bispecific therapeutic antibodies through the blood-brain barrier. FRONTIERS IN DRUG DELIVERY 2023; 3:1227816. [PMID: 37583474 PMCID: PMC10426772 DOI: 10.3389/fddev.2023.1227816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Therapeutic antibody drug development is a rapidly growing sector of the pharmaceutical industry. However, antibody drug development for the brain is a technical challenge, and therapeutic antibodies for the central nervous system account for ~3% of all such agents. The principal obstacle to antibody drug development for brain or spinal cord is the lack of transport of large molecule biologics across the blood-brain barrier (BBB). Therapeutic antibodies can be made transportable through the blood-brain barrier by the re-engineering of the therapeutic antibody as a BBB-penetrating bispecific antibody (BSA). One arm of the BSA is the therapeutic antibody and the other arm of the BSA is a transporting antibody. The transporting antibody targets an exofacial epitope on a BBB receptor, and this enables receptor-mediated transcytosis (RMT) of the BSA across the BBB. Following BBB transport, the therapeutic antibody then engages the target receptor in brain. RMT systems at the BBB that are potential conduits to the brain include the insulin receptor (IR), the transferrin receptor (TfR), the insulin-like growth factor receptor (IGFR) and the leptin receptor. Therapeutic antibodies have been re-engineered as BSAs that target the insulin receptor, TfR, or IGFR RMT systems at the BBB for the treatment of Alzheimer's disease and Parkinson's disease.
Collapse
|
3
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Pardridge WM. Blood-brain barrier delivery for lysosomal storage disorders with IgG-lysosomal enzyme fusion proteins. Adv Drug Deliv Rev 2022; 184:114234. [PMID: 35307484 DOI: 10.1016/j.addr.2022.114234] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/25/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
The majority of lysosomal storage diseases affect the brain. Treatment of the brain with intravenous enzyme replacement therapy is not successful, because the recombinant lysosomal enzymes do not cross the blood-brain barrier (BBB). Biologic drugs, including lysosomal enzymes, can be re-engineered for BBB delivery as IgG-enzyme fusion proteins. The IgG domain of the fusion protein is a monoclonal antibody directed against an endogenous receptor-mediated transporter at the BBB, such as the insulin receptor or the transferrin receptor. This receptor transports the IgG across the BBB, in parallel with the endogenous receptor ligand, and the IgG acts as a molecular Trojan horse to ferry into brain the lysosomal enzyme genetically fused to the IgG. The IgG-enzyme fusion protein is bi-functional and retains both high affinity binding for the BBB receptor, and high lysosomal enzyme activity. IgG-lysosomal enzymes are presently in clinical trials for treatment of the brain in Mucopolysaccharidosis.
Collapse
|
5
|
Meta-analysis of cognitive and behavioral tests in leptin- and leptin receptor-deficient mice. Neurosci Res 2020; 170:217-235. [PMID: 33316303 DOI: 10.1016/j.neures.2020.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/06/2020] [Accepted: 11/11/2020] [Indexed: 01/19/2023]
Abstract
Leptin is a hormone produced by adipocytes that regulates food intake and metabolism. Leptin-related gene-deficient mice, such as db/db and ob/ob mice, are widely used to study diabetes and its related diseases. However, broad effects of leptin appear to cause variability in behavioral test results. We performed a meta-analysis of major behavioral tests in db/db and ob/ob mice. These mice exhibited significant impairments in the Morris water maze, forced swim, novel object recognition, Y-maze, tail suspension, and light-dark box tests, whereas the elevated plus maze and open field tests did not reveal significant changes. We also performed correlation and regression analyses between the animals' performances and the experimental protocols and conditions. The memory-related tests were characterized by the correlations of their results with animal age, while the performances in the elevated plus-maze and forced swim tests were affected by the width of the devices used. In conclusion, db/db and ob/ob mice mainly exhibit memory deficits and depression-like behavior, although experimenters should be aware of animal age and device size in conducting experiments.
Collapse
|
6
|
Pardridge WM. Brain Delivery of Nanomedicines: Trojan Horse Liposomes for Plasmid DNA Gene Therapy of the Brain. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:602236. [PMID: 35047884 PMCID: PMC8757841 DOI: 10.3389/fmedt.2020.602236] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Non-viral gene therapy of the brain is enabled by the development of plasmid DNA brain delivery technology, which requires the engineering and manufacturing of nanomedicines that cross the blood-brain barrier (BBB). The development of such nanomedicines is a multi-faceted problem that requires progress at multiple levels. First, the type of nanocontainer, e.g., nanoparticle or liposome, which encapsulates the plasmid DNA, must be developed. Second, the type of molecular Trojan horse, e.g., peptide or receptor-specific monoclonal antibody (MAb), must be selected for incorporation on the surface of the nanomedicine, as this Trojan horse engages specific receptors expressed on the BBB, and the brain cell membrane, to trigger transport of the nanomedicine from blood into brain cells beyond the BBB. Third, the plasmid DNA must be engineered without bacterial elements, such as antibiotic resistance genes, to enable administration to humans; the plasmid DNA must also be engineered with tissue-specific gene promoters upstream of the therapeutic gene, to insure gene expression in the target organ with minimal off-target expression. Fourth, upstream manufacturing of the nanomedicine must be developed and scalable so as to meet market demand for the target disease, e.g., annual long-term treatment of 1,000 patients with an orphan disease, short term treatment of 10,000 patients with malignant glioma, or 100,000 patients with new onset Parkinson's disease. Fifth, downstream manufacturing problems, such as nanomedicine lyophilization, must be solved to ensure the nanomedicine has a commercially viable shelf-life for treatment of CNS disease in humans.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
7
|
Fujita Y, Yamashita T. The Effects of Leptin on Glial Cells in Neurological Diseases. Front Neurosci 2019; 13:828. [PMID: 31447640 PMCID: PMC6692660 DOI: 10.3389/fnins.2019.00828] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022] Open
Abstract
It is known that various endocrine modulators, including leptin and ghrelin, have neuroprotective roles in neurological diseases. Leptin is a hormone produced by adipocytes and was originally identified as a gene related to obesity in mice. The leptin receptors in the hypothalamus are the main target for the homeostatic regulation of body weight. Recent studies have demonstrated that leptin receptors are also expressed in other regions of the central nervous system (CNS), such as the hippocampus, cerebral cortex, and spinal cord. Accordingly, these studies identified the involvement of leptin in the regulation of neuronal survival and neural development. Furthermore, leptin has been shown to have neuroprotective functions in animal models of neurological diseases and demyelination. These observations also suggest that dysregulation of leptin signaling may be involved in the association between neurodegeneration and obesity. In this review, we summarize novel functions of leptin in animal models of neurodegenerative diseases. Specifically, we focus on the emerging evidence for the role of leptin in non-neuronal cells in the CNS, including astrocytes, microglia, and oligodendrocytes. Understanding leptin-mediated neuroprotective signals and molecular mechanisms underlying remyelination will be helpful to establish therapeutic strategies against neurological diseases.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.,Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
8
|
Yoo S, Cha D, Kim DW, Hoang TV, Blackshaw S. Tanycyte-Independent Control of Hypothalamic Leptin Signaling. Front Neurosci 2019; 13:240. [PMID: 30941008 PMCID: PMC6433882 DOI: 10.3389/fnins.2019.00240] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/01/2019] [Indexed: 12/29/2022] Open
Abstract
Leptin is secreted by adipocytes to regulate appetite and body weight. Recent studies have reported that tanycytes actively transport circulating leptin across the brain barrier into the hypothalamus, and are required for normal levels of hypothalamic leptin signaling. However, direct evidence for leptin receptor (LepR) expression is lacking, and the effect of tanycyte-specific deletion of LepR has not been investigated. In this study, we analyze the expression and function of the tanycytic LepR in mice. Using single-molecule fluorescent in situ hybridization (smfISH), RT-qPCR, single-cell RNA sequencing (scRNA-Seq), and selective deletion of the LepR in tanycytes, we are unable to detect expression of LepR in the tanycytes. Tanycyte-specific deletion of LepR likewise did not affect leptin-induced pSTAT3 expression in hypothalamic neurons, regardless of whether leptin was delivered by intraperitoneal or intracerebroventricular injection. Finally, we use activity-regulated scRNA-Seq (act-Seq) to comprehensively profile leptin-induced changes in gene expression in all cell types in mediobasal hypothalamus. Clear evidence for leptin signaling is only seen in endothelial cells and subsets of neurons, although virtually all cell types show leptin-induced changes in gene expression. We thus conclude that LepR expression in tanycytes is either absent or undetectably low, that tanycytes do not directly regulate hypothalamic leptin signaling through a LepR-dependent mechanism, and that leptin regulates gene expression in diverse hypothalamic cell types through both direct and indirect mechanisms.
Collapse
Affiliation(s)
- Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - David Cha
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States.,Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, Johns Hopkins University, Baltimore, MD, United States.,Center for Human Systems Biology, Johns Hopkins University, Baltimore, MD, United States.,School of Medicine, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
9
|
Yuan X, Caron A, Wu H, Gautron L. Leptin Receptor Expression in Mouse Intracranial Perivascular Cells. Front Neuroanat 2018; 12:4. [PMID: 29410615 PMCID: PMC5787097 DOI: 10.3389/fnana.2018.00004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/08/2018] [Indexed: 12/28/2022] Open
Abstract
Past studies have suggested that non-neuronal brain cells express the leptin receptor. However, the identity and distribution of these leptin receptor-expressing non-neuronal brain cells remain debated. This study assessed the distribution of the long form of the leptin receptor (LepRb) in non-neuronal brain cells using a reporter mouse model in which LepRb-expressing cells are permanently marked by tdTomato fluorescent protein (LepRb-CretdTomato). Double immunohistochemistry revealed that, in agreement with the literature, the vast majority of tdTomato-tagged cells across the mouse brain were neurons (i.e., based on immunoreactivity for NeuN). Non-neuronal structures also contained tdTomato-positive cells, including the choroid plexus and the perivascular space of the meninges and, to a lesser extent, the brain. Based on morphological criteria and immunohistochemistry, perivascular cells were deduced to be mainly pericytes. Notably, tdTomato-positive cells were immunoreactive for vitronectin and platelet derived growth factor receptor beta (PDGFBR). In situ hybridization studies confirmed that most tdTomato-tagged perivascular cells were enriched in leptin receptor mRNA (all isoforms). Using qPCR studies, we confirmed that the mouse meninges were enriched in Leprb and, to a greater extent, the short isoforms of the leptin receptor. Interestingly, qPCR studies further demonstrated significantly altered expression for Vtn and Pdgfrb in the meninges and hypothalamus of LepRb-deficient mice. Collectively, our data demonstrate that the only intracranial non-neuronal cells that express LepRb in the adult mouse are cells that form the blood-brain barrier, including, most notably, meningeal perivascular cells. Our data suggest that pericytic leptin signaling plays a role in the integrity of the intracranial perivascular space and, consequently, may provide a link between obesity and numerous brain diseases.
Collapse
Affiliation(s)
- Xuefeng Yuan
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alexandre Caron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Laurent Gautron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
10
|
Teixeira TM, da Costa DC, Resende AC, Soulage CO, Bezerra FF, Daleprane JB. Activation of Nrf2-Antioxidant Signaling by 1,25-Dihydroxycholecalciferol Prevents Leptin-Induced Oxidative Stress and Inflammation in Human Endothelial Cells. J Nutr 2017; 147:506-513. [PMID: 28250190 DOI: 10.3945/jn.116.239475] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/26/2016] [Accepted: 02/08/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Obesity is associated with hyperleptinemia and endothelial dysfunction. Hyperleptinemia has been reported to induce both oxidative stress and inflammation by increasing reactive oxygen species production.Objective: The objective of this study was to determine the effects of 1,25-dihydroxycholecalciferol [1,25(OH)2D3] against leptin-induced oxidative stress and inflammation in human endothelial cells.Methods: Small interfering RNA (siRNA) were used to knock down the expression of vitamin D receptor (VDR) in human umbilical vein endothelial cells (HUVECs). HUVECs were pretreated for 4 h with physiologic (10-10 M) or supraphysiologic (10-7 M) concentrations of 1,25(OH)2D3 and exposed to leptin (10 ng/mL). Superoxide anion production and translocation of nuclear factor (erythroid-derived 2)-like 2 (NRF2) and nuclear transcription factor κB (NF-κB) subunit p65 to the nucleus and the activation of their target genes were quantified.Results: Pretreatment of HUVECs with 1,25(OH)2D3 prevented the leptin-induced increase in superoxide anion production (P < 0.05). Pretreatment with 1,25(OH)2D3 further increased NRF2 translocation to the nucleus (by 3-fold; P < 0.05) and increased mRNA expression of superoxide dismutase 2 (SOD2; by 2-fold), glutathione peroxidase (GPX; by 3-fold), NAD(P)H dehydrogenase (quinone) 1 (NQO1; by 4-fold), and heme oxygenase 1 (HMOX1; by 2-fold) (P < 0.05). Leptin doubled the translocation of NF-κB (P < 0.05) to the nucleus and increased (P < 0.05) the upregulation of vascular inflammatory mediators such as monocyte chemoattractant protein 1 (MCP1; by 1-fold), transforming growth factor β (TGF β by 1-fold), and vascular cell adhesion molecule 1 (VCAM1; by 4-fold) (P < 0.05), which were prevented (P < 0.05) by pretreatment with 1,25(OH)2D3 Protective effects of 1,25(OH)2D3 were confirmed to be VDR dependent by using VDR siRNA.Conclusion: Pretreatment with 1,25(OH)2D3 in the presence of a high concentration of leptin has a beneficial effect on HUVECs through the regulation of mediators of antioxidant activity and inflammation.
Collapse
Affiliation(s)
| | | | - Angela C Resende
- Pharmacology, Rio de Janeiro State University, Rio de Janeiro, Brazil; and
| | | | | | | |
Collapse
|
11
|
Shpakov AO. The brain leptin signaling system and its functional state in metabolic syndrome and type 2 diabetes mellitus. J EVOL BIOCHEM PHYS+ 2016; 52:177-195. [DOI: 10.1134/s0022093016030017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
12
|
Brain signaling systems in the Type 2 diabetes and metabolic syndrome: promising target to treat and prevent these diseases. Future Sci OA 2015; 1:FSO25. [PMID: 28031898 PMCID: PMC5137856 DOI: 10.4155/fso.15.23] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The changes in the brain signaling systems play an important role in etiology and pathogenesis of Type 2 diabetes mellitus (T2DM) and metabolic syndrome (MS), being a possible cause of these diseases. Therefore, their restoration at the early stages of T2DM and MS can be regarded as a promising way to treat and prevent these diseases and their complications. The data on the functional state of the brain signaling systems regulated by insulin, IGF-1, leptin, dopamine, serotonin, melanocortins and glucagon-like peptide-1, in T2DM and MS, are analyzed. The pharmacological approaches to restoration of these systems and improvement of insulin sensitivity, energy expenditure, lipid metabolism, and to prevent diabetic complications are discussed.
Collapse
|
13
|
Ghantous CM, Kobeissy FH, Soudani N, Rahman FA, Al-Hariri M, Itani HA, Sabra R, Zeidan A. Mechanical stretch-induced vascular hypertrophy occurs through modulation of leptin synthesis-mediated ROS formation and GATA-4 nuclear translocation. Front Pharmacol 2015; 6:240. [PMID: 26557089 PMCID: PMC4615939 DOI: 10.3389/fphar.2015.00240] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/05/2015] [Indexed: 12/11/2022] Open
Abstract
Background: Obesity and hypertension are associated with increased leptin production contributing to cardiovascular remodeling. Mechanisms involving mechanical stretch-induced leptin production and the cross talk between signaling pathways leading to vascular remodeling have not been fully elucidated. Methods and Results: Rat portal vein (RPV) organ culture was used to investigate the effect of mechanical stretch on leptin protein expression in vascular smooth muscle cells (VSMCs). Moreover, the involvement of reactive oxygen species (ROS), the RhoA/ROCK pathway, actin cytoskeleton dynamics and the transcriptional factor GATA-4 activation in mechanical stretch-induced vascular remodeling were investigated. Stretching the RPV for 1 or 24 h significantly increased leptin protein level and ROS formation in VSMCs, which was prevented by 1 h pretreatment with the ROCK inhibitor Y-27632 and the actin cytoskeleton depolymerization agent cytochalasin D. Moreover, Western blotting and immunohistochemistry revealed that mechanical stretch or treatment with 3.1 nmol/L leptin for 24 h significantly increased actin polymerization, as reflected by an increase in the F-actin to G-actin ratio. Increases in blood vessels’ wet weight and [3H]-leucine incorporation following a 24 h treatment with conditioned media from cultured stretched RPVs indicated RPV hypertrophy. This effect was prevented by 1 h pretreatment with anti-leptin antibody, indicating leptin’s crucial role in promoting VSMC hypertrophy. As an index of GATA-4 activation, GATA-4 nuclear translocation was assessed by immunohistochemistry method. Pretreating VSMC with leptin for 1 h significantly activated GATA-4 nuclear translocation, which was potently attenuated by the NADPH oxidase inhibitor apocynin, Y-27632, and cytochalasin D. Conclusion: Our results demonstrate that ROS formation, RhoA/ROCK pathway, and GATA-4 activation play a pivotal role in mechanical stretch-induced leptin synthesis leading to VSMC remodeling.
Collapse
Affiliation(s)
- Crystal M Ghantous
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, American University of Beirut , Beirut, Lebanon
| | - Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut , Beirut, Lebanon
| | - Nadia Soudani
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, American University of Beirut , Beirut, Lebanon
| | - Farah A Rahman
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, American University of Beirut , Beirut, Lebanon
| | - Mustafa Al-Hariri
- Department of Biochemistry and Molecular Genetics, American University of Beirut , Beirut, Lebanon
| | - Hana A Itani
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine , Nashville, TN, USA
| | - Ramzi Sabra
- Department of Pharmacology and Toxicology, American University of Beirut , Beirut, Lebanon
| | - Asad Zeidan
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, American University of Beirut , Beirut, Lebanon
| |
Collapse
|
14
|
Balland E, Cowley MA. New insights in leptin resistance mechanisms in mice. Front Neuroendocrinol 2015; 39:59-65. [PMID: 26410445 DOI: 10.1016/j.yfrne.2015.09.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 11/18/2022]
Abstract
Leptin resistance is one of the main challenges of obesity. To date, two levels of resistance have been identified, first a decreased rate of leptin uptake into the brain and secondly a diminished central response to leptin. New findings have identified the mechanisms of leptin transport and demonstrated that it can be rescued in obesity, but it did not overcome the problem of central resistance. Alteration in the actions of leptin following diet-induced obesity (DIO) appears to be a multifactorial condition. Several phosphatases are inhibiting leptin signaling pathways in a pathological way. Besides, hypothalamic inflammation alters the neuronal circuits that control metabolism. Recent studies describing both mechanisms (inhibition of leptin signaling and inflammation), have provided key insights to potential new targets for treatment. However, recent data showing that DIO mice may conserve a cellular and physiological response to endogenous leptin, highlights the need to redefine the concept of "leptin resistance".
Collapse
Affiliation(s)
- Eglantine Balland
- Department of Physiology, Monash Obesity and Diabetes Institute, Monash University, Clayton, VIC 3800, Australia.
| | - Michael A Cowley
- Department of Physiology, Monash Obesity and Diabetes Institute, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
15
|
Gerstberger R. Eckhart Simon, a German physiologist's connections with physiology in Japan. Temperature (Austin) 2015; 2:316-9. [PMID: 27227034 PMCID: PMC4843913 DOI: 10.1080/23328940.2015.1066922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/23/2015] [Accepted: 06/23/2015] [Indexed: 11/04/2022] Open
Abstract
Eckhart Simon's research in the fields of Thermal and Regulatory Physiology is distinguished by exemplary relations with Thermal Physiology in Japan. The essay outlines the fields in which he has cooperated with Japanese physiologists. A list of joint publications documents their contributions to Eckhart Simon's and his department's scientific achievements.
Collapse
Affiliation(s)
- Rüdiger Gerstberger
- Department of Veterinary Physiology and Biochemistry; Justus-Liebig University Giessen ; Giessen, Germany
| |
Collapse
|
16
|
Aguilar-Valles A, Inoue W, Rummel C, Luheshi GN. Obesity, adipokines and neuroinflammation. Neuropharmacology 2015; 96:124-34. [PMID: 25582291 DOI: 10.1016/j.neuropharm.2014.12.023] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 12/14/2022]
Abstract
Global levels of obesity are reaching epidemic proportions, leading to a dramatic increase in incidence of secondary diseases and the significant economic burden associated with their treatment. These comorbidities include diabetes, cardiovascular disease, and some psychopathologies, which have been linked to a low-grade inflammatory state. Obese individuals exhibit an increase in circulating inflammatory mediators implicated as the underlying cause of these comorbidities. A number of these molecules are also manufactured and released by white adipose tissue (WAT), in direct proportion to tissue mass and are collectively known as adipokines. In the current review we focused on the role of two of the better-studied members of this family namely, leptin and adiponectin, with particular emphasis on their role in neuro-immune interactions, neuroinflammation and subsequent brain diseases. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Argel Aguilar-Valles
- Department of Neuroscience, Université de Montréal and Goodman Cancer Centre, Department of Biochemistry, McGill University, Montréal, Canada
| | - Wataru Inoue
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Christoph Rummel
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Giamal N Luheshi
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec H4H 1R3, Canada.
| |
Collapse
|
17
|
Ghantous CM, Azrak Z, Hanache S, Abou-Kheir W, Zeidan A. Differential Role of Leptin and Adiponectin in Cardiovascular System. Int J Endocrinol 2015; 2015:534320. [PMID: 26064110 PMCID: PMC4433709 DOI: 10.1155/2015/534320] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 04/23/2015] [Indexed: 02/07/2023] Open
Abstract
Leptin and adiponectin are differentially expressed adipokines in obesity and cardiovascular diseases. Leptin levels are directly associated with adipose tissue mass, while adiponectin levels are downregulated in obesity. Although significantly produced by adipocytes, leptin is also produced by vascular smooth muscle cells and cardiomyocytes. Plasma leptin concentrations are elevated in cases of cardiovascular diseases, such as hypertension, congestive heart failure, and myocardial infarction. As for the event of left ventricular hypertrophy, researchers have been stirring controversy about the role of leptin in this form of cardiac remodeling. In this review, we discuss how leptin has been shown to play an antihypertrophic role in the development of left ventricular hypertrophy through in vitro experiments, population-based cross-sectional studies, and longitudinal cohort studies. Conversely, we also examine how leptin may actually promote left ventricular hypertrophy using in vitro analysis and human-based univariate and multiple linear stepwise regression analysis. On the other hand, as opposed to leptin's generally detrimental effects on the cardiovascular system, adiponectin is a cardioprotective hormone that reduces left ventricular and vascular hypertrophy, oxidative stress, and inflammation. In this review, we also highlight adiponectin signaling and its protective actions on the cardiovascular system.
Collapse
Affiliation(s)
- C. M. Ghantous
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
| | - Z. Azrak
- Department of Pharmacology and Toxicology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
| | - S. Hanache
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
| | - W. Abou-Kheir
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
| | - A. Zeidan
- Department of Anatomy, Cell biology and Physiology, American University of Beirut, DTS-255, P.O. Box 11-0236, Beirut 1107-2020, Lebanon
- *A. Zeidan:
| |
Collapse
|
18
|
Jung CH, Kim MS. Molecular mechanisms of central leptin resistance in obesity. Arch Pharm Res 2013; 36:201-7. [DOI: 10.1007/s12272-013-0020-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 12/24/2012] [Indexed: 02/06/2023]
|
19
|
Strazielle N, Ghersi-Egea JF. Physiology of blood-brain interfaces in relation to brain disposition of small compounds and macromolecules. Mol Pharm 2013; 10:1473-91. [PMID: 23298398 DOI: 10.1021/mp300518e] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The brain develops and functions within a strictly controlled environment resulting from the coordinated action of different cellular interfaces located between the blood and the extracellular fluids of the brain, which include the interstitial fluid and the cerebrospinal fluid (CSF). As a correlate, the delivery of pharmacologically active molecules and especially macromolecules to the brain is challenged by the barrier properties of these interfaces. Blood-brain interfaces comprise both the blood-brain barrier located at the endothelium of the brain microvessels and the blood-CSF barrier located at the epithelium of the choroid plexuses. Although both barriers develop extensive surface areas of exchange between the blood and the neuropil or the CSF, the molecular fluxes across these interfaces are tightly regulated. Cerebral microvessels acquire a barrier phenotype early during cerebral vasculogenesis under the influence of the Wnt/β-catenin pathway, and of recruited pericytes. Later in development, astrocytes also play a role in blood-brain barrier maintenance. The tight choroid plexus epithelium develops very early during embryogenesis. It is specified by various signaling molecules from the embryonic dorsal midline, such as bone morphogenic proteins, and grows under the influence of Sonic hedgehog protein. Tight junctions at each barrier comprise a distinctive set of claudins from the pore-forming and tightening categories that determine their respective paracellular barrier characteristics. Vesicular traffic is limited in the cerebral endothelium and abundant in the choroidal epithelium, yet without evidence of active fluid phase transcytosis. Inorganic ion transport is highly regulated across the barriers. Small organic compounds such as nutrients, micronutrients and hormones are transported into the brain by specific solute carriers. Other bioactive metabolites, lipophilic toxic xenobiotics or pharmacological agents are restrained from accumulating in the brain by several ATP-binding cassette efflux transporters, multispecific solute carriers, and detoxifying enzymes. These various molecular effectors differently distribute between the two barriers. Receptor-mediated endocytotic and transcytotic mechanisms are active in the barriers. They enable brain penetration of selected polypeptides and proteins, or inversely macromolecule efflux as it is the case for immnoglobulins G. An additional mechanism specific to the BCSFB mediates the transport of selected plasma proteins from blood into CSF in the developing brain. All these mechanisms could be explored and manipulated to improve macromolecule delivery to the brain.
Collapse
Affiliation(s)
- N Strazielle
- Brain-i, Lyon Neuroscience Research Center, Lyon, France.
| | | |
Collapse
|
20
|
Rummel C, Inoue W, Poole S, Luheshi GN. Leptin regulates leukocyte recruitment into the brain following systemic LPS-induced inflammation. Mol Psychiatry 2010; 15:523-34. [PMID: 19773811 DOI: 10.1038/mp.2009.98] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The appetite suppressing hormone leptin has emerged as an important modulator of immune function and is now considered to be a critical link between energy balance and host defense responses to pathogens. These 'adaptive' responses can, in situations of severe and sustained systemic inflammation, lead to adverse effects including brain damage that is partly mediated by neutrophil recruitment into the brain. We examined the contribution of leptin to this process in leptin-deficient (ob/ob), -resistant (db/db) and wild-type (WT) mice injected intraperitoneally with a septic dose of lipopolysaccharide (LPS). This treatment induced a dramatic increase in the number of neutrophils entering the brain of WT mice, an effect that was almost totally abolished in the mutant mice and correlated with a significant reduction in the mRNA levels of interleukin-1beta, intracellular adhesion molecule-1 and neutrophil-specific chemokines. These effects were reversed with leptin replenishment in ob/ob mice leading to recovery of neutrophil recruitment into the brain. Moreover, 48 h food deprivation in WT mice, which decreased circulating leptin levels, attenuated the LPS-induced neutrophil recruitment as did a single injection of an anti-leptin antiserum 4 h before LPS treatment in WT mice. These results provide the first demonstration that leptin has a critical role in leukocyte recruitment to the brain following severe systemic inflammation with possible implications for individuals with altered leptin levels such as during obesity or starvation.
Collapse
Affiliation(s)
- C Rummel
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
21
|
Mitchell SE, Nogueiras R, Morris A, Tovar S, Grant C, Cruickshank M, Rayner DV, Dieguez C, Williams LM. Leptin receptor gene expression and number in the brain are regulated by leptin level and nutritional status. J Physiol 2009; 587:3573-85. [PMID: 19491239 DOI: 10.1113/jphysiol.2009.173328] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Hormone potency depends on receptor availability, regulated via gene expression and receptor trafficking. To ascertain how central leptin receptors are regulated, the effects of leptin challenge, high-fat diet, fasting and refeeding were measured on leptin receptor number and gene expression. These were measured using quantitative (125)I-labelled leptin in vitro autoradiography and in situ hybridisation, respectively. Ob-R (all forms of leptin receptor) expression in the choroid plexus (CP) was unchanged by high-fat diet or leptin challenge, whereas fasting increased but refeeding failed to decrease expression. (125)I-labelled leptin binding to the CP was increased by fasting and returned to basal levels on refeeding. (125)I-Labelled leptin was reduced by leptin challenge and increased by high-fat feeding. Ob-Rb (signalling form) in the arcuate (ARC) and ventromedial (VMH) nuclei was increased after fasting and decreased by refeeding. Leptin challenge increased Ob-Rb expression in the ARC, but not after high-fat feeding. In general, changes in gene expression in the ARC and VMH appeared to be largely due to changes in area rather than density of labelling, indicating that the number of cells expressing Ob-Rb was the parameter that contributed most to these changes. Leptin stimulation of suppressor of cytokine signalling 3 (SOCS3), a marker of stimulation of the Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway, was unchanged after high-fat diet. Thus, early loss of leptin sensitivity after high-fat feeding is unrelated to down-regulation of leptin receptor expression or number and does not involve the JAK/STAT pathway. The effect of leptin to decrease (125)I-labelled leptin binding and the loss of ability of leptin to up-regulate Ob-Rb expression in the ARC after high-fat feeding offer potential mechanisms for the development of leptin insensitivity in response to both hyperleptinaemia and high-fat diet.
Collapse
Affiliation(s)
- Sharon E Mitchell
- Obesity and Metabolic Health Division, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Synthesized and released by the adipose tissue, leptin is the widely studied 167-amino acid hormonal protein product of the obesity gene. Originally leptin was defined in association with satiety and energy balance and claimed to be an anti-obesity factor that functioned via a feedback effect from adipocytes to hypothalamus. There is a growing body of evidence that emphasizes the importance of leptin in the regulation of food intake and body weight in animals and humans, alike. Other research findings point out that it plays a role in the regulation of the metabolism, sexual development, reproduction, hematopoiesis, immunity, gastrointestinal functions, sympathetic activation, and angiogenesis. The aim of this review is to evaluate the relation between leptin and the central nervous system (CNS).
Collapse
Affiliation(s)
- Yusuf Ziya Ziylan
- Department of Physiology, Istanbul Medical School, Istanbul University, Capa, Istanbul, Turkey
| | | | | |
Collapse
|
23
|
Fan W, Yanase T, Nishi Y, Chiba S, Okabe T, Nomura M, Yoshimatsu H, Kato S, Takayanagi R, Nawata H. Functional potentiation of leptin-signal transducer and activator of transcription 3 signaling by the androgen receptor. Endocrinology 2008; 149:6028-36. [PMID: 18703637 DOI: 10.1210/en.2008-0431] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hypogonadism is associated with increased fat mass and dysregulation of metabolic homeostasis in men. Our previous study revealed that androgen receptor (AR)-null male mice (ARL-/Y) develop late-onset obesity and are leptin-resistant. The present study evaluated how hypothalamic AR contributes to central leptin-signal transducer and activator of transcription 3 (STAT3) signaling. We evaluated leptin action in wild-type and ARL-/Y mice, the anatomic co-relationship between AR and leptin signaling in the hypothalamus, and the effects of AR on leptin-mediated STAT3 transactivation and nuclear translocation. AR deletion in male mice results in a weaker leptin-induced suppression of food intake and body weight drop even before the onset of overt obesity. In wild-type male but not female mice, AR was highly expressed in various hypothalamic nuclei that also expressed the long-form leptin receptor (OBRB) and co-resided with OBRB directly in the arcuate neurons. In vitro, AR significantly enhanced STAT3-mediated transcription of leptin target genes including POMC and SOCS3. This effect relied on the AR N-terminal activation function-1 (AF-1) domain and was specific to AR in that none of the other sex steroid hormone receptors tested showed similar effects. AR enhanced the low concentrations of leptin-induced STAT3 nuclear translocation in vitro, and ARL-/Y mice receiving leptin had impaired STAT3 nuclear localization in the arcuate neurons. These findings indicate that AR in the hypothalamus functions as a regulator of central leptin-OBRB-STAT3 signaling and has a physiological role in energy homeostasis and metabolic regulation in male mice.
Collapse
Affiliation(s)
- WuQiang Fan
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Rummel C, Inoue W, Sachot C, Poole S, Hübschle T, Luheshi GN. Selective contribution of interleukin-6 and leptin to brain inflammatory signals induced by systemic LPS injection in mice. J Comp Neurol 2008; 511:373-95. [DOI: 10.1002/cne.21850] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Signore AP, Zhang F, Weng Z, Gao Y, Chen J. Leptin neuroprotection in the CNS: mechanisms and therapeutic potentials. J Neurochem 2008; 106:1977-90. [PMID: 18466320 PMCID: PMC2634657 DOI: 10.1111/j.1471-4159.2008.05457.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Leptin is well known as a hormone important in the central control of appetitive behaviors via receptor-mediated actions in the hypothalamus, where leptin adjusts food intake to maintain homeostasis with the body's energy stores. Recent evidence has shown that leptin and its receptors are widespread in the CNS and may provide neuronal survival signals. This review summarizes our current knowledge of how leptin functions in the brain and then focuses on the ability of leptin to mitigate neuronal damage in experimental models of human neurological disorders. Damage to the brain by acute events such as stroke, or long-term loss of neurons associated with neurodegenerative diseases, including Parkinson's and Alzheimer's disease, may be amenable to treatment using leptin to limit death of susceptible cells. Leptin-mediated pro-survival signaling is now known to prevent the death of neurons in these models. The signaling cascades that leptin generates are shared by other neuroprotective molecules including insulin and erythropoietin, and are thus a component of the neurotrophic effects mediated by endogenous hormones. Coupled with evidence that leptin dysregulation in human disease also results in enhanced neuronal susceptibility to damage, development of leptin as a therapeutic methodology is an attractive and viable possibility.
Collapse
Affiliation(s)
- Armando P. Signore
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Feng Zhang
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Zhongfang Weng
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - YanQing Gao
- State Key Laboratory of Medical Neurobiology, Fudan University School of Medicine, Shanghai, China 200032
| | - Jun Chen
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
- State Key Laboratory of Medical Neurobiology, Fudan University School of Medicine, Shanghai, China 200032
- Geriatric Research, Educational and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
26
|
Frontini A, Bertolotti P, Tonello C, Valerio A, Nisoli E, Cinti S, Giordano A. Leptin-dependent STAT3 phosphorylation in postnatal mouse hypothalamus. Brain Res 2008; 1215:105-115. [PMID: 18485333 DOI: 10.1016/j.brainres.2008.03.078] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Revised: 03/18/2008] [Accepted: 03/26/2008] [Indexed: 01/30/2023]
Abstract
Leptin, a hormone produced by adipose tissue, reduces food intake and boosts energy expenditure via activation of the JAK2-STAT3 signalling pathway in adult mammal hypothalamic neurons. It is found in blood early after birth, peaking around postnatal day (P) 10. The hypothalamus of neonatal mice administered intraperitoneal leptin (3 mg/kg of body weight) was investigated for phospho-STAT3-positive cells to gain insights into the timing of maturation of the leptin signal transduction system. Leptin responsiveness was first detected in arcuate nucleus, where it was faint at P1 and evident from P5. It was then identified in medial preoptic area, anterior hypothalamus, retrochiasmatic area, dorsomedial nucleus and premammillary nucleus from P7, and in ventromedial nucleus and lateral hypothalamus from P11. From P13 onwards, hypothalamic P-STAT3 staining was indistinguishable from that of adult mice. Significant hypothalamic STAT3 activation was also detected by Western blotting at P11 and P15. The level of activation seen in adults was comparable to that observed at P15 although, remarkably, leptin-induced feeding reduction is observed only after the fourth postnatal week. Neuronal and glial markers and double-labelling immunohistochemistry showed that leptin-stimulated hypothalamic cells that had already reached their final position in a given area or nucleus were neurons; however, leptin responsiveness preceded positivity for the neuronal markers, suggesting a not fully differentiated status. Interestingly, leptin also increased P-STAT3 and c-Fos immunoreactivity in a distinctive and transient (from P5 to P13) cell population found in the dorsal part of the third ventricle and in subependymal position. These cells did not express mature or immature neuronal or glial markers. Their ultrastructural appearance, though suggestive of differentiating cells, was not conclusive for a specific phenotype.
Collapse
Affiliation(s)
- Andrea Frontini
- Institute of Normal Human Morphology, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Angiogenesis, the formation of new blood vessels from the pre-existing vasculature, is a complex multistage process regulated by a number of signal transduction pathways. Accumulating evidence suggests that signal transducer and activator of transcription (STATs), mainly STAT3, play an important role in angiogenesis under both physiological and pathological conditions in addition to cell survival, proliferation, differentiation, and oncogenesis. STAT3, as a critical multifunctional mediator, regulates many aspects of angiogenesis at the transcriptional level. This review will highlight the pivotal role of STAT3 in well-studied tumorous angiogenesis and cardiac angiogenesis, and summarize various potential mechanisms utilized by STAT3 to regulate the transcriptional activation of VEGF.
Collapse
Affiliation(s)
- Zhong Chen
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, P. R. China
| | | |
Collapse
|
28
|
Xie Q, Chen XC, Gong Y, Gu YX. Leptin plays a role in ruptured human brain arteriovenous malformations. ACTA NEUROCHIRURGICA. SUPPLEMENT 2008; 105:221-224. [PMID: 19066113 DOI: 10.1007/978-3-211-09469-3_42] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
INTRODUCTION Intracerebral hemorrhage (ICH) is one of the most common clinical manifestations of human brain arteriovenous malformation (BAVM). However, the hemorrhagic mechanism of BAVM is still unclear. Leptin, first discovered in obesity research, has not been systematically studied in BAVM and ICH. We investigated expression and effect of leptin on human BAVM. METHODS Specimens were obtained from 6 BAVM patients, who had been divided into either hemorrhagic or non-hemorrhagic groups. Leptin, leptin receptor, and signal transducers and activators of transcription-3 (STAT3) were analyzed by different methods, such as gene chips, reverse transcription-polymerase chain reaction (RT-PCR), immunohistochemistry, and Western blot. Perinidal brain tissue around each BAVM served as control. RESULTS Gene chips and RT-PCR found transcriptional leptin raised at least 2 levels in hemorrhagic BAVM. Immunohistochemical slices also showed higher expression of leptin, leptin receptor, and STAT3 on nidus part of hemorrhagic BAVM than non-hemorrhagic ones. On Western blot analysis, hemorrhagic BAVMs had higher levels of leptin (p < 0.01). CONCLUSIONS The transcriptional and translational levels of leptin, leptin receptor, and STAT3 were higher in hemorrhagic BAVM, suggesting that leptin may play an important role in the hemorrhagic mechanism of BAVM.
Collapse
Affiliation(s)
- Q Xie
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
29
|
Weng Z, Signore AP, Gao Y, Wang S, Zhang F, Hastings T, Yin XM, Chen J. Leptin protects against 6-hydroxydopamine-induced dopaminergic cell death via mitogen-activated protein kinase signaling. J Biol Chem 2007; 282:34479-91. [PMID: 17895242 DOI: 10.1074/jbc.m705426200] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The death of midbrain dopaminergic neurons in sporadic Parkinson disease is of unknown etiology but may involve altered growth factor signaling. The present study showed that leptin, a centrally acting hormone secreted by adipocytes, rescued dopaminergic neurons, reversed behavioral asymmetry, and restored striatal catecholamine levels in the unilateral 6-hydroxydopamine (6-OHDA) mouse model of dopaminergic cell death. In vitro studies using the murine dopaminergic cell line MN9D showed that leptin attenuated 6-OHDA-induced apoptotic markers, including caspase-9 and caspase-3 activation, internucleosomal DNA fragmentation, and cytochrome c release. ERK1/2 phosphorylation (pERK1/2) was found to be critical for mediating leptin-induced neuroprotection, because inhibition of the MEK pathway blocked both the pERK1/2 response and the pro-survival effect of leptin in cultures. Knockdown of the downstream messengers JAK2 or GRB2 precluded leptin-induced pERK1/2 activation and neuroprotection. Leptin/pERK1/2 signaling involved phosphorylation and nuclear localization of CREB (pCREB), a well known survival factor for dopaminergic neurons. Leptin induced a marked MEK-dependent increase in pCREB that was essential for neuroprotection following 6-OHDA toxicity. Transfection of a dominant negative MEK protein abolished leptin-enhanced pCREB formation, whereas a dominant negative CREB or decoy oligonucleotide diminished both pCREB binding to its target DNA sequence and MN9D survival against 6-OHDA toxicity. Moreover, in the substantia nigra of mice, leptin treatment increased the levels of pERK1/2, pCREB, and the downstream gene product BDNF, which were reversed by the MEK inhibitor PD98059. Collectively, these data provide evidence that leptin prevents the degeneration of dopaminergic neurons by 6-OHDA and may prove useful in the treatment of Parkinson disease.
Collapse
Affiliation(s)
- Zhongfang Weng
- Department of Neurology, and Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Tu H, Pan W, Feucht L, Kastin AJ. Convergent trafficking pattern of leptin after endocytosis mediated by ObRa-ObRd. J Cell Physiol 2007; 212:215-22. [PMID: 17323382 DOI: 10.1002/jcp.21020] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The cellular effects of leptin are dependent on the receptor subtypes that mediate the signaling and fate of endocytosed leptin inside the cells. In this study, we examined the differences in receptor expression, endocytosis, intracellular degradation, and exocytosis of a trace amount of leptin in cells overexpressing ObRb and short forms of the leptin receptor. The relative contribution of proteasomes and lysosomes in the intracellular fate of leptin was also determined. There were three unusual findings: (1) all receptor subtypes could mediate the binding and endocytosis of leptin, although ObRb was expressed at a lower level than ObRa, ObRc, and ObRd after transient transfection. This indicates that ObRb can be a transporting receptor. (2) Once internalized, the intracellular degradation pattern and exocytosis of leptin were independent of the receptor subtype. (3) Endocytosed leptin could remain intact for at least 1 h. This stability was further enhanced by inhibition of lysosomal activity. Thus, the intracellular pool of intact leptin may allow prolonged biological functions for this adipokine.
Collapse
Affiliation(s)
- Hong Tu
- Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | | | | | | |
Collapse
|
31
|
Sachot C, Rummel C, Bristow AF, Luheshi GN. The role of the vagus nerve in mediating the long-term anorectic effects of leptin. J Neuroendocrinol 2007; 19:250-61. [PMID: 17355316 DOI: 10.1111/j.1365-2826.2006.01528.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Leptin, the product of the obese (ob) gene, is mainly known for its regulatory role of energy balance by direct activation of hypothalamic receptors. Recently, its function in the acute control of food intake was additionally attributed to activation of the vagus nerve to regulate meal termination. Whether vagal afferent neurones are involved in longer term effects of leptin on food intake, however, remains undetermined. Using vagotomised (VGX) rats, we sought to clarify the contributions of vagal afferents in mediating the long-lasting effect of leptin on appetite suppression. Intraperitoneal (i.p.) injection of leptin (3.5 mg/kg) attenuated food intake at 4, 6, 8 and 24 h and body weight at 24 h postinjection in SHAM-operated rats; however, this response was not abrogated by vagotomy. In a separate study using immunohistochemistry, we observed leptin-induced Fos expression in the nucleus tractus solitarii, a brain structure where vagal afferent fibres terminate. This signal was not attenuated in VGX animals compared to the SHAM group. Moreover, leptin treatment led to a similar level of nuclear STAT3 translocation, a marker of leptin signalling, in the hypothalami of SHAM and VGX animals. In addition to the effects of leptin, vagotomy surgery itself resulted in a decrease of 24 h food intake. Analyses of brains from saline-treated VGX animals revealed a significant induction of Fos in the nucleus tractus solitarii and changes in agouti-related peptide and pro-opiomelanocortin mRNA expression in the hypothalamus compared to their SHAM counterparts, indicating that the vagotomy surgery itself induced a modification of brain activity in areas involved in regulating appetite. Collectively, our data suggest that vagal afferents do not constitute a major route of mediating the regulatory effect of leptin on food intake over a period of several hours.
Collapse
Affiliation(s)
- C Sachot
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
32
|
Voss T, Barth SW, Rummel C, Gerstberger R, Hübschle T, Roth J. STAT3 and COX-2 activation in the guinea-pig brain during fever induced by the Toll-like receptor-3 agonist polyinosinic:polycytidylic acid. Cell Tissue Res 2007; 328:549-61. [PMID: 17345100 DOI: 10.1007/s00441-007-0386-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2006] [Accepted: 01/26/2007] [Indexed: 01/09/2023]
Abstract
Intra-arterial injections of synthetic double-stranded RNA (polyinosinic:polycytidylic acid, PIPC) at a dose of 500 microg/kg evoked pronounced fever in guinea-pigs. PIPC-induced fever could be antagonized by treatment with the non-selective cyclooxygenase (COX) inhibitor diclofenac and was, in part, attenuated by the administration of the selective COX-2-inhibitor nimesulide (dose: 5 mg/kg for both COX inhibitors). We further investigated whether direct activation of brain cells during PIPC-induced fever could be demonstrated. Using radioactive in situ hybridization, we demonstrated that treatment with PIPC resulted in an upregulation of COX-2 and interleukin-1 beta mRNA in the guinea-pig brain. Thus, COX-2-specific hybridization signals seemed to be mainly associated with brain blood vessels. Intra-arterial injections of PIPC further induced the pronounced nuclear translocation of the transcription factor STAT3 in the endothelium of various fore- and hindbrain areas and in the meninges. In brain structures that lacked a tight blood-brain barrier, i.e. the sensory circumventricular organs (area postrema, vascular organ of laminae terminalis, subfornical organ), the astrocytes and a population of still undetermined cellular phenotype also showed marked STAT3 activation in response to PIPC. The Toll-like receptor-3 agonist PIPC therefore caused a similar activation of brain cells as that reported for other experimental models of systemic inflammation.
Collapse
Affiliation(s)
- Thilo Voss
- Institut für Veterinär-Physiologie, Justus-Liebig-Universität Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Mütze J, Roth J, Gerstberger R, Hübschle T. Nuclear translocation of the transcription factor STAT5 in the rat brain after systemic leptin administration. Neurosci Lett 2007; 417:286-91. [PMID: 17353091 DOI: 10.1016/j.neulet.2007.02.074] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 01/26/2007] [Accepted: 02/20/2007] [Indexed: 10/23/2022]
Abstract
Leptin binding to its functional receptor stimulates JAK-STAT-signaling pathway, which finally results in activation and nuclear translocation of transcription factors of the signal transducer and activator of transcription (STAT) family, namely of STAT3. Here we report for the first time that systemic treatment with leptin (5 mg/kg; intraperitoneal injection) also increased the number of nuclear STAT5 signals in the hypothalamus. In particular, the entire arcuate nucleus (ARC), the ventral premammilary nucleus (PMV), and the supraoptic nucleus (SO) showed an enhanced nuclear STAT5 translocation in response to leptin when compared to saline, 120 min after the respective injection. Co-localization studies revealed that a high percentage of those STAT5-responsive cells proved to be neurons. In addition, some astrocytes within the ARC showed nuclear STAT5 signals. The functional relevance of leptin-induced nuclear STAT5 activation in hypothalamic cells still has to be determined.
Collapse
Affiliation(s)
- Jörg Mütze
- Department of Veterinary-Physiology, Justus-Liebig-University Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | | | | | | |
Collapse
|
34
|
Tarantino G, Saldalamacchia G, Conca P, Arena A. Non-alcoholic fatty liver disease: further expression of the metabolic syndrome. J Gastroenterol Hepatol 2007; 22:293-303. [PMID: 17295757 DOI: 10.1111/j.1440-1746.2007.04824.x] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease has been associated with metabolic disorders, including central obesity, dyslipidemia, hypertension and hyperglycemia. Metabolic syndrome, obesity, and insulin resistance are major risk factors in the pathogenesis of non-alcoholic fatty liver disease. Non-alcoholic fatty liver disease refers to a wide spectrum of liver damage, ranging from simple steatosis to non-alcoholic steatohepatitis, advanced fibrosis and cirrhosis.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical and Experimental Medicine, Federico II University Medical School, Naples, Italy.
| | | | | | | |
Collapse
|
35
|
Walker CD, Long H, Williams S, Richard D. Long-lasting effects of elevated neonatal leptin on rat hippocampal function, synaptic proteins and NMDA receptor subunits. J Neurosci Res 2007; 85:816-28. [PMID: 17245750 DOI: 10.1002/jnr.21173] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The high circulating levels of leptin in neonatal rodents do not seem to be regulating energy balance at this age, but rather may play an important role for brain development. We tested the hypothesis that high neonatal leptin levels modify hippocampal function and production of synaptic proteins with possible long-term consequences on long-term potentiation (LTP) in adulthood. We first showed that in postnatal day (PND) 10 neonates, acute leptin treatment functionally activated leptin receptors (ObR) in the CA1 and DG regions of the hippocampus through the induction of phosphoERK1/2, but not phosphoSTAT3 protein although both phospho-proteins were induced in the arcuate nucleus. We next examined whether chronic leptin administration (3 mg/kg BW, intraperitoneally) during the first 2 weeks of life (postnatal day, PND 2-14) produces a functional signal in the hippocampus that alters the expression of NMDA receptor subunits (NR1, NR2A, NR2B), synaptic proteins and LTP in the short and long-term. In PND 10 as in adults (PND 70) rats, chronic leptin treatment increased NR1 expression in the hippocampus while reducing NR2B protein levels. Elevated hippocampal concentrations of synapsin2A and synaptophysin were detected during leptin treatment on PND 10 suggesting increased neurotransmitter release. In adults, only SNAP-25 expression was increased after neonatal leptin treatment. LTP was reduced dramatically by leptin treatment in preweaning rats although the changes did not persist until adulthood. Elevated exposure to leptin during a critical period of neonatal hippocampal development might serve to enhance NMDA-dependent functions other than LTP and have important effects on synaptogenesis and neurotransmitter release.
Collapse
Affiliation(s)
- Claire-Dominique Walker
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, Montreal, Quebec, Canada.
| | | | | | | |
Collapse
|
36
|
Inoue W, Poole S, Bristow AF, Luheshi GN. Leptin induces cyclooxygenase-2 via an interaction with interleukin-1beta in the rat brain. Eur J Neurosci 2006; 24:2233-45. [PMID: 17074047 DOI: 10.1111/j.1460-9568.2006.05105.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In addition to its central effects on appetite regulation, leptin has been implicated in immune function and inflammation. Previous data suggested that leptin acts as an inflammatory signal within the brain, as exogenously administered leptin induced fever, a typical brain-regulated inflammatory response. The present study aimed to delineate the inflammatory actions and cellular targets of leptin in the brain by examining its effects on the expression of interleukin (IL)-1beta and cyclooxygenase (COX)-2, two important inflammatory components of the fever response. Intracerebroventricular injection of leptin (5 microg/rat) induced IL-1beta and COX-2 mRNA and protein in the hypothalamus between 1 and 3 h after treatment as determined by reverse transcription-polymerase chain reaction and immunohistochemistry. Coinjection of IL-1 receptor antagonist (100 microg/rat, intracerebroventricular) attenuated leptin-induced COX-2, whereas IL-1 receptor antagonist had no effect on endogenous IL-1beta levels, suggesting that leptin induces COX-2 via, at least partly, IL-1beta action. IL-1beta protein expression was induced in macrophages in the meningis and perivascular space after leptin treatment, whereas COX-2 induction was observed in endothelial cells, indicating the roles for these non-neuronal cells in mediating inflammatory actions of leptin. In addition, neutralization of endogenous circulating leptin with anti-leptin antiserum attenuated intraperitoneal lipopolysaccharide (100 microg/kg)-induced brain IL-1beta and COX-2 upregulation, suggesting that leptin indeed acts as an inflammatory signal to the brain during systemic inflammation. These findings are in contrast to the effects of leptin on appetite regulation where it is believed to act primarily on neurons, thus presenting a distinct anatomical basis for the inflammatory and appetite regulatory actions of leptin in the brain.
Collapse
Affiliation(s)
- Wataru Inoue
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada
| | | | | | | |
Collapse
|