1
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2025; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [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: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
2
|
Kumari S, Kamiya A, Karnik SS, Rohilla S, Dubey SK, Taliyan R. Novel Gene Therapy Approaches for Targeting Neurodegenerative Disorders: Focusing on Delivering Neurotrophic Genes. Mol Neurobiol 2025; 62:386-411. [PMID: 38856793 DOI: 10.1007/s12035-024-04260-y] [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: 12/06/2023] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
Abstract
Neurodegenerative illnesses (NDDs) like Alzheimer's, Parkinson's, amyotrophic lateral sclerosis, spinal muscular atrophy, and Huntington's disease have demonstrated considerable potential for gene therapy as a viable therapeutic intervention. NDDs are marked by the decline of neurons, resulting in changes in both behavior and pathology within the body. Strikingly, only symptomatic management is available without a cure for the NDDs. There is an unmet need for a permanent therapeutic approach. Many studies have been going on to target the newer therapeutic molecular targets for NDDs including gene-based therapy. Gene therapy has the potential to provide therapeutic benefits to a large number of patients with NDDs by offering mechanisms including neuroprotection, neuro-restoration, and rectification of pathogenic pathways. Gene therapy is a medical approach that aims to modify the biological characteristics of living cells by controlling the expression of specific genes in certain neurological disorders. Despite being the most complex and well-protected organ in the human body, there is clinical evidence to show that it is possible to specifically target the central nervous system (CNS). This provides hope for the prospective application of gene therapy in treating NDDs in the future. There are several advanced techniques available for using viral or non-viral vectors to deliver the therapeutic gene to the afflicted region. Neurotrophic factors (NTF) in the brain are crucial for the development, differentiation, and survival of neurons in the CNS, making them important in the context of various neurological illnesses. Gene delivery of NTF has the potential to be used as a therapeutic approach for the treatment of neurological problems in the brain. This review primarily focuses on the methodologies employed for delivering the genes of different NTFs to treat neurological disorders. These techniques are currently being explored as a viable therapeutic approach for neurodegenerative diseases. The article exclusively addresses gene delivery approaches and does not cover additional therapy strategies for NDDs. Gene therapy offers a promising alternative treatment for NDDs by stimulating neuronal growth instead of solely relying on symptom relief from drugs and their associated adverse effects. It can serve as a long-lasting and advantageous treatment choice for the management of NDDs. The likelihood of developing NDDs increases with age as a result of neuronal degradation in the brain. Gene therapy is an optimal approach for promoting neuronal growth through the introduction of nerve growth factor genes.
Collapse
Affiliation(s)
- Shobha Kumari
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Aayush Kamiya
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sanika Sanjay Karnik
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sumedha Rohilla
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | | | - Rajeev Taliyan
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India.
| |
Collapse
|
3
|
McClarty BM, Rodriguez G, Dong H. Class 1 histone deacetylases differentially modulate memory and synaptic genes in a spatial and temporal manner in aged and APP/PS1 mice. Brain Res 2024; 1837:148951. [PMID: 38642789 PMCID: PMC11182336 DOI: 10.1016/j.brainres.2024.148951] [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: 01/23/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Epigenetics plays a vital role in aging and Alzheimer's disease (AD); however, whether epigenetic alterations during aging can initiate AD and exacerbate AD progression remains unclear. In this study, using 3-, 12- and 18- month-old APP/PS1 mice and age matched WT littermates, we conducted a series of memory tests, measured synapse-related gene expression, class 1 histone deacetylases (HDACs) abundance, and H3K9ac levels at target gene promoters in the hippocampus and prefrontal cortex (PFC). Our results showed impaired recognition and long-term spatial memory in 18-month-old WT mice and impaired recognition, short-term working, and long-term spatial reference memory in 12-and 18- month-old APP/PS1 mice. These memory impairments are associated with changes of synapse-related gene (nr2a, glur1, glur2, psd95) expression, HDAC abundance, and H3K9ac levels; more specifically, increased HDAC2 was associated with synapse-related gene expression changes through modulation of H3K9ac at the gene promoters during aging and AD progression in the hippocampus. Conversely, increased HDAC3 was associated with synapse-related gene expression changes through modulation of H3K9ac at the gene promoters during AD progression in the PFC. These findings suggest memory impairments in aging and AD may associated with a differential HDAC modulation of synapse-related gene expression in the brain.
Collapse
Affiliation(s)
- Bryan M McClarty
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA.
| |
Collapse
|
4
|
Harvey J. Novel Leptin-Based Therapeutic Strategies to Limit Synaptic Dysfunction in Alzheimer's Disease. Int J Mol Sci 2024; 25:7352. [PMID: 39000459 PMCID: PMC11242278 DOI: 10.3390/ijms25137352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Accumulation of hyper-phosphorylated tau and amyloid beta (Aβ) are key pathological hallmarks of Alzheimer's disease (AD). Increasing evidence indicates that in the early pre-clinical stages of AD, phosphorylation and build-up of tau drives impairments in hippocampal excitatory synaptic function, which ultimately leads to cognitive deficits. Consequently, limiting tau-related synaptic abnormalities may have beneficial effects in AD. There is now significant evidence that the hippocampus is an important brain target for the endocrine hormone leptin and that leptin has pro-cognitive properties, as activation of synaptic leptin receptors markedly influences higher cognitive processes including learning and memory. Clinical studies have identified a link between the circulating leptin levels and the risk of AD, such that AD risk is elevated when leptin levels fall outwith the physiological range. This has fuelled interest in targeting the leptin system therapeutically. Accumulating evidence supports this possibility, as numerous studies have shown that leptin has protective effects in a variety of models of AD. Recent findings have demonstrated that leptin has beneficial effects in the preclinical stages of AD, as leptin prevents the early synaptic impairments driven by tau protein and amyloid β. Here we review recent findings that implicate the leptin system as a potential novel therapeutic target in AD.
Collapse
Affiliation(s)
- Jenni Harvey
- Department of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
5
|
Carbone G, Bencivenga L, Santoro MA, De Lucia N, Palaia ME, Ercolano E, Scognamiglio F, Edison P, Ferrara N, Vitale DF, Rengo G, Femminella GD. Impact of serum leptin and adiponectin levels on brain infarcts in patients with mild cognitive impairment and Alzheimer's disease: a longitudinal analysis. Front Endocrinol (Lausanne) 2024; 15:1389014. [PMID: 38686200 PMCID: PMC11056582 DOI: 10.3389/fendo.2024.1389014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/27/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction The adipokines leptin and adiponectin have been associated with atherosclerosis and the risk of cerebral infarcts. Pre-clinical studies, however, suggest a protective role against ischemic brain damage. In this study we analyzed the relationship between serum leptin and adiponectin levels and the onset or progression of brain infarcts in subjects with mild cognitive impairment (MCI) and Alzheimer's disease (AD). Methods All data were extracted from the ADNI database. The final population included 566 subjects, with 58 healthy controls, 396 MCI and 112 AD. All patients with available serum leptin and adiponectin levels at baseline were selected. Demographics, neuropsychological test results, CSF biomarkers, regional brain metabolism with FDG-PET data and the number of brain infarcts on longitudinal MRI scans were extracted. Results Leptin levels were significantly lower in patients with MCI than controls at baseline, while adiponectin levels were not different between the groups. Multivariate logistic regression analysis at baseline for the presence of brain infarcts showed a predictive value for leptin but not for adiponectin. Multivariate longitudinal analysis showed that age was the only significant predictor of brain infarcts development at 15-year follow-up, while serum leptin and adiponectin levels did not play a role in this population. Discussion The evidence on the pathogenetic or protective role of adipokines on ischemic brain damage is mixed. In this MCI and AD population, serum leptin and adiponectin were not associated with the development of brain infarcts; therefore, these results do not support the use of adipokines as biomarkers of cerebrovascular pathology in this population.
Collapse
Affiliation(s)
- Giovanni Carbone
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
| | - Maria Angela Santoro
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
| | - Natascia De Lucia
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, “Federico II” University, Naples, Italy
| | - Maria Emiliana Palaia
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
| | - Erica Ercolano
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
| | | | - Paul Edison
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Nicola Ferrara
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
| | | | - Giuseppe Rengo
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
- Laboratorio di fisiopatologia del sistema neurovegetativo, Istituti Clinici Scientifici Maugeri Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) - Scientific Institute of Telese Terme, Telese Terme, BN, Italy
| | - Grazia Daniela Femminella
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
Neto A, Fernandes A, Barateiro A. The complex relationship between obesity and neurodegenerative diseases: an updated review. Front Cell Neurosci 2023; 17:1294420. [PMID: 38026693 PMCID: PMC10665538 DOI: 10.3389/fncel.2023.1294420] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is a global epidemic, affecting roughly 30% of the world's population and predicted to rise. This disease results from genetic, behavioral, societal, and environmental factors, leading to excessive fat accumulation, due to insufficient energy expenditure. The adipose tissue, once seen as a simple storage depot, is now recognized as a complex organ with various functions, including hormone regulation and modulation of metabolism, inflammation, and homeostasis. Obesity is associated with a low-grade inflammatory state and has been linked to neurodegenerative diseases like multiple sclerosis (MS), Alzheimer's (AD), and Parkinson's (PD). Mechanistically, reduced adipose expandability leads to hypertrophic adipocytes, triggering inflammation, insulin and leptin resistance, blood-brain barrier disruption, altered brain metabolism, neuronal inflammation, brain atrophy, and cognitive decline. Obesity impacts neurodegenerative disorders through shared underlying mechanisms, underscoring its potential as a modifiable risk factor for these diseases. Nevertheless, further research is needed to fully grasp the intricate connections between obesity and neurodegeneration. Collaborative efforts in this field hold promise for innovative strategies to address this complex relationship and develop effective prevention and treatment methods, which also includes specific diets and physical activities, ultimately improving quality of life and health.
Collapse
Affiliation(s)
- Alexandre Neto
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Adelaide Fernandes
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Barateiro
- Central Nervous System, Blood and Peripheral Inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
7
|
Ma J, Hou YH, Liao ZY, Ma Z, Zhang XX, Wang JL, Zhu YB, Shan HL, Wang PY, Li CB, Lv YL, Wei YL, Dou JZ. Neuroprotective Effects of Leptin on the APP/PS1 Alzheimer's Disease Mouse Model: Role of Microglial and Neuroinflammation. Degener Neurol Neuromuscul Dis 2023; 13:69-79. [PMID: 37905186 PMCID: PMC10613410 DOI: 10.2147/dnnd.s427781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/05/2023] [Indexed: 11/02/2023] Open
Abstract
Background Microglia are closely linked to Alzheimer's disease (AD) many years ago; however, the pathological mechanisms of AD remain unclear. The purpose of this study was to determine whether leptin affected microglia in the hippocampus of young and aged male APP/PS1 mice. Objective In a transgenic model of AD, we investigated the association between intraperitoneal injection of leptin and microglia. Methods We intraperitoneal injection of leptin (1mg/kg) every day for one week and analyzed inflammatory markers in microglia in the hippocampus of adult (6 months) and aged (12 months) APP/PS1 mice. Results In all leptin treatment group, the brain Aβ levels were decrease. We found increased levels of IL-1β, IL-6 and microglial activation in the hippocampus of adult mice. Using aged mice as an experimental model for chronic neuroinflammation and leptin resistance, the number of Iba-1+ microglia and the levels of IL-1β/IL-6 in the hippocampus were greatly increased as compared to the adult. But between the leptin treatment and un-treatment, there were no difference. Conclusion Leptin signaling would regulate the activation of microglia and the release of inflammatory factors, but it is not the only underlying mechanism in the neuroprotective effects of AD pathogenesis.
Collapse
Affiliation(s)
- Jing Ma
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Yi-Hui Hou
- Department of Neurology, Chengde Medical University Affiliated Hospital, School of Medicine, Chengde Medical University, Chengde, People’s Republic of China
| | - Zhe-Yan Liao
- Department of Neurology, Chengde Medical University Affiliated Hospital, School of Medicine, Chengde Medical University, Chengde, People’s Republic of China
| | - Zheng Ma
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Xiao-Xuan Zhang
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Jian-Li Wang
- Department of Hepatobiliary Surgery, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Yun-Bo Zhu
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Hai-Lei Shan
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Ping-Yue Wang
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Cheng-Bo Li
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Ying-Lei Lv
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Yi-Lan Wei
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| | - Jie-Zhi Dou
- Department of Neurology, Chengde Medical University Affiliated Hospital, Chengde Medical University, Chengde, People’s Republic of China
| |
Collapse
|
8
|
Reddy I, Yadav Y, Dey CS. Cellular and Molecular Regulation of Exercise-A Neuronal Perspective. Cell Mol Neurobiol 2023; 43:1551-1571. [PMID: 35986789 PMCID: PMC11412429 DOI: 10.1007/s10571-022-01272-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
Abstract
The beneficial effects of exercise on the proper functioning of the body have been firmly established. Multi-systemic metabolic regulation of exercise is the consequence of multitudinous changes that occur at the cellular level. The exercise responsome comprises all molecular entities including exerkines, miRNA species, growth factors, signaling proteins that are elevated and activated by physical exercise. Exerkines are secretory molecules released by organs such as skeletal muscle, adipose tissue, liver, and gut as a function of acute/chronic exercise. Exerkines such as FNDC5/irisin, Cathepsin B, Adiponectin, and IL-6 circulate through the bloodstream, cross the blood-brain barrier, and modulate the expression of important signaling molecules such as AMPK, SIRT1, PGC1α, BDNF, IGF-1, and VEGF which further contribute to improved energy metabolism, glucose homeostasis, insulin sensitivity, neurogenesis, synaptic plasticity, and overall well-being of the body and brain. These molecules are also responsible for neuroprotective adaptations that exercise confers on the brain and potentially ameliorate neurodegeneration. This review aims to detail important cellular and molecular species that directly or indirectly mediate exercise-induced benefits in the body, with an emphasis on the central nervous system.
Collapse
Affiliation(s)
- Ishitha Reddy
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Yamini Yadav
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India.
| |
Collapse
|
9
|
Corrigan RR, Labrador L, Grizzanti J, Mey M, Piontkivska H, Casadesús G. Neuroprotective Mechanisms of Amylin Receptor Activation, Not Antagonism, in the APP/PS1 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2023; 91:1495-1514. [PMID: 36641678 DOI: 10.3233/jad-221057] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Amylin, a pancreatic amyloid peptide involved in energy homeostasis, is increasingly studied in the context of Alzheimer's disease (AD) etiology. To date, conflicting pathogenic and neuroprotective roles for this peptide and its analogs for AD pathogenesis have been described. OBJECTIVE Whether the benefits of amylin are associated with peripheral improvement of metabolic tone/function or directly through the activation of central amylin receptors is also unknown and downstream signaling mechanisms of amylin receptors are major objectives of this study. METHODS To address these questions more directly we delivered the amylin analog pramlintide systemically (IP), at previously identified therapeutic doses, while centrally (ICV) inhibiting the receptor using an amylin receptor antagonist (AC187), at doses known to impact CNS function. RESULTS Here we show that pramlintide improved cognitive function independently of CNS receptor activation and provide transcriptomic data that highlights potential mechanisms. Furthermore, we show than inhibition of the amylin receptor increased amyloid-beta pathology in female APP/PS1 mice, an effect than was mitigated by peripheral delivery of pramlintide. Through transcriptomic analysis of pramlintide therapy in AD-modeled mice we found sexual dimorphic modulation of neuroprotective mechanisms: oxidative stress protection in females and membrane stability and reduced neuronal excitability markers in males. CONCLUSION These data suggest an uncoupling of functional and pathology-related events and highlighting a more complex receptor system and pharmacological relationship that must be carefully studied to clarify the role of amylin in CNS function and AD.
Collapse
Affiliation(s)
| | - Luis Labrador
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - John Grizzanti
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Megan Mey
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Helen Piontkivska
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Gemma Casadesús
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Grasso P. Harnessing the Power of Leptin: The Biochemical Link Connecting Obesity, Diabetes, and Cognitive Decline. Front Aging Neurosci 2022; 14:861350. [PMID: 35527735 PMCID: PMC9072663 DOI: 10.3389/fnagi.2022.861350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/21/2022] [Indexed: 12/02/2022] Open
Abstract
In this review, the current understanding of leptin’s role in energy balance, glycemic regulation, and cognitive function is examined, and its involvement in maintaining the homeostatic “harmony” of these physiologies is explored. The effects of exercise on circulating leptin levels are summarized, and the results of clinical application of leptin to metabolic disease and neurologic dysfunction are reviewed. Finally, pre-clinical evidence is presented which suggests that synthetic peptide leptin mimetics may be useful in resolving not only the leptin resistance associated with common obesity and other elements of metabolic syndrome, but also the peripheral insulin resistance characterizing type 2 diabetes mellitus, and the central insulin resistance associated with certain neurologic deficits in humans.
Collapse
Affiliation(s)
- Patricia Grasso
- Department of Medicine, Albany Medical College, Albany, NY, United States
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
- *Correspondence: Patricia Grasso,
| |
Collapse
|
11
|
Serrano ME, Kim E, Petrinovic MM, Turkheimer F, Cash D. Imaging Synaptic Density: The Next Holy Grail of Neuroscience? Front Neurosci 2022; 16:796129. [PMID: 35401097 PMCID: PMC8990757 DOI: 10.3389/fnins.2022.796129] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The brain is the central and most complex organ in the nervous system, comprising billions of neurons that constantly communicate through trillions of connections called synapses. Despite being formed mainly during prenatal and early postnatal development, synapses are continually refined and eliminated throughout life via complicated and hitherto incompletely understood mechanisms. Failure to correctly regulate the numbers and distribution of synapses has been associated with many neurological and psychiatric disorders, including autism, epilepsy, Alzheimer’s disease, and schizophrenia. Therefore, measurements of brain synaptic density, as well as early detection of synaptic dysfunction, are essential for understanding normal and abnormal brain development. To date, multiple synaptic density markers have been proposed and investigated in experimental models of brain disorders. The majority of the gold standard methodologies (e.g., electron microscopy or immunohistochemistry) visualize synapses or measure changes in pre- and postsynaptic proteins ex vivo. However, the invasive nature of these classic methodologies precludes their use in living organisms. The recent development of positron emission tomography (PET) tracers [such as (18F)UCB-H or (11C)UCB-J] that bind to a putative synaptic density marker, the synaptic vesicle 2A (SV2A) protein, is heralding a likely paradigm shift in detecting synaptic alterations in patients. Despite their limited specificity, novel, non-invasive magnetic resonance (MR)-based methods also show promise in inferring synaptic information by linking to glutamate neurotransmission. Although promising, all these methods entail various advantages and limitations that must be addressed before becoming part of routine clinical practice. In this review, we summarize and discuss current ex vivo and in vivo methods of quantifying synaptic density, including an evaluation of their reliability and experimental utility. We conclude with a critical assessment of challenges that need to be overcome before successfully employing synaptic density biomarkers as diagnostic and/or prognostic tools in the study of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Eugene Kim
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Marija M Petrinovic
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Diana Cash
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| |
Collapse
|
12
|
Dodiya HB, Lutz HL, Weigle IQ, Patel P, Michalkiewicz J, Roman-Santiago CJ, Zhang CM, Liang Y, Srinath A, Zhang X, Xia J, Olszewski M, Zhang X, Schipma MJ, Chang EB, Tanzi RE, Gilbert JA, Sisodia SS. Gut microbiota-driven brain Aβ amyloidosis in mice requires microglia. J Exp Med 2022; 219:e20200895. [PMID: 34854884 PMCID: PMC8647415 DOI: 10.1084/jem.20200895] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/16/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
We previously demonstrated that lifelong antibiotic (ABX) perturbations of the gut microbiome in male APPPS1-21 mice lead to reductions in amyloid β (Aβ) plaque pathology and altered phenotypes of plaque-associated microglia. Here, we show that a short, 7-d treatment of preweaned male mice with high-dose ABX is associated with reductions of Aβ amyloidosis, plaque-localized microglia morphologies, and Aβ-associated degenerative changes at 9 wk of age in male mice only. More importantly, fecal microbiota transplantation (FMT) from transgenic (Tg) or WT male donors into ABX-treated male mice completely restored Aβ amyloidosis, plaque-localized microglia morphologies, and Aβ-associated degenerative changes. Transcriptomic studies revealed significant differences between vehicle versus ABX-treated male mice and FMT from Tg mice into ABX-treated mice largely restored the transcriptome profiles to that of the Tg donor animals. Finally, colony-stimulating factor 1 receptor (CSF1R) inhibitor-mediated depletion of microglia in ABX-treated male mice failed to reduce cerebral Aβ amyloidosis. Thus, microglia play a critical role in driving gut microbiome-mediated alterations of cerebral Aβ deposition.
Collapse
Affiliation(s)
- Hemraj B. Dodiya
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Holly L. Lutz
- Department of Pediatrics and Scripps Institution of Oceanography, University of California, San Diego, San Diego, CA
| | - Ian Q. Weigle
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Priyam Patel
- Center for Genetic Medicine, Northwestern University, Chicago, IL
| | | | | | | | - Yingxia Liang
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Abhinav Srinath
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Xulun Zhang
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Jessica Xia
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Monica Olszewski
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Xiaoqiong Zhang
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | | | - Eugene B. Chang
- Department of Digestive Diseases, The University of Chicago, Chicago, IL
| | | | - Jack A. Gilbert
- Department of Pediatrics and Scripps Institution of Oceanography, University of California, San Diego, San Diego, CA
| | | |
Collapse
|
13
|
Corrigan RR, Piontkivska H, Casadesus G. Amylin Pharmacology in Alzheimer's Disease Pathogenesis and Treatment. Curr Neuropharmacol 2022; 20:1894-1907. [PMID: 34852745 PMCID: PMC9886804 DOI: 10.2174/1570159x19666211201093147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022] Open
Abstract
The metabolic peptide hormone amylin, in concert with other metabolic peptides like insulin and leptin, has an important role in metabolic homeostasis and has been intimately linked to Alzheimer's disease (AD). Interestingly, this pancreatic amyloid peptide is known to self-aggregate much like amyloid-beta and has been reported to be a source of pathogenesis in both Type II diabetes mellitus (T2DM) and Alzheimer's disease. The traditional "gain of toxic function" properties assigned to amyloid proteins are, however, contrasted by several reports highlighting neuroprotective effects of amylin and a recombinant analog, pramlintide, in the context of these two diseases. This suggests that pharmacological therapies aimed at modulating the amylin receptor may be therapeutically beneficial for AD development, as they already are for T2DMM. However, the nature of amylin receptor signaling is highly complex and not well studied in the context of CNS function. Therefore, to begin to address this pharmacological paradox in amylin research, the goal of this review is to summarize the current research on amylin signaling and CNS functions and critically address the paradoxical nature of this hormone's signaling in the context of AD pathogenesis.
Collapse
Affiliation(s)
| | | | - Gemma Casadesus
- Address correspondence to this author at the Department of Pharmacology and Therapeutics, University of Florida, PO Box 100495. Gainesville, FL32610 USA; Tel: 352-294-5346; E-mail:
| |
Collapse
|
14
|
Clark KA, Shin AC, Sirivelu MP, MohanKumar RC, Maddineni SR, Ramachandran R, MohanKumar PS, MohanKumar SMJ. Evaluation of the Central Effects of Systemic Lentiviral-Mediated Leptin Delivery in Streptozotocin-Induced Diabetic Rats. Int J Mol Sci 2021; 22:ijms222413197. [PMID: 34947993 PMCID: PMC8703968 DOI: 10.3390/ijms222413197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Type 1 diabetes (T1D) is characterized by hyperphagia, hyperglycemia and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We have reported previously that daily leptin injections help to alleviate these symptoms. Therefore, we hypothesized that leptin gene therapy could help to normalize the neuroendocrine dysfunction seen in T1D. Adult male Sprague Dawley rats were injected i.v. with a lentiviral vector containing the leptin gene or green fluorescent protein. Ten days later, they were injected with the vehicle or streptozotocin (STZ). HPA function was assessed by measuring norepinephrine (NE) levels in the paraventricular nucleus (PVN) and serum corticosterone (CS). Treatment with the leptin lentiviral vector (Lepvv) increased leptin and insulin levels in non-diabetic rats, but not in diabetic animals. There was a significant reduction in blood glucose levels in diabetic rats due to Lepvv treatment. Both NE levels in the PVN and serum CS were reduced in diabetic rats treated with Lepvv. Results from this study provide evidence that leptin gene therapy in STZ-induced diabetic rats was able to partially normalize some of the neuroendocrine abnormalities, but studies with higher doses of the Lepvv are needed to develop this into a viable option for treating T1D.
Collapse
MESH Headings
- Animals
- Corticosterone/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Disease Models, Animal
- Genetic Therapy
- Genetic Vectors/administration & dosage
- Injections, Intravenous
- Lentivirus/genetics
- Leptin/genetics
- Male
- Norepinephrine/metabolism
- Paraventricular Hypothalamic Nucleus/metabolism
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Kimberly A. Clark
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
| | - Andrew C. Shin
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA;
| | - Madhu P. Sirivelu
- Pathobiology and Diagnostic Investigation, Michigan State University, E. Lansing, MI 48824, USA;
| | - Ramya C. MohanKumar
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
| | - Sreenivasa R. Maddineni
- Department of Poultry Science, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (S.R.M.); (R.R.)
| | - Ramesh Ramachandran
- Department of Poultry Science, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (S.R.M.); (R.R.)
| | - Puliyur S. MohanKumar
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
- Pathobiology and Diagnostic Investigation, Michigan State University, E. Lansing, MI 48824, USA;
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
- Department of Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Sheba M. J. MohanKumar
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
- Department of Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
- Correspondence: ; Tel.: +1-706-542-1945
| |
Collapse
|
15
|
Tournissac M, Leclerc M, Valentin-Escalera J, Vandal M, Bosoi CR, Planel E, Calon F. Metabolic determinants of Alzheimer's disease: A focus on thermoregulation. Ageing Res Rev 2021; 72:101462. [PMID: 34534683 DOI: 10.1016/j.arr.2021.101462] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a complex age-related neurodegenerative disease, associated with central and peripheral metabolic anomalies, such as impaired glucose utilization and insulin resistance. These observations led to a considerable interest not only in lifestyle-related interventions, but also in repurposing insulin and other anti-diabetic drugs to prevent or treat dementia. Body temperature is the oldest known metabolic readout and mechanisms underlying its maintenance fail in the elderly, when the incidence of AD rises. This raises the possibility that an age-associated thermoregulatory deficit contributes to energy failure underlying AD pathogenesis. Brown adipose tissue (BAT) plays a central role in thermogenesis and maintenance of body temperature. In recent years, the modulation of BAT activity has been increasingly demonstrated to regulate energy expenditure, insulin sensitivity and glucose utilization, which could also provide benefits for AD. Here, we review the evidence linking thermoregulation, BAT and insulin-related metabolic defects with AD, and we propose mechanisms through which correcting thermoregulatory impairments could slow the progression and delay the onset of AD.
Collapse
|
16
|
Yang Y, Chen W, Wang X, Ge W. Impact of mitochondrial aldehyde dehydrogenase 2 on cognitive impairment in the AD model mouse. Acta Biochim Biophys Sin (Shanghai) 2021; 53:837-847. [PMID: 33954430 DOI: 10.1093/abbs/gmab057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is one of the major life-threatening diseases for the elderly because neither pathogenesis nor effective treatment is available. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) has been shown to reduce the cell-damaging aldehydes in response to reactive oxygen species (ROS). However, whether it plays a role in AD remains elusive. In the present study, we found that ALDH2 overexpression significantly improved the cognitive function of the AD mouse. Behavioral analyses of ALDH2-overexpressing APP/PS1 AD mice showed that the learning and cognitive abilities were significantly higher in these mice than in the control group APP/PS1 mice. Further open-field behavior experiments showed the same results. At the cellular level, ALDH2 protects nerve cells. HT22 cells were challenged with Aβ to establish an AD cell model, in the presence or absence of the ALDH2 activator Alda-1 and ALDH2 inhibitor Daidzin. Incubation with 50 μM Aβ for 24 h significantly reduced HT22 cell survival and cell viability, the effects of which were attenuated by the ALDH2 activator Alda-1 (50 μM). Aβ challenge promoted apoptosis and upregulated caspase3 level but suppressed Bcl-2 level, and the upregulated caspase3 level was reversed by the ALDH-2 agonist Alda-1. Aβ-induced clonal ball abnormal was reversed by Alda-1. Aβ altered the mitochondria geometry evidenced by vacuolar degeneration and membrane rupture, whereas Alda-1 changed the Aβ-induced mitochondria geometry anomalies. Moreover, superoxide anion and toxic 4-hydroxy-nonanal (4-HNE) and ROS increased by Aβ challenge were reversed by Alda-1. Meanwhile, Aβ-induced ATP reduction was reversed by Alda-1. Taken together, ALDH2 overexpression significantly improves the cognitive function of the AD mice. Furthermore, our results suggested that ALDH2 protects against Aβ hippocampal neuronal toxicity possibly through alleviating toxic aldehydes and ROS, as well as increasing ATP production to preserve mitochondrial integrity and reduce neuronal apoptosis.
Collapse
Affiliation(s)
- Ying Yang
- Department of General Practice, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Wei Chen
- Department of General Practice, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Wei Ge
- Department of General Practice, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
17
|
Rojas M, Chávez-Castillo M, Pirela D, Parra H, Nava M, Chacín M, Angarita L, Añez R, Salazar J, Ortiz R, Durán Agüero S, Gravini-Donado M, Bermúdez V, Díaz-Camargo E. Metabolic Syndrome: Is It Time to Add the Central Nervous System? Nutrients 2021; 13:nu13072254. [PMID: 34208833 PMCID: PMC8308252 DOI: 10.3390/nu13072254] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/05/2021] [Accepted: 06/09/2021] [Indexed: 12/28/2022] Open
Abstract
Metabolic syndrome (MS) is a set of cardio-metabolic risk factors that includes central obesity, hyperglycemia, hypertension, and dyslipidemias. The syndrome affects 25% of adults worldwide. The definition of MS has evolved over the last 80 years, with various classification systems and criteria, whose limitations and benefits are currently the subject of some controversy. Likewise, hypotheses regarding the etiology of MS add more confusion from clinical and epidemiological points of view. The leading suggestion for the pathophysiology of MS is insulin resistance (IR). IR can affect multiple tissues and organs, from the classic “triumvirate” (myocyte, adipocyte, and hepatocyte) to possible effects on organs considered more recently, such as the central nervous system (CNS). Mild cognitive impairment (MCI) and Alzheimer’s disease (AD) may be clinical expressions of CNS involvement. However, the association between MCI and MS is not understood. The bidirectional relationship that seems to exist between these factors raises the questions of which phenomenon occurs first and whether MCI can be a precursor of MS. This review explores shared pathophysiological mechanisms between MCI and MS and establishes a hypothesis of a possible MCI role in the development of IR and the appearance of MS.
Collapse
Affiliation(s)
- Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | | | - Daniela Pirela
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 08002, Colombia;
| | - Lissé Angarita
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andrés Bello, Sede Concepción 4260000, Chile;
| | - Roberto Añez
- Departamento de Endocrinología y Nutrición, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain;
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.R.); (D.P.); (H.P.); (M.N.); (J.S.)
| | - Rina Ortiz
- Posgrado, Carrera de Medicina, Universidad Católica de Cuenca, Cantón de Cuenca 010101, Ecuador;
| | - Samuel Durán Agüero
- Facultad de Ciencias Para el Cuidado de la Salud, Universidad San Sebastián, Los Leones 8420524, Chile;
| | - Marbel Gravini-Donado
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Barranquilla 080002, Colombia;
| | - Valmore Bermúdez
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia;
| | - Edgar Díaz-Camargo
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540006, Colombia;
- Correspondence:
| |
Collapse
|
18
|
Tedeschi DV, da Cunha AF, Cominetti MR, Pedroso RV. Efficacy of Gene Therapy to Restore Cognition in Alzheimer's Disease: A Systematic Review. Curr Gene Ther 2021; 21:246-257. [PMID: 33494678 DOI: 10.2174/1566523221666210120091146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the main cause of dementia and it is a progressive neurogenerative disease characterized by the accumulation of neurofibrillary tangles and senile plaques. There is currently no cure; however, some treatments are available to slow down the progression of the disease, including gene therapy, which has been investigated to have great potential for the treatment of AD. OBJECTIVE The aim of this review was to identify the efficacy of gene therapy to restore cognition in AD. METHODS A systematic review was carried out using papers published up to May 2020 and available in the Web of Science, Scopus, and Medline/PUBMED databases. Articles were considered for inclusion if they were original researches that investigated the effects of gene therapy on cognition in AD. The methodological quality of the selected studies was evaluated using the Risk of Bias Tool for Animal Intervention Studies (SYRCLE's Rob tool) and the Jadad Scale. RESULTS Most preclinical studies obtained positive results in improving memory and learning in mice that underwent treatment with gene therapy. On the other hand, clinical studies have obtained inconclusive results related to the delivery methods of the viral vector used in gene therapy. CONCLUSION Gene therapy has shown a great potential for the treatment of AD in preclinical trials, but results should be interpreted with caution since preclinical studies presented limitations to predict the efficacy of the treatment outcome in humans.
Collapse
Affiliation(s)
- Desyrre V Tedeschi
- Federal University of Sao Carlos, Rodovia Washington Luis Km 310 - 13565-905, Sao Carlos SP, Brazil
| | - Anderson F da Cunha
- Department of Genetics and Evolution, Federal University of Sao Carlos, Rodovia Washington Luis Km 310 - 13565- 905, Sao Carlos SP, Brazil
| | - Márcia R Cominetti
- Department of Gerontology, Federal University of Sao Carlos, Rodovia Washington Luis Km 310 - 13565-905, Sao Carlos SP, Brazil
| | - Renata Valle Pedroso
- Department of Gerontology, Federal University of Sao Carlos, Rodovia Washington Luis Km 310 - 13565-905, Sao Carlos SP, Brazil
| |
Collapse
|
19
|
Hamilton K, Harvey J. The Neuronal Actions of Leptin and the Implications for Treating Alzheimer's Disease. Pharmaceuticals (Basel) 2021; 14:ph14010052. [PMID: 33440796 PMCID: PMC7827292 DOI: 10.3390/ph14010052] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
It is widely accepted that the endocrine hormone leptin controls food intake and energy homeostasis via activation of leptin receptors expressed on hypothalamic arcuate neurons. The hippocampal formation also displays raised levels of leptin receptor expression and accumulating evidence indicates that leptin has a significant impact on hippocampal synaptic function. Thus, cellular and behavioural studies support a cognitive enhancing role for leptin as excitatory synaptic transmission, synaptic plasticity and glutamate receptor trafficking at hippocampal Schaffer collateral (SC)-CA1 synapses are regulated by leptin, and treatment with leptin enhances performance in hippocampus-dependent memory tasks. Recent studies indicate that hippocampal temporoammonic (TA)-CA1 synapses are also a key target for leptin. The ability of leptin to regulate TA-CA1 synapses has important functional consequences as TA-CA1 synapses are implicated in spatial and episodic memory processes. Moreover, degeneration is initiated in the TA pathway at very early stages of Alzheimer's disease, and recent clinical evidence has revealed links between plasma leptin levels and the incidence of Alzheimer's disease (AD). Additionally, accumulating evidence indicates that leptin has neuroprotective actions in various AD models, whereas dysfunctions in the leptin system accelerate AD pathogenesis. Here, we review the data implicating the leptin system as a potential novel target for AD, and the evidence that boosting the hippocampal actions of leptin may be beneficial.
Collapse
|
20
|
Leptin enhances adult neurogenesis and reduces pathological features in a transgenic mouse model of Alzheimer's disease. Neurobiol Dis 2020; 148:105219. [PMID: 33301880 DOI: 10.1016/j.nbd.2020.105219] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 01/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common dementia worldwide and is characterized by the presence of senile plaques by amyloid-beta (Aβ) and neurofibrillary tangles of hyperphosphorylated Tau protein. These changes lead to progressive neuronal degeneration and dysfunction, resulting in severe brain atrophy and cognitive deficits. With the discovery that neurogenesis persists in the adult mammalian brain, including brain regions affected by AD, studies of the use of neural stem cells (NSCs) for the treatment of neurodegenerative diseases to repair or prevent neuronal cell loss have increased. Here we demonstrate that leptin administration increases the neurogenic process in the dentate gyrus of the hippocampus as well as in the subventricular zone of lateral ventricles of adult and aged mice. Chronic treatment with leptin increased NSCs proliferation with significant effects on proliferation and differentiation of newborn cells. The expression of the long form of the leptin receptor, LepRb, was detected in the neurogenic niches by reverse qPCR and immunohistochemistry. Moreover, leptin modulated astrogliosis, microglial cell number and the formation of senile plaques. Additionally, leptin led to attenuation of Aβ-induced neurodegeneration and superoxide anion production as revealed by Fluoro-Jade B and dihydroethidium staining. Our study contributes to the understanding of the effects of leptin in the brain that may lead to the development of new therapies to treat Alzheimer's disease.
Collapse
|
21
|
Salazar AM, Leisgang AM, Ortiz AA, Murtishaw AS, Kinney JW. Alterations of GABA B receptors in the APP/PS1 mouse model of Alzheimer's disease. Neurobiol Aging 2020; 97:129-143. [PMID: 33232936 DOI: 10.1016/j.neurobiolaging.2020.10.013] [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: 01/15/2020] [Revised: 09/08/2020] [Accepted: 10/16/2020] [Indexed: 01/20/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the progressive decline of memory and cognitive function. The disease is characterized by the presence of amyloid plaques, tau tangles, altered inflammatory signaling, and alterations in numerous neurotransmitter signaling systems, including γ-aminobutyric acid (GABA). Given the extensive role of GABA in regulating neuronal activity, a careful investigation of GABA-related changes is needed. Further, given persistent inflammation has been demonstrated to drive AD pathology, the presence of GABA B receptor expressed on glia that serve a role regulation of the immune response adds to potential implications of altered GABA in AD. There has not previously been a systematic evaluation of GABA-related changes in an amyloid model of AD that specifically focuses on examining changes in GABA B receptors. In the present study, we examined alterations in several GABA-specific targets in the APP/PS1 mouse model at different ages. In the 4-month-old cohort, no significant deficits in spatial learning and memory or alterations in any of the GABAergic targets were observed compared with wild-type controls. However, we identified significant alterations in several GABA-related targets in the 6-month-old cohort that exhibited spatial learning deficits that include changes in glutamic acid decarboxylase 65, GABA transporter type 3, and GABA B receptors protein and mRNA levels. This was the same cohort at which learning and memory deficits and significant amyloid pathology was observed. Overall, our study provides evidence of altered GABAergic signaling in an amyloid model of AD at a time point consistent with AD-related deficits.
Collapse
Affiliation(s)
- Arnold M Salazar
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Amanda M Leisgang
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Andrew A Ortiz
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Andrew S Murtishaw
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Jefferson W Kinney
- Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA.
| |
Collapse
|
22
|
Robison LS, Gannon OJ, Thomas MA, Salinero AE, Abi-Ghanem C, Poitelon Y, Belin S, Zuloaga KL. Role of sex and high-fat diet in metabolic and hypothalamic disturbances in the 3xTg-AD mouse model of Alzheimer's disease. J Neuroinflammation 2020; 17:285. [PMID: 32993686 PMCID: PMC7526387 DOI: 10.1186/s12974-020-01956-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Background Hypothalamic dysfunction occurs early in the clinical course of Alzheimer’s disease (AD), likely contributing to disturbances in feeding behavior and metabolic function that are often observed years prior to the onset of cognitive symptoms. Late-life weight loss and low BMI are associated with increased risk of dementia and faster progression of disease. However, high-fat diet and metabolic disease (e.g., obesity, type 2 diabetes), particularly in mid-life, are associated with increased risk of AD, as well as exacerbated AD pathology and behavioral deficits in animal models. In the current study, we explored possible relationships between hypothalamic function, diet/metabolic status, and AD. Considering the sex bias in AD, with women representing two-thirds of AD patients, we sought to determine whether these relationships vary by sex. Methods WT and 3xTg-AD male and female mice were fed a control (10% fat) or high-fat (HF 60% fat) diet from ~ 3–7 months of age, then tested for metabolic and hypothalamic disturbances. Results On control diet, male 3xTg-AD mice displayed decreased body weight, reduced fat mass, hypoleptinemia, and mild systemic inflammation, as well as increased expression of gliosis- and inflammation-related genes in the hypothalamus (Iba1, GFAP, TNF-α, IL-1β). In contrast, female 3xTg-AD mice on control diet displayed metabolic disturbances opposite that of 3xTg-AD males (increased body and fat mass, impaired glucose tolerance). HF diet resulted in expected metabolic alterations across groups (increased body and fat mass; glucose intolerance; increased plasma insulin and leptin, decreased ghrelin; nonalcoholic fatty liver disease-related pathology). HF diet resulted in the greatest weight gain, adiposity, and glucose intolerance in 3xTg-AD females, which were associated with markedly increased hypothalamic expression of GFAP and IL-1β, as well as GFAP labeling in several hypothalamic nuclei that regulate energy balance. In contrast, HF diet increased diabetes markers and systemic inflammation preferentially in AD males but did not exacerbate hypothalamic inflammation in this group. Conclusions These findings provide further evidence for the roles of hypothalamic and metabolic dysfunction in AD, which in the 3xTg-AD mouse model appears to be dependent on both sex and diet.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Melissa A Thomas
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Yannick Poitelon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Sophie Belin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
23
|
Mejido DC, Peny JA, Vieira MN, Ferreira ST, De Felice FG. Insulin and leptin as potential cognitive enhancers in metabolic disorders and Alzheimer's disease. Neuropharmacology 2020; 171:108115. [PMID: 32344008 DOI: 10.1016/j.neuropharm.2020.108115] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 03/30/2020] [Accepted: 04/20/2020] [Indexed: 02/08/2023]
|
24
|
Guo Q, Xu S, Yang P, Wang P, Lu S, Sheng D, Qian K, Cao J, Lu W, Zhang Q. A dual-ligand fusion peptide improves the brain-neuron targeting of nanocarriers in Alzheimer's disease mice. J Control Release 2020; 320:347-362. [DOI: 10.1016/j.jconrel.2020.01.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
|
25
|
Hirschstein Z, Novakovic ZM, Grasso P. MA-[D-Leu-4]-OB3, a Small Molecule Synthetic Peptide Leptin Mimetic, Normalizes Glucose Tolerance and Episodic Memory in a Mouse Model of Type 2 Diabetes Mellitus and Alzheimer’s Disease-Like Cognitive Impairment. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09995-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
26
|
Anti-ageing gene therapy: Not so far away? Ageing Res Rev 2019; 56:100977. [PMID: 31669577 DOI: 10.1016/j.arr.2019.100977] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/31/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022]
Abstract
Improving healthspan is the main objective of anti-ageing research. Currently, innovative gene therapy-based approaches seem to be among the most promising for preventing and treating chronic polygenic pathologies, including age-related ones. The gene-based therapy allows to modulate the genome architecture using both direct (e.g., by gene editing) and indirect (e.g., by viral or non-viral vectors) approaches. Nevertheless, considering the extraordinary complexity of processes involved in ageing and ageing-related diseases, the effectiveness of these therapeutic options is often unsatisfactory and limited by their side-effects. Thus, clinical implementation of such applications is certainly a long-time process that will require many translation phases for addressing challenges. However, after overcoming these issues, their implementation in clinical practice may obviously provide new possibilities in anti-ageing medicine. Here, we review and discuss recent advances in this rapidly developing research field.
Collapse
|
27
|
Potential exerkines for physical exercise-elicited pro-cognitive effects: Insight from clinical and animal research. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 147:361-395. [PMID: 31607361 DOI: 10.1016/bs.irn.2019.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A sedentary lifestyle is now known as a critical risk factor for accelerated aging-related neurodegenerative disorders. In contract, having regular physical exercise has opposite effects. Clinical findings have suggested that physical exercise can promote brain plasticity, particularly the hippocampus and the prefrontal cortex, that are important for learning and memory and mood regulations. However, the underlying mechanisms are still unclear. Animal studies reveal that the effects of physical exercise on promoting neuroplasticity could be mediated by different exerkines derived from the peripheral system and the brain itself. This book chapter summarizes the recent evidence from clinical and pre-clinical studies showing the emerging mediators for exercise-promoted brain health, including myokines secreted from skeletal muscles, adipokines from adipose tissues, and other factors secreted from the bone and liver.
Collapse
|
28
|
Lloret A, Monllor P, Esteve D, Cervera-Ferri A, Lloret MA. Obesity as a Risk Factor for Alzheimer's Disease: Implication of Leptin and Glutamate. Front Neurosci 2019; 13:508. [PMID: 31191220 PMCID: PMC6540965 DOI: 10.3389/fnins.2019.00508] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/02/2019] [Indexed: 12/22/2022] Open
Abstract
Obesity is known to induce leptin and insulin resistance. Leptin is a peptide hormone synthesized in adipose tissue that mainly regulates food intake. It has been shown that insulin stimulates the production of leptin when adipocytes are exposed to glucose to encourage satiety; while leptin, via a negative feedback, decreases the insulin release and enhances tissue sensitivity to it, leading to glucose uptake for energy utilization or storage. Therefore, resistance to insulin is closely related to leptin resistance. Obesity in middle age has also been related to Alzheimer's disease (AD). In recent years, the relation between impaired leptin signaling pathway and the onset of AD has been studied. In all this context the role of the blood brain barrier (BBB) is crucial. Slow excitotoxicity happens in AD due to an excess of the neurotransmitter glutamate. Since leptin has been shown to regulate N-methyl-D-aspartate (NMDA) receptors, we want to review the link between these pathological pathways, and how they are affected by other AD triggering factors and its role in the onset of AD.
Collapse
Affiliation(s)
- Ana Lloret
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain
| | - Paloma Monllor
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain
| | - Daniel Esteve
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain
| | - Ana Cervera-Ferri
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Maria-Angeles Lloret
- Department of Clinic Neurophysiology, University Clinic Hospital of Valencia, Valencia, Spain
| |
Collapse
|
29
|
Leptin in hippocampus mediates benefits of mild exercise by an antioxidant on neurogenesis and memory. Proc Natl Acad Sci U S A 2019; 116:10988-10993. [PMID: 31085646 DOI: 10.1073/pnas.1815197116] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Regular exercise and dietary supplements with antioxidants each have the potential to improve cognitive function and attenuate cognitive decline, and, in some cases, they enhance each other. Our current results reveal that low-intensity exercise (mild exercise, ME) and the natural antioxidant carotenoid astaxanthin (AX) each have equivalent beneficial effects on hippocampal neurogenesis and memory function. We found that the enhancement by ME combined with AX in potentiating hippocampus-based plasticity and cognition is mediated by leptin (LEP) made and acting in the hippocampus. In assessing the combined effects upon wild-type (WT) mice undergoing ME with or without an AX diet for four weeks, we found that, when administrated alone, ME and AX separately enhanced neurogenesis and spatial memory, and when combined they were at least additive in their effects. DNA microarray and bioinformatics analyses revealed not only the up-regulation of an antioxidant gene, ABHD3, but also that the up-regulation of LEP gene expression in the hippocampus of WT mice with ME alone is further enhanced by AX. Together, they also increased hippocampal LEP (h-LEP) protein levels and enhanced spatial memory mediated through AKT/STAT3 signaling. AX treatment also has direct action on human neuroblastoma cell lines to increase cell viability associated with increased LEP expression. In LEP-deficient mice (ob/ob), chronic infusion of LEP into the lateral ventricles restored the synergy. Collectively, our findings suggest that not only h-LEP but also exogenous LEP mediates effects of ME on neural functions underlying memory, which is further enhanced by the antioxidant AX.
Collapse
|
30
|
Zou X, Zhong L, Zhu C, Zhao H, Zhao F, Cui R, Gao S, Li B. Role of Leptin in Mood Disorder and Neurodegenerative Disease. Front Neurosci 2019; 13:378. [PMID: 31130833 PMCID: PMC6510114 DOI: 10.3389/fnins.2019.00378] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
The critical regulatory role of leptin in the neuroendocrine system has been widely reported. Significantly, leptin can improve learning and memory, affect hippocampal synaptic plasticity, exert neuroprotective efficacy and reduce the risk of several neuropsychiatric diseases. In terms of depression, leptin could modulate the levels of neurotransmitters, neurotrophic factors and reverse the dysfunction in the hypothalamic-pituitary-adrenal axis (HPA). At the same time, leptin affects neurological diseases during the regulation of metabolic homeostasis. With regards to neurodegenerative diseases, leptin can affect them via neuroprotection, mainly including Alzheimer's disease and Parkinson's disease. This review will summarize the mechanisms of leptin signaling within the neuroendocrine system with respect to these diseases and discuss the therapeutic potential of leptin.
Collapse
Affiliation(s)
- Xiaohan Zou
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Lili Zhong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Cuilin Zhu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Haisheng Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Shuohui Gao
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Guo Q, Zheng X, Yang P, Pang X, Qian K, Wang P, Xu S, Sheng D, Wang L, Cao J, Lu W, Zhang Q, Jiang X. Small interfering RNA delivery to the neurons near the amyloid plaques for improved treatment of Alzheimer׳s disease. Acta Pharm Sin B 2019; 9:590-603. [PMID: 31193846 PMCID: PMC6543096 DOI: 10.1016/j.apsb.2018.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 09/29/2018] [Accepted: 11/15/2018] [Indexed: 02/02/2023] Open
Abstract
Gene therapy represents a promising treatment for the Alzheimer׳s disease (AD). However, gene delivery specific to brain lesions through systemic administration remains big challenge. In our previous work, we have developed an siRNA nanocomplex able to be specifically delivered to the amyloid plaques through surface modification with both CGN peptide for the blood–brain barrier (BBB) penetration and QSH peptide for β-amyloid binding. But, whether the as-designed nanocomplex could indeed improve the gene accumulation in the impaired neuron cells and ameliorate AD-associated symptoms remains further study. Herein, we prepared the nanocomplexes with an siRNA against β-site amyloid precursor protein-cleaving enzyme 1 (BACE1), the rate-limiting enzyme of Aβ production, as the therapeutic siRNA of AD. The nanocomplexes exhibited high distribution in the Aβ deposits-enriched hippocampus, especially in the neurons near the amyloid plaques after intravenous administration. In APP/PS1 transgenic mice, the nanocomplexes down-regulated BACE1 in both mRNA and protein levels, as well as Aβ and amyloid plaques to the level of wild-type mice. Moreover, the nanocomplexes significantly increased the level of synaptophysin and rescued memory loss of the AD transgenic mice without hematological or histological toxicity. Taken together, this work presented direct evidences that the design of precise gene delivery to the AD lesions markedly improves the therapeutic outcome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Wei Lu
- Corresponding authors. Tel.: +86 21 519980068; fax: +86 21 51980067.
| | - Qizhi Zhang
- Corresponding authors. Tel.: +86 21 519980068; fax: +86 21 51980067.
| | | |
Collapse
|
32
|
Zhou F, Chen S. Effects of Gender and Other Confounding Factors on Leptin Concentrations in Alzheimer's Disease: Evidence from the Combined Analysis of 27 Case-Control Studies. J Alzheimers Dis 2019; 62:477-486. [PMID: 29439354 DOI: 10.3233/jad-170983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Leptin, as a link between fat mass and the brain, has been reported to be associated with gender. The gender differences in leptin levels between Alzheimer's disease (AD) and healthy elderly controls are inconclusive so far. To quantitatively summarize the leptin data available from female and male patients with AD, we searched PubMed and EMBASE for articles published from inception to July 20, 2017. Data were extracted from 27 studies, consisting of 3,014 participants. The pooled results showed that the overall leptin levels were lower in AD (Hedges' g = -0.481; p = 0.002) than in controls, and the leptin levels in whole blood and serum were decreased with moderate and large effect sizes (g = -0.677, -0.839; respectively; both of p-values <0.001) in AD compared with controls. In blood, there were significantly lower concentrations of leptin in female AD than in female controls (g = -0.590; p = 0.014), but not in male case-control group (g = -0.666; p = 0.067). Meta-regression analysis demonstrated that the decreased extent of leptin levels in AD paralleled the degree of the severity of dementia symptoms, as well as the alterations of body mass index (p-values ≤0.002). The findings provide strong evidence that 1) the blood concentrations of leptin are lower in female AD patients than in female controls; and 2) the greater the severity of dementia symptoms, the greater the decreases in the blood leptin levels. But more future investigations on the blood leptin levels in male AD patients is warranted.
Collapse
Affiliation(s)
- Futao Zhou
- College of Medicine and Health, Lishui University, Lishui Zhejiang, China
| | - Shuangrong Chen
- College of Engineering, Lishui University, Lishui Zhejiang, China
| |
Collapse
|
33
|
Forny-Germano L, De Felice FG, Vieira MNDN. The Role of Leptin and Adiponectin in Obesity-Associated Cognitive Decline and Alzheimer's Disease. Front Neurosci 2019; 12:1027. [PMID: 30692905 PMCID: PMC6340072 DOI: 10.3389/fnins.2018.01027] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022] Open
Abstract
Cross-talk between adipose tissue and central nervous system (CNS) underlies the increased risk of obese people to develop brain diseases such as cognitive and mood disorders. Detailed mechanisms for how peripheral changes caused by adipose tissue accumulation in obesity impact the CNS to cause brain dysfunction are poorly understood. Adipokines are a large group of substances secreted by the white adipose tissue to regulate a wide range of homeostatic processes including, but not limited to, energy metabolism and immunity. Obesity is characterized by a generalized change in the levels of circulating adipokines due to abnormal accumulation and dysfunction of adipose tissue. Altered adipokine levels underlie complications of obesity as well as the increased risk for the development of obesity-related comorbidities such as type 2 diabetes, cardiovascular and neurodegenerative diseases. Here, we review the literature for the role of adipokines as key mediators of the communication between periphery and CNS in health and disease. We will focus on the actions of leptin and adiponectin, two of the most abundant and well studied adipokines, in the brain, with particular emphasis on how altered signaling of these adipokines in obesity may lead to cognitive dysfunction and augmented risk for Alzheimer's disease. A better understanding of adipokine biology in brain disorders may prove of major relevance to diagnostic, prevention and therapy.
Collapse
Affiliation(s)
- Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda G. De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Centre for Neuroscience Studies, Department of Psychiatry, Queen’s University, Kingston, ON, Canada
| | | |
Collapse
|
34
|
Hiller AJ, Ishii M. Disorders of Body Weight, Sleep and Circadian Rhythm as Manifestations of Hypothalamic Dysfunction in Alzheimer's Disease. Front Cell Neurosci 2018; 12:471. [PMID: 30568576 PMCID: PMC6289975 DOI: 10.3389/fncel.2018.00471] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022] Open
Abstract
While cognitive decline and memory loss are the major clinical manifestations of Alzheimer’s disease (AD), they are now recognized as late features of the disease. Recent failures in clinical drug trials highlight the importance of evaluating and treating patients with AD as early as possible and the difficulties in developing effective therapies once the disease progresses. Since the pathological hallmarks of AD including the abnormal aggregation of amyloid-beta (Aβ) and tau can occur decades before any significant cognitive decline in the preclinical stage of AD, it is important to identify the earliest clinical manifestations of AD and elucidate their underlying cellular and molecular mechanisms. Importantly, metabolic and non-cognitive manifestations of AD such as weight loss and alterations of peripheral metabolic signals can occur before the onset of cognitive symptoms and worsen with disease progression. Accumulating evidence suggests that the major culprit behind these early metabolic and non-cognitive manifestations of AD is AD pathology causing dysfunction of the hypothalamus, a brain region critical for integrating peripheral signals with essential homeostatic physiological functions. Here, we aim to highlight recent developments that address the role of AD pathology in the development of hypothalamic dysfunction associated with metabolic and non-cognitive manifestations seen in AD. Understanding the mechanisms underlying hypothalamic dysfunction in AD could give key new insights into the development of novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Abigail J Hiller
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Makoto Ishii
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States.,Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| |
Collapse
|
35
|
McGregor G, Harvey J. Regulation of Hippocampal Synaptic Function by the Metabolic Hormone, Leptin: Implications for Health and Neurodegenerative Disease. Front Cell Neurosci 2018; 12:340. [PMID: 30386207 PMCID: PMC6198461 DOI: 10.3389/fncel.2018.00340] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/13/2018] [Indexed: 11/13/2022] Open
Abstract
The role of the endocrine hormone leptin in controlling energy homeostasis in the hypothalamus are well documented. However the CNS targets for leptin are not restricted to the hypothalamus as a high density of leptin receptors are also expressed in several parts of the brain involved in higher cognitive functions including the hippocampus. Numerous studies have identified that in the hippocampus, leptin has cognitive enhancing actions as exogenous application of this hormone facilitates hippocampal-dependent learning and memory, whereas lack or insensitivity to leptin results in significant memory deficits. Leptin also markedly influences some of the main cellular changes that are involved in learning and memory including NMDA-receptor dependent synaptic plasticity and glutamate receptor trafficking. Like other metabolic hormones, there is a significant decline in neuronal sensitivity to leptin during the ageing process. Indeed, the capacity of leptin to modulate the functioning of hippocampal synapses is substantially reduced in aged compared to adult tissue. Clinical studies have also identified an association between circulating leptin levels and the risk of certain neurodegenerative disorders such as Alzheimer’s disease (AD). In view of this, targeting leptin and/or its receptor/signaling mechanisms may be an innovative approach for developing therapies to treat AD. In support of this, accumulating evidence indicates that leptin has cognitive enhancing and neuroprotective actions in various models of AD. Here we assess recent evidence that supports an important regulatory role for leptin at hippocampal CA1 synapses, and we discuss how age-related alterations in this hormonal system influences neurodegenerative disease.
Collapse
Affiliation(s)
- Gemma McGregor
- Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Jenni Harvey
- Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
36
|
Sasmita AO. Current viral-mediated gene transfer research for treatment of Alzheimer’s disease. Biotechnol Genet Eng Rev 2018; 35:26-45. [DOI: 10.1080/02648725.2018.1523521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Andrew Octavian Sasmita
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Mora-Muñoz L, Guerrero-Naranjo A, Rodríguez-Jimenez EA, Mastronardi CA, Velez-van-Meerbeke A. Leptin: role over central nervous system in epilepsy. BMC Neurosci 2018; 19:51. [PMID: 30185147 PMCID: PMC6126011 DOI: 10.1186/s12868-018-0453-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/29/2018] [Indexed: 02/05/2023] Open
Abstract
Adipose tissue is a dynamic organ with different effects on the body. Many of these effects are mediated by leptin, a hormone strongly involved in regulation of feeding and energy metabolism. It has an important role as a mediator of neuronal excitatory activity and higher brain functions. The aim of this study was to review the association between leptin and cerebral neuronal function, in particular its anticonvulsant or convulsant effects and the possible therapeutic role for treating epilepsy. For this purpose, the databases Pubmed, Science Direct, Elsevier, ResearchGate and Scielo were searched to identify experimental studies, reviews and systematic review articles, published in English, Spanish or Portuguese. Experimental studies and the presence of leptin receptors in nervous system sites other than the hypothalamus suggest an influence on higher brain functions. Indeed several animal studies have demonstrated a role of these channels in epileptiform activity as both anticonvulsive and convulsive effects have been found. The reason for these discrepancies is unclear but provides clear evidence of a potential role of leptin and leptin therapy in epileptiform activity. The association between leptin and brain function demonstrates the importance of peripheral metabolic hormones on central nervous system and opens a new way for the development of novel therapeutic interventions in diseases like epilepsy. Nevertheless further investigations are important to clarify the dynamics and diverse actions of leptin on excitatory regulation in the brain.
Collapse
Affiliation(s)
- Laura Mora-Muñoz
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Cra 24 No 63C-69, Bogotá, Colombia
| | | | | | | | - Alberto Velez-van-Meerbeke
- Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Cra 24 No 63C-69, Bogotá, Colombia.
| |
Collapse
|
38
|
King A, Brain A, Hanson K, Dittmann J, Vickers J, Fernandez-Martos C. Disruption of leptin signalling in a mouse model of Alzheimer's disease. Metab Brain Dis 2018; 33:1097-1110. [PMID: 29546689 DOI: 10.1007/s11011-018-0203-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/15/2018] [Indexed: 12/12/2022]
Abstract
Disruption of leptin signalling has been implicated as playing a role in the development of Alzheimer's disease (AD). Leptin has previously been shown to be affected by amyloid-beta (Aβ)-related signalling; however, pathways that link leptin to the disease pathogenesis have not been determined. To characterize the association between increasing age-dependent Aβ levels with leptin signalling and the vulnerable brain regions in AD, we assessed the mRNA and protein expression profile of leptin and leptin receptor (Ob-Rb) at 9 and 18-month-age in APP/PS1 mice. Immunohistochemical labelling demonstrated that leptin and Ob-Rb proteins were localised to neocortical and hippocampal neurons in APP/PS1 and wildtype (WT) mice. Neuronal leptin and Ob-Rb immunolabelling was more prominent in the neocortex of both groups at 9 month of age, while, at 18 months, labelling was reduced in the hippocampus of APP/PS1 mice relative to WT. Immunoblotting analysis demonstrated decreased hippocampal leptin levels, concomitantly with an increased Ob-Rb levels, in APP/PS1 mice compared with WT controls at 18 month of age. While no leptin mRNA was found in either of the groups analysed, Ob-Rb mRNA was significantly decreased in the hippocampus of APP/PS1 mice at both ages analysed. In addition, a significant decreased protein kinase B (Akt) activity concomitantly with an upregulation of suppressor of cytokine signaling-3 (SOCS3) and protein-tyrosine phosphatase 1B (PTP1B) transcripts was present. Thus, these results collectively indicate alterations of leptin signalling in the hippocampus of APP/PS1 mice, providing novel insights about the pathways that could link aberrant leptin signaling to the pathological changes of AD.
Collapse
Affiliation(s)
- Anna King
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Private Bag 143, Hobart, TAS, 7000, Australia
| | - Anna Brain
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Private Bag 143, Hobart, TAS, 7000, Australia
| | - Kelsey Hanson
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Private Bag 143, Hobart, TAS, 7000, Australia
| | - Justin Dittmann
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Private Bag 143, Hobart, TAS, 7000, Australia
| | - James Vickers
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Private Bag 143, Hobart, TAS, 7000, Australia
| | - Carmen Fernandez-Martos
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Private Bag 143, Hobart, TAS, 7000, Australia.
| |
Collapse
|
39
|
Chen L, Huang Z, Du Y, Fu M, Han H, Wang Y, Dong Z. Capsaicin Attenuates Amyloid-β-Induced Synapse Loss and Cognitive Impairments in Mice. J Alzheimers Dis 2018; 59:683-694. [PMID: 28671132 DOI: 10.3233/jad-170337] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of progressive cognitive impairment in the aged. The aggregation of the amyloid β-protein (Aβ) is a hallmark of AD and is linked to synapse loss and cognitive impairment. Capsaicin, a specific agonist of the transient receptor potential vanilloid 1 (TRPV1), has been proven to ameliorate stress-induced AD-like pathological and cognitive impairments, but it is unclear whether TRPV1 activation can affect cognitive and synaptic functions in Aβ-induced mouse model of AD. In this study, we investigated the effects of TRPV1 activation on spatial memory and synaptic plasticity in mice treated with Aβ. To induce AD-like pathological and cognitive impairments, adult C57Bl/6 mice were microinjected with Aβ42 (100 μM, 2.5 μl/mouse, i.c.v.). Two weeks after Aβ42 microinjection, spatial learning and memory as well as hippocampal long-term potentiation (LTP) were examined. The results showed that Aβ42 microinjection significantly impaired spatial learning and memory in the Morris water maze and novel object recognition tests compared with controls. These behavioral changes were accompanied by synapse loss and impaired LTP in the CA1 area of hippocampus. More importantly, daily capsaicin (1 mg/kg, i.p.) treatment throughout the experiment dramatically improved spatial learning and memory and synaptic function, as reflected by enhanced hippocampal LTP and reduced synapse loss, whereas the TRPV1 antagonist capsazepine (1 mg/kg, i.p.) treatment had no effects on cognitive and synaptic function in Aβ42-treated mice. These results indicate that TRPV1 activation by capsaicin rescues cognitive deficit in the Aβ42-induced mouse model of AD both structurely and functionally.
Collapse
Affiliation(s)
- Long Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Zhilin Huang
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yehong Du
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Min Fu
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Huili Han
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yutian Wang
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
40
|
Down Syndrome, Obesity, Alzheimer's Disease, and Cancer: A Brief Review and Hypothesis. Brain Sci 2018; 8:brainsci8040053. [PMID: 29587359 PMCID: PMC5924389 DOI: 10.3390/brainsci8040053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/15/2018] [Accepted: 03/22/2018] [Indexed: 12/19/2022] Open
Abstract
Down syndrome (trisomy 21), a complex mix of physical, mental, and biochemical issues, includes an increased risk of Alzheimer’s disease and childhood leukemia, a decreased risk of other tumors, and a high frequency of overweight/obesity. Certain features related to the third copy of chromosome 21 (which carries the APP gene and several anti-angiogenesis genes) create an environment favorable for Alzheimer’s disease and unfavorable for cancer. This environment may be enhanced by two bioactive compounds from fat cells, leptin, and adiponectin. This paper outlines these fat-related disease mechanisms and suggests new avenues of research to reduce disease risk in Down syndrome.
Collapse
|
41
|
McGregor G, Harvey J. Food for thought: Leptin regulation of hippocampal function and its role in Alzheimer's disease. Neuropharmacology 2017; 136:298-306. [PMID: 28987937 DOI: 10.1016/j.neuropharm.2017.09.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/27/2017] [Accepted: 09/30/2017] [Indexed: 01/08/2023]
Abstract
Accumulating evidence indicates that diet and body weight are important factors associated with Alzheimer's disease (AD), with a significant increase in AD risk linked to mid-life obesity, and weight loss frequently occurring in the early stages of AD. This has fuelled interest in the hormone leptin, as it is an important hypothalamic regulator of food intake and body weight, but leptin also markedly influences the functioning of the hippocampus; a key brain region that degenerates in AD. Increasing evidence indicates that leptin has cognitive enhancing properties as it facilitates the cellular events that underlie hippocampal-dependent learning and memory. However, significant reductions in leptin's capacity to regulate hippocampal synaptic function occurs with age and dysfunctions in the leptin system are associated with an increased risk of AD. Moreover, leptin is a potential novel target in AD as leptin treatment has beneficial effects in various models of AD. Here we summarise recent advances in leptin neurobiology with particular focus on regulation of hippocampal synaptic function by leptin and the implications of this for neurodegenerative disorders like AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | - Jenni Harvey
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
42
|
Franx BAA, Arnoldussen IAC, Kiliaan AJ, Gustafson DR. Weight Loss in Patients with Dementia: Considering the Potential Impact of Pharmacotherapy. Drugs Aging 2017; 34:425-436. [PMID: 28478593 DOI: 10.1007/s40266-017-0462-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Unintentional body weight loss is common in patients with dementia and is linked to cognitive impairment and poorer disease outcomes. It is proposed that some dementia medications with market approval, while aiming to improve cognitive and functional outcomes of a patient with dementia, are associated with reported body weight or body mass index loss. This review presents evidence in the published literature on body weight loss in dementia, describes selected theories behind body weight loss, evaluates the potential impact of approved dementia pharmacotherapies on body weight, considers the potential role for medical foods, understands the potential influence of treatments for neuropsychiatric symptoms and signs, and finally, summarizes this important area.
Collapse
Affiliation(s)
- Bart A A Franx
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ilse A C Arnoldussen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Amanda J Kiliaan
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Deborah R Gustafson
- Department of Neurology, Section for NeuroEpidemiology, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 1213, Brooklyn, NY, 11203, USA. .,Neuropsychiatric Epidemiology Unit (EPINEP), Institute for Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden. .,Department of Health and Education, University of Skövde, Skövde, Sweden.
| |
Collapse
|
43
|
Abstract
Our understanding of adipose tissue as an endocrine organ has been transformed over the last 20 years. During this time, a number of adipocyte-derived factors or adipokines have been identified. This article will review evidence for how adipokines acting via the central nervous system (CNS) regulate normal physiology and disease pathology. The reported CNS-mediated effects of adipokines are varied and include the regulation of energy homeostasis, autonomic nervous system activity, the reproductive axis, neurodevelopment, cardiovascular function, and cognition. Due to the wealth of information available and the diversity of their known functions, the archetypal adipokines leptin and adiponectin will be focused on extensively. Other adipokines with established CNS actions will also be discussed. Due to the difficulties associated with studying CNS function on a molecular level in humans, the majority of our knowledge, and as such the studies described in this paper, comes from work in experimental animal models; however, where possible the relevant data from human studies are also highlighted. © 2017 American Physiological Society. Compr Physiol 7:1359-1406, 2017.
Collapse
Affiliation(s)
- Craig Beall
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Lydia Hanna
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Kate L J Ellacott
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| |
Collapse
|
44
|
Yuruyen M, Gultekin G, Batun GC, Yavuzer H, Akcan FE, Doventas A, Emul M. Does plasma phoenixin level associate with cognition? Comparison between subjective memory complaint, mild cognitive impairment, and mild Alzheimer's disease. Int Psychogeriatr 2017; 29:1543-1550. [PMID: 28552081 DOI: 10.1017/s1041610217000825] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Alteration in energy expenditure or metabolism is the most accused risk issue for the onset and for the course of neurodegenerative cognitive disorders. Neuropeptides are suggested to be related with learning and memory. Phoenixin (PNX) is the most recently reported neuropeptide and we aimed to compare the plasma level in people with subjective memory complaints, patients with mild cognitive impairment, and mild Alzheimer's disease (AD). METHODS Ninety two participants enrolled in the study. After screening tests, all participants were assessed with a neuropsychological battery for further cognitive evaluations. We used ELISA kit to assay the level of Human PNX. RESULTS Patients with AD were significantly older than people in subjective memory complaint group (p = 0.02). There was no significant difference between groups according to gender (p = 0.435). Mean plasma PNX level was not significantly different between groups (p = 0.279). Mean plasma PNX level in MCI group was positively correlated with BMI (r = 0.402 and p = 0.028), serum HDL level (r = 0.454 and p = 0.012), blood systolic pressure (r = 0.428 and p = 0.018) and negatively correlated with logical memory (r=-0.335 and p=0.031). The mean plasma PNX level was positively correlated with immediate recall in subjective memory complaint group (r = 0.417 and p = 0.034). CONCLUSION This study is the first studying the association of plasma PNX level and cognitive complaints or decline. The knowledge about the role, interaction, and physiological functions of PNX is lacking. Lower plasma PNX level might be important in prodromal stages as MCI and the predictive role of PNX should be investigated in further studies.
Collapse
Affiliation(s)
- Mehmet Yuruyen
- Department of Internal Medicine, Medical School of Cerrahpasa, Istanbul University, Istanbul, Turkey
| | - Gozde Gultekin
- Department of Psychiatry, Medical School of Cerrahpasa, Istanbul University, Istanbul, Turkey
| | - Gizem Cetiner Batun
- Department of Psychiatry, Medical School of Cerrahpasa, Istanbul University, Istanbul, Turkey
| | - Hakan Yavuzer
- Department of Internal Medicine, Medical School of Cerrahpasa, Istanbul University, Istanbul, Turkey
| | - Fundan Engin Akcan
- Department of Psychiatry, Medical School of Cerrahpasa, Istanbul University, Istanbul, Turkey
| | - Alper Doventas
- Department of Internal Medicine, Medical School of Cerrahpasa, Istanbul University, Istanbul, Turkey
| | - Murat Emul
- Department of Psychiatry, Medical School of Cerrahpasa, Istanbul University, Istanbul, Turkey
| |
Collapse
|
45
|
Mainardi M, Spinelli M, Scala F, Mattera A, Fusco S, D'Ascenzo M, Grassi C. Loss of Leptin-Induced Modulation of Hippocampal Synaptic Trasmission and Signal Transduction in High-Fat Diet-Fed Mice. Front Cell Neurosci 2017; 11:225. [PMID: 28804449 PMCID: PMC5532388 DOI: 10.3389/fncel.2017.00225] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/14/2017] [Indexed: 12/16/2022] Open
Abstract
Hippocampal plasticity is triggered by a variety of stimuli including sensory inputs, neurotrophins and inflammation. Leptin, whose primary function is to regulate food intake and energy expenditure, has been recently shown to affect hippocampal neurogenesis and plasticity. Interestingly, mice fed a high-fat diet (HFD) exhibit impaired hippocampal function, but the underlying mechanisms are poorly understood. To address this issue, we compared leptin responsiveness of hippocampal neurons in control and HFD-fed mice by combining single-cell electrophysiology and biochemical assays. We found that leptin modulated spontaneous and evoked synaptic transmission in control, but not HFD, mice. This functional impairment was paralleled by blunted activation of STAT-3, one of the key signal transduction pathways controlled by the fully functional isoform of the leptin receptor, ObRb. In addition, SOCS-3 expression was non-responsive to leptin, indicating that modulation of negative feedback impinging on ObRb was also altered. Our results advance the understanding of leptin action on hippocampal plasticity and, more importantly, suggest that leptin resistance is a key determinant of hippocampal dysfunction associated with hypercaloric diet.
Collapse
Affiliation(s)
- Marco Mainardi
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Matteo Spinelli
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Federico Scala
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Andrea Mattera
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Salvatore Fusco
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Marcello D'Ascenzo
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Universita Cattolica del Sacro CuoreRome, Italy
| |
Collapse
|
46
|
Peng Y, Zhou L, Cao Y, Chen P, Chen Y, Zong D, Ouyang R. Relation between serum leptin levels, lipid profiles and neurocognitive deficits in Chinese OSAHS patients. Int J Neurosci 2017; 127:981-987. [PMID: 28117613 DOI: 10.1080/00207454.2017.1286654] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The aim of this study was to compare serum leptin, apolipoprotein A1 (ApoA1), apolipoprotein J (ApoJ) and apolipoprotein H (ApoH) levels in males with obstructive sleep apnea and hypopnea syndrome (OSAHS) to those in healthy control subjects and to examine the possible relation between neurocognitive performance and these factors/serum markers in the subjects. METHODS In this observational, cross-sectional study, a full-night polysomnography and sensitive neuropsychological assessment were performed on 50 newly diagnosed Chinese male patients and 30 healthy subjects. Fasting blood samples were used to measure leptin and ApoA1, ApoH and ApoJ levels using ELISA. RESULTS Compared with normal control subjects, OSAHS patients have significantly lower levels of ApoA1 and higher levels of leptin, ApoH and ApoJ. After adjustment for age, years of education, body mass index (BMI) and apnea-hypopnea index, leptin and ApoA1 were associated with global cognitive function, and leptin level was positively correlated with inhibition reaction time. ApoJ was negatively correlated with visual reproduction and logical memory performance. Multiple regression analysis shows that from age, BMI, education year, biomarker levels and the parameters of PSG, only the variables of leptin and education year added to the prediction of the Montreal cognitive assessment score in a statistically significant way. CONCLUSIONS Abnormal expression of leptin and apolipoproteins and poor performance on neuropsychological tests were observed in patients with OSAHS. There is also an association between serum leptin, ApoA1, and ApoJ levels and cognitive performance in the patients.
Collapse
Affiliation(s)
- Yating Peng
- a Department of Respiratory Medicine , Respiratory Disease Research Institute, The Second Xiangya Hospital, Central South University , Changsha , China
| | - Li Zhou
- a Department of Respiratory Medicine , Respiratory Disease Research Institute, The Second Xiangya Hospital, Central South University , Changsha , China
| | - Yuping Cao
- b Mental Health Institute, The Second Xiangya Hospital, Central South University , Changsha , China
| | - Ping Chen
- a Department of Respiratory Medicine , Respiratory Disease Research Institute, The Second Xiangya Hospital, Central South University , Changsha , China
| | - Yan Chen
- a Department of Respiratory Medicine , Respiratory Disease Research Institute, The Second Xiangya Hospital, Central South University , Changsha , China
| | - Dandan Zong
- a Department of Respiratory Medicine , Respiratory Disease Research Institute, The Second Xiangya Hospital, Central South University , Changsha , China
| | - Ruoyun Ouyang
- a Department of Respiratory Medicine , Respiratory Disease Research Institute, The Second Xiangya Hospital, Central South University , Changsha , China
| |
Collapse
|
47
|
Bartolome F, Antequera D, Tavares E, Pascual C, Maldonado R, Camins A, Carro E. Obesity and neuroinflammatory phenotype in mice lacking endothelial megalin. J Neuroinflammation 2017; 14:26. [PMID: 28143489 PMCID: PMC5282716 DOI: 10.1186/s12974-017-0800-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The multiligand receptor megalin controls the brain uptake of a number of ligands, including insulin and leptin. Despite the role of megalin in the transport of these metabolically relevant hormones, the role of megalin at the blood-brain-barrier (BBB) has not yet been explored in the context of metabolic regulation. METHODS Here we investigate the role of brain endothelial megalin in energy metabolism and leptin signaling using an endothelial cell-specific megalin deficient (EMD) mouse model. RESULTS We found megalin is important to protect mice from developing obesity and metabolic syndrome when mice are fed a normal chow diet. EMD mice developed neuroinflammation, by triggering several pro-inflammatory cytokines, displayed reduced neurogenesis and mitochondrial deregulation. CONCLUSIONS These results implicate brain endothelial megalin expression in obesity-related metabolic changes through the leptin signaling pathway proposing a potential link between obesity and neurodegeneration.
Collapse
Affiliation(s)
- Fernando Bartolome
- Neurodegenerative Disorders Group, Instituto de Investigacion Hospital 12 de Octubre (i + 12), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Desiree Antequera
- Neurodegenerative Disorders Group, Instituto de Investigacion Hospital 12 de Octubre (i + 12), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Eva Tavares
- Clinical and Experimental Pharmacology Research Unit, Valme University Hospital, Seville, Spain
| | - Consuelo Pascual
- Neurodegenerative Disorders Group, Instituto de Investigacion Hospital 12 de Octubre (i + 12), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Rosario Maldonado
- Clinical and Experimental Pharmacology Research Unit, Valme University Hospital, Seville, Spain
| | - Antoni Camins
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain.,Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina de la UB (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Eva Carro
- Neurodegenerative Disorders Group, Instituto de Investigacion Hospital 12 de Octubre (i + 12), Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain.
| |
Collapse
|
48
|
Fernandez-Martos CM, Atkinson RAK, Chuah MI, King AE, Vickers JC. Combination treatment with leptin and pioglitazone in a mouse model of Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2016; 3:92-106. [PMID: 29067321 PMCID: PMC5651376 DOI: 10.1016/j.trci.2016.11.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Combination therapy approaches may be necessary to address the many facets of pathologic change in the brain in Alzheimer's disease (AD). The drugs leptin and pioglitazone have previously been shown individually to have neuroprotective and anti-inflammatory actions, respectively, in animal models. METHODS We studied the impact of combined leptin and pioglitazone treatment in 6-month-old APP/PS1 (APPswe/PSEN1dE9) transgenic AD mouse model. RESULTS We report that an acute 2-week treatment with combined leptin and pioglitazone resulted in a reduction of spatial memory deficits (Y maze) and brain β-amyloid levels (soluble β-amyloid and amyloid plaque burden) relative to vehicle-treated animals. Combination treatment was also associated with amelioration in plaque-associated neuritic pathology and synapse loss, and also a significantly reduced neocortical glial response. DISCUSSION Combination therapy with leptin and pioglitazone ameliorates pathologic changes in APP/PS1 mice and may represent a potential treatment approach for AD.
Collapse
Affiliation(s)
- Carmen M Fernandez-Martos
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - Rachel A K Atkinson
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - Meng I Chuah
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
49
|
Bolze F, Bast A, Mocek S, Morath V, Yuan D, Rink N, Schlapschy M, Zimmermann A, Heikenwalder M, Skerra A, Klingenspor M. Treatment of diet-induced lipodystrophic C57BL/6J mice with long-acting PASylated leptin normalises insulin sensitivity and hepatic steatosis by promoting lipid utilisation. Diabetologia 2016; 59:2005-12. [PMID: 27272237 DOI: 10.1007/s00125-016-4004-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
Abstract
AIMS/HYPOTHESIS Recombinant leptin offers a viable treatment for lipodystrophy (LD) syndromes. However, due to its short plasma half-life, leptin replacement therapy requires at least daily subcutaneous (s.c.) injections. Here, we optimised this treatment strategy in LD mice by using a novel leptin version with extended plasma half-life using PASylation technology. METHODS A long-acting leptin version was prepared by genetic fusion with a 600 residue polypeptide made of Pro, Ala and Ser (PASylation), which enlarges the hydrodynamic volume and, thus, retards renal filtration, allowing less frequent injection. LD was induced in C57BL/6J mice by feeding a diet supplemented with conjugated linoleic acid (CLA). Chronic and acute effects of leptin treatment were assessed by evaluating plasma insulin levels, insulin tolerance, histological liver sections, energy expenditure, energy intake and body composition. RESULTS In a cohort of female mice, 4 nmol PAS-leptin (applied via four s.c. injections every 3 days) successfully alleviated the CLA-induced LD phenotype, which was characterised by hyperinsulinaemia, insulin intolerance and hepatosteatosis. The same injection regimen had no measurable effect when unmodified recombinant leptin was administered at an equivalent dose. In a cohort of LD males, a single s.c. injection of PAS-leptin did not affect energy expenditure but inhibited food intake and promoted a shift in fuel selection towards preferential fat oxidation, which mechanistically substantiates the metabolic improvements. CONCLUSIONS/INTERPRETATION The excellent pharmacological properties render PASylated leptin an agent of choice for refining both animal studies and therapeutic strategies in the context of LD syndromes and beyond.
Collapse
Affiliation(s)
- Florian Bolze
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Andrea Bast
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Sabine Mocek
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Volker Morath
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354, Freising (Weihenstephan), Germany
| | - Detian Yuan
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Rink
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Martin Schlapschy
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354, Freising (Weihenstephan), Germany
| | - Anika Zimmermann
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354, Freising (Weihenstephan), Germany.
- XL-protein GmbH, Freising, Germany.
| | - Martin Klingenspor
- Lehrstuhl für Molekulare Ernährungsmedizin and Else Kröner-Fresenius Center, Technische Universität München, Gregor-Mendel-Str. 2, 85354, Freising (Weihenstephan), Germany.
- Research Center for Nutrition and Food Science (ZIEL), Technische Universität München, Freising (Weihenstephan), Germany.
| |
Collapse
|
50
|
McGuire MJ, Ishii M. Leptin Dysfunction and Alzheimer's Disease: Evidence from Cellular, Animal, and Human Studies. Cell Mol Neurobiol 2016; 36:203-17. [PMID: 26993509 DOI: 10.1007/s10571-015-0282-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/01/2015] [Indexed: 12/13/2022]
Abstract
There is accumulating evidence from epidemiological studies that changes in body weight are associated with Alzheimer's disease (AD) from mid-life obesity increasing the risk of developing AD to weight loss occurring at the earliest stages of AD. Therefore, factors that regulate body weight are likely to influence the development and progression of AD. The adipocyte-derived hormone leptin has emerged as a major regulator of body weight mainly by activating hypothalamic neural circuits. Leptin also has several pleotropic effects including regulating cognitive function and having neuroprotective effects, suggesting a potential link between leptin and AD. Here, we will examine the relationship between leptin and AD by reviewing the recent evidence from cellular and animal models to human studies. We present a model where leptin has a bidirectional role in AD. Not only can alterations in leptin levels and function worsen cognitive decline and progression of AD pathology, but AD pathology, in of itself, can disrupt leptin signaling, which together would lead to a downward spiral of progressive neurodegeneration and worsening body weight and systemic metabolic deficits. Collectively, these studies serve as a framework to highlight the importance of understanding the molecular mechanisms underlying the body weight and systemic metabolic deficits in AD, which has the potential to open new avenues that may ultimately lead to novel therapeutic targets and diagnostic tools.
Collapse
Affiliation(s)
- Matthew J McGuire
- Feil Family Brain and Mind Research Institute, Joan and Sanford I. Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Makoto Ishii
- Feil Family Brain and Mind Research Institute, Joan and Sanford I. Weill Medical College of Cornell University, New York, NY, 10065, USA.
| |
Collapse
|