Review article
Diabetes, insulin and new therapeutic strategies for Parkinson’s disease: Focus on glucagon-like peptide-1 receptor agonists

https://doi.org/10.1016/j.yfrne.2021.100914Get rights and content

Highlights

  • Epidemiological studies show a relationship between Parkinson’s disease and Diabetes.

  • Recent data support repurposing antidiabetic treatments for Parkinson’s disease.

  • Insulin and GLP1 agonists are a promising strategy against Parkinson’s disease.

Abstract

Parkinson's disease and diabetes mellitus are two chronic disorders associated with aging that are becoming increasingly prevalent worldwide. Parkinson is a multifactorial progressive condition with no available disease modifying treatments at the moment. Over the last few years there is growing interest in the relationship between diabetes (and impaired insulin signaling) and neurodegenerative diseases, as well as the possible benefit of antidiabetic treatments as neuroprotectors, even in non-diabetic patients. Insulin regulates essential functions in the brain such as neuronal survival, autophagy of toxic proteins, synaptic plasticity, neurogenesis, oxidative stress and neuroinflammation. We review the existing epidemiological, experimental and clinical evidence that supports the interplay between insulin and neurodegeneration in Parkinson's disease, as well as the role of antidiabetic treatments in this disease.

Introduction

Parkinson’s disease (PD) is the second most frequent neurodegenerative disorder, with an increasing prevalence in elderly population (about 1% in people over 65 years old). Although its etiology is still not well understood, multiple factors are known to play a role in the nigrostriatal pathway degeneration, both genetic and environmental. The special vulnerability of dopaminergic neurons to oxidative stress has led to growing research in inflammatory mechanisms to achieve possible neuroprotective strategies. Diabetes mellitus (DM) is well known to be an inflammatory condition, and has also been linked to neurodegenerative diseases through abnormal insulin pathways (Fiory et al., 2019). The results of epidemiological studies searching for evidence of a relationship between PD and DM are controversial. However, strong evidence suggests that DM might be a risk factor for PD, and that it might be limited to those patients with long diabetic disease duration (over 10 years) (De Pablo-Fernandez et al., 2017). Also, DM has been considered as a modifying factor for motor and non-motor symptoms of PD (Pagano et al., 2018). Accordingly, it is essential to clarify if a very frequent comorbidity as DM can influence the development or prognosis of PD, as well as the role of the available treatments for DM in neurodegeneration.

Section snippets

Parkinson and diabetes: epidemiological evidence

PD and DM are two well known aging-associated disorders. More importantly, DM is considered an epidemic in modern society. Therefore, it is important to determine if DM or insulin resistance involves an increased risk to develop PD or a worse prognosis. However, a large number of controversial studies have been reported, which will be analyzed in the following paragraphs.

A meta-analysis (Cereda et al., 2011) published in 2011 reviewed the relationship between pre-existing DM and PD, concluding

Parkinson and insulin

The above-mentioned epidemiological evidence is supported by molecular evidences. DM and neurodegeneration share several pathological pathways, such as oxidative stress, mitochondrial dysfunction and neuro-inflammation. Furthermore, significant evidence supports the role of insulin resistance in the interplay between DM and PD. The existence of common molecular mechanisms encourages the use of drug repurposing treatments to modify disease progression in parkinsonian patients.

Glucose uptake in

Diabetes treatment repurposing in Parkinson’s disease

Considering the evidence about the impact of insulin resistance in neurodegeneration, it seems reasonable to wonder about the role of antidiabetic drugs in the central nervous system and neurodegenerative disorders such as PD or Alzheimer’s disease. A population-based, longitudinal, cohort study (Brauer et al., 2020) reported different PD incidence among T2DM patients depending on the antidiabetic treatment received, as well as a higher risk in untreated diabetic patients. Taking into account

Treatment repurposing: evidence from human clinical trials

After obtaining positive results from experimental data, it is vital to determine if these treatments are also effective against PD in humans. In the following paragraphs we will summarize major results in clinical trials on treatment repurposing using the above-mentioned drugs for PD.

In 2013 Aviles-Olmos et al. (2013a) developed a single-blind trial with 45 moderate non-diabetic PD patients to assess exenatide treatment in PD for a 12 month period. This trial was designed as a proof of concept

Levodopa and antidiabetics

A great concern when repurposing treatments is whether they may interact with other necessary drugs required for the patients. In PD, levodopa is the most effective antiparkinsonian drug and the current gold standard treatment, so it is essential to know if repurposed treatments may modify its function. There are several animal models and some clinical data reporting a possible benefit in motor function of different antidiabetic drugs when administered with levodopa, as we will describe in this

Conclusions. Synthesis and future directions

Traditionally, patients with PD are considered as patients with a neurologic disease that may also suffer other comorbidities. The pathophysiology of PD is very complex, and is not limited to central nervous system structures. We need to comprehend PD as a systemic disease, where non-neurologic disorders can determine the severity or even the development of neurodegeneration. This can help us not only to provide a better treatment against the disease, but also to identify individuals with

Funding

Grant sponsor: Spanish Ministry of Health (PI17/00828 and CIBERNED). Grant sponsor: Galician Government (XUGA, ED431C 2018/10, ED431G/05). Grant sponsor: FEDER (Regional European Development Fund).

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (185)

  • E.A. Elbassuoni et al.

    Mechanism of the neuroprotective effect of GLP-1 in a rat model of Parkinson’s with pre-existing diabetes

    Neurochem. Int.

    (2019)
  • P. Feng et al.

    Two novel dual GLP-1/GIP receptor agonists are neuroprotective in the MPTP mouse model of Parkinson’s disease

    Neuropharmacology

    (2018)
  • J.M. Fine et al.

    Intranasal delivery of low-dose insulin ameliorates motor dysfunction and dopaminergic cell death in a 6-OHDA rat model of Parkinson’s Disease

    Neurosci. Lett.

    (2020)
  • S. Gao et al.

    Alpha-synuclein overexpression negatively regulates insulin receptor substrate 1 by activating mTORC1/S6K1 signaling

    Int. J. Biochem. Cell Biol.

    (2015)
  • Y. Gao et al.

    Dietary sugars, not lipids, drive hypothalamic inflammation

    Mol. Metab.

    (2017)
  • S. Goodenough et al.

    Glycogen synthase kinase 3β links neuroprotection by 17β-estradiol to key Alzheimer processes

    Neuroscience

    (2005)
  • H.H. Hansen et al.

    Characterization of liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, in rat partial and full nigral 6-hydroxydopamine lesion models of Parkinson’s disease

    Brain Res.

    (2016)
  • X. Hou et al.

    Metformin reduces intracellular reactive oxygen species levels by upregulating expression of the antioxidant thioredoxin via the AMPK-FOXO3 pathway

    Biochem. Biophys. Res. Commun.

    (2010)
  • J.H. Jhamandas et al.

    Actions of B-Amyloid Protein on Human Neurons Are Expressed through the Amylin Receptor

    Am. J. Pathol.

    (2011)
  • C. Ji et al.

    A novel dual GLP-1 and GIP receptor agonist is neuroprotective in the MPTP mouse model of Parkinson’s disease by increasing expression of BNDF

    Brain Res.

    (2016)
  • A.M. Kabel et al.

    Linagliptin potentiates the effect of L-dopa on the behavioural, biochemical and immunohistochemical changes in experimentally-induced Parkinsonism: Role of toll-like receptor 4, TGF-β1, NF-κB and glucagon-like peptide 1

    Physiol. Behav.

    (2018)
  • N. Katila et al.

    Metformin lowers α-synuclein phosphorylation and upregulates neurotrophic factor in the MPTP mouse model of Parkinson’s disease

    Neuropharmacology

    (2017)
  • S. Kim et al.

    Exendin-4 protects dopaminergic neurons by inhibition of microglial activation and matrix metalloproteinase-3 expression in an animal model of Parkinson’s disease

    J. Endocrinol.

    (2009)
  • F.J. Kong et al.

    Liraglutide ameliorates cognitive decline by promoting autophagy via the AMP-activated protein kinase/mammalian target of rapamycin pathway in a streptozotocin-induced mouse model of diabetes

    Neuropharmacology

    (2018)
  • V. Kotagal et al.

    Diabetes is associated with postural instability and gait difficulty in Parkinson disease

    Park. Relat. Disord.

    (2013)
  • E.Y. Lee et al.

    Rosiglitazone, a PPAR-γ agonist, protects against striatal dopaminergic neurodegeneration induced by 6-OHDA lesions in the substantia nigra of rats

    Toxicol. Lett.

    (2012)
  • T. Li et al.

    Neuroprotection of GLP-1/GIP receptor agonist via inhibition of mitochondrial stress by AKT/JNK pathway in a Parkinson’s disease model

    Life Sci.

    (2020)
  • Y. Li et al.

    D-Ala2-GIP-glu-PAL is neuroprotective in a chronic Parkinson’s disease mouse model and increases BNDF expression while reducing neuroinflammation and lipid peroxidation

    Eur. J. Pharmacol.

    (2017)
  • Y. Li et al.

    Neuroprotective effects of a GIP analogue in the MPTP Parkinson’s disease mouse model

    Neuropharmacology

    (2016)
  • G. Lietzau et al.

    Dipeptidyl peptidase-4 inhibitors and sulfonylureas prevent the progressive impairment of the nigrostriatal dopaminergic system induced by diabetes during aging

    Neurobiol. Aging

    (2020)
  • Y.A. Lim et al.

    Human but not rat amylin shares neurotoxic properties with Abeta42 in long-term hippocampal and cortical cultures

    FEBS Lett.

    (2008)
  • J. Liu et al.

    Enhanced AMPA Receptor Trafficking Mediates the Anorexigenic Effect of Endogenous Glucagon-like Peptide-1 in the Paraventricular Hypothalamus Article Enhanced AMPA Receptor Trafficking Mediates the Anorexigenic Effect of Endogenous Glucagon- like Peptide-1 in the Paraventricular Hypothalamus

    Neuron

    (2017)
  • W. Liu et al.

    Neuroprotective effects of lixisenatide and liraglutide in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease

    Neuroscience

    (2015)
  • R.M. Abdelsalam et al.

    Neuroprotective effects of vildagliptin in rat rotenone Parkinsons disease model role of RAGE-NFκB and Nrf2-antioxidant signaling pathways

    J. Neurochem.

    (2015)
  • A. Abedini et al.

    The receptor for advanced glycation endproducts is a mediator of toxicity by IAPP and other proteotoxic aggregates: Establishing and exploiting common ground for novel amyloidosis therapies

    Protein Sci.

    (2018)
  • A. Abuirmeileh et al.

    Exendin-4 treatment enhances L-DOPA evoked release of striatal dopamine and decreases dyskinetic movements in the 6-hydoxydopamine lesioned rat

    J. ofPharmacy Pharmacol.

    (2012)
  • E. Alvarez et al.

    The expression of GLP-1 receptor mRNA and protein allows the effect of GLP-1 on glucose metabolism in the human hypothalamus and brainstem

    J. Neurochem.

    (2005)
  • D. Athauda et al.

    Insulin resistance and Parkinson’s disease: a new target for disease modification?

    Prog Neurobiol Oct-Nov

    (2016)
  • D. Athauda et al.

    Utility of Neuronal-Derived Exosomes to Examine Molecular Mechanisms That Affect Motor Function in Patients with Parkinson Disease: A Secondary Analysis of the Exenatide-PD Trial

    JAMA Neurol.

    (2019)
  • D. Athauda et al.

    Post hoc analysis of the Exenatide-PD trial—Factors that predict response

    Eur. J. Neurosci.

    (2019)
  • D. Athauda et al.

    What effects might exenatide have on non-motor symptoms in Parkinson’s disease: A post Hoc analysis

    J. Parkinsons. Dis.

    (2018)
  • I. Aviles-Olmos et al.

    Exenatide and the treatment of patients with Parkinson’s disease

    J Clin Invest

    (2013)
  • I. Aviles-Olmos et al.

    Motor and Cognitive Advantages Persist 12 Months after Exenatide Exposure in Parkinson’s Disease

    J. Parkinsons. Dis.

    (2015)
  • I. Aviles-Olmos et al.

    Parkinson’s disease, insulin resistance and novel agents of neuroprotection

    Brain

    (2013)
  • G.A. Badawi et al.

    Sitagliptin and Liraglutide Modulate L-dopa Effect and Attenuate Dyskinetic Movements in Rotenone-Lesioned Rats

    Neurotox. Res.

    (2019)
  • G.A. Badawi et al.

    Sitagliptin and liraglutide reversed nigrostriatal degeneration of rodent brain in rotenone-induced Parkinson’s disease

    Inflammopharmacology

    (2017)
  • F. Bassil et al.

    Insulin resistance and exendin-4 treatment for multiple system atrophy

    Brain

    (2017)
  • W.M. Berhanu et al.

    Inter-Species Cross-Seeding: Stability and Assembly of Rat - Human Amylin Aggregates

    PLoS ONE

    (2014)
  • G. Bertilsson et al.

    Peptide Hormone Exendin-4 Stimulates Subventricular Zone Neurogenesis in the Adult Rodent Brain and Induces Recovery in an Animal Model of Parkinson’s Disease

    J. Neurosci. Res.

    (2008)
  • A. Biosa et al.

    Diabetes Mellitus as a Risk Factor for Parkinson’s Disease: a Molecular Point of View

    Mol. Neurobiol.

    (2018)
  • Cited by (12)

    • AT1 receptor autoantibodies mediate effects of metabolic syndrome on dopaminergic vulnerability

      2023, Brain, Behavior, and Immunity
      Citation Excerpt :

      This is consistent with a recent study, showing significant increase of AT1-AA and ACE2-AA levels in Parkinson’s disease patients, which was related to the dopaminergic degeneration and neurodegeneration-related neuroinflammation (Labandeira et al., 2022a). An important number of experimental and clinical studies have related MetS (Leehey et al., 2017; Nam et al., 2018; Park et al., 2021; Zhang and Tian, 2014) and several of the MetS components such as obesity (Chen et al., 2014; Hu et al., 2006), diabetes (Labandeira et al., 2021), hypercholesterolemia (Hu et al., 2008) and hypertension (Chen et al., 2019) with higher risk of Parkinson’s disease, and risk of Parkinson’s disease may gradually increase with the number of MetS components involved (Nam et al., 2018). Controversial findings have also been reported (Hu, 2010; Saaksjarvi et al., 2015).

    • Type 2 diabetes mellitus-associated cognitive dysfunction: Advances in potential mechanisms and therapies

      2022, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      Sitagliptin therapy for 6 months was associated with improved cognitive function in elderly diabetes patients with and without AD (Isik et al., 2017). Moreover, the neuroprotective effect of DPP-4Is has also been observed in PD and HD experimental models (Badawi et al., 2017; Labandeira et al., 2021; Lietzau et al., 2020). In clinical settings, DPP-4Is have beneficial effects on baseline nigrostriatal dopamine degeneration and improve long-term motor outcomes in T2D patients with PD (Jeong et al., 2021).

    • Repositioning and development of new treatments for neurodegenerative diseases: Focus on neuroinflammation

      2022, European Journal of Pharmacology
      Citation Excerpt :

      While some of them suggest that DM is a risk factor for PD (De Pablo-Fernandez et al., 2018; Schernhammer et al., 2011; Yang et al., 2017), others failed to show this association (Palacios et al., 2011; Simon et al., 2007). The connection between neurodegenerative disorders and DM is not yet fully understood; however, both diseases share common pathological pathways, including inflammation, oxidative stress, and mitochondrial dysfunction (Labandeira et al., 2021). Furthermore, the phenomenon of insulin resistance, recognized as a central feature of DM2, has been proposed to occur also in the brain and play a crucial role in the neurodegeneration (Arnold et al., 2018; Aviles-Olmos et al., 2013b).

    View all citing articles on Scopus
    View full text