검색
검색 팝업 닫기

Ex) Article Title, Author, Keywords

Article

Split Viewer

Review

DTT 2024; 3(1): 83-93

Published online March 31, 2024

https://doi.org/10.58502/DTT.24.0030

Copyright © The Pharmaceutical Society of Korea.

Anti-Inflammatory Effects of Dipeptidyl Peptidase-4 Inhibitors and Their Therapeutic Application for Parkinson's Disease

Yeojin Bang , Soung-Hee Moon, Sumin Lee, Hyun Jin Choi

College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea

Correspondence to:Hyun Jin Choi, hjchoi3@cha.ac.kr

Received: October 31, 2023; Revised: February 26, 2024; Accepted: February 26, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Parkinson’s disease (PD) is characterized by a progressive loss of dopaminergic neurons, leading to a decline in dopamine levels and the manifestation of motor and non-motor symptoms. The current treatments primarily focus on symptom management, leaving the need for disease-modifying therapies unmet. Emerging research has shown that neuroinflammation plays a pivotal role in the pathogenesis and progression of PD. Dipeptidyl peptidase-4 (DPP-4) inhibitors, originally developed for diabetes treatment, represent a novel avenue of investigation in the context of PD treatment. Beyond their established role in glycemic control, DPP-4 inhibitors have shown promise in modulating inflammatory responses within the central nervous system. This indicates their potential application in controlling neuroinflammation in PD, which in turn, could impact disease progression and neurodegeneration. This review summarized the current understanding of the role of DPP-4 inhibitors in neuroinflammation and their potential to modulate PD progression. Understanding the intricate interplay between neuroinflammation and dopaminergic neuron degeneration could pave the way for a new era of disease-modifying strategies, and DPP-4 inhibitors could offer a potential avenue to address both inflammation and neurodegeneration in PD.

Keywordsdipeptidyl peptidase-4 inhibitors, neuroinflammation, neuroprotection, parkinson’s disease

Parkinson’s disease (PD) is a complex neurodegenerative disorder characterized by a progressive loss of dopaminergic neurons in the substantia nigra and a significant reduction in dopamine levels in the striatum of the brain (Vaillancourt and Mitchell 2020), ultimately leading to the development of both non-motor and motor symptoms (Bang et al. 2021). This dopaminergic deficit is responsible for the motor symptoms observed in PD, such as resting tremors, bradykinesia (slowness of movement), rigidity, and postural instability (Moustafa et al. 2016).

Available PD treatments primarily target the modulation of dopamine signaling to alleviate motor symptoms and improve the quality of life for patients. Levodopa, the precursor of dopamine, is commonly prescribed to replenish dopamine levels in the brain, providing symptomatic relief (Ide et al. 2020). Additionally, dopamine agonists, monoamine oxidase inhibitors, and catechol-O-methyl transferase inhibitors are utilized to enhance dopaminergic activity or reduce dopamine metabolism. Despite their effectiveness in managing motor symptoms, these agents do not halt the progressive dopaminergic neurodegeneration at the core of PD. In addition, patients often experience motor fluctuations and dyskinesia as the disease progresses, which significantly challenges the management of symptoms.

Given the central role of dopaminergic neurodegeneration in PD, researchers and clinicians have sought to develop disease-modifying treatments that can decelerate or halt the process. Despite significant efforts and a better understanding of the pathogenesis of PD, the development of effective disease-modifying therapies remains challenging. Various therapeutic approaches targeting pathways involved in protein aggregation, mitochondrial function, neuroinflammation, adult neurogenesis, and neuroprotection have been explored (Lim et al. 2018; Troncoso-Escudero et al. 2018; Moon et al. 2022). Among these, emerging research has highlighted the intricate role of neuroinflammation in the pathogenesis of PD. Neuroinflammation, which involves the activation of glial cells and release of pro-inflammatory cytokines, has garnered increasing attention due to its potential contribution to neuronal damage and PD progression. With the growing recognition of the involvement of neuroinflammation in PD, the development of drugs capable of regulating neuroinflammation to manage PD has gained traction.

This review aims to provide a comprehensive overview of the role of neuroinflammation in PD pathology and highlight the importance of developing inflammation-modulating drugs to treat PD. By understanding the intricate interplay between inflammation and neurodegeneration, we can better address the unmet need for disease-modifying therapies in PD management.

Emerging evidence from studies based on animal models and humans suggests that the immune system and neuroinflammation play crucial roles in the pathogenesis of PD (Table 1). Imaging techniques, such as positron emission tomography, have enabled the visualization of neuroinflammation in patients with PD, providing valuable insights into disease progression (Bartels et al. 2010; Kang et al. 2018; Lavisse et al. 2021; Doot et al. 2022).

Table 1 Pathological and inflammatory alterations in parkinson’s disease

GroupRegulation in PDReferences
PD patientsPD (n = 5), DAT (n = 9), cerebro vascular accidents (n = 2), without premortem neurologic disease (n = 7)↑ Lewy bodies in SN of PD patients
↑ GFAP+, HLA-DR+ cells in SN of PD patients
McGeer et al. (1988)
De novo PD (n = 22), sporadic AD (n = 11)↑ IL-6 and IL-1β in CSF of PD patientsBlum-Degen et al. (1995)
Healthy participants (n = 22), early-stage PD (n = 24)↑ Th1/total T cells, IL-10, IL-17A in blood of PD patientsLiu et al. (2022)
LRRK2-G2019S-PD patients (n = 33), idiopathic PD patients (n = 33)↑ Rest tremor, postural instability, freezing of gait, dyskinesias, UPSIT score, depression and bristol scale in LRRK2-G2019S PD patients
↓ Hyposmia and action tremor in LRRK2-G2019S PD patients
Gaig et al. (2014)
PD (n = 4)↑ CD68 in olfactory bulb of PD patientsVroon et al. (2007)
PD animal modelsNeurotoxinLPS rats (10 μg/4 μL, 2 days and 4 weeks)↑ iNOS, TNF-α and IL-6 in SN of LPS administrated rats
↑ α-syn in SN of LPS administrated rats
Choi et al. (2010)
LPS mice (5 μg/1 μL, stereotaxic)↓ TH+ neurons in SN of LPS administrated WT mice
↑ Isolectin B4 and LRRK2 in SN of LPS administrated WT mice
↑ TH+ neurons in SN of LPS administrated LRRK2−/− mice
↓ Isolectin B4 and LRRK2 in SN of LPS administrated LRRK2−/− mice
Moehle et al. (2012)
MPTP mice (20 mg/kg, i.p., 4 times at 2 h intervals, 1, 3 and 5 days)↑ IL-1β, IL-1α and IL-1ra in SN, striatum and olfactory bulb of MPTP administrated miceVroon et al. (2007)
Adβgal (stereotaxic, 7, 21, 40, 60 days)↓ TH+ neurons in SN of Adβgal administrated rats
↑ IL-1β, GFAP and CD68 in SN of Adβgal administrated rats
Ferrari et al. (2006)
MACO rats↑ DJ-1 in brain of cerebral I/R injury rats
↑ TNF-α, IL-1β and IL-6 in brain of cerebral I/R injury rats
Peng et al. (2020)
NLRP3A350V mice, Parkinflx/flx mice, Casp1+/−/parkin+/flx mice (adged 8-10 weeks)↑ NLRP3, cleaved caspase 1 and Iba-1 in SNpc of AAV GFP-Cre injected Parkinflx/flx mice and NLRP3A350V mice
↓ TH+ neurons in SNpc of Parkinflx/flx mice and NLRP3A350V mice
Panicker et al. (2022)
TransgenicNlrp3−/−, Cx3CrlCreER, Nlrp3D301NneoR mice (aged 9-12 weeks)↓ Motor function in MPTP administrated Nlrp3+/+ mice
↓ TH+ neurons and dopamine in SN of MPTP administrated Nlrp3+/+ mice
↑ Iba-1, IL-1β and ASC in SN of MPTP administrated Nlrp3+/+ mice
Lee et al. (2019)
LRRK2G2019S, LRRK2−/− mice↑ LRRK2 in brain of human α-syn fibrils administrated miceXu et al. (2022)
Parkin−/−, PINK1−/− mice↑ IL-12 and IL-13 in serum of Parkin−/− and PINK1−/− mice
↑ CXCL1, CCL2 and CCL4 in serum of Parkin−/− and PINK1−/− mice
Sliter et al. (2018)
DJ-1-deficient mice (Park7−/−)↓ TNF-α, IL-1β and IL-23p19 in striatum of Park7−/− miceNakamura et al. (2021)
t-SCI rats↑ DJ-1 in t-SCI in brain of rats
↑ NLRP3, cleaved caspase 1, IL-1β, IL-18 and MMP9 in t-SCI in brain of rats
Cai et al. (2022)

This table provides an overview of pathological features and inflammatory alterations in both PD patients and animal models. For details and references, see main text. ASC, apoptosis-associated speck like protein containing a CARD; AD, Alzheimer’s disease; CCL, chemokine (C-C motif) ligand; CD68, cluster of differentiation; COX-2, cyclooxygenase 2; CSF, cerebrospinal fluid; CXCL1, chemokine (C-X-C motif) ligand 1; DAT, dopamine transporter; GFAP, glial fibrillary acidic protein; HLA-DR, human leukocyte antigen-DR; IL, interleukin; iNOS, inducible nitric oxide synthase; I/R, ischemia/reperfusion; LPS, lipopolysaccharide; LRRK2, leucine rich repeat kinase 2; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MMP9, matrix metallopeptidase 9; NLRP3, nucleotide-binding oligomerization domain leucine rich repeat and pyrin domain containing 3; PBMC, peripheral blood mononuclear cells; PD, Parkinson’s disease; SN, substantia nigra; TH, tyrosine hydroxylase; Th1, type 1 T helper; TNF, tumor necrosis factor; t-SCI, traumatic spinal cord injury; UPSIT, University of Pennsylvania smell identification test; WT, wild type.


Microglia are specialized immune cells found within the central nervous system (CNS) and are considered the main form of active immune defense in the CNS (Filiano et al. 2015). Their ability to recognize certain patterns, through receptors known as pattern-recognition receptors, enables them to detect molecules linked to pathogens and damage, leading to microglial stimulation (Akira et al. 2006). Microgliosis, the continuous activation of microglia with a pro-inflammatory nature, has been identified as a contributing factor in various neurodegenerative diseases, including PD. Early evidence has shown that people diagnosed with both PD and PD combined with dementia exhibit a high concentration of activated microglia in their substantia nigra (McGeer et al. 1988). The activation of microglia, astrogliosis, and the infiltration of activated immune cells from the periphery contribute to harmful changes in the brain, resulting in neuronal death and the advancement of PD (Hirsch and Hunot 2009). Additionally, the number of microglia and their activation state correlate with α-synuclein deposition in the pathway involved in dopaminergic neuron loss (Croisier et al. 2005; Choi et al. 2010). Moreover, inflammatory cytokines like interleukin-1beta (IL-1β) and IL-6, which are released upon microglial activation, have been identified in the nigrostriatal pathway of brains affected by PD in both humans and corresponding animal models (Ferrari et al. 2006). These mediators exacerbate dopamine impairment and even lead to neuronal death (Blum-Degen et al. 1995; Vroon et al. 2007). In addition, according to recent research, early stages of PD are associated with microglial activation in the putamen and peripheral T helper 1 phenotypic bias (Liu et al. 2022). Consequently, peripheral adaptive immunity may indirectly result in microglial activation in the neurodegenerative process involved in PD (Liu et al. 2022).

The NOD-, LRR- and pyrin domain containing 3 (NLRP3) inflammasome is emerging as a pivotal component in understanding the neuroinflammatory processes involved in PD. This intracellular multiprotein complex, primarily known for its role in initiating innate immune responses, has recently been connected to the intricate mechanisms underlying PD pathogenesis. Upon activation, NLRP3 leads to the cleavage of pro-caspase-1 into active caspase-1, thereby enabling the conversion of pro-IL-1β into their active form (Franchi et al. 2009). These pro-inflammatory cytokines contribute to a chronic inflammatory state within the brain. This phenomenon activates the NLRP3 inflammasome and may also further exacerbate neuroinflammatory responses, establishing a feedback loop that sustains microgliosis (Heneka et al. 2018). Furthermore, dysregulation of the NLRP3 inflammasome has been linked to dopaminergic neuronal loss, a defining pathological hallmark of PD (Lee et al. 2019; Panicker et al. 2022). Some preclinical trials investigating the use of NLRP3 inhibitors to attenuate neuroinflammation and ameliorate neurodegenerative symptoms have yielded promising results (Coll et al. 2015; Lonnemann et al. 2020; Xue et al. 2021; Zeng et al. 2021). These associations emphasize the potential of NLRP3 as a therapeutic target.

Several studies have reported a unique link between certain genes associated with PD and the immune response of CNS cells. The leucine-rich repeat kinase 2 (LRRK2) gene has emerged as a pivotal component in PD; not only is it one of the most commonly mutated genes in familial PD, but variations in LRRK2 are also linked with sporadic PD cases (Zimprich et al. 2004; Gaig et al. 2014; Cookson 2017). LRRK2 encodes a protein kinase that is involved in a multitude of cellular processes, including autophagy, vesicle trafficking, and immune response. In PD, LRRK2 mutations lead to an aberrant activation of microglial cells. Reducing LRRK2 levels through knockdown or inhibiting its kinase activity in primary microglia leads to a decrease in the release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and IL-1β (Moehle et al. 2012; Russo et al. 2015). Moreover, mutated LRRK2 may enhance α-synuclein pathology, further exacerbating the inflammatory response (Bieri et al. 2019; Xu et al. 2022). The Phosphatase and tensin homolog-induced putative kinase 1 (PINK1) gene has been identified as a critical component in the development and progression of PD, specifically in connection with neuroinflammation. In healthy neurons, PINK1 monitors mitochondrial health and when damage occurs, it triggers mitophagy, the process involved in the selective degradation of damaged mitochondria. Mutations in the PINK1 gene can disrupt this protective mechanism, leading to an accumulation of damaged mitochondria within the cell. Mitochondrial accumulation can trigger a cascade of pro-inflammatory signals, inducing microglial activation, and the release of inflammatory mediators such as TNFα and IL-1β (Sliter et al. 2018). The activated microglia, in turn, can cause further damage to the neurons, creating a vicious cycle of neuroinflammation that results in the neurodegenerative process observed in PD. PINK1 has also been associated with immune responses outside the CNS, like alterations in the peripheral immune cells of patients with PD with PINK1 mutations, indicating a broader systemic involvement in inflammatory regulation (Zhou et al. 2019a). The DJ-1 gene (Parkinson disease protein 7, PARK7), associated with PD, is primarily expressed in astrocytes and microglia, and it has been investigated for its potential link to inflammation. In the context of ischemic stroke, DJ-1 has been shown to have anti-inflammatory properties by suppressing the production of inflammatory cytokines such as TNF-α, IL-1β, and IL-18 (Peng et al. 2020; Nakamura et al. 2021). Moreover, recent research has indicated that DJ-1 reduces neuroinflammation and the associated impairment of the blood-spinal cord barrier by inhibiting the activation of the NLRP3 inflammasome through the suppressor of cytokine signaling 1/Ras-related C3 botulinum toxin substrate 1/reactive oxygen species signaling pathway in a rat model of traumatic spinal cord injury (Cai et al. 2022).

DPP-4 is a serine exopeptidase involved in the degradation of incretin hormones, including glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide. DPP-4 is widely distributed in several organs, including the liver, lungs, intestinal epithelium, placenta, kidney, renal proximal tubules, and neurons, as well as in body fluids including the cerebrospinal fluid (Green et al. 2006; Kosaraju et al. 2013). In addition to its crucial role in the regulation of blood glucose levels, which are commonly used in the treatment of type 2 diabetes mellitus (T2DM), DPP-4 can be directly associated with the modulation of various non-incretin related processes, including inflammatory responses and neurophysiological functions (Aertgeerts et al. 2004).

Several reports have demonstrated that DPP-4 inhibitors modulate the activation of immune cells in adipose tissues (Shirakawa et al. 2011), and the inhibition of DPP-4 activity reduces the expression and activity of the macrophage marker F4/80 (Klein et al. 2014). The DPP-4 inhibitors, sitagliptin and NVPDPP728, exhibit significant potential in repressing the expression of NLRP3 and IL-1β in human macrophages (Dai et al. 2014). In patients with T2DM, a meta-analysis by Xie et al. confirmed that DPP-4 inhibitors exhibit substantial anti-inflammatory effects. The study, encompassing 22 trials with 1595 patients with T2DM, revealed that DPP-4 inhibitor therapy is notably linked to reductions in C-reactive protein, TNF-α, IL-6, and IL-1β levels (Xie et al. 2023). Sitagliptin is a DPP-4 inhibitor with anti-inflammatory and antioxidant properties, and effectively alleviates intestinal inflammation associated with severe acute pancreatitis (SAP), both in vitro and in vivo. Sitagliptin treatment leads to the upregulation of nuclear factor erythroid-derived 2-like 2 (Nrf2) expression while concurrently downregulating NF-κB expression in mice with SAP, thereby contributing significantly to its therapeutic efficacy (Zhou et al. 2019b). Moreover, for severe acute pancreatitis-related acute lung injury, sitagliptin is a promising therapeutic agent that activates the p62-kelch like ECH associated protein 1-Nrf2 signaling pathway, leading to Nrf2 nuclear translocation (Kong et al. 2021). Vildagliptin effectively suppresses the generation of reactive oxygen species (ROS), the production of vascular inflammatory factors, and monocyte adhesion to endothelial cells, potentially through the inhibition of toll-like receptor 4 and NF-κB nuclear signals (Wiciński et al. 2020).

It is not yet certain whether the anti-inflammatory effects of DPP4 inhibitors are mediated through the accumulation of GLP1 or if they are independent of GLP1 receptor activation and instead directly related to the inhibition of DPP4 activity.

Lots of studies have demonstrated that the anti-inflammatory potential of DPP-4 inhibitors could be associated with the accumulation of GLP-1 and GLP-2, the two main targets of DPP-4; GLP-1 and GLP-2 activate their receptors and increase intracellular cyclic adenosine monophosphate, which activates their downstream pathways involved in the inhibition of the neuroinflammatory response (Flock et al. 2007). Linagliptin has been implicated in mitigating Aβ-induced neurotoxicity through GLP-1-mediated pathways, evidenced by the upregulation of insulin and insulin-like growth factor-1 (IGF-1) gene expression (Sedighi et al. 2021). Ide et al. found that linagliptin improved cognitive impairment in streptozotocin-induced diabetic mice by reducing proinflammatory cytokines, as well as inhibiting microglial activation. These effects were observed independently of glucose-lowering, suggesting a direct neuroprotective effect of linagliptin beyond its antidiabetic properties. This neuroprotective effect occurs independently of changes in blood glucose levels, suggesting a GLP-1-mediated mechanism. Additionally, in a rotenone-induced rat model of PD, linagliptin treatment increased striatal dopamine and active GLP-1 levels in the brain, indicating GLP-1’s involvement in its neuroprotective effects. Moreover, linagliptin attenuated rotenone-induced alterations in inflammatory markers like TNF-α. These findings underscore the potential of linagliptin’s anti-inflammatory and neuroprotective actions mediated through GLP-1-dependent mechanisms.

On the other hand, there are evidences indicating that changes in the activity of DPP4 itself are associated with inflammatory responses, suggesting that the anti-inflammatory activity of DPP4 inhibitors could occur independently of GLP1 accumulation. In facts, DPP-4 expression is altered in inflammation and/or immunity-associated diseases and acts as a regulator of immune cells (Trzaskalski et al. 2020). DPP-4 is involved in the maturation and phenotypic differentiation of T-cells, and therefore, in immune regulation (Al-Badri et al. 2018). Ghorpade et al. (2018) demonstrated the interaction between the liver and visceral adipose tissues (VAT), wherein obesity triggers hepatocytes to produce DPP-4, contributing to inflammation in adipose tissue macrophages (ATMs). In obese mice, DPP-4 synthesis by hepatocytes, along with the plasma factor Xa, triggers inflammation in visceral ATMs. Moreover, silencing DPP-4 expression in hepatocytes alleviates VAT inflammation and insulin resistance (Ghorpade et al. 2018). Renal protection has been a focal point of DPP-4 inhibitor studies, revealing pathways independent of GLP-1. Notably, DPP-4 inhibitors such as linagliptin and vildagliptin have demonstrated renoprotective effects mediated by substrates beyond GLP-1 (Kanasaki et al. 2014; Shi et al. 2016). These include the upregulation of Stromal Cell-Derived Factor-1 (SDF-1), modulation of microRNAs (miR-29 and miR-200a), and downregulation of Transforming Growth Factor-β (TGF-β) signaling. It should be noted that while several studies have highlighted the anti-inflammatory effects of DPP-4 inhibitors in neurodegenerative diseases, there remains a lack of conclusive evidence regarding their actions through GLP-1 independent pathways. In conjunction with these findings, reports on the anti-inflammatory effects of DPP4 inhibitors in the brain suggest that the anti-inflammatory effects of DPP4 inhibitors in neuroinflammatory lesions may be attributable not only to GLP1 accumulation but also to the inhibition of DPP4 enzyme activity itself. However, further research is needed to elucidate the underlying mechanisms by which DPP4 enzyme activity inhibition specifically contributes to the regulation of inflammatory responses.

Multiple in vitro studies have demonstrated that DPP-4 inhibitors exhibit a neuroprotective effect on neuronal cells (Abdelsalam and Safar 2015; Wiciński et al. 2018; Zhang et al. 2020). When exposed to neurotoxic stimuli, such as oxidative stress, inflammation, or excitotoxicity, neuronal cells treated with DPP-4 inhibitors exhibited enhanced viability compared to untreated cells. This finding suggests a potential role for DPP-4 inhibitors in preserving neuronal health and countering neurodegenerative processes. The experimental findings indicating the neuroprotective of DPP-4 inhibitors in various neuronal cell damage models, including those related to neurodegenerative diseases, have been emphasized (Table 2). Both in vitro and in vivo studies using animal models have revealed that DPP-4 inhibitors possess antioxidant properties and can mitigate oxidative damage in neurons. DPP-4 inhibitors can protect neurons from oxidative injury by scavenging free radicals and enhancing endogenous antioxidant defenses, potentially slowing the progression of neurodegenerative disorders. Several preclinical studies employing animal models of cognitive impairment have shown that DPP-4 inhibitors can ameliorate cognitive deficits. These improvements in cognitive function are associated with increased synaptic plasticity, enhanced neurogenesis, and improved cerebral blood flow. Moreover, Sitagliptin was found to improve memory deficits in PD rats by upregulating brain-derived neurotrophic factor to prevent neuronal and dendritic spine loss (Li et al. 2018). All these findings suggest that DPP-4 inhibitors might attenuate the cognitive decline observed in neurodegenerative diseases.

Table 2 Neuroprotective effects of DPP-4 inhibitors

DrugModelEffectsReferences
Pathological changesMOA
LinagliptinSH-SY5Y cells treated Aβ1-42↑ Cell viability↓ Inflammation; ↓ TNF-α, IL-1β and IL-6, ↑ Wnt1, pCREB, and PKCεSedighi et al. (2021)
Rotenone mice (1.5 mg/kg, s.c., 21 days)↑ Locomotor activity
↑ DA level in striatum
↓ Oxidative stress; ↑ SOD, catalase and GSH in brain homogenate
↓ Inflammation; ↓ TNF-α level in brain homogenate
Abhangi and Patel (2022)
Rotenone rats (2 mg/kg, s.c., 28 days)↑ Locomotor activity
↓ Catalepsy
↓ Inflammation; ↓ TNF-α, IL-6, and HIF1-α, ↑ DJ-1 in the midbrain and striatum
↓ Apoptosis; ↓ caspase-3 in the midbrain and striatum
ElGamal et al. (2023)
Focal Ischemic Stroke in Hyperglycemic mice↓ Neuronal cell death
↑ Locomotor Activity
↓ Inflammation; ↓ Iba-1, IL-1β and IL-6
↓ Apoptosis; ↓ caspase-3 and Bcl-2, ↑ p-Akt, p-mTOR, and Bax
Zhang et al. (2020)
HFD TauP301S mice PS19 transgenic mice↑ Cognitive function
No significant correlation in phosphorylated tau in hippocampus
Nakaoku et al. (2019)
VildagliptinMPTP mice (30 mg/kg, i.p., 14 days)↑ Locomotor activity
↑ TH+ neurons in SNpc
↑ TH density in striatum
↓ Apoptosis; ↓ caspase-3 and Bax/Bcl-2 ratio, ↓ p-ERK/ERK and p-JNK/JNK in striatum
Autophagy; No significant in LC3B and LC3B-II in striatum and SNpc
Pariyar et al. (2022)
SH-SY5Y cells treated MPP+↑ Cell viability↓ Apoptosis; ↑ p-Akt/Akt, ↓ p-ERK/ERK and p-JNK/JNK
↓ Autophagy; ↓ LC3B-II
Rotenone rats (1.5 mg/kg, s.c., 21 days)↑ Locomotor activity
↑ DA in striatum
↓ Body weight loss
↓ Inflammation; ↓ MPO, ICAM, TNF-α and iNOS in striatum
↓ Apoptosis; ↓ caspase-3, cytochrome c, TBARS and Nrf2 in striatum
Abdelsalam and Safar (2015)
AlCl3 rats (100 mg/kg, oral, 60 days)↑ Locomotor activity
↑ Cognitive function
↓ Aβ1-42 in hippocampus
↓ Oxidative stress; ↓ FOXO1 in hippocampus
↓ Inflammation; ↓ TNF-α in hippocampus
↓ Apoptosis; ↓ Bax and caspase-3, ↑ Bcl-2 in hippocampus
↑ Klotho, p-AKT and p-ERK, ↓ STAT3 and JAK2 in hippocampus
Yossef et al. (2020)
AlogliptinRotenone rats (1.5 mg/kg, i.p., 21 days)↑ Locomotor activity
↑ TH density in SNpc
↓ Inflammasome; ↓ Iba-1, caspase-1, TLR-4, NLRP3, HMGB1 and IL-1β in striatumSafar et al. (2021)
SaxagliptinSTZ-induced AD rats (3 mg/kg, ICV, 3 days)↑ Cognitive function
↓ Aβ1-42 levels in hippocampus
↓ Total tau levels in hippocampus
↓ Inflammation; ↓ TNF-α and IL-1β in hippocampusKosaraju et al. (2013)
Aβ-induced AD rats (300 pmol/day, stereotaxic, 15 days)↑ Cognitive function↓ Oxidative stress; ↓ MDA, ↑ SOD, catalase, GSH, Nrf2 and HO-1Li et al. (2018)
OmarigliptinLPS rats (250 μg/kg, i.p., 7 days)No significant in locomotor activity
↑ Cognitive function
↓ Oxidative stress; ↑ Nrf2 and HO-1 in hippocampus
↓ Inflammation; ↓ TNF-α, IL-8, TGF-β1, TLR-4 and NLRP3, ↑ IL-10 in hippocampus
↑ Autophagy; ↓ p-AKT and GSK-3β, ↑ beclin-1 in hippocampus
Kabel et al. (2022)
SitagliptinRotenone rats (3 mg/kg,s.c., 10 days)↑ Locomotor activity
↑ TH+ neurons in SNpc
↑ DA level in striatum
↓ Apoptosis; ↓ Bax, ↑ Bcl-2 in SN
↑ GDNF level in SN
↓ Inflammation; ↓TGF-β1, IL-1β and IL-6 in SN
Badawi et al. (2017)
6-OHDA rats (8 μg/rat, stereotaxic)↑ Cognitive function
↑ TH levels in SN
↑ Spine density in hippocampal CA1
↑ BDNF level in brainLi et al. (2018)

This table provides an overview of the neuroprotective effects of DPP-4 inhibitors with underlying pathological and mechanical changes in PD in both animal cell lines models. For details and references, see main text. AD, Alzheimer’s disease; Aβ, amyloid β; Bcl-2, B cell lymphoma 2; CA1, cornu ammonis; CREB, cAMP-response element binding protein; DA, dopamine; ERK, extracellular signal-regulated kinase; FOXO1, forkhead box protein O1; GSH, glutathione; GSK-3β, glycogen synthase kinase 3 beta; HIF1-α, hypoxia-inducible factor 1-alpha; HFD, high fat diet; HMGB1, high-mobility group box 1; HO-1, heme oxygenase-1; Iba-1, ionized calcium-binding adapter molecule-1; ICAM, intercellular adhesion molecule-1; ICV, intracerebroventricular; IL-1β, interleukin-1β; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; JAK2, Janus kinase; JNK, c-Jun N-terminal kinase; LC3B, microtubule-associated protein 1A/1B-light chain 3B; LPS, lipopolysaccharide; MDA, malondialdehyde; MPO, Myeloperoxidase; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NLRP3, nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3; Nrf2, Nuclear factor erythroid-2-related factor 2; PKCε, protein kinase C; mTOR, mammalian target of rapamycin; SN, substantia nigra; SNpc, substantia nigra pars compacta; SOD, superoxide dismutase; STAT3, signal transducer and activator of transcription; STZ, streptozotocin; TBARS, thiobarbituric acid reactive substances; TGF-β1, transforming growth factor-β1; TH, tyrosine hydroxylase; TLR-4, toll-like receptor-4; TNF-a, tumor necrosis factor-α.


In several experimental studies using animal models, DPP-4 inhibitors have been shown to reduce neuroinflammation by suppressing microglial activation and inhibiting the production of pro-inflammatory cytokines in the brain. Accumulated evidence has revealed that DPP-4 inhibitors suppress neuroinflammation by reducing TNF-α and IL-1β levels, thereby contributing to the amelioration of the pathology associated with several neurodegenerative diseases. Exendin-4 and linagliptin treatments have induced microglial polarization in the anti-inflammatory M2 phenotype and reduced pro-inflammatory cytokine secretion in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced PD mouse model (Yu et al. 2023). Pretreatment with linagliptin also attenuates amyloid-β (Aβ)-induced inflammation and cytotoxicity by suppressing the release of IL-6 in human SH-SY5Y neuroblastoma cells exposed to Aβ cytotoxicity (Sedighi et al. 2021). Linagliptin dose-dependently improved cognitive impairment and reduced the levels of soluble Aβ (1-42), TNF-α, IL-1β, IL-6, acetylcholinesterase, and oxidative/nitrosative stress markers in a rat model of Alzheimer’s disease induced by intracerebroventricular streptozotocin treatment (Siddiqui et al. 2023). Vildagliptin effectively improves motor performance, reduces striatal dopamine depletion, and suppresses inflammatory mediators like NF-κB and TNF-α in a rat model of rotenone-induced PD (Abdelsalam and Safar 2015). Sitagliptin also reversed nigrostriatal degeneration, improved motor performance, and decreased the levels of IL-1β, IL-6, and TGF-β1 in the rat brain in a rotenone-induced model of PD (Badawi et al. 2017). Sitagliptin, both alone and in combination with pregabalin, alleviates acute epileptogenesis induced by pentylenetetrazole in mice. The findings demonstrated that sitagliptin exerted substantial anti-inflammatory effects by modulating the levels of inflammatory factors like Nrf2 and NF-κb (Nader et al. 2018). Omarigliptin exhibits anti-inflammatory effects by inhibiting NF-κB activation, leading to a reduction in nitric oxide (NO) production and the expression of inducible NO synthase (iNOS) in a model of neurotoxin-induced toxicity in PC12 cells, which mimics PD (Gouda and Cho 2022).

Preclinical studies have shown that DPP-4 inhibitors may protect dopaminergic neurons from degeneration, enhance neuroplasticity, and attenuate neuroinflammation (Table 2). These findings have sparked interest in exploring the therapeutic potential of DPP-4 inhibitors in PD, focusing on dopaminergic neuron degeneration.

Numerous clinical trials have assessed the neurorestorative potential of antidiabetic drugs in preclinical models of PD, yielding positive outcomes. The most extensively reported potential for repurposing antidiabetic drugs as treatment for PD lies in GLP1 receptor agonists. Specifically, clinical trial (NCT01971242) (University College London, 2016) are underway for exenatide, a GLP-1 receptor agonist, as a treatment for PD. These clinical trials have evaluated the impact of exenatide in patients with PD, utilizing a randomized, double-blind, and placebo-controlled design, wherein participants received weekly subcutaneous injections of either exenatide or a placebo alongside their standard PD treatment (Athauda et al. 2017). The outcomes of these trials provided insights into whether the neuroprotective effects of exenatide, previously observed in preclinical models, could translate into positive outcomes in a clinical setting (Athauda et al. 2017). The therapeutic potential of GLP-1 agonist lixisenatide in PD has also garnered significant interest following promising results from clinical trials (NCT03439943) (University College London, 2023). Participants receiving lixisenatide showed less disability, as assessed by the Movement Disorder Society-Unified Parkinson’s disease rating scale (MDS-UPDRS), compared to those receiving placebo. Importantly, improvements in the MDS-UPDRS scores were observed in both ON and OFF states, indicating a potential disease-modifying effect of lixisenatide beyond the amplification of pharmacological impacts of existing medications. Lin et al. reported that diabetic patients treated with dipeptidyl peptidase-4 (DPP-4) inhibitors, particularly vildagliptin, exhibited a significantly lower risk of PD compared to those using other oral antidiabetic drugs (Lin et al. 2023). Additionally, Jeong et al. demonstrated that treatment with DPP-4 inhibitors in diabetic patients with PD was associated with higher baseline nigrostriatal dopamine transporter availability and slower longitudinal increase in levodopa-equivalent dose, suggesting potential beneficial effects on motor outcomes in this population (Jeong et al. 2021). Furthermore, given that DPP-4 inhibitors are small molecules, they are more practical for use in patients with PD compared to larger peptide-based molecules like GLP-1 receptor agonists. Despite their mechanism primarily involving the augmentation of GLP-1 signaling, which may confer anti-inflammatory effects, the inhibition of DPP-4 itself could also contribute to additional anti-inflammatory effects. This dual mechanism suggests that the use of DPP-4 inhibitors in PD patients may offer comprehensive neuroprotective effects.

In summary, the current state of therapeutics for PD highlights a significant demand for innovative disease-modifying treatments. This review discusses the current understanding of the anti-inflammatory properties of DPP-4 inhibitors and their potential as a novel therapeutic strategy for PD (Fig. 1). Elucidating the mechanisms through which DPP-4 inhibitors influence neuroinflammation may offer new prospects for disease-modifying therapies in PD. However, it is crucial to conduct further research, including clinical trials, to confirm the validity of the preclinical findings discussed in this review and ascertain the safety and effectiveness of DPP-4 inhibitors in treating PD.

Figure 1.Role of DPP4 inhibitors in neuroinflammation.
  1. Abhangi KV, Patel JI (2022) Neuroprotective effects of linagliptin in a rotenone-induced rat model of Parkinson's disease. Indian J Pharmacol 54:46-50. doi: 10.4103/ijp.IJP_384_20
    Pubmed KoreaMed CrossRef
  2. Abdelsalam RM, Safar MM (2015) Neuroprotective effects of vildagliptin in rat rotenone Parkinson's disease model: role of RAGE-NFκB and Nrf2-antioxidant signaling pathways. J Neurochem 133:700-707. doi: 10.1111/jnc.13087
    Pubmed CrossRef
  3. Aertgeerts K, Ye S, Tennant MG, Kraus ML, Rogers J, Sang BC, Skene RJ, Webb DR, Prasad GS (2004) Crystal structure of human dipeptidyl peptidase IV in complex with a decapeptide reveals details on substrate specificity and tetrahedral intermediate formation. Protein Sci 13:412-421. doi: 10.1110/ps.03460604
    Pubmed KoreaMed CrossRef
  4. Akira S, Uematsu S, Takeuchi O (2006) Pathogen recognition and innate immunity. Cell 124:783-801. doi: 10.1016/j.cell.2006.02.015
    Pubmed CrossRef
  5. Al-Badri G, Leggio GM, Musumeci G, Marzagalli R, Drago F, Castorina A (2018) Tackling dipeptidyl peptidase IV in neurological disorders. Neural Regen Res 13:26-34. doi: 10.4103/1673-5374.224365
    Pubmed KoreaMed CrossRef
  6. Athauda D, Maclagan K, Skene SS, Bajwa-Joseph M, Letchford D, Chowdhury K, Hibbert S, Budnik N, Zampedri L, Dickson J, Li Y, Aviles-Olmos I, Warner TT, Limousin P, Lees AJ, Greig NH, Tebbs S, Foltynie T (2017) Exenatide once weekly versus placebo in Parkinson's disease: a randomised, double-blind, placebo-controlled trial. Lancet 390:1664-1675. doi: 10.1016/S0140-6736(17)31585-4
    Pubmed CrossRef
  7. Badawi GA, Abd El Fattah MA, Zaki HF, El Sayed MI (2017) Sitagliptin and liraglutide reversed nigrostriatal degeneration of rodent brain in rotenone-induced Parkinson's disease. Inflammopharmacology 25:369-382. doi: 10.1007/s10787-017-0331-6
    Pubmed CrossRef
  8. Bang Y, Lim J, Choi HJ (2021) Recent advances in the pathology of prodromal non-motor symptoms olfactory deficit and depression in Parkinson's disease: clues to early diagnosis and effective treatment. Arch Pharm Res 44:588-604. doi: 10.1007/s12272-021-01337-3
    Pubmed KoreaMed CrossRef
  9. Bartels AL, Willemsen AT, Doorduin J, de Vries EF, Dierckx RA, Leenders KL (2010) [11C]-PK11195 PET: quantification of neuroinflammation and a monitor of anti-inflammatory treatment in Parkinson's disease?. Parkinsonism Relat Disord 16:57-59. doi: 10.1016/j.parkreldis.2009.05.005
    Pubmed CrossRef
  10. Bieri G, Brahic M, Bousset L, Couthouis J, Kramer NJ, Ma R, Nakayama L, Monbureau M, Defensor E, Schüle B, Shamloo M, Melki R, Gitler AD (2019) LRRK2 modifies α-syn pathology and spread in mouse models and human neurons. Acta Neuropathol 137:961-980. doi: 10.1007/s00401-019-01995-0
    Pubmed KoreaMed CrossRef
  11. Blum-Degen D, Müller T, Kuhn W, Gerlach M, Przuntek H, Riederer P (1995) Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer's and de novo Parkinson's disease patients. Neurosci Lett 202:17-20. doi: 10.1016/0304-3940(95)12192-7
    Pubmed CrossRef
  12. Cai L, Gao L, Zhang G, Zeng H, Wu X, Tan X, Qian C, Chen G (2022) DJ-1 alleviates neuroinflammation and the related blood-spinal cord barrier destruction by suppressing NLRP3 inflammasome activation via SOCS1/Rac1/ROS pathway in a rat model of traumatic spinal cord injury. J Clin Med 11:3716. doi: 10.3390/jcm11133716
    Pubmed KoreaMed CrossRef
  13. Choi DY, Zhang J, Bing G (2010) Aging enhances the neuroinflammatory response and alpha-synuclein nitration in rats. Neurobiol Aging 31:1649-1653. doi: 10.1016/j.neurobiolaging.2008.09.010
    Pubmed CrossRef
  14. Coll RC, Robertson AA, Chae JJ, Higgins SC, Muñoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS, Haneklaus M, Sutton CE, Núñez G, Latz E, Kastner DL, Mills KH, Masters SL, Schroder K, Cooper MA, O'Neill LA (2015) A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med 21:248-255. doi: 10.1038/nm.3806
    Pubmed KoreaMed CrossRef
  15. Cookson MR (2017) Mechanisms of mutant LRRK2 neurodegeneration. Adv Neurobiol 14:227-239. doi: 10.1007/978-3-319-49969-7_12
    Pubmed CrossRef
  16. Croisier E, Moran LB, Dexter DT, Pearce RK, Graeber MB (2005) Microglial inflammation in the parkinsonian substantia nigra: relationship to alpha-synuclein deposition. J Neuroinflammation 2:14. doi: 10.1186/1742-2094-2-14
    Pubmed KoreaMed CrossRef
  17. Dai Y, Dai D, Wang X, Ding Z, Mehta JL (2014) DPP-4 inhibitors repress NLRP3 inflammasome and interleukin-1beta via GLP-1 receptor in macrophages through protein kinase C pathway. Cardiovasc Drugs Ther 28:425-432. doi: 10.1007/s10557-014-6539-4
    Pubmed CrossRef
  18. Doot RK, Young AJ, Nasrallah IM, Wetherill RR, Siderowf A, Mach RH, Dubroff JG (2022) [18F]NOS PET brain imaging suggests elevated neuroinflammation in idiopathic Parkinson's disease. Cells 11:3081. doi: 10.3390/cells11193081
    Pubmed KoreaMed CrossRef
  19. ElGamal RZ, Tadros MG, Menze ET (2023) Linagliptin counteracts rotenone's toxicity in non-diabetic rat model of Parkinson's disease: insights into the neuroprotective roles of DJ-1, SIRT-1/Nrf-2 and implications of HIF1-α. Eur J Pharmacol 941:175498. doi: 10.1016/j.ejphar.2023.175498
    Pubmed CrossRef
  20. Ferrari CC, Pott Godoy MC, Tarelli R, Chertoff M, Depino AM, Pitossi FJ (2006) Progressive neurodegeneration and motor disabilities induced by chronic expression of IL-1beta in the substantia nigra. Neurobiol Dis 24:183-193. doi: 10.1016/j.nbd.2006.06.013
    Pubmed CrossRef
  21. Filiano AJ, Gadani SP, Kipnis J (2015) Interactions of innate and adaptive immunity in brain development and function. Brain Res 1617:18-27. doi: 10.1016/j.brainres.2014.07.050
    Pubmed KoreaMed CrossRef
  22. Flock G, Baggio LL, Longuet C, Drucker DJ (2007) Incretin receptors for glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide are essential for the sustained metabolic actions of vildagliptin in mice. Diabetes 56:3006-3013. doi: 10.2337/db07-0697
    Pubmed CrossRef
  23. Franchi L, Eigenbrod T, Muñoz-Planillo R, Nuñez G (2009) The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol 10:241-247. doi: 10.1038/ni.1703
    Pubmed KoreaMed CrossRef
  24. Gaig C, Vilas D, Infante J, Sierra M, García-Gorostiaga I, Buongiorno M, Ezquerra M, Martí MJ, Valldeoriola F, Aguilar M, Calopa M, Hernandez-Vara J, Tolosa E (2014) Nonmotor symptoms in LRRK2 G2019S associated Parkinson's disease. PLoS One 9:e108982. doi: 10.1371/journal.pone.0108982
    Pubmed KoreaMed CrossRef
  25. Ghorpade DS, Ozcan L, Zheng Z, Nicoloro SM, Shen Y, Chen E, Blüher M, Czech MP, Tabas I (2018) Hepatocyte-secreted DPP4 in obesity promotes adipose inflammation and insulin resistance. Nature 555:673-677. doi: 10.1038/nature26138
    Pubmed KoreaMed CrossRef
  26. Gouda NA, Cho J (2022) Omarigliptin mitigates 6-hydroxydopamine- or rotenone-induced oxidative toxicity in PC12 cells by antioxidant, anti-inflammatory, and anti-apoptotic actions. Antioxidants (Basel) 11:1940. doi: 10.3390/antiox11101940
    Pubmed KoreaMed CrossRef
  27. Green BD, Irwin N, Flatt PR (2006) Pituitary adenylate cyclase-activating peptide (PACAP): assessment of dipeptidyl peptidase IV degradation, insulin-releasing activity and antidiabetic potential. Peptides 27:1349-1358. doi: 10.1016/j.peptides.2005.11.010
    Pubmed CrossRef
  28. Heneka MT, McManus RM, Latz E (2018) Inflammasome signalling in brain function and neurodegenerative disease. Nat Rev Neurosci 19:610-621. doi: 10.1038/s41583-018-0055-7. Erratum in: (2019) Nat Rev Neurosci 20:187. doi: 10.1038/s41583-019-0137-1
    Pubmed CrossRef
  29. Hirsch EC, Hunot S (2009) Neuroinflammation in Parkinson's disease: a target for neuroprotection?. Lancet Neurol 8:382-397. doi: 10.1016/S1474-4422(09)70062-6
    Pubmed CrossRef
  30. Ide M, Sonoda N, Inoue T, Kimura S, Minami Y, Makimura H, Hayashida E, Hyodo F, Yamato M, Takayanagi R, Inoguchi T (2020) The dipeptidyl peptidase-4 inhibitor, linagliptin, improves cognitive impairment in streptozotocin-induced diabetic mice by inhibiting oxidative stress and microglial activation. PLoS One 15:e0228750. doi: 10.1371/journal.pone.0228750
    Pubmed KoreaMed CrossRef
  31. Jeong SH, Chung SJ, Yoo HS, Hong N, Jung JH, Baik K, Lee YH, Sohn YH, Lee PH (2021) Beneficial effects of dipeptidyl peptidase-4 inhibitors in diabetic Parkinson's disease. Brain 144:1127-1137. doi: 10.1093/brain/awab015
    Pubmed CrossRef
  32. Kabel AM, Arab HH, Atef A, Estfanous RS (2022) Omarigliptin/galangin combination mitigates lipopolysaccharide-induced neuroinflammation in rats: involvement of glucagon-like peptide-1, toll-like receptor-4, apoptosis and Akt/GSK-3β signaling. Life Sci 295:120396. doi: 10.1016/j.lfs.2022.120396
    Pubmed CrossRef
  33. Kanasaki K, Shi S, Kanasaki M, He J, Nagai T, Nakamura Y, Ishigaki Y, Kitada M, ivastava SP Sr, Koya D (2014) Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes 63:2120-2131. doi: 10.2337/db13-1029
    Pubmed CrossRef
  34. Kang Y, Mozley PD, Verma A, Schlyer D, Henchcliffe C, Gauthier SA, Chiao PC, He B, Nikolopoulou A, Logan J, Sullivan JM, Pryor KO, Hesterman J, Kothari PJ, Vallabhajosula S (2018) Noninvasive PK11195-PET image analysis techniques can detect abnormal cerebral microglial activation in Parkinson's disease. J Neuroimaging 28:496-505. doi: 10.1111/jon.12519
    Pubmed KoreaMed CrossRef
  35. Klein T, Fujii M, Sandel J, Shibazaki Y, Wakamatsu K, Mark M, Yoneyama H (2014) Linagliptin alleviates hepatic steatosis and inflammation in a mouse model of non-alcoholic steatohepatitis. Med Mol Morphol 47:137-149. doi: 10.1007/s00795-013-0053-9
    Pubmed CrossRef
  36. Kong L, Deng J, Zhou X, Cai B, Zhang B, Chen X, Chen Z, Wang W (2021) Sitagliptin activates the p62-Keap1-Nrf2 signalling pathway to alleviate oxidative stress and excessive autophagy in severe acute pancreatitis-related acute lung injury. Cell Death Dis 12:928. doi: 10.1038/s41419-021-04227-0
    Pubmed KoreaMed CrossRef
  37. Kosaraju J, Gali CC, Khatwal RB, Dubala A, Chinni S, Holsinger RM, Madhunapantula VS, Muthureddy Nataraj SK, Basavan D (2013) Saxagliptin: a dipeptidyl peptidase-4 inhibitor ameliorates streptozotocin induced Alzheimer's disease. Neuropharmacology 72:291-300. doi: 10.1016/j.neuropharm.2013.04.008
    Pubmed CrossRef
  38. Lavisse S, Goutal S, Wimberley C, Tonietto M, Bottlaender M, Gervais P, Kuhnast B, Peyronneau MA, Barret O, Lagarde J, Sarazin M, Hantraye P, Thiriez C, Remy P (2021) Increased microglial activation in patients with Parkinson disease using [18F]-DPA714 TSPO PET imaging. Parkinsonism Relat Disord 82:29-36. doi: 10.1016/j.parkreldis.2020.11.011
    Pubmed CrossRef
  39. Lee E, Hwang I, Park S, Hong S, Hwang B, Cho Y, Son J, Yu JW (2019) MPTP-driven NLRP3 inflammasome activation in microglia plays a central role in dopaminergic neurodegeneration. Cell Death Differ 26:213-228. doi: 10.1038/s41418-018-0124-5
    Pubmed KoreaMed CrossRef
  40. Li J, Zhang S, Li C, Li M, Ma L (2018) Sitagliptin rescues memory deficits in Parkinsonian rats via upregulating BDNF to prevent neuron and dendritic spine loss. Neurol Res 40:736-743. doi: 10.1080/01616412.2018.1474840
    Pubmed CrossRef
  41. Li Y, Tian Q, Li Z, Dang M, Lin Y, Hou X (2019) Activation of Nrf2 signaling by sitagliptin and quercetin combination against β-amyloid induced Alzheimer's disease in rats. Drug Dev Res 80:837-845. doi: 10.1002/ddr.21567
    Pubmed CrossRef
  42. Lim J, Bang Y, Choi HJ (2018) Abnormal hippocampal neurogenesis in Parkinson's disease: relevance to a new therapeutic target for depression with Parkinson's disease. Arch Pharm Res 41:943-954. doi: 10.1007/s12272-018-1063-x
    Pubmed CrossRef
  43. Lin YH, Hsu CC, Liu JS, Chang KC, Huang JA (2023) Use of dipeptidyl peptidase-4 inhibitors was associated with a lower risk of Parkinson's disease in diabetic patients. Sci Rep 13:22489. doi: 10.1038/s41598-023-49870-z
    Pubmed KoreaMed CrossRef
  44. Liu SY, Qiao HW, Song TB, Liu XL, Yao YX, Zhao CS, Barret O, Xu SL, Cai YN, Tamagnan GD, Sossi V, Lu J, Chan P (2022) Brain microglia activation and peripheral adaptive immunity in Parkinson's disease: a multimodal PET study. J Neuroinflammation 19:209. doi: 10.1186/s12974-022-02574-z
    Pubmed KoreaMed CrossRef
  45. Lonnemann N, Hosseini S, Marchetti C, Skouras DB, Stefanoni D, D'Alessandro A, Dinarello CA, Korte M (2020) The NLRP3 inflammasome inhibitor OLT1177 rescues cognitive impairment in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 117:32145-32154. doi: 10.1073/pnas.2009680117
    Pubmed KoreaMed CrossRef
  46. McGeer PL, Itagaki S, Boyes BE, McGeer EG (1988) Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology 38:1285-1291. doi: 10.1212/wnl.38.8.1285
    Pubmed CrossRef
  47. Moehle MS, Webber PJ, Tse T, Sukar N, Standaert DG, DeSilva TM, Cowell RM, West AB (2012) LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci 32:1602-1611. doi: 10.1523/JNEUROSCI.5601-11.2012. Erratum in: (2022) J Neurosci 42:938. doi: 10.1523/JNEUROSCI.2507-21.2021
    Pubmed KoreaMed CrossRef
  48. Moon SH, Kwon Y, Huh YE, Choi HJ (2022) Trehalose ameliorates prodromal non-motor deficits and aberrant protein accumulation in a rotenone-induced mouse model of Parkinson's disease. Arch Pharm Res 45:417-432. doi: 10.1007/s12272-022-01386-2
    Pubmed CrossRef
  49. Moustafa AA, Chakravarthy S, Phillips JR, Gupta A, Keri S, Polner B, Frank MJ, Jahanshahi M (2016) Motor symptoms in Parkinson's disease: a unified framework. Neurosci Biobehav Rev 68:727-740. doi: 10.1016/j.neubiorev.2016.07.010
    Pubmed CrossRef
  50. Nader MA, Ateyya H, El-Shafey M, El-Sherbeeny NA (2018) Sitagliptin enhances the neuroprotective effect of pregabalin against pentylenetetrazole-induced acute epileptogenesis in mice: Implication of oxidative, inflammatory, apoptotic and autophagy pathways. Neurochem Int 115:11-23. doi: 10.1016/j.neuint.2017.10.006
    Pubmed CrossRef
  51. Nakamura K, Sakai S, Tsuyama J, Nakamura A, Otani K, Kurabayashi K, Yogiashi Y, Masai H, Shichita T (2021) Extracellular DJ-1 induces sterile inflammation in the ischemic brain. PLoS Biol 19:e3000939. doi: 10.1371/journal.pbio.3000939
    Pubmed KoreaMed CrossRef
  52. Nakaoku Y, Saito S, Yamamoto Y, Maki T, Takahashi R, Ihara M (2019) The dipeptidyl peptidase-4 inhibitor linagliptin ameliorates high-fat induced cognitive decline in tauopathy model mice. Int J Mol Sci 20:2539. doi: 10.3390/ijms20102539
    Pubmed KoreaMed CrossRef
  53. Panicker N, Kam TI, Wang H, Neifert S, Chou SC, Kumar M, Brahmachari S, Jhaldiyal A, Hinkle JT, Akkentli F, Mao X, Xu E, Karuppagounder SS, Hsu ET, Kang SU, Pletnikova O, Troncoso J, Dawson VL, Dawson TM (2022) Neuronal NLRP3 is a parkin substrate that drives neurodegeneration in Parkinson's disease. Neuron 110:2422-2437.e9. doi: 10.1016/j.neuron.2022.05.009
    Pubmed KoreaMed CrossRef
  54. Pariyar R, Bastola T, Lee DH, Seo J (2022) Neuroprotective effects of the DPP4 inhibitor vildagliptin in in vivo and in vitro models of Parkinson's disease. Int J Mol Sci 23:2388. doi: 10.3390/ijms23042388
    Pubmed KoreaMed CrossRef
  55. Peng L, Zhou Y, Jiang N, Wang T, Zhu J, Chen Y, Li L, Zhang J, Yu S, Zhao Y (2020) DJ-1 exerts anti-inflammatory effects and regulates NLRX1-TRAF6 via SHP-1 in stroke. J Neuroinflammation 17:81. doi: 10.1186/s12974-020-01764-x
    Pubmed KoreaMed CrossRef
  56. Russo I, Berti G, Plotegher N, Bernardo G, Filograna R, Bubacco L, Greggio E (2015) Leucine-rich repeat kinase 2 positively regulates inflammation and down-regulates NF-κB p50 signaling in cultured microglia cells. J Neuroinflammation 12:230. doi: 10.1186/s12974-015-0449-7. Erratum in: (2016) J Neuroinflammation 13:70. doi: 10.1186/s12974-016-0535-5
    Pubmed KoreaMed CrossRef
  57. Safar MM, Abdelkader NF, Ramadan E, Kortam MA, Mohamed AF (2021) Novel mechanistic insights towards the repositioning of alogliptin in Parkinson's disease. Life Sci 287:120132. doi: 10.1016/j.lfs.2021.120132
    Pubmed CrossRef
  58. Sedighi M, Baluchnejadmojarad T, Roghani M (2021) Linagliptin protects human SH-SY5Y neuroblastoma cells against amyloid-β cytotoxicity via the activation of Wnt1 and suppression of IL-6 release. Iran Biomed J 25:343-348. doi: 10.52547/ibj.25.5.343
    CrossRef
  59. Shi S, Kanasaki K, Koya D (2016) Linagliptin but not Sitagliptin inhibited transforming growth factor-β2-induced endothelial DPP-4 activity and the endothelial-mesenchymal transition. Biochem Biophys Res Commun 471:184-190. doi: 10.1016/j.bbrc.2016.01.154
    Pubmed CrossRef
  60. Shirakawa J, Amo K, Ohminami H, Orime K, Togashi Y, Ito Y, Tajima K, Koganei M, Sasaki H, Takeda E, Terauchi Y (2011) Protective effects of dipeptidyl peptidase-4 (DPP-4) inhibitor against increased β cell apoptosis induced by dietary sucrose and linoleic acid in mice with diabetes. J Biol Chem 286:25467-25476. doi: 10.1074/jbc.M110.217216
    Pubmed KoreaMed CrossRef
  61. Siddiqui N, Ali J, Parvez S, Najmi AK, Akhtar M (2023) Neuroprotective role of DPP-4 inhibitor Linagliptin against neurodegeneration, neuronal insulin resistance and neuroinflammation induced by intracerebroventricular streptozotocin in rat model of Alzheimer's disease. Neurochem Res 48:2714-2730. doi: 10.1007/s11064-023-03924-w
    Pubmed CrossRef
  62. Sliter DA, Martinez J, Hao L, Chen X, Sun N, Fischer TD, Burman JL, Li Y, Zhang Z, Narendra DP, Cai H, Borsche M, Klein C, Youle RJ (2018) Parkin and PINK1 mitigate STING-induced inflammation. Nature 561:258-262. doi: 10.1038/s41586-018-0448-9
    Pubmed KoreaMed CrossRef
  63. Troncoso-Escudero P, Parra A, Nassif M, Vidal RL (2018) Outside in: unraveling the role of neuroinflammation in the progression of Parkinson's disease. Front Neurol 9:860. doi: 10.3389/fneur.2018.00860
    Pubmed KoreaMed CrossRef
  64. Trzaskalski NA, Fadzeyeva E, Mulvihill EE (2020) Dipeptidyl peptidase-4 at the interface between inflammation and metabolism. Clin Med Insights Endocrinol Diabetes 13:1179551420912972. doi: 10.1177/1179551420912972
    Pubmed KoreaMed CrossRef
  65. University College, London (2016) Trial of Exenatide for Parkinson’s disease. National Library of Medicine. https://classic.clinicaltrials.gov/ct2/show/NCT01971242 Accessed 1 December 2023.
  66. University Hospital, Toulouse (2023) Study to evaluate the effect of lixisenatide in patient with Parkinson’s disease (LixiPark). National Library of Medicine. https://clinicaltrials.gov/study/NCT03439943 Accessed 1 December 2023.
  67. Vaillancourt DE, Mitchell T (2020) Parkinson's disease progression in the substantia nigra: location, location, location. Brain 143:2628-2630. doi: 10.1093/brain/awaa252
    Pubmed KoreaMed CrossRef
  68. Vroon A, Drukarch B, Bol JG, Cras P, Brevé JJ, Allan SM, Relton JK, Hoogland PV, Van Dam AM (2007) Neuroinflammation in Parkinson's patients and MPTP-treated mice is not restricted to the nigrostriatal system: microgliosis and differential expression of interleukin-1 receptors in the olfactory bulb. Exp Gerontol 42:762-771. doi: 10.1016/j.exger.2007.04.010
    Pubmed CrossRef
  69. Wiciński M, Górski K, Wódkiewicz E, Walczak M, Nowaczewska M, Malinowski B (2020) Vasculoprotective effects of vildagliptin. Focus on atherogenesis. Int J Mol Sci 21:2275. doi: 10.3390/ijms21072275
    Pubmed KoreaMed CrossRef
  70. Wiciński M, Wódkiewicz E, Słupski M, Walczak M, Socha M, Malinowski B, Pawlak-Osińska K (2018) Neuroprotective activity of sitagliptin via reduction of neuroinflammation beyond the incretin effect: focus on Alzheimer's disease. Biomed Res Int 2018:6091014. doi: 10.1155/2018/6091014
    Pubmed KoreaMed CrossRef
  71. Xie D, Wang Q, Huang W, Zhao L (2023) Dipeptidyl-peptidase-4 inhibitors have anti-inflammatory effects in patients with type 2 diabetes. Eur J Clin Pharmacol 79:1291-1301. doi: 10.1007/s00228-023-03541-0
    Pubmed CrossRef
  72. Xu E, Boddu R, Abdelmotilib HA, Sokratian A, Kelly K, Liu Z, Bryant N, Chandra S, Carlisle SM, Lefkowitz EJ, Harms AS, Benveniste EN, Yacoubian TA, Volpicelli-Daley LA, Standaert DG, West AB (2022) Pathological α-synuclein recruits LRRK2 expressing pro-inflammatory monocytes to the brain. Mol Neurodegener 17:7. doi: 10.1186/s13024-021-00509-5
    Pubmed KoreaMed CrossRef
  73. Xue Y, Li R, Fang P, Ye ZQ, Zhao Y, Zhou Y, Zhang KQ, Li L (2021) NLRP3 inflammasome inhibitor cucurbitacin B suppresses gout arthritis in mice. J Mol Endocrinol 67:27-40. doi: 10.1530/JME-20-0305
    Pubmed CrossRef
  74. Yossef RR, Al-Yamany MF, Saad MA, El-Sahar AE (2020) Neuroprotective effects of vildagliptin on drug induced Alzheimer's disease in rats with metabolic syndrome: role of hippocampal klotho and AKT signaling pathways. Eur J Pharmacol 889:173612. doi: 10.1016/j.ejphar.2020.173612
    Pubmed CrossRef
  75. Yu HY, Sun T, Wang Z, Li H, Xu D, An J, Wen LL, Li JY, Li W, Feng J (2023) Exendin-4 and linagliptin attenuate neuroinflammation in a mouse model of Parkinson's disease. Neural Regen Res 18:1818-1826. doi: 10.4103/1673-5374.360242
    Pubmed KoreaMed CrossRef
  76. Zeng W, Wu D, Sun Y, Suo Y, Yu Q, Zeng M, Gao Q, Yu B, Jiang X, Wang Y (2021) The selective NLRP3 inhibitor MCC950 hinders atherosclerosis development by attenuating inflammation and pyroptosis in macrophages. Sci Rep 11:19305. doi: 10.1038/s41598-021-98437-3
    Pubmed KoreaMed CrossRef
  77. Zhang G, Kim S, Gu X, Yu SP, Wei L (2020) DPP-4 inhibitor linagliptin is neuroprotective in hyperglycemic mice with stroke via the AKT/mTOR pathway and anti-apoptotic effects. Neurosci Bull 36:407-418. doi: 10.1007/s12264-019-00446-w
    Pubmed KoreaMed CrossRef
  78. Zhou J, Yang R, Zhang Z, Liu Q, Zhang Y, Wang Q, Yuan H (2019a) Mitochondrial protein PINK1 positively regulates RLR signaling. Front Immunol 10:1069. doi: 10.3389/fimmu.2019.01069
    Pubmed KoreaMed CrossRef
  79. Zhou X, Wang W, Wang C, Zheng C, Xu X, Ni X, Hu S, Cai B, Sun L, Shi K, Chen B, Zhou M, Chen G (2019b) DPP4 inhibitor attenuates severe acute pancreatitis-associated intestinal inflammation via Nrf2 signaling. Oxid Med Cell Longev 2019:6181754. doi: 10.1155/2019/6181754
    Pubmed KoreaMed CrossRef
  80. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, Stoessl AJ, Pfeiffer RF, Patenge N, Carbajal IC, Vieregge P, Asmus F, Müller-Myhsok B, Dickson DW, Meitinger T, Strom TM, Wszolek ZK, Gasser T (2004) Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 44:601-607. doi: 10.1016/j.neuron.2004.11.005
    Pubmed CrossRef

Article

Review

DTT 2024; 3(1): 83-93

Published online March 31, 2024 https://doi.org/10.58502/DTT.24.0030

Copyright © The Pharmaceutical Society of Korea.

Anti-Inflammatory Effects of Dipeptidyl Peptidase-4 Inhibitors and Their Therapeutic Application for Parkinson's Disease

Yeojin Bang , Soung-Hee Moon, Sumin Lee, Hyun Jin Choi

College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea

Correspondence to:Hyun Jin Choi, hjchoi3@cha.ac.kr

Received: October 31, 2023; Revised: February 26, 2024; Accepted: February 26, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Parkinson’s disease (PD) is characterized by a progressive loss of dopaminergic neurons, leading to a decline in dopamine levels and the manifestation of motor and non-motor symptoms. The current treatments primarily focus on symptom management, leaving the need for disease-modifying therapies unmet. Emerging research has shown that neuroinflammation plays a pivotal role in the pathogenesis and progression of PD. Dipeptidyl peptidase-4 (DPP-4) inhibitors, originally developed for diabetes treatment, represent a novel avenue of investigation in the context of PD treatment. Beyond their established role in glycemic control, DPP-4 inhibitors have shown promise in modulating inflammatory responses within the central nervous system. This indicates their potential application in controlling neuroinflammation in PD, which in turn, could impact disease progression and neurodegeneration. This review summarized the current understanding of the role of DPP-4 inhibitors in neuroinflammation and their potential to modulate PD progression. Understanding the intricate interplay between neuroinflammation and dopaminergic neuron degeneration could pave the way for a new era of disease-modifying strategies, and DPP-4 inhibitors could offer a potential avenue to address both inflammation and neurodegeneration in PD.

Keywords: dipeptidyl peptidase-4 inhibitors, neuroinflammation, neuroprotection, parkinson&rsquo,s disease

Introduction

Parkinson’s disease (PD) is a complex neurodegenerative disorder characterized by a progressive loss of dopaminergic neurons in the substantia nigra and a significant reduction in dopamine levels in the striatum of the brain (Vaillancourt and Mitchell 2020), ultimately leading to the development of both non-motor and motor symptoms (Bang et al. 2021). This dopaminergic deficit is responsible for the motor symptoms observed in PD, such as resting tremors, bradykinesia (slowness of movement), rigidity, and postural instability (Moustafa et al. 2016).

Available PD treatments primarily target the modulation of dopamine signaling to alleviate motor symptoms and improve the quality of life for patients. Levodopa, the precursor of dopamine, is commonly prescribed to replenish dopamine levels in the brain, providing symptomatic relief (Ide et al. 2020). Additionally, dopamine agonists, monoamine oxidase inhibitors, and catechol-O-methyl transferase inhibitors are utilized to enhance dopaminergic activity or reduce dopamine metabolism. Despite their effectiveness in managing motor symptoms, these agents do not halt the progressive dopaminergic neurodegeneration at the core of PD. In addition, patients often experience motor fluctuations and dyskinesia as the disease progresses, which significantly challenges the management of symptoms.

Given the central role of dopaminergic neurodegeneration in PD, researchers and clinicians have sought to develop disease-modifying treatments that can decelerate or halt the process. Despite significant efforts and a better understanding of the pathogenesis of PD, the development of effective disease-modifying therapies remains challenging. Various therapeutic approaches targeting pathways involved in protein aggregation, mitochondrial function, neuroinflammation, adult neurogenesis, and neuroprotection have been explored (Lim et al. 2018; Troncoso-Escudero et al. 2018; Moon et al. 2022). Among these, emerging research has highlighted the intricate role of neuroinflammation in the pathogenesis of PD. Neuroinflammation, which involves the activation of glial cells and release of pro-inflammatory cytokines, has garnered increasing attention due to its potential contribution to neuronal damage and PD progression. With the growing recognition of the involvement of neuroinflammation in PD, the development of drugs capable of regulating neuroinflammation to manage PD has gained traction.

This review aims to provide a comprehensive overview of the role of neuroinflammation in PD pathology and highlight the importance of developing inflammation-modulating drugs to treat PD. By understanding the intricate interplay between inflammation and neurodegeneration, we can better address the unmet need for disease-modifying therapies in PD management.

Neuroinflammation in PD

Emerging evidence from studies based on animal models and humans suggests that the immune system and neuroinflammation play crucial roles in the pathogenesis of PD (Table 1). Imaging techniques, such as positron emission tomography, have enabled the visualization of neuroinflammation in patients with PD, providing valuable insights into disease progression (Bartels et al. 2010; Kang et al. 2018; Lavisse et al. 2021; Doot et al. 2022).

Table 1 . Pathological and inflammatory alterations in parkinson’s disease.

GroupRegulation in PDReferences
PD patientsPD (n = 5), DAT (n = 9), cerebro vascular accidents (n = 2), without premortem neurologic disease (n = 7)↑ Lewy bodies in SN of PD patients
↑ GFAP+, HLA-DR+ cells in SN of PD patients
McGeer et al. (1988)
De novo PD (n = 22), sporadic AD (n = 11)↑ IL-6 and IL-1β in CSF of PD patientsBlum-Degen et al. (1995)
Healthy participants (n = 22), early-stage PD (n = 24)↑ Th1/total T cells, IL-10, IL-17A in blood of PD patientsLiu et al. (2022)
LRRK2-G2019S-PD patients (n = 33), idiopathic PD patients (n = 33)↑ Rest tremor, postural instability, freezing of gait, dyskinesias, UPSIT score, depression and bristol scale in LRRK2-G2019S PD patients
↓ Hyposmia and action tremor in LRRK2-G2019S PD patients
Gaig et al. (2014)
PD (n = 4)↑ CD68 in olfactory bulb of PD patientsVroon et al. (2007)
PD animal modelsNeurotoxinLPS rats (10 μg/4 μL, 2 days and 4 weeks)↑ iNOS, TNF-α and IL-6 in SN of LPS administrated rats
↑ α-syn in SN of LPS administrated rats
Choi et al. (2010)
LPS mice (5 μg/1 μL, stereotaxic)↓ TH+ neurons in SN of LPS administrated WT mice
↑ Isolectin B4 and LRRK2 in SN of LPS administrated WT mice
↑ TH+ neurons in SN of LPS administrated LRRK2−/− mice
↓ Isolectin B4 and LRRK2 in SN of LPS administrated LRRK2−/− mice
Moehle et al. (2012)
MPTP mice (20 mg/kg, i.p., 4 times at 2 h intervals, 1, 3 and 5 days)↑ IL-1β, IL-1α and IL-1ra in SN, striatum and olfactory bulb of MPTP administrated miceVroon et al. (2007)
Adβgal (stereotaxic, 7, 21, 40, 60 days)↓ TH+ neurons in SN of Adβgal administrated rats
↑ IL-1β, GFAP and CD68 in SN of Adβgal administrated rats
Ferrari et al. (2006)
MACO rats↑ DJ-1 in brain of cerebral I/R injury rats
↑ TNF-α, IL-1β and IL-6 in brain of cerebral I/R injury rats
Peng et al. (2020)
NLRP3A350V mice, Parkinflx/flx mice, Casp1+/−/parkin+/flx mice (adged 8-10 weeks)↑ NLRP3, cleaved caspase 1 and Iba-1 in SNpc of AAV GFP-Cre injected Parkinflx/flx mice and NLRP3A350V mice
↓ TH+ neurons in SNpc of Parkinflx/flx mice and NLRP3A350V mice
Panicker et al. (2022)
TransgenicNlrp3−/−, Cx3CrlCreER, Nlrp3D301NneoR mice (aged 9-12 weeks)↓ Motor function in MPTP administrated Nlrp3+/+ mice
↓ TH+ neurons and dopamine in SN of MPTP administrated Nlrp3+/+ mice
↑ Iba-1, IL-1β and ASC in SN of MPTP administrated Nlrp3+/+ mice
Lee et al. (2019)
LRRK2G2019S, LRRK2−/− mice↑ LRRK2 in brain of human α-syn fibrils administrated miceXu et al. (2022)
Parkin−/−, PINK1−/− mice↑ IL-12 and IL-13 in serum of Parkin−/− and PINK1−/− mice
↑ CXCL1, CCL2 and CCL4 in serum of Parkin−/− and PINK1−/− mice
Sliter et al. (2018)
DJ-1-deficient mice (Park7−/−)↓ TNF-α, IL-1β and IL-23p19 in striatum of Park7−/− miceNakamura et al. (2021)
t-SCI rats↑ DJ-1 in t-SCI in brain of rats
↑ NLRP3, cleaved caspase 1, IL-1β, IL-18 and MMP9 in t-SCI in brain of rats
Cai et al. (2022)

This table provides an overview of pathological features and inflammatory alterations in both PD patients and animal models. For details and references, see main text. ASC, apoptosis-associated speck like protein containing a CARD; AD, Alzheimer’s disease; CCL, chemokine (C-C motif) ligand; CD68, cluster of differentiation; COX-2, cyclooxygenase 2; CSF, cerebrospinal fluid; CXCL1, chemokine (C-X-C motif) ligand 1; DAT, dopamine transporter; GFAP, glial fibrillary acidic protein; HLA-DR, human leukocyte antigen-DR; IL, interleukin; iNOS, inducible nitric oxide synthase; I/R, ischemia/reperfusion; LPS, lipopolysaccharide; LRRK2, leucine rich repeat kinase 2; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MMP9, matrix metallopeptidase 9; NLRP3, nucleotide-binding oligomerization domain leucine rich repeat and pyrin domain containing 3; PBMC, peripheral blood mononuclear cells; PD, Parkinson’s disease; SN, substantia nigra; TH, tyrosine hydroxylase; Th1, type 1 T helper; TNF, tumor necrosis factor; t-SCI, traumatic spinal cord injury; UPSIT, University of Pennsylvania smell identification test; WT, wild type..



Microglia are specialized immune cells found within the central nervous system (CNS) and are considered the main form of active immune defense in the CNS (Filiano et al. 2015). Their ability to recognize certain patterns, through receptors known as pattern-recognition receptors, enables them to detect molecules linked to pathogens and damage, leading to microglial stimulation (Akira et al. 2006). Microgliosis, the continuous activation of microglia with a pro-inflammatory nature, has been identified as a contributing factor in various neurodegenerative diseases, including PD. Early evidence has shown that people diagnosed with both PD and PD combined with dementia exhibit a high concentration of activated microglia in their substantia nigra (McGeer et al. 1988). The activation of microglia, astrogliosis, and the infiltration of activated immune cells from the periphery contribute to harmful changes in the brain, resulting in neuronal death and the advancement of PD (Hirsch and Hunot 2009). Additionally, the number of microglia and their activation state correlate with α-synuclein deposition in the pathway involved in dopaminergic neuron loss (Croisier et al. 2005; Choi et al. 2010). Moreover, inflammatory cytokines like interleukin-1beta (IL-1β) and IL-6, which are released upon microglial activation, have been identified in the nigrostriatal pathway of brains affected by PD in both humans and corresponding animal models (Ferrari et al. 2006). These mediators exacerbate dopamine impairment and even lead to neuronal death (Blum-Degen et al. 1995; Vroon et al. 2007). In addition, according to recent research, early stages of PD are associated with microglial activation in the putamen and peripheral T helper 1 phenotypic bias (Liu et al. 2022). Consequently, peripheral adaptive immunity may indirectly result in microglial activation in the neurodegenerative process involved in PD (Liu et al. 2022).

The NOD-, LRR- and pyrin domain containing 3 (NLRP3) inflammasome is emerging as a pivotal component in understanding the neuroinflammatory processes involved in PD. This intracellular multiprotein complex, primarily known for its role in initiating innate immune responses, has recently been connected to the intricate mechanisms underlying PD pathogenesis. Upon activation, NLRP3 leads to the cleavage of pro-caspase-1 into active caspase-1, thereby enabling the conversion of pro-IL-1β into their active form (Franchi et al. 2009). These pro-inflammatory cytokines contribute to a chronic inflammatory state within the brain. This phenomenon activates the NLRP3 inflammasome and may also further exacerbate neuroinflammatory responses, establishing a feedback loop that sustains microgliosis (Heneka et al. 2018). Furthermore, dysregulation of the NLRP3 inflammasome has been linked to dopaminergic neuronal loss, a defining pathological hallmark of PD (Lee et al. 2019; Panicker et al. 2022). Some preclinical trials investigating the use of NLRP3 inhibitors to attenuate neuroinflammation and ameliorate neurodegenerative symptoms have yielded promising results (Coll et al. 2015; Lonnemann et al. 2020; Xue et al. 2021; Zeng et al. 2021). These associations emphasize the potential of NLRP3 as a therapeutic target.

Several studies have reported a unique link between certain genes associated with PD and the immune response of CNS cells. The leucine-rich repeat kinase 2 (LRRK2) gene has emerged as a pivotal component in PD; not only is it one of the most commonly mutated genes in familial PD, but variations in LRRK2 are also linked with sporadic PD cases (Zimprich et al. 2004; Gaig et al. 2014; Cookson 2017). LRRK2 encodes a protein kinase that is involved in a multitude of cellular processes, including autophagy, vesicle trafficking, and immune response. In PD, LRRK2 mutations lead to an aberrant activation of microglial cells. Reducing LRRK2 levels through knockdown or inhibiting its kinase activity in primary microglia leads to a decrease in the release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and IL-1β (Moehle et al. 2012; Russo et al. 2015). Moreover, mutated LRRK2 may enhance α-synuclein pathology, further exacerbating the inflammatory response (Bieri et al. 2019; Xu et al. 2022). The Phosphatase and tensin homolog-induced putative kinase 1 (PINK1) gene has been identified as a critical component in the development and progression of PD, specifically in connection with neuroinflammation. In healthy neurons, PINK1 monitors mitochondrial health and when damage occurs, it triggers mitophagy, the process involved in the selective degradation of damaged mitochondria. Mutations in the PINK1 gene can disrupt this protective mechanism, leading to an accumulation of damaged mitochondria within the cell. Mitochondrial accumulation can trigger a cascade of pro-inflammatory signals, inducing microglial activation, and the release of inflammatory mediators such as TNFα and IL-1β (Sliter et al. 2018). The activated microglia, in turn, can cause further damage to the neurons, creating a vicious cycle of neuroinflammation that results in the neurodegenerative process observed in PD. PINK1 has also been associated with immune responses outside the CNS, like alterations in the peripheral immune cells of patients with PD with PINK1 mutations, indicating a broader systemic involvement in inflammatory regulation (Zhou et al. 2019a). The DJ-1 gene (Parkinson disease protein 7, PARK7), associated with PD, is primarily expressed in astrocytes and microglia, and it has been investigated for its potential link to inflammation. In the context of ischemic stroke, DJ-1 has been shown to have anti-inflammatory properties by suppressing the production of inflammatory cytokines such as TNF-α, IL-1β, and IL-18 (Peng et al. 2020; Nakamura et al. 2021). Moreover, recent research has indicated that DJ-1 reduces neuroinflammation and the associated impairment of the blood-spinal cord barrier by inhibiting the activation of the NLRP3 inflammasome through the suppressor of cytokine signaling 1/Ras-related C3 botulinum toxin substrate 1/reactive oxygen species signaling pathway in a rat model of traumatic spinal cord injury (Cai et al. 2022).

Anti-Inflammatory Potential of Dipeptidyl Peptidase-4 (DPP-4) Inhibitors

DPP-4 is a serine exopeptidase involved in the degradation of incretin hormones, including glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide. DPP-4 is widely distributed in several organs, including the liver, lungs, intestinal epithelium, placenta, kidney, renal proximal tubules, and neurons, as well as in body fluids including the cerebrospinal fluid (Green et al. 2006; Kosaraju et al. 2013). In addition to its crucial role in the regulation of blood glucose levels, which are commonly used in the treatment of type 2 diabetes mellitus (T2DM), DPP-4 can be directly associated with the modulation of various non-incretin related processes, including inflammatory responses and neurophysiological functions (Aertgeerts et al. 2004).

Several reports have demonstrated that DPP-4 inhibitors modulate the activation of immune cells in adipose tissues (Shirakawa et al. 2011), and the inhibition of DPP-4 activity reduces the expression and activity of the macrophage marker F4/80 (Klein et al. 2014). The DPP-4 inhibitors, sitagliptin and NVPDPP728, exhibit significant potential in repressing the expression of NLRP3 and IL-1β in human macrophages (Dai et al. 2014). In patients with T2DM, a meta-analysis by Xie et al. confirmed that DPP-4 inhibitors exhibit substantial anti-inflammatory effects. The study, encompassing 22 trials with 1595 patients with T2DM, revealed that DPP-4 inhibitor therapy is notably linked to reductions in C-reactive protein, TNF-α, IL-6, and IL-1β levels (Xie et al. 2023). Sitagliptin is a DPP-4 inhibitor with anti-inflammatory and antioxidant properties, and effectively alleviates intestinal inflammation associated with severe acute pancreatitis (SAP), both in vitro and in vivo. Sitagliptin treatment leads to the upregulation of nuclear factor erythroid-derived 2-like 2 (Nrf2) expression while concurrently downregulating NF-κB expression in mice with SAP, thereby contributing significantly to its therapeutic efficacy (Zhou et al. 2019b). Moreover, for severe acute pancreatitis-related acute lung injury, sitagliptin is a promising therapeutic agent that activates the p62-kelch like ECH associated protein 1-Nrf2 signaling pathway, leading to Nrf2 nuclear translocation (Kong et al. 2021). Vildagliptin effectively suppresses the generation of reactive oxygen species (ROS), the production of vascular inflammatory factors, and monocyte adhesion to endothelial cells, potentially through the inhibition of toll-like receptor 4 and NF-κB nuclear signals (Wiciński et al. 2020).

Anti-Inflammatory Signaling Mechanism of DPP-4 Inhibitors

It is not yet certain whether the anti-inflammatory effects of DPP4 inhibitors are mediated through the accumulation of GLP1 or if they are independent of GLP1 receptor activation and instead directly related to the inhibition of DPP4 activity.

Lots of studies have demonstrated that the anti-inflammatory potential of DPP-4 inhibitors could be associated with the accumulation of GLP-1 and GLP-2, the two main targets of DPP-4; GLP-1 and GLP-2 activate their receptors and increase intracellular cyclic adenosine monophosphate, which activates their downstream pathways involved in the inhibition of the neuroinflammatory response (Flock et al. 2007). Linagliptin has been implicated in mitigating Aβ-induced neurotoxicity through GLP-1-mediated pathways, evidenced by the upregulation of insulin and insulin-like growth factor-1 (IGF-1) gene expression (Sedighi et al. 2021). Ide et al. found that linagliptin improved cognitive impairment in streptozotocin-induced diabetic mice by reducing proinflammatory cytokines, as well as inhibiting microglial activation. These effects were observed independently of glucose-lowering, suggesting a direct neuroprotective effect of linagliptin beyond its antidiabetic properties. This neuroprotective effect occurs independently of changes in blood glucose levels, suggesting a GLP-1-mediated mechanism. Additionally, in a rotenone-induced rat model of PD, linagliptin treatment increased striatal dopamine and active GLP-1 levels in the brain, indicating GLP-1’s involvement in its neuroprotective effects. Moreover, linagliptin attenuated rotenone-induced alterations in inflammatory markers like TNF-α. These findings underscore the potential of linagliptin’s anti-inflammatory and neuroprotective actions mediated through GLP-1-dependent mechanisms.

On the other hand, there are evidences indicating that changes in the activity of DPP4 itself are associated with inflammatory responses, suggesting that the anti-inflammatory activity of DPP4 inhibitors could occur independently of GLP1 accumulation. In facts, DPP-4 expression is altered in inflammation and/or immunity-associated diseases and acts as a regulator of immune cells (Trzaskalski et al. 2020). DPP-4 is involved in the maturation and phenotypic differentiation of T-cells, and therefore, in immune regulation (Al-Badri et al. 2018). Ghorpade et al. (2018) demonstrated the interaction between the liver and visceral adipose tissues (VAT), wherein obesity triggers hepatocytes to produce DPP-4, contributing to inflammation in adipose tissue macrophages (ATMs). In obese mice, DPP-4 synthesis by hepatocytes, along with the plasma factor Xa, triggers inflammation in visceral ATMs. Moreover, silencing DPP-4 expression in hepatocytes alleviates VAT inflammation and insulin resistance (Ghorpade et al. 2018). Renal protection has been a focal point of DPP-4 inhibitor studies, revealing pathways independent of GLP-1. Notably, DPP-4 inhibitors such as linagliptin and vildagliptin have demonstrated renoprotective effects mediated by substrates beyond GLP-1 (Kanasaki et al. 2014; Shi et al. 2016). These include the upregulation of Stromal Cell-Derived Factor-1 (SDF-1), modulation of microRNAs (miR-29 and miR-200a), and downregulation of Transforming Growth Factor-β (TGF-β) signaling. It should be noted that while several studies have highlighted the anti-inflammatory effects of DPP-4 inhibitors in neurodegenerative diseases, there remains a lack of conclusive evidence regarding their actions through GLP-1 independent pathways. In conjunction with these findings, reports on the anti-inflammatory effects of DPP4 inhibitors in the brain suggest that the anti-inflammatory effects of DPP4 inhibitors in neuroinflammatory lesions may be attributable not only to GLP1 accumulation but also to the inhibition of DPP4 enzyme activity itself. However, further research is needed to elucidate the underlying mechanisms by which DPP4 enzyme activity inhibition specifically contributes to the regulation of inflammatory responses.

Neuroprotective Effect of DPP-4 Inhibitors

Multiple in vitro studies have demonstrated that DPP-4 inhibitors exhibit a neuroprotective effect on neuronal cells (Abdelsalam and Safar 2015; Wiciński et al. 2018; Zhang et al. 2020). When exposed to neurotoxic stimuli, such as oxidative stress, inflammation, or excitotoxicity, neuronal cells treated with DPP-4 inhibitors exhibited enhanced viability compared to untreated cells. This finding suggests a potential role for DPP-4 inhibitors in preserving neuronal health and countering neurodegenerative processes. The experimental findings indicating the neuroprotective of DPP-4 inhibitors in various neuronal cell damage models, including those related to neurodegenerative diseases, have been emphasized (Table 2). Both in vitro and in vivo studies using animal models have revealed that DPP-4 inhibitors possess antioxidant properties and can mitigate oxidative damage in neurons. DPP-4 inhibitors can protect neurons from oxidative injury by scavenging free radicals and enhancing endogenous antioxidant defenses, potentially slowing the progression of neurodegenerative disorders. Several preclinical studies employing animal models of cognitive impairment have shown that DPP-4 inhibitors can ameliorate cognitive deficits. These improvements in cognitive function are associated with increased synaptic plasticity, enhanced neurogenesis, and improved cerebral blood flow. Moreover, Sitagliptin was found to improve memory deficits in PD rats by upregulating brain-derived neurotrophic factor to prevent neuronal and dendritic spine loss (Li et al. 2018). All these findings suggest that DPP-4 inhibitors might attenuate the cognitive decline observed in neurodegenerative diseases.

Table 2 . Neuroprotective effects of DPP-4 inhibitors.

DrugModelEffectsReferences
Pathological changesMOA
LinagliptinSH-SY5Y cells treated Aβ1-42↑ Cell viability↓ Inflammation; ↓ TNF-α, IL-1β and IL-6, ↑ Wnt1, pCREB, and PKCεSedighi et al. (2021)
Rotenone mice (1.5 mg/kg, s.c., 21 days)↑ Locomotor activity
↑ DA level in striatum
↓ Oxidative stress; ↑ SOD, catalase and GSH in brain homogenate
↓ Inflammation; ↓ TNF-α level in brain homogenate
Abhangi and Patel (2022)
Rotenone rats (2 mg/kg, s.c., 28 days)↑ Locomotor activity
↓ Catalepsy
↓ Inflammation; ↓ TNF-α, IL-6, and HIF1-α, ↑ DJ-1 in the midbrain and striatum
↓ Apoptosis; ↓ caspase-3 in the midbrain and striatum
ElGamal et al. (2023)
Focal Ischemic Stroke in Hyperglycemic mice↓ Neuronal cell death
↑ Locomotor Activity
↓ Inflammation; ↓ Iba-1, IL-1β and IL-6
↓ Apoptosis; ↓ caspase-3 and Bcl-2, ↑ p-Akt, p-mTOR, and Bax
Zhang et al. (2020)
HFD TauP301S mice PS19 transgenic mice↑ Cognitive function
No significant correlation in phosphorylated tau in hippocampus
Nakaoku et al. (2019)
VildagliptinMPTP mice (30 mg/kg, i.p., 14 days)↑ Locomotor activity
↑ TH+ neurons in SNpc
↑ TH density in striatum
↓ Apoptosis; ↓ caspase-3 and Bax/Bcl-2 ratio, ↓ p-ERK/ERK and p-JNK/JNK in striatum
Autophagy; No significant in LC3B and LC3B-II in striatum and SNpc
Pariyar et al. (2022)
SH-SY5Y cells treated MPP+↑ Cell viability↓ Apoptosis; ↑ p-Akt/Akt, ↓ p-ERK/ERK and p-JNK/JNK
↓ Autophagy; ↓ LC3B-II
Rotenone rats (1.5 mg/kg, s.c., 21 days)↑ Locomotor activity
↑ DA in striatum
↓ Body weight loss
↓ Inflammation; ↓ MPO, ICAM, TNF-α and iNOS in striatum
↓ Apoptosis; ↓ caspase-3, cytochrome c, TBARS and Nrf2 in striatum
Abdelsalam and Safar (2015)
AlCl3 rats (100 mg/kg, oral, 60 days)↑ Locomotor activity
↑ Cognitive function
↓ Aβ1-42 in hippocampus
↓ Oxidative stress; ↓ FOXO1 in hippocampus
↓ Inflammation; ↓ TNF-α in hippocampus
↓ Apoptosis; ↓ Bax and caspase-3, ↑ Bcl-2 in hippocampus
↑ Klotho, p-AKT and p-ERK, ↓ STAT3 and JAK2 in hippocampus
Yossef et al. (2020)
AlogliptinRotenone rats (1.5 mg/kg, i.p., 21 days)↑ Locomotor activity
↑ TH density in SNpc
↓ Inflammasome; ↓ Iba-1, caspase-1, TLR-4, NLRP3, HMGB1 and IL-1β in striatumSafar et al. (2021)
SaxagliptinSTZ-induced AD rats (3 mg/kg, ICV, 3 days)↑ Cognitive function
↓ Aβ1-42 levels in hippocampus
↓ Total tau levels in hippocampus
↓ Inflammation; ↓ TNF-α and IL-1β in hippocampusKosaraju et al. (2013)
Aβ-induced AD rats (300 pmol/day, stereotaxic, 15 days)↑ Cognitive function↓ Oxidative stress; ↓ MDA, ↑ SOD, catalase, GSH, Nrf2 and HO-1Li et al. (2018)
OmarigliptinLPS rats (250 μg/kg, i.p., 7 days)No significant in locomotor activity
↑ Cognitive function
↓ Oxidative stress; ↑ Nrf2 and HO-1 in hippocampus
↓ Inflammation; ↓ TNF-α, IL-8, TGF-β1, TLR-4 and NLRP3, ↑ IL-10 in hippocampus
↑ Autophagy; ↓ p-AKT and GSK-3β, ↑ beclin-1 in hippocampus
Kabel et al. (2022)
SitagliptinRotenone rats (3 mg/kg,s.c., 10 days)↑ Locomotor activity
↑ TH+ neurons in SNpc
↑ DA level in striatum
↓ Apoptosis; ↓ Bax, ↑ Bcl-2 in SN
↑ GDNF level in SN
↓ Inflammation; ↓TGF-β1, IL-1β and IL-6 in SN
Badawi et al. (2017)
6-OHDA rats (8 μg/rat, stereotaxic)↑ Cognitive function
↑ TH levels in SN
↑ Spine density in hippocampal CA1
↑ BDNF level in brainLi et al. (2018)

This table provides an overview of the neuroprotective effects of DPP-4 inhibitors with underlying pathological and mechanical changes in PD in both animal cell lines models. For details and references, see main text. AD, Alzheimer’s disease; Aβ, amyloid β; Bcl-2, B cell lymphoma 2; CA1, cornu ammonis; CREB, cAMP-response element binding protein; DA, dopamine; ERK, extracellular signal-regulated kinase; FOXO1, forkhead box protein O1; GSH, glutathione; GSK-3β, glycogen synthase kinase 3 beta; HIF1-α, hypoxia-inducible factor 1-alpha; HFD, high fat diet; HMGB1, high-mobility group box 1; HO-1, heme oxygenase-1; Iba-1, ionized calcium-binding adapter molecule-1; ICAM, intercellular adhesion molecule-1; ICV, intracerebroventricular; IL-1β, interleukin-1β; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; JAK2, Janus kinase; JNK, c-Jun N-terminal kinase; LC3B, microtubule-associated protein 1A/1B-light chain 3B; LPS, lipopolysaccharide; MDA, malondialdehyde; MPO, Myeloperoxidase; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NLRP3, nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3; Nrf2, Nuclear factor erythroid-2-related factor 2; PKCε, protein kinase C; mTOR, mammalian target of rapamycin; SN, substantia nigra; SNpc, substantia nigra pars compacta; SOD, superoxide dismutase; STAT3, signal transducer and activator of transcription; STZ, streptozotocin; TBARS, thiobarbituric acid reactive substances; TGF-β1, transforming growth factor-β1; TH, tyrosine hydroxylase; TLR-4, toll-like receptor-4; TNF-a, tumor necrosis factor-α..



In several experimental studies using animal models, DPP-4 inhibitors have been shown to reduce neuroinflammation by suppressing microglial activation and inhibiting the production of pro-inflammatory cytokines in the brain. Accumulated evidence has revealed that DPP-4 inhibitors suppress neuroinflammation by reducing TNF-α and IL-1β levels, thereby contributing to the amelioration of the pathology associated with several neurodegenerative diseases. Exendin-4 and linagliptin treatments have induced microglial polarization in the anti-inflammatory M2 phenotype and reduced pro-inflammatory cytokine secretion in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced PD mouse model (Yu et al. 2023). Pretreatment with linagliptin also attenuates amyloid-β (Aβ)-induced inflammation and cytotoxicity by suppressing the release of IL-6 in human SH-SY5Y neuroblastoma cells exposed to Aβ cytotoxicity (Sedighi et al. 2021). Linagliptin dose-dependently improved cognitive impairment and reduced the levels of soluble Aβ (1-42), TNF-α, IL-1β, IL-6, acetylcholinesterase, and oxidative/nitrosative stress markers in a rat model of Alzheimer’s disease induced by intracerebroventricular streptozotocin treatment (Siddiqui et al. 2023). Vildagliptin effectively improves motor performance, reduces striatal dopamine depletion, and suppresses inflammatory mediators like NF-κB and TNF-α in a rat model of rotenone-induced PD (Abdelsalam and Safar 2015). Sitagliptin also reversed nigrostriatal degeneration, improved motor performance, and decreased the levels of IL-1β, IL-6, and TGF-β1 in the rat brain in a rotenone-induced model of PD (Badawi et al. 2017). Sitagliptin, both alone and in combination with pregabalin, alleviates acute epileptogenesis induced by pentylenetetrazole in mice. The findings demonstrated that sitagliptin exerted substantial anti-inflammatory effects by modulating the levels of inflammatory factors like Nrf2 and NF-κb (Nader et al. 2018). Omarigliptin exhibits anti-inflammatory effects by inhibiting NF-κB activation, leading to a reduction in nitric oxide (NO) production and the expression of inducible NO synthase (iNOS) in a model of neurotoxin-induced toxicity in PC12 cells, which mimics PD (Gouda and Cho 2022).

Therapeutic Potential of DPP-4 Inhibitors in PD

Preclinical studies have shown that DPP-4 inhibitors may protect dopaminergic neurons from degeneration, enhance neuroplasticity, and attenuate neuroinflammation (Table 2). These findings have sparked interest in exploring the therapeutic potential of DPP-4 inhibitors in PD, focusing on dopaminergic neuron degeneration.

Numerous clinical trials have assessed the neurorestorative potential of antidiabetic drugs in preclinical models of PD, yielding positive outcomes. The most extensively reported potential for repurposing antidiabetic drugs as treatment for PD lies in GLP1 receptor agonists. Specifically, clinical trial (NCT01971242) (University College London, 2016) are underway for exenatide, a GLP-1 receptor agonist, as a treatment for PD. These clinical trials have evaluated the impact of exenatide in patients with PD, utilizing a randomized, double-blind, and placebo-controlled design, wherein participants received weekly subcutaneous injections of either exenatide or a placebo alongside their standard PD treatment (Athauda et al. 2017). The outcomes of these trials provided insights into whether the neuroprotective effects of exenatide, previously observed in preclinical models, could translate into positive outcomes in a clinical setting (Athauda et al. 2017). The therapeutic potential of GLP-1 agonist lixisenatide in PD has also garnered significant interest following promising results from clinical trials (NCT03439943) (University College London, 2023). Participants receiving lixisenatide showed less disability, as assessed by the Movement Disorder Society-Unified Parkinson’s disease rating scale (MDS-UPDRS), compared to those receiving placebo. Importantly, improvements in the MDS-UPDRS scores were observed in both ON and OFF states, indicating a potential disease-modifying effect of lixisenatide beyond the amplification of pharmacological impacts of existing medications. Lin et al. reported that diabetic patients treated with dipeptidyl peptidase-4 (DPP-4) inhibitors, particularly vildagliptin, exhibited a significantly lower risk of PD compared to those using other oral antidiabetic drugs (Lin et al. 2023). Additionally, Jeong et al. demonstrated that treatment with DPP-4 inhibitors in diabetic patients with PD was associated with higher baseline nigrostriatal dopamine transporter availability and slower longitudinal increase in levodopa-equivalent dose, suggesting potential beneficial effects on motor outcomes in this population (Jeong et al. 2021). Furthermore, given that DPP-4 inhibitors are small molecules, they are more practical for use in patients with PD compared to larger peptide-based molecules like GLP-1 receptor agonists. Despite their mechanism primarily involving the augmentation of GLP-1 signaling, which may confer anti-inflammatory effects, the inhibition of DPP-4 itself could also contribute to additional anti-inflammatory effects. This dual mechanism suggests that the use of DPP-4 inhibitors in PD patients may offer comprehensive neuroprotective effects.

Conclusion

In summary, the current state of therapeutics for PD highlights a significant demand for innovative disease-modifying treatments. This review discusses the current understanding of the anti-inflammatory properties of DPP-4 inhibitors and their potential as a novel therapeutic strategy for PD (Fig. 1). Elucidating the mechanisms through which DPP-4 inhibitors influence neuroinflammation may offer new prospects for disease-modifying therapies in PD. However, it is crucial to conduct further research, including clinical trials, to confirm the validity of the preclinical findings discussed in this review and ascertain the safety and effectiveness of DPP-4 inhibitors in treating PD.

Figure 1. Role of DPP4 inhibitors in neuroinflammation.

Conflict of interest

The authors declare that they have no conflict of interest.

Acknowledgements

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (NRF-2021R1A2C1013180, RS-2023-00246151).

Fig 1.

Figure 1.Role of DPP4 inhibitors in neuroinflammation.
Drug Targets and Therapeutics 2024; 3: 83-93https://doi.org/10.58502/DTT.24.0030

Table 1 Pathological and inflammatory alterations in parkinson’s disease

GroupRegulation in PDReferences
PD patientsPD (n = 5), DAT (n = 9), cerebro vascular accidents (n = 2), without premortem neurologic disease (n = 7)↑ Lewy bodies in SN of PD patients
↑ GFAP+, HLA-DR+ cells in SN of PD patients
McGeer et al. (1988)
De novo PD (n = 22), sporadic AD (n = 11)↑ IL-6 and IL-1β in CSF of PD patientsBlum-Degen et al. (1995)
Healthy participants (n = 22), early-stage PD (n = 24)↑ Th1/total T cells, IL-10, IL-17A in blood of PD patientsLiu et al. (2022)
LRRK2-G2019S-PD patients (n = 33), idiopathic PD patients (n = 33)↑ Rest tremor, postural instability, freezing of gait, dyskinesias, UPSIT score, depression and bristol scale in LRRK2-G2019S PD patients
↓ Hyposmia and action tremor in LRRK2-G2019S PD patients
Gaig et al. (2014)
PD (n = 4)↑ CD68 in olfactory bulb of PD patientsVroon et al. (2007)
PD animal modelsNeurotoxinLPS rats (10 μg/4 μL, 2 days and 4 weeks)↑ iNOS, TNF-α and IL-6 in SN of LPS administrated rats
↑ α-syn in SN of LPS administrated rats
Choi et al. (2010)
LPS mice (5 μg/1 μL, stereotaxic)↓ TH+ neurons in SN of LPS administrated WT mice
↑ Isolectin B4 and LRRK2 in SN of LPS administrated WT mice
↑ TH+ neurons in SN of LPS administrated LRRK2−/− mice
↓ Isolectin B4 and LRRK2 in SN of LPS administrated LRRK2−/− mice
Moehle et al. (2012)
MPTP mice (20 mg/kg, i.p., 4 times at 2 h intervals, 1, 3 and 5 days)↑ IL-1β, IL-1α and IL-1ra in SN, striatum and olfactory bulb of MPTP administrated miceVroon et al. (2007)
Adβgal (stereotaxic, 7, 21, 40, 60 days)↓ TH+ neurons in SN of Adβgal administrated rats
↑ IL-1β, GFAP and CD68 in SN of Adβgal administrated rats
Ferrari et al. (2006)
MACO rats↑ DJ-1 in brain of cerebral I/R injury rats
↑ TNF-α, IL-1β and IL-6 in brain of cerebral I/R injury rats
Peng et al. (2020)
NLRP3A350V mice, Parkinflx/flx mice, Casp1+/−/parkin+/flx mice (adged 8-10 weeks)↑ NLRP3, cleaved caspase 1 and Iba-1 in SNpc of AAV GFP-Cre injected Parkinflx/flx mice and NLRP3A350V mice
↓ TH+ neurons in SNpc of Parkinflx/flx mice and NLRP3A350V mice
Panicker et al. (2022)
TransgenicNlrp3−/−, Cx3CrlCreER, Nlrp3D301NneoR mice (aged 9-12 weeks)↓ Motor function in MPTP administrated Nlrp3+/+ mice
↓ TH+ neurons and dopamine in SN of MPTP administrated Nlrp3+/+ mice
↑ Iba-1, IL-1β and ASC in SN of MPTP administrated Nlrp3+/+ mice
Lee et al. (2019)
LRRK2G2019S, LRRK2−/− mice↑ LRRK2 in brain of human α-syn fibrils administrated miceXu et al. (2022)
Parkin−/−, PINK1−/− mice↑ IL-12 and IL-13 in serum of Parkin−/− and PINK1−/− mice
↑ CXCL1, CCL2 and CCL4 in serum of Parkin−/− and PINK1−/− mice
Sliter et al. (2018)
DJ-1-deficient mice (Park7−/−)↓ TNF-α, IL-1β and IL-23p19 in striatum of Park7−/− miceNakamura et al. (2021)
t-SCI rats↑ DJ-1 in t-SCI in brain of rats
↑ NLRP3, cleaved caspase 1, IL-1β, IL-18 and MMP9 in t-SCI in brain of rats
Cai et al. (2022)

This table provides an overview of pathological features and inflammatory alterations in both PD patients and animal models. For details and references, see main text. ASC, apoptosis-associated speck like protein containing a CARD; AD, Alzheimer’s disease; CCL, chemokine (C-C motif) ligand; CD68, cluster of differentiation; COX-2, cyclooxygenase 2; CSF, cerebrospinal fluid; CXCL1, chemokine (C-X-C motif) ligand 1; DAT, dopamine transporter; GFAP, glial fibrillary acidic protein; HLA-DR, human leukocyte antigen-DR; IL, interleukin; iNOS, inducible nitric oxide synthase; I/R, ischemia/reperfusion; LPS, lipopolysaccharide; LRRK2, leucine rich repeat kinase 2; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MMP9, matrix metallopeptidase 9; NLRP3, nucleotide-binding oligomerization domain leucine rich repeat and pyrin domain containing 3; PBMC, peripheral blood mononuclear cells; PD, Parkinson’s disease; SN, substantia nigra; TH, tyrosine hydroxylase; Th1, type 1 T helper; TNF, tumor necrosis factor; t-SCI, traumatic spinal cord injury; UPSIT, University of Pennsylvania smell identification test; WT, wild type.


Table 2 Neuroprotective effects of DPP-4 inhibitors

DrugModelEffectsReferences
Pathological changesMOA
LinagliptinSH-SY5Y cells treated Aβ1-42↑ Cell viability↓ Inflammation; ↓ TNF-α, IL-1β and IL-6, ↑ Wnt1, pCREB, and PKCεSedighi et al. (2021)
Rotenone mice (1.5 mg/kg, s.c., 21 days)↑ Locomotor activity
↑ DA level in striatum
↓ Oxidative stress; ↑ SOD, catalase and GSH in brain homogenate
↓ Inflammation; ↓ TNF-α level in brain homogenate
Abhangi and Patel (2022)
Rotenone rats (2 mg/kg, s.c., 28 days)↑ Locomotor activity
↓ Catalepsy
↓ Inflammation; ↓ TNF-α, IL-6, and HIF1-α, ↑ DJ-1 in the midbrain and striatum
↓ Apoptosis; ↓ caspase-3 in the midbrain and striatum
ElGamal et al. (2023)
Focal Ischemic Stroke in Hyperglycemic mice↓ Neuronal cell death
↑ Locomotor Activity
↓ Inflammation; ↓ Iba-1, IL-1β and IL-6
↓ Apoptosis; ↓ caspase-3 and Bcl-2, ↑ p-Akt, p-mTOR, and Bax
Zhang et al. (2020)
HFD TauP301S mice PS19 transgenic mice↑ Cognitive function
No significant correlation in phosphorylated tau in hippocampus
Nakaoku et al. (2019)
VildagliptinMPTP mice (30 mg/kg, i.p., 14 days)↑ Locomotor activity
↑ TH+ neurons in SNpc
↑ TH density in striatum
↓ Apoptosis; ↓ caspase-3 and Bax/Bcl-2 ratio, ↓ p-ERK/ERK and p-JNK/JNK in striatum
Autophagy; No significant in LC3B and LC3B-II in striatum and SNpc
Pariyar et al. (2022)
SH-SY5Y cells treated MPP+↑ Cell viability↓ Apoptosis; ↑ p-Akt/Akt, ↓ p-ERK/ERK and p-JNK/JNK
↓ Autophagy; ↓ LC3B-II
Rotenone rats (1.5 mg/kg, s.c., 21 days)↑ Locomotor activity
↑ DA in striatum
↓ Body weight loss
↓ Inflammation; ↓ MPO, ICAM, TNF-α and iNOS in striatum
↓ Apoptosis; ↓ caspase-3, cytochrome c, TBARS and Nrf2 in striatum
Abdelsalam and Safar (2015)
AlCl3 rats (100 mg/kg, oral, 60 days)↑ Locomotor activity
↑ Cognitive function
↓ Aβ1-42 in hippocampus
↓ Oxidative stress; ↓ FOXO1 in hippocampus
↓ Inflammation; ↓ TNF-α in hippocampus
↓ Apoptosis; ↓ Bax and caspase-3, ↑ Bcl-2 in hippocampus
↑ Klotho, p-AKT and p-ERK, ↓ STAT3 and JAK2 in hippocampus
Yossef et al. (2020)
AlogliptinRotenone rats (1.5 mg/kg, i.p., 21 days)↑ Locomotor activity
↑ TH density in SNpc
↓ Inflammasome; ↓ Iba-1, caspase-1, TLR-4, NLRP3, HMGB1 and IL-1β in striatumSafar et al. (2021)
SaxagliptinSTZ-induced AD rats (3 mg/kg, ICV, 3 days)↑ Cognitive function
↓ Aβ1-42 levels in hippocampus
↓ Total tau levels in hippocampus
↓ Inflammation; ↓ TNF-α and IL-1β in hippocampusKosaraju et al. (2013)
Aβ-induced AD rats (300 pmol/day, stereotaxic, 15 days)↑ Cognitive function↓ Oxidative stress; ↓ MDA, ↑ SOD, catalase, GSH, Nrf2 and HO-1Li et al. (2018)
OmarigliptinLPS rats (250 μg/kg, i.p., 7 days)No significant in locomotor activity
↑ Cognitive function
↓ Oxidative stress; ↑ Nrf2 and HO-1 in hippocampus
↓ Inflammation; ↓ TNF-α, IL-8, TGF-β1, TLR-4 and NLRP3, ↑ IL-10 in hippocampus
↑ Autophagy; ↓ p-AKT and GSK-3β, ↑ beclin-1 in hippocampus
Kabel et al. (2022)
SitagliptinRotenone rats (3 mg/kg,s.c., 10 days)↑ Locomotor activity
↑ TH+ neurons in SNpc
↑ DA level in striatum
↓ Apoptosis; ↓ Bax, ↑ Bcl-2 in SN
↑ GDNF level in SN
↓ Inflammation; ↓TGF-β1, IL-1β and IL-6 in SN
Badawi et al. (2017)
6-OHDA rats (8 μg/rat, stereotaxic)↑ Cognitive function
↑ TH levels in SN
↑ Spine density in hippocampal CA1
↑ BDNF level in brainLi et al. (2018)

This table provides an overview of the neuroprotective effects of DPP-4 inhibitors with underlying pathological and mechanical changes in PD in both animal cell lines models. For details and references, see main text. AD, Alzheimer’s disease; Aβ, amyloid β; Bcl-2, B cell lymphoma 2; CA1, cornu ammonis; CREB, cAMP-response element binding protein; DA, dopamine; ERK, extracellular signal-regulated kinase; FOXO1, forkhead box protein O1; GSH, glutathione; GSK-3β, glycogen synthase kinase 3 beta; HIF1-α, hypoxia-inducible factor 1-alpha; HFD, high fat diet; HMGB1, high-mobility group box 1; HO-1, heme oxygenase-1; Iba-1, ionized calcium-binding adapter molecule-1; ICAM, intercellular adhesion molecule-1; ICV, intracerebroventricular; IL-1β, interleukin-1β; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; JAK2, Janus kinase; JNK, c-Jun N-terminal kinase; LC3B, microtubule-associated protein 1A/1B-light chain 3B; LPS, lipopolysaccharide; MDA, malondialdehyde; MPO, Myeloperoxidase; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NLRP3, nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3; Nrf2, Nuclear factor erythroid-2-related factor 2; PKCε, protein kinase C; mTOR, mammalian target of rapamycin; SN, substantia nigra; SNpc, substantia nigra pars compacta; SOD, superoxide dismutase; STAT3, signal transducer and activator of transcription; STZ, streptozotocin; TBARS, thiobarbituric acid reactive substances; TGF-β1, transforming growth factor-β1; TH, tyrosine hydroxylase; TLR-4, toll-like receptor-4; TNF-a, tumor necrosis factor-α.


References

  1. Abhangi KV, Patel JI (2022) Neuroprotective effects of linagliptin in a rotenone-induced rat model of Parkinson's disease. Indian J Pharmacol 54:46-50. doi: 10.4103/ijp.IJP_384_20
    Pubmed KoreaMed CrossRef
  2. Abdelsalam RM, Safar MM (2015) Neuroprotective effects of vildagliptin in rat rotenone Parkinson's disease model: role of RAGE-NFκB and Nrf2-antioxidant signaling pathways. J Neurochem 133:700-707. doi: 10.1111/jnc.13087
    Pubmed CrossRef
  3. Aertgeerts K, Ye S, Tennant MG, Kraus ML, Rogers J, Sang BC, Skene RJ, Webb DR, Prasad GS (2004) Crystal structure of human dipeptidyl peptidase IV in complex with a decapeptide reveals details on substrate specificity and tetrahedral intermediate formation. Protein Sci 13:412-421. doi: 10.1110/ps.03460604
    Pubmed KoreaMed CrossRef
  4. Akira S, Uematsu S, Takeuchi O (2006) Pathogen recognition and innate immunity. Cell 124:783-801. doi: 10.1016/j.cell.2006.02.015
    Pubmed CrossRef
  5. Al-Badri G, Leggio GM, Musumeci G, Marzagalli R, Drago F, Castorina A (2018) Tackling dipeptidyl peptidase IV in neurological disorders. Neural Regen Res 13:26-34. doi: 10.4103/1673-5374.224365
    Pubmed KoreaMed CrossRef
  6. Athauda D, Maclagan K, Skene SS, Bajwa-Joseph M, Letchford D, Chowdhury K, Hibbert S, Budnik N, Zampedri L, Dickson J, Li Y, Aviles-Olmos I, Warner TT, Limousin P, Lees AJ, Greig NH, Tebbs S, Foltynie T (2017) Exenatide once weekly versus placebo in Parkinson's disease: a randomised, double-blind, placebo-controlled trial. Lancet 390:1664-1675. doi: 10.1016/S0140-6736(17)31585-4
    Pubmed CrossRef
  7. Badawi GA, Abd El Fattah MA, Zaki HF, El Sayed MI (2017) Sitagliptin and liraglutide reversed nigrostriatal degeneration of rodent brain in rotenone-induced Parkinson's disease. Inflammopharmacology 25:369-382. doi: 10.1007/s10787-017-0331-6
    Pubmed CrossRef
  8. Bang Y, Lim J, Choi HJ (2021) Recent advances in the pathology of prodromal non-motor symptoms olfactory deficit and depression in Parkinson's disease: clues to early diagnosis and effective treatment. Arch Pharm Res 44:588-604. doi: 10.1007/s12272-021-01337-3
    Pubmed KoreaMed CrossRef
  9. Bartels AL, Willemsen AT, Doorduin J, de Vries EF, Dierckx RA, Leenders KL (2010) [11C]-PK11195 PET: quantification of neuroinflammation and a monitor of anti-inflammatory treatment in Parkinson's disease?. Parkinsonism Relat Disord 16:57-59. doi: 10.1016/j.parkreldis.2009.05.005
    Pubmed CrossRef
  10. Bieri G, Brahic M, Bousset L, Couthouis J, Kramer NJ, Ma R, Nakayama L, Monbureau M, Defensor E, Schüle B, Shamloo M, Melki R, Gitler AD (2019) LRRK2 modifies α-syn pathology and spread in mouse models and human neurons. Acta Neuropathol 137:961-980. doi: 10.1007/s00401-019-01995-0
    Pubmed KoreaMed CrossRef
  11. Blum-Degen D, Müller T, Kuhn W, Gerlach M, Przuntek H, Riederer P (1995) Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer's and de novo Parkinson's disease patients. Neurosci Lett 202:17-20. doi: 10.1016/0304-3940(95)12192-7
    Pubmed CrossRef
  12. Cai L, Gao L, Zhang G, Zeng H, Wu X, Tan X, Qian C, Chen G (2022) DJ-1 alleviates neuroinflammation and the related blood-spinal cord barrier destruction by suppressing NLRP3 inflammasome activation via SOCS1/Rac1/ROS pathway in a rat model of traumatic spinal cord injury. J Clin Med 11:3716. doi: 10.3390/jcm11133716
    Pubmed KoreaMed CrossRef
  13. Choi DY, Zhang J, Bing G (2010) Aging enhances the neuroinflammatory response and alpha-synuclein nitration in rats. Neurobiol Aging 31:1649-1653. doi: 10.1016/j.neurobiolaging.2008.09.010
    Pubmed CrossRef
  14. Coll RC, Robertson AA, Chae JJ, Higgins SC, Muñoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS, Haneklaus M, Sutton CE, Núñez G, Latz E, Kastner DL, Mills KH, Masters SL, Schroder K, Cooper MA, O'Neill LA (2015) A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med 21:248-255. doi: 10.1038/nm.3806
    Pubmed KoreaMed CrossRef
  15. Cookson MR (2017) Mechanisms of mutant LRRK2 neurodegeneration. Adv Neurobiol 14:227-239. doi: 10.1007/978-3-319-49969-7_12
    Pubmed CrossRef
  16. Croisier E, Moran LB, Dexter DT, Pearce RK, Graeber MB (2005) Microglial inflammation in the parkinsonian substantia nigra: relationship to alpha-synuclein deposition. J Neuroinflammation 2:14. doi: 10.1186/1742-2094-2-14
    Pubmed KoreaMed CrossRef
  17. Dai Y, Dai D, Wang X, Ding Z, Mehta JL (2014) DPP-4 inhibitors repress NLRP3 inflammasome and interleukin-1beta via GLP-1 receptor in macrophages through protein kinase C pathway. Cardiovasc Drugs Ther 28:425-432. doi: 10.1007/s10557-014-6539-4
    Pubmed CrossRef
  18. Doot RK, Young AJ, Nasrallah IM, Wetherill RR, Siderowf A, Mach RH, Dubroff JG (2022) [18F]NOS PET brain imaging suggests elevated neuroinflammation in idiopathic Parkinson's disease. Cells 11:3081. doi: 10.3390/cells11193081
    Pubmed KoreaMed CrossRef
  19. ElGamal RZ, Tadros MG, Menze ET (2023) Linagliptin counteracts rotenone's toxicity in non-diabetic rat model of Parkinson's disease: insights into the neuroprotective roles of DJ-1, SIRT-1/Nrf-2 and implications of HIF1-α. Eur J Pharmacol 941:175498. doi: 10.1016/j.ejphar.2023.175498
    Pubmed CrossRef
  20. Ferrari CC, Pott Godoy MC, Tarelli R, Chertoff M, Depino AM, Pitossi FJ (2006) Progressive neurodegeneration and motor disabilities induced by chronic expression of IL-1beta in the substantia nigra. Neurobiol Dis 24:183-193. doi: 10.1016/j.nbd.2006.06.013
    Pubmed CrossRef
  21. Filiano AJ, Gadani SP, Kipnis J (2015) Interactions of innate and adaptive immunity in brain development and function. Brain Res 1617:18-27. doi: 10.1016/j.brainres.2014.07.050
    Pubmed KoreaMed CrossRef
  22. Flock G, Baggio LL, Longuet C, Drucker DJ (2007) Incretin receptors for glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide are essential for the sustained metabolic actions of vildagliptin in mice. Diabetes 56:3006-3013. doi: 10.2337/db07-0697
    Pubmed CrossRef
  23. Franchi L, Eigenbrod T, Muñoz-Planillo R, Nuñez G (2009) The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol 10:241-247. doi: 10.1038/ni.1703
    Pubmed KoreaMed CrossRef
  24. Gaig C, Vilas D, Infante J, Sierra M, García-Gorostiaga I, Buongiorno M, Ezquerra M, Martí MJ, Valldeoriola F, Aguilar M, Calopa M, Hernandez-Vara J, Tolosa E (2014) Nonmotor symptoms in LRRK2 G2019S associated Parkinson's disease. PLoS One 9:e108982. doi: 10.1371/journal.pone.0108982
    Pubmed KoreaMed CrossRef
  25. Ghorpade DS, Ozcan L, Zheng Z, Nicoloro SM, Shen Y, Chen E, Blüher M, Czech MP, Tabas I (2018) Hepatocyte-secreted DPP4 in obesity promotes adipose inflammation and insulin resistance. Nature 555:673-677. doi: 10.1038/nature26138
    Pubmed KoreaMed CrossRef
  26. Gouda NA, Cho J (2022) Omarigliptin mitigates 6-hydroxydopamine- or rotenone-induced oxidative toxicity in PC12 cells by antioxidant, anti-inflammatory, and anti-apoptotic actions. Antioxidants (Basel) 11:1940. doi: 10.3390/antiox11101940
    Pubmed KoreaMed CrossRef
  27. Green BD, Irwin N, Flatt PR (2006) Pituitary adenylate cyclase-activating peptide (PACAP): assessment of dipeptidyl peptidase IV degradation, insulin-releasing activity and antidiabetic potential. Peptides 27:1349-1358. doi: 10.1016/j.peptides.2005.11.010
    Pubmed CrossRef
  28. Heneka MT, McManus RM, Latz E (2018) Inflammasome signalling in brain function and neurodegenerative disease. Nat Rev Neurosci 19:610-621. doi: 10.1038/s41583-018-0055-7. Erratum in: (2019) Nat Rev Neurosci 20:187. doi: 10.1038/s41583-019-0137-1
    Pubmed CrossRef
  29. Hirsch EC, Hunot S (2009) Neuroinflammation in Parkinson's disease: a target for neuroprotection?. Lancet Neurol 8:382-397. doi: 10.1016/S1474-4422(09)70062-6
    Pubmed CrossRef
  30. Ide M, Sonoda N, Inoue T, Kimura S, Minami Y, Makimura H, Hayashida E, Hyodo F, Yamato M, Takayanagi R, Inoguchi T (2020) The dipeptidyl peptidase-4 inhibitor, linagliptin, improves cognitive impairment in streptozotocin-induced diabetic mice by inhibiting oxidative stress and microglial activation. PLoS One 15:e0228750. doi: 10.1371/journal.pone.0228750
    Pubmed KoreaMed CrossRef
  31. Jeong SH, Chung SJ, Yoo HS, Hong N, Jung JH, Baik K, Lee YH, Sohn YH, Lee PH (2021) Beneficial effects of dipeptidyl peptidase-4 inhibitors in diabetic Parkinson's disease. Brain 144:1127-1137. doi: 10.1093/brain/awab015
    Pubmed CrossRef
  32. Kabel AM, Arab HH, Atef A, Estfanous RS (2022) Omarigliptin/galangin combination mitigates lipopolysaccharide-induced neuroinflammation in rats: involvement of glucagon-like peptide-1, toll-like receptor-4, apoptosis and Akt/GSK-3β signaling. Life Sci 295:120396. doi: 10.1016/j.lfs.2022.120396
    Pubmed CrossRef
  33. Kanasaki K, Shi S, Kanasaki M, He J, Nagai T, Nakamura Y, Ishigaki Y, Kitada M, ivastava SP Sr, Koya D (2014) Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes 63:2120-2131. doi: 10.2337/db13-1029
    Pubmed CrossRef
  34. Kang Y, Mozley PD, Verma A, Schlyer D, Henchcliffe C, Gauthier SA, Chiao PC, He B, Nikolopoulou A, Logan J, Sullivan JM, Pryor KO, Hesterman J, Kothari PJ, Vallabhajosula S (2018) Noninvasive PK11195-PET image analysis techniques can detect abnormal cerebral microglial activation in Parkinson's disease. J Neuroimaging 28:496-505. doi: 10.1111/jon.12519
    Pubmed KoreaMed CrossRef
  35. Klein T, Fujii M, Sandel J, Shibazaki Y, Wakamatsu K, Mark M, Yoneyama H (2014) Linagliptin alleviates hepatic steatosis and inflammation in a mouse model of non-alcoholic steatohepatitis. Med Mol Morphol 47:137-149. doi: 10.1007/s00795-013-0053-9
    Pubmed CrossRef
  36. Kong L, Deng J, Zhou X, Cai B, Zhang B, Chen X, Chen Z, Wang W (2021) Sitagliptin activates the p62-Keap1-Nrf2 signalling pathway to alleviate oxidative stress and excessive autophagy in severe acute pancreatitis-related acute lung injury. Cell Death Dis 12:928. doi: 10.1038/s41419-021-04227-0
    Pubmed KoreaMed CrossRef
  37. Kosaraju J, Gali CC, Khatwal RB, Dubala A, Chinni S, Holsinger RM, Madhunapantula VS, Muthureddy Nataraj SK, Basavan D (2013) Saxagliptin: a dipeptidyl peptidase-4 inhibitor ameliorates streptozotocin induced Alzheimer's disease. Neuropharmacology 72:291-300. doi: 10.1016/j.neuropharm.2013.04.008
    Pubmed CrossRef
  38. Lavisse S, Goutal S, Wimberley C, Tonietto M, Bottlaender M, Gervais P, Kuhnast B, Peyronneau MA, Barret O, Lagarde J, Sarazin M, Hantraye P, Thiriez C, Remy P (2021) Increased microglial activation in patients with Parkinson disease using [18F]-DPA714 TSPO PET imaging. Parkinsonism Relat Disord 82:29-36. doi: 10.1016/j.parkreldis.2020.11.011
    Pubmed CrossRef
  39. Lee E, Hwang I, Park S, Hong S, Hwang B, Cho Y, Son J, Yu JW (2019) MPTP-driven NLRP3 inflammasome activation in microglia plays a central role in dopaminergic neurodegeneration. Cell Death Differ 26:213-228. doi: 10.1038/s41418-018-0124-5
    Pubmed KoreaMed CrossRef
  40. Li J, Zhang S, Li C, Li M, Ma L (2018) Sitagliptin rescues memory deficits in Parkinsonian rats via upregulating BDNF to prevent neuron and dendritic spine loss. Neurol Res 40:736-743. doi: 10.1080/01616412.2018.1474840
    Pubmed CrossRef
  41. Li Y, Tian Q, Li Z, Dang M, Lin Y, Hou X (2019) Activation of Nrf2 signaling by sitagliptin and quercetin combination against β-amyloid induced Alzheimer's disease in rats. Drug Dev Res 80:837-845. doi: 10.1002/ddr.21567
    Pubmed CrossRef
  42. Lim J, Bang Y, Choi HJ (2018) Abnormal hippocampal neurogenesis in Parkinson's disease: relevance to a new therapeutic target for depression with Parkinson's disease. Arch Pharm Res 41:943-954. doi: 10.1007/s12272-018-1063-x
    Pubmed CrossRef
  43. Lin YH, Hsu CC, Liu JS, Chang KC, Huang JA (2023) Use of dipeptidyl peptidase-4 inhibitors was associated with a lower risk of Parkinson's disease in diabetic patients. Sci Rep 13:22489. doi: 10.1038/s41598-023-49870-z
    Pubmed KoreaMed CrossRef
  44. Liu SY, Qiao HW, Song TB, Liu XL, Yao YX, Zhao CS, Barret O, Xu SL, Cai YN, Tamagnan GD, Sossi V, Lu J, Chan P (2022) Brain microglia activation and peripheral adaptive immunity in Parkinson's disease: a multimodal PET study. J Neuroinflammation 19:209. doi: 10.1186/s12974-022-02574-z
    Pubmed KoreaMed CrossRef
  45. Lonnemann N, Hosseini S, Marchetti C, Skouras DB, Stefanoni D, D'Alessandro A, Dinarello CA, Korte M (2020) The NLRP3 inflammasome inhibitor OLT1177 rescues cognitive impairment in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 117:32145-32154. doi: 10.1073/pnas.2009680117
    Pubmed KoreaMed CrossRef
  46. McGeer PL, Itagaki S, Boyes BE, McGeer EG (1988) Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology 38:1285-1291. doi: 10.1212/wnl.38.8.1285
    Pubmed CrossRef
  47. Moehle MS, Webber PJ, Tse T, Sukar N, Standaert DG, DeSilva TM, Cowell RM, West AB (2012) LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci 32:1602-1611. doi: 10.1523/JNEUROSCI.5601-11.2012. Erratum in: (2022) J Neurosci 42:938. doi: 10.1523/JNEUROSCI.2507-21.2021
    Pubmed KoreaMed CrossRef
  48. Moon SH, Kwon Y, Huh YE, Choi HJ (2022) Trehalose ameliorates prodromal non-motor deficits and aberrant protein accumulation in a rotenone-induced mouse model of Parkinson's disease. Arch Pharm Res 45:417-432. doi: 10.1007/s12272-022-01386-2
    Pubmed CrossRef
  49. Moustafa AA, Chakravarthy S, Phillips JR, Gupta A, Keri S, Polner B, Frank MJ, Jahanshahi M (2016) Motor symptoms in Parkinson's disease: a unified framework. Neurosci Biobehav Rev 68:727-740. doi: 10.1016/j.neubiorev.2016.07.010
    Pubmed CrossRef
  50. Nader MA, Ateyya H, El-Shafey M, El-Sherbeeny NA (2018) Sitagliptin enhances the neuroprotective effect of pregabalin against pentylenetetrazole-induced acute epileptogenesis in mice: Implication of oxidative, inflammatory, apoptotic and autophagy pathways. Neurochem Int 115:11-23. doi: 10.1016/j.neuint.2017.10.006
    Pubmed CrossRef
  51. Nakamura K, Sakai S, Tsuyama J, Nakamura A, Otani K, Kurabayashi K, Yogiashi Y, Masai H, Shichita T (2021) Extracellular DJ-1 induces sterile inflammation in the ischemic brain. PLoS Biol 19:e3000939. doi: 10.1371/journal.pbio.3000939
    Pubmed KoreaMed CrossRef
  52. Nakaoku Y, Saito S, Yamamoto Y, Maki T, Takahashi R, Ihara M (2019) The dipeptidyl peptidase-4 inhibitor linagliptin ameliorates high-fat induced cognitive decline in tauopathy model mice. Int J Mol Sci 20:2539. doi: 10.3390/ijms20102539
    Pubmed KoreaMed CrossRef
  53. Panicker N, Kam TI, Wang H, Neifert S, Chou SC, Kumar M, Brahmachari S, Jhaldiyal A, Hinkle JT, Akkentli F, Mao X, Xu E, Karuppagounder SS, Hsu ET, Kang SU, Pletnikova O, Troncoso J, Dawson VL, Dawson TM (2022) Neuronal NLRP3 is a parkin substrate that drives neurodegeneration in Parkinson's disease. Neuron 110:2422-2437.e9. doi: 10.1016/j.neuron.2022.05.009
    Pubmed KoreaMed CrossRef
  54. Pariyar R, Bastola T, Lee DH, Seo J (2022) Neuroprotective effects of the DPP4 inhibitor vildagliptin in in vivo and in vitro models of Parkinson's disease. Int J Mol Sci 23:2388. doi: 10.3390/ijms23042388
    Pubmed KoreaMed CrossRef
  55. Peng L, Zhou Y, Jiang N, Wang T, Zhu J, Chen Y, Li L, Zhang J, Yu S, Zhao Y (2020) DJ-1 exerts anti-inflammatory effects and regulates NLRX1-TRAF6 via SHP-1 in stroke. J Neuroinflammation 17:81. doi: 10.1186/s12974-020-01764-x
    Pubmed KoreaMed CrossRef
  56. Russo I, Berti G, Plotegher N, Bernardo G, Filograna R, Bubacco L, Greggio E (2015) Leucine-rich repeat kinase 2 positively regulates inflammation and down-regulates NF-κB p50 signaling in cultured microglia cells. J Neuroinflammation 12:230. doi: 10.1186/s12974-015-0449-7. Erratum in: (2016) J Neuroinflammation 13:70. doi: 10.1186/s12974-016-0535-5
    Pubmed KoreaMed CrossRef
  57. Safar MM, Abdelkader NF, Ramadan E, Kortam MA, Mohamed AF (2021) Novel mechanistic insights towards the repositioning of alogliptin in Parkinson's disease. Life Sci 287:120132. doi: 10.1016/j.lfs.2021.120132
    Pubmed CrossRef
  58. Sedighi M, Baluchnejadmojarad T, Roghani M (2021) Linagliptin protects human SH-SY5Y neuroblastoma cells against amyloid-β cytotoxicity via the activation of Wnt1 and suppression of IL-6 release. Iran Biomed J 25:343-348. doi: 10.52547/ibj.25.5.343
    CrossRef
  59. Shi S, Kanasaki K, Koya D (2016) Linagliptin but not Sitagliptin inhibited transforming growth factor-β2-induced endothelial DPP-4 activity and the endothelial-mesenchymal transition. Biochem Biophys Res Commun 471:184-190. doi: 10.1016/j.bbrc.2016.01.154
    Pubmed CrossRef
  60. Shirakawa J, Amo K, Ohminami H, Orime K, Togashi Y, Ito Y, Tajima K, Koganei M, Sasaki H, Takeda E, Terauchi Y (2011) Protective effects of dipeptidyl peptidase-4 (DPP-4) inhibitor against increased β cell apoptosis induced by dietary sucrose and linoleic acid in mice with diabetes. J Biol Chem 286:25467-25476. doi: 10.1074/jbc.M110.217216
    Pubmed KoreaMed CrossRef
  61. Siddiqui N, Ali J, Parvez S, Najmi AK, Akhtar M (2023) Neuroprotective role of DPP-4 inhibitor Linagliptin against neurodegeneration, neuronal insulin resistance and neuroinflammation induced by intracerebroventricular streptozotocin in rat model of Alzheimer's disease. Neurochem Res 48:2714-2730. doi: 10.1007/s11064-023-03924-w
    Pubmed CrossRef
  62. Sliter DA, Martinez J, Hao L, Chen X, Sun N, Fischer TD, Burman JL, Li Y, Zhang Z, Narendra DP, Cai H, Borsche M, Klein C, Youle RJ (2018) Parkin and PINK1 mitigate STING-induced inflammation. Nature 561:258-262. doi: 10.1038/s41586-018-0448-9
    Pubmed KoreaMed CrossRef
  63. Troncoso-Escudero P, Parra A, Nassif M, Vidal RL (2018) Outside in: unraveling the role of neuroinflammation in the progression of Parkinson's disease. Front Neurol 9:860. doi: 10.3389/fneur.2018.00860
    Pubmed KoreaMed CrossRef
  64. Trzaskalski NA, Fadzeyeva E, Mulvihill EE (2020) Dipeptidyl peptidase-4 at the interface between inflammation and metabolism. Clin Med Insights Endocrinol Diabetes 13:1179551420912972. doi: 10.1177/1179551420912972
    Pubmed KoreaMed CrossRef
  65. University College, London (2016) Trial of Exenatide for Parkinson’s disease. National Library of Medicine. https://classic.clinicaltrials.gov/ct2/show/NCT01971242 Accessed 1 December 2023.
  66. University Hospital, Toulouse (2023) Study to evaluate the effect of lixisenatide in patient with Parkinson’s disease (LixiPark). National Library of Medicine. https://clinicaltrials.gov/study/NCT03439943 Accessed 1 December 2023.
  67. Vaillancourt DE, Mitchell T (2020) Parkinson's disease progression in the substantia nigra: location, location, location. Brain 143:2628-2630. doi: 10.1093/brain/awaa252
    Pubmed KoreaMed CrossRef
  68. Vroon A, Drukarch B, Bol JG, Cras P, Brevé JJ, Allan SM, Relton JK, Hoogland PV, Van Dam AM (2007) Neuroinflammation in Parkinson's patients and MPTP-treated mice is not restricted to the nigrostriatal system: microgliosis and differential expression of interleukin-1 receptors in the olfactory bulb. Exp Gerontol 42:762-771. doi: 10.1016/j.exger.2007.04.010
    Pubmed CrossRef
  69. Wiciński M, Górski K, Wódkiewicz E, Walczak M, Nowaczewska M, Malinowski B (2020) Vasculoprotective effects of vildagliptin. Focus on atherogenesis. Int J Mol Sci 21:2275. doi: 10.3390/ijms21072275
    Pubmed KoreaMed CrossRef
  70. Wiciński M, Wódkiewicz E, Słupski M, Walczak M, Socha M, Malinowski B, Pawlak-Osińska K (2018) Neuroprotective activity of sitagliptin via reduction of neuroinflammation beyond the incretin effect: focus on Alzheimer's disease. Biomed Res Int 2018:6091014. doi: 10.1155/2018/6091014
    Pubmed KoreaMed CrossRef
  71. Xie D, Wang Q, Huang W, Zhao L (2023) Dipeptidyl-peptidase-4 inhibitors have anti-inflammatory effects in patients with type 2 diabetes. Eur J Clin Pharmacol 79:1291-1301. doi: 10.1007/s00228-023-03541-0
    Pubmed CrossRef
  72. Xu E, Boddu R, Abdelmotilib HA, Sokratian A, Kelly K, Liu Z, Bryant N, Chandra S, Carlisle SM, Lefkowitz EJ, Harms AS, Benveniste EN, Yacoubian TA, Volpicelli-Daley LA, Standaert DG, West AB (2022) Pathological α-synuclein recruits LRRK2 expressing pro-inflammatory monocytes to the brain. Mol Neurodegener 17:7. doi: 10.1186/s13024-021-00509-5
    Pubmed KoreaMed CrossRef
  73. Xue Y, Li R, Fang P, Ye ZQ, Zhao Y, Zhou Y, Zhang KQ, Li L (2021) NLRP3 inflammasome inhibitor cucurbitacin B suppresses gout arthritis in mice. J Mol Endocrinol 67:27-40. doi: 10.1530/JME-20-0305
    Pubmed CrossRef
  74. Yossef RR, Al-Yamany MF, Saad MA, El-Sahar AE (2020) Neuroprotective effects of vildagliptin on drug induced Alzheimer's disease in rats with metabolic syndrome: role of hippocampal klotho and AKT signaling pathways. Eur J Pharmacol 889:173612. doi: 10.1016/j.ejphar.2020.173612
    Pubmed CrossRef
  75. Yu HY, Sun T, Wang Z, Li H, Xu D, An J, Wen LL, Li JY, Li W, Feng J (2023) Exendin-4 and linagliptin attenuate neuroinflammation in a mouse model of Parkinson's disease. Neural Regen Res 18:1818-1826. doi: 10.4103/1673-5374.360242
    Pubmed KoreaMed CrossRef
  76. Zeng W, Wu D, Sun Y, Suo Y, Yu Q, Zeng M, Gao Q, Yu B, Jiang X, Wang Y (2021) The selective NLRP3 inhibitor MCC950 hinders atherosclerosis development by attenuating inflammation and pyroptosis in macrophages. Sci Rep 11:19305. doi: 10.1038/s41598-021-98437-3
    Pubmed KoreaMed CrossRef
  77. Zhang G, Kim S, Gu X, Yu SP, Wei L (2020) DPP-4 inhibitor linagliptin is neuroprotective in hyperglycemic mice with stroke via the AKT/mTOR pathway and anti-apoptotic effects. Neurosci Bull 36:407-418. doi: 10.1007/s12264-019-00446-w
    Pubmed KoreaMed CrossRef
  78. Zhou J, Yang R, Zhang Z, Liu Q, Zhang Y, Wang Q, Yuan H (2019a) Mitochondrial protein PINK1 positively regulates RLR signaling. Front Immunol 10:1069. doi: 10.3389/fimmu.2019.01069
    Pubmed KoreaMed CrossRef
  79. Zhou X, Wang W, Wang C, Zheng C, Xu X, Ni X, Hu S, Cai B, Sun L, Shi K, Chen B, Zhou M, Chen G (2019b) DPP4 inhibitor attenuates severe acute pancreatitis-associated intestinal inflammation via Nrf2 signaling. Oxid Med Cell Longev 2019:6181754. doi: 10.1155/2019/6181754
    Pubmed KoreaMed CrossRef
  80. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, Stoessl AJ, Pfeiffer RF, Patenge N, Carbajal IC, Vieregge P, Asmus F, Müller-Myhsok B, Dickson DW, Meitinger T, Strom TM, Wszolek ZK, Gasser T (2004) Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 44:601-607. doi: 10.1016/j.neuron.2004.11.005
    Pubmed CrossRef

Stats or Metrics

Share this article on :

  • line