Skip to main content

REVIEW article

Front. Neurosci., 17 December 2020
Sec. Neuropharmacology
This article is part of the Research Topic Adenosinergic System and Central Nervous System Disorders View all 11 articles

Caffeine and Parkinson’s Disease: Multiple Benefits and Emerging Mechanisms

  • 1Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
  • 2State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
  • 3Department of Biochemistry, Medical College, Jiaxing University, Jiaxing, China

Parkinson’s disease (PD) is the second most common neurodegenerative disorder, characterized by dopaminergic neurodegeneration, motor impairment and non-motor symptoms. Epidemiological and experimental investigations into potential risk factors have firmly established that dietary factor caffeine, the most-widely consumed psychoactive substance, may exerts not only neuroprotective but a motor and non-motor (cognitive) benefits in PD. These multi-benefits of caffeine in PD are supported by convergence of epidemiological and animal evidence. At least six large prospective epidemiological studies have firmly established a relationship between increased caffeine consumption and decreased risk of developing PD. In addition, animal studies have also demonstrated that caffeine confers neuroprotection against dopaminergic neurodegeneration using PD models of mitochondrial toxins (MPTP, 6-OHDA, and rotenone) and expression of α-synuclein (α-Syn). While caffeine has complex pharmacological profiles, studies with genetic knockout mice have clearly revealed that caffeine’s action is largely mediated by the brain adenosine A2A receptor (A2AR) and confer neuroprotection by modulating neuroinflammation and excitotoxicity and mitochondrial function. Interestingly, recent studies have highlighted emerging new mechanisms including caffeine modulation of α-Syn degradation with enhanced autophagy and caffeine modulation of gut microbiota and gut-brain axis in PD models. Importantly, since the first clinical trial in 2003, United States FDA has finally approved clinical use of the A2AR antagonist istradefylline for the treatment of PD with OFF-time in Sept. 2019. To realize therapeutic potential of caffeine in PD, genetic study of caffeine and risk genes in human population may identify useful pharmacogenetic markers for predicting individual responses to caffeine in PD clinical trials and thus offer a unique opportunity for “personalized medicine” in PD.

Introduction

Parkinson’s disease (PD) is the second most common neurodegenerative disorder characterized by clinical presentation of motor impairment and non-motor symptoms. The hallmarks of PD pathology are selective degeneration of dopaminergic neurons in the midbrain and by the prominent α-Syn-containing proteinaceous inclusions, Lewy body (Dauer and Przedborski, 2003; Kalia and Kalia, 2015; Postuma and Berg, 2016; Wong and Krainc, 2017). PD affect 1% of the world population age 65 and older, but with the aging population and increases in life expectancy in modern society, the number of PD cases is projected to double by 2030 (Dorsey et al., 2007; Lees et al., 2009). Dopamine replacement, such as L-dopa, is the mainstay treatment to control motor symptoms (Rascol et al., 2002; Lang and Obeso, 2004; Goetz et al., 2005), but chronic L-dopa treatment is associated with loss of its efficacy and the onset of debilitating motor complications (dyskinesia, wearing-off and ON-OFF) (Ahlskog and Muenter, 2001). Furthermore, with the discovery of α-Syn as the main pathogenesis gene and the main component of Lewy body (Lashuel et al., 2013; Kalia and Kalia, 2015; Wong and Krainc, 2017), wide-spread distribution of α-Syn aggregates in the brain provide the pathological basis for non-motor symptoms of PD (e.g., cognitive dysfunction, fatigue, balance impairment, sleep disturbance, autonomic dysfunction) which are increasingly recognized as a key component of the disease (Chaudhuri et al., 2006; Postuma and Berg, 2016). Importantly, despite of the extensive studies, there are no successful treatments currently available that can slow or halt this chronic neurodegeneration (Shoulson, 1998; Mattson, 2004; Olanow, 2004; Lopez-Diego and Weiner, 2008; Mestre et al., 2009).

In the absence of an effective disease modifying treatment to slow down or hold PD course, epidemiological and experimental investigations into potential risk factors (such as dietary factor caffeine) that may modify PD pathology and confer therapeutic benefits become compelling. Caffeine is probably the most widely psychoactive substance and regularly consumed by >50% of the world’s adult population, largely for its psychostimulant (and cognitive enhancement) effect, reducing fatigue and enhancing performance. The regular human consumption of caffeine was previously estimated to be 70–350 mg/person/day or 5 to 8 mg/kg/day (equivalent to three couples of coffee) which is absorbed by the small intestine within 45 min of ingestion and produces peak plasma concentration of 0.25 to 2 mg/l (or approximately 1 to 10 μM) within 1–2 h, distributes throughout all bodily tissues and produces overall psychostimulant effects, plasma caffeine levels are usually in the range of 2–10 mg/L in coffee drinkers (Nehlig et al., 1992; Fredholm et al., 1999b). Caffeine-containing drinks, such as coffee, tea, and cola, are very popular; as of 2014, 85% of American adults consumed some form of caffeine daily, consuming 164 mg on average (Mitchell et al., 2014). Caffeine intake doses determined not to be associated with adverse effects by Health Canada (comparators: 400 mg/day for adults [10 g for lethality], 300 mg/day for pregnant women, and 2.5 mg/kg/day for children and adolescents), 2.5 mg caffeine/kg body weight/day remains an appropriate recommendation (Wikoff et al., 2017). In healthy adults, caffeine’s half-life is between 3 and 7 h, caffeine is metabolized in the liver by the cytochrome P450 oxidase enzyme system, in particular, by the CYP1A2 isozyme, into three dimethylxanthines, including paraxanthine, theobromine, and theophylline, each of which has its own effects on the human body. Caffeine was found to present in all rat tissues after administration for 10 days and accumulated for 25 days. The caffeine level was high in brain, liver and kidney and widely distributed and accumulated in these organs (Che et al., 2012). Because caffeine is both water- and lipid-soluble, it readily crosses the blood brain barrier. Once in the brain, caffeine may act at multiple molecular targets to produce complex pharmacological actions, ranging from adenosine receptor antagonism, to phosphodiesterase inhibition, to GABA receptor blockade and calcium release (Fredholm et al., 1999a). However, genetic knockout studies have demonstrated that caffeine’s primary action in the brain is as an antagonist of adenosine receptors (mainly A2AR) (Chen et al., 2013).

In this review, we first summarized the neuroprotective, motor and cognitive benefits of caffeine in PD patients and PD models. We then described the potential mechanisms underlying caffeine’s protective effects, including modulation of neuroinflammation and of newly emerging mechanisms associated with autophagy and gut microbiota. Lastly, we discussed possible genetic polymorphisms of caffeine-associated genes in influencing caffeine drug responses and its clinical implications.

Multiple Benefits of Caffeine in PD Treatment

Potential Neuroprotective Effects of Caffeine in PD Patients and PD Models

The first evidence for the potential neuroprotective effect of caffeine came from the Honolulu Heart Program, a large prospective study of 8004 Japanese-American men over a 30 years follow-up study. The study revealed that daily consumption of 784 mg/kg or more of coffee during the mid-life reduce the risk for developing PD at age of 65-year old by 5-folds compared to non-coffee drinkers after age- and smoking-adjustment (Ross et al., 2000). This inverse relationship between consumption of caffeine and the risk of developing PD was further supported by the Health Professionals’ Follow-Up Study and the Nurses’ Health Study – involving 47,351 men and 88,565 women, and subsequently also by at least three additional large prospective studies, including Finnish Mobile Clinic Health Examination Survey (Saaksjarvi et al., 2008) and the NeuroGenetics Research Consortium (Powers et al., 2008) and Danish case-control study involving idiopathic PD (Kyrozis et al., 2013). A meta-analysis of 13 study involving total 901,764 participants for coffee intake found a non-linear relationship was found between coffee intake and PD risk, with maximum protection effect at approximately 3 cups/day (Qi and Li, 2014). Systemic analysis of 120 observation studies have firmly established that regular human consumption of caffeine is associated with reduced risk for PD (Ross et al., 2000; Ascherio et al., 2001, 2003; Saaksjarvi et al., 2008; Grosso et al., 2017) and does not impose significant adverse effects on the cardiovascular system, bone status, or the incidence of cancer (Fredholm et al., 1999b; Winkelmayer et al., 2005; Higdon and Frei, 2006; van Dam et al., 2006; Cadden et al., 2007; Daly, 2007). Interestingly, this inverse relationship of coffee consumption and risker for developing PD is largely attributed to caffeine since that the consumption of caffeinated (but not decaffeinated) coffee is associated with the reduced risk of developing PD in the Health Professional Follow-up Study (Ascherio et al., 2001). Notably, this inverse relationship is strong and consistent in men in the Health Professional Follow-up Study (Grosso et al., 2017) and in postmenopausal women who never used hormone replacement therapy in the Cancer Prevention Study II Nutrition Cohort (CPS II-Nutrition) (Palacios et al., 2012), but uncertain in women and postmenopausal women ever users of hormone replacement therapy. Thus, caffeine and estrogen interaction modify the risk of developing PD. More recently, the Harvard Biomarkers Study (HBS) conducted a cross-sectional, case-control study in 566 subjects consisting of idiopathic PD patients and healthy controls, which highlighted the robustness of lower caffeine intake and plasma urate levels as factors inversely associated with idiopathic PD (Bakshi et al., 2020).

Furthermore, the neuroprotective effect of caffeine from epidemiological investigation is further supported by mounting evidence from animal studies demonstrating that caffeine confers neuroprotection against dopaminergic neurodegeneration in neurotoxin PD models using mitochondrial toxins (MPTP, 6-OHDA and rotenone) (Chen et al., 2001; Ikeda et al., 2002; Xu et al., 2002; Aguiar et al., 2006) and α-Syn transmission mouse model through intracerebral injection of α-Syn fibers (Luan et al., 2018). For example, acute or chronic treatment with caffeine attenuates MPTP-induced dopaminergic neurotoxicity and neurodegeneration (Chen et al., 2001; Xu et al., 2002). It is interesting to note that in a chronic MPTP infusion model of PD caffeine can still confer protection against dopamine neurodegeneration even when caffeine was administered after the onset of the neurodegenerative process (i.e., 14 days after MPTP infusion) (Sonsalla et al., 2012). Furthermore, our recent study demonstrated that caffeine can protect against A53T mutant α-Syn induced pathological alterations in intact animals using the α-Syn fibril model of PD, an effect associated with the enhanced activity of autophagy (specifically macroautophagy and CMA) (Luan et al., 2018) (see below for the more discussion).

Importantly, similar to caffeine, pharmacological blockade or genetic deletion of adenosine A2A receptor (A2AR, the main pharmacological target of caffeine in the brain) also protected against dopaminergic neurodegeneration in animal models of PD (Chen et al., 2001; Ikeda et al., 2002; Kachroo and Schwarzschild, 2012), suggesting that the protective effects of caffeine are likely due to its action on the A2AR. Moreover, recent studies also show that A2AR modulated α-Syn aggregation and toxicity in SH-SY5Y cells (Ferreira et al., 2017a), and A2AR blockade rescued synaptic and cognitive deficits in α-Syn transgenic mice model of PD (Ferreira et al., 2017b). These animal studies provide a neurobiological basis for the inverse relationship between caffeine consumption and the reduced risk of developing PD, and support the clinical potential for caffeine and A2AR antagonists as a disease-modifying drug target for PD.

Two important caveats need to be taken into the consideration. First, caffeine exerts synergistic neuroprotection with eicosanoyl-5-hydroxytryptamide (EHT) in animal models of PD, co-administration of these two compounds of coffee orally have synergistic effects in protecting the brain against α-Syn mediated toxicity through maintenance of protein phosphatase 2A in an active state (Yan et al., 2018). Second, in methamphetamine-induced neurotoxicity both in vitro and in vivo, caffeine has been shown to increase toxicity of methamphetamine in SH-SY5Y neuroblastoma cell line through inhibition of autophagy (Pitaksalee et al., 2015) and potentiate 3,4-methylenedioxymethamphetamine (MDMA) -induced dopamine neuron degeneration in substantia nigra pars compacta, possibly involving an increase in dopamine release and excess ROS generation (Sinchai et al., 2011; Frau et al., 2016). Therefore, caffeine has been consistently shown to exert a neuroprotective effect in multiple neurotoxin (including MPTP, 6-OHDA, rotenone) and alpha-synuclein models of PD, but has been reported to exacerbate methamphetamine-induced neurotoxicity. Additional studies are needed to clarify these neurotoxicity context-dependent effects of caffeine.

Motor Benefit of Caffeine in PD Patients and PD Models

The symptomatic effect of caffeine in PD was first tested in 1970s (Shoulson and Chase, 1975), but has been revisited by several clinical studies recently. The motor benefit of caffeine were documented in a pilot open-label, 6-week dose-escalation study (Altman et al., 2011) and a 6-week randomized controlled trial of caffeine (200–400 mg daily) involving 61 PD patients (Postuma et al., 2012). These clinical studies suggest that caffeine improved objective motor deficits in PD with the reduced total Unified PD Rating Scale score and the objective motor component. Furthermore, coffee consumption (>336 mg/day) is associated with the reduced hazard ratio for the development of dyskinesia compared with subjects who consumed <112 mg/day in the Comparison of the Agonist Pramipexole with Levodopa on Motor Complications of Parkinson’s Disease (CALM-PD) and CALM Cohort extension studies (Wills et al., 2013). Based on these positive findings, caffeine was recently investigated for motor and disease-modification involving 121 PD patients PD in a phase 3, 5-years (planned), two-arm, double-blind RCT, with a primary outcome focused on motor symptoms and disease-modification as a secondary outcome1. Unfortunately, with the primary outcome analysis after 6 months demonstrating no significant symptomatic benefit of caffeine (200 mg twice daily) (Postuma et al., 2017), the study was terminated earlier than the planned.

Based on the concentrated expression of A2AR in the striatum and the A2AR is the key molecular target of caffeine, caffeine (and A2AR antagonists) has been proposed 20 years ago to improve motor activity in PD (Garcao et al., 2013; Morato et al., 2017). Indeed, preclinical studies using rodent and non-human primate models of PD demonstrate motor benefits caffeine and A2AR antagonists in PD (Richardson et al., 1997; Chen, 2003; Schwarzschild et al., 2006; Jenner et al., 2009), leading to clinical pursuit of A2AR antagonists as a leading non-dopaminergic treatment for motor deficits in PD. Since 2001, more than 25 clinical trials were conducted to evaluate the safety and clinical efficacy of A2AR antagonists in PD patients; among these, eight double blind, placebo controlled, phase IIb and III trials of istradefylline (KW-6002, > 4000 PD patients), one phase IIb trial with preladenant (SCH 420814, 253 PD patients), one phase IIb trial with tozadenant (337 PD patients), all reported motor benefits in advanced PD patients as an add-on therapy with L-DOPA (Hauser et al., 2011). The culmination of the two decade-long clinical studies of the effects of istradefylline in more than 4,000 PD finally led to the U.S. Food and Drug Administration (FDA) approval of the A2AR antagonist Nourianz® (istradefylline) developed by Kyowa Hakko-Kirin Inc., Japan, as an add-on treatment to levodopa in Parkinson’s disease (PD) with “OFF” episodes in August 2019. Istradefylline is the first non-dopaminergic drug approved by United States FDA for PD in the last two decades and this approval paves the way to foster novel therapeutic opportunities for A2AR antagonists including caffeine for neuroprotection or reversal of mood and cognitive deficits in PD.

Caffeine and Cognitive Improvement in PD

Parkinson’s disease is primarily characterized by cardinal motor symptoms but cognitive changes also occur both in the early and later stages of the disease process. In fact, ∼30% of PD patients have dementia and an additional 25% of non-demented PD patients developed mild cognitive impairments that are characterized by frontostriatal cognitive deficits such as alterations in executive function, attention, working and episodic memory (Majbour and El-Agnaf, 2017). Convergent evidence from human and animal studies supports the existence of DA-dependent cognitive deficits in PD (Lewis et al., 2003). The cognitive symptoms seen early in PD include the decreased executive function (e.g., planning and decision making), working memory deficit and impaired procedural learning, leading to cognitive inflexibility that are largely attributed to the frontostriatal dysfunction (Knowlton et al., 1996; Kehagia et al., 2010). Early cognitive deficits are extremely troubling to patients and reduce their quality of life. Currently there is no effective treatment for mild cognitive impairments in PD. In this regard, it is important to note that at least six longitudinal studies support an inverse relationship between caffeine consumption and decreased memory impairments associated with aging as well as a reduced risk of developing dementia and Alzheimer’s disease, including the Maastricht Aging Study (Hameleers et al., 2000; van Boxtel et al., 2003), the Canadian Study of Health and Aging (CSHA) (Lindsay et al., 2002), the FINE study (van Gelder et al., 2007), the French Three Cities study (Ritchie et al., 2007), the Cardiovascular risk factors, Aging and Dementia (CAIDE) study (Eskelinen et al., 2009), the Honolulu-Asia Aging Study (Gelber et al., 2011). Furthermore, in a cross-sectional study involving 196 early-stage, treatment-naïve PD patients, coffee drinking was significantly associated with a reduced severity of the mood/cognition domain of NMSS in patients with PD (p = 0.003) (Cho et al., 2018). These epidemiological findings raise the possibility of caffeine as therapeutic treatment for cognitive impairments in PD.

Indeed, preclinical studies with A2AR antagonist effect on cognition in normal and MPTP-treated non-human primates (NHP) provide the experimental evidence that A2AR antagonists including caffeine can improve cognitive impairments in PD models (Chen et al., 2013; Chen, 2014). Recent preclinical studies in rodents and non-human primates demonstrated that A2AR antagonists not only enhance working memory (Zhou et al., 2009), reversal learning (Wei et al., 2011), set-shifting (Mingote et al., 2008), goal-directed behavior (Li et al., 2016), and Pavlovian conditioning (Wei et al., 2014) in normal animals, but also reverse working memory impairments in animal models of PD (Ko et al., 2016) and Huntington’s disease (Li et al., 2015), traumatic brain injury (Ning et al., 2013, 2015, 2019) as well as Alzheimer’s disease (AD) (Dall’Igna et al., 2007; Cunha and Agostinho, 2010; Laurent et al., 2014; Faivre et al., 2018). Furthermore, we recently demonstrated a pro-cognitive effect in normal as well as MPTP-treated Cynomolgus monkeys (Li et al., 2018b). The demonstrated treatment paradigm of the A2AR antagonist KW6002 for spatial working memory enhancement in non-human primate model of PD provide required preclinical data to facilitate the design of clinical trial of A2AR antagonists including caffeine for cognitive benefit in PD patients (Li et al., 2018b). Notably, recent clinical trials of A2AR antagonists for motor benefits in PD did not evaluate their possible effects on cognitive impairment in PD patients (Chase et al., 2003; Aarsland et al., 2010). With the approval of istradefylline, it will now be possible to evaluate the ability of A2AR antagonists to reverse cognitive deficits in PD patients in clinical phase IV trials.

Mechanisms of Neuroprotection by Caffeine in PD

Multiple mechanisms have been proposed to account for the neuroprotective effects of caffeine, including modulation of glutamatergic excitotoxicity and neuroinflammation via adenosine receptors (Chen et al., 2013). Furthermore, recent investigation into the autophagy and gut microbiota in PD pathogenesis raise the exciting possibilities that caffeine may modify autophagy (through metabolism-related action of caffeine) and gut microbiome (with caffeine direct action on gut microbiota) to influence PD development.

Caffeine Modulates Neuroinflammation in PD

Neuroinflammation is critically involved in the pathogenesis of PD. Increasing evidence showed that neuroinflammation response regulated by reactive microglia played an important role in the neurodegeneration of DA neurons (Hirsch and Hunot, 2009; Tansey and Goldberg, 2010; Hirsch et al., 2012). α-Syn, in extracellular aggregated form, can bind to Toll-like receptor 2 (TLR2), CD11b receptors and integrin β1 subunit on microglia to trigger massive microglial activation and neuroinflammation, contributing to consequent neuronal death (Lee et al., 2010; Tansey and Goldberg, 2010; Fellner et al., 2013; Yasuda et al., 2013; Sacino et al., 2014). The involvement of neuroinflammation in PD was further suggested by the observation of the increased number of reactive microglial cells and an upregulation of major histocompatibility complex class II (MHC-II) in PD patients (McGeer et al., 1988). Caffeine can exert an anti-neuroinflammatory effect under various pathological conditions. Caffeine administration (daily intraperitoneal injection) reduces lipopolysaccharide (LPS)-induced microglia activation in three regions of the hippocampus, in a dose-dependent manner (Brothers et al., 2010) and abrogate LPS-induced neuroinflammation, and synaptic dysfunction in adult mouse brains (Badshah et al., 2019). As a critical neuroprotective factor in PD, caffeine may control microglia-mediated neuroinflammatory response associated PD (Madeira et al., 2017). Indeed, daily intraperitoneal administration of caffeine attenuates microglia reactivity and prevents blood-brain barrier (BBB) dysregulation in the MPTP mouse model, leading to decreased dopaminergic neuronal loss (Xu et al., 2002; Chen et al., 2008). Furthermore, even when introduced in the later phases of the neurodegenerative process, caffeine is also able to attenuate the inflammatory process and microglial cell expression of CD68 (a marker of reactive microglia), which suggests its ability to arrest or delay neuroinflammation and neurodegeneration (Chen et al., 2008). Consistent with this view, by using an α-Syn fibril model of PD, we recently found that chronic caffeine treatment attenuated α-Syn-induced microglial activation and astrogliosis in the striatum in mice (Luan et al., 2018). In addition, caffeine has been shown to protect dopaminergic neurons by activation of the anti-oxidative signaling pathways, such as the nuclear factor erythroid 2-related factor 2 (Nrf2)-Keap1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1 alpha) (Zhou et al., 2019).

The mechanisms underlying anti-neuroinflammation by caffeine may involve the antagonism of the A2AR, the major molecular target (Chen et al., 2001; Kalda et al., 2006). A2ARs affords neuroprotection through the control of microglia reactivity and neuroinflammation. Notably, pharmacological blockade or genetic deletion of A2AR produces similar anti-neuroinflammatory and neuroprotective effects as with caffeine in several experimental models of PD (Chen et al., 2001; Ikeda et al., 2002; Kalda et al., 2006; Hu et al., 2016; Luan et al., 2018). For example, enhanced reactive astrogliosis and NF-κB p65 activation around the injection site in hippocampus in an α-Syn transmission mouse model of PD, and these α-Syn-triggered neuroinflammatory responses were largely prevented in A2AR KO mice (Hu et al., 2016). In the well-established α-Syn fibril model of PD, chronic caffeine treatment largely reverted the α-Syn-induced microglial activation and astrogliosis in the striatum in mice (Brambilla et al., 2003; Boison et al., 2010; Luan et al., 2018). Moreover, in A2AR antagonists also control neuroinflammation (through p38), of synaptopathy (Canas et al., 2009) and β-amyloid processing (Cao et al., 2009) in AD models. Thus, caffeine may exert anti-neuroinflammation and neuroprotection effect in PD by targeting the A2ARs.

Caffeine May Modulate PD Pathology by Regulating Autophagy Activity

Autophagy is a high conserved defense and protective mechanism in eukaryotic cell to achieve degradation of abnormal proteins and damaged organelles by three types of autophagy processes: microautophagy, macroautophagy and chaperone-mediated autophagy (CMA). PD is essentially a protein misfolding disease or conformational disease; characterized by abnormal accumulation and aggregation of misfolded α-Syn (Kalia and Kalia, 2015). Increasing evidence demonstrates that aberrant regulation of autophagy contributes to the aggregation of α-Syn and α-Syn-induced neurodegeneration in PD (Ebrahimi-Fakhari et al., 2012; Poehler et al., 2014; Xilouri et al., 2016; Hou et al., 2020; Senkevich and Gan-Or, 2020). First, several studies found that alteration of autophagy-lysosome pathway activity, including both CMA and macroautophagy, existed in brains of postmortem PD patients and experimental models of PD (Alvarez-Erviti et al., 2010; Cerri and Blandini, 2019; Lamonaca and Volta, 2020; Qin et al., 2020; Wan et al., 2020), revealing the direct correlation between autophagy and pathological process of PD. Second, abnormally aggregated α-Syn inclusions are mainly degraded by autophagy (Webb et al., 2003) and defects or deficiency of autophagy can lead to accumulation of intracellular misfolded amyloid α-Syn aggregates, thus causally linking autophagy to PD pathological process (Xilouri et al., 2016; Guo et al., 2020). Third, α-Syn overexpression can impede autophagy by reducing autophagosome formation in human Neuroblastoma SH-SY5Y (Nascimento et al., 2020), and contribute to many different pathologies seen in PD (Winslow and Rubinsztein, 2011). Notably, the pathogenic A53T and A30P α-Syn mutants selectively bound to lysosome-associated membrane protein type 2A (LAMP2A) with high affinity, blocked CMA activity at the LAMP2A level, ultimately resulting in complete damage to this pathway (Cuervo et al., 2004). Collectively, autophagy plays an important role in the pathogenesis of PD, thus targeting autophagy may represent a promising strategy for treatment of PD (Rivero-Rios et al., 2016; Moors et al., 2017; Lu et al., 2020).

Both caffeine and A2AR signaling can regulate autophagy activity under different conditions in several cell types (Sinha et al., 2014; Liu et al., 2016). Caffeine can induce macroautophagy caused by a starvation response and confer a cytocidal effect on Zygosaccharomyces bailii food spoilage yeast) in combination with benzoic acid (Winter et al., 2008). Higher concentrations of caffeine (10 mM) enhance autophagic flux in various tumor cell lines (HeLa, SH-SY5Y, and PC12D cells). Caffeine can induce apoptosis by enhancement of autophagy in PC12D cells through PI3K/Akt/mTOR/p70S6 signaling pathway (Saiki et al., 2011). Using both genetic and pharmacological inhibitors of autophagy, researchers directly linked caffeine-induced autophagy with oxidative lipid metabolism both in HepG2 cells and in mice liver; and further demonstrated that autophagy was associated with caffeine-induced hepatic fat clearance in a mouse model of non-alcoholic fatty liver disease, indicating that caffeine surprisingly is a potent stimulator of hepatic autophagic flux (Ray, 2013; Sinha et al., 2014). Accordingly, caffeine-enhanced autophagic flux in hepatic stellate cells was stimulated by endoplasmic reticulum (ER) stress via the IRE1-α signaling pathway, and autophagy trigged by caffeine instigated cell apoptosis (Li et al., 2017). Acute high-caffeine exposure also significantly reduced skeletal myotube diameter by increasing autophagic flux in differentiated C2C12 mouse skeletal myoblasts cells (Bloemberg and Quadrilatero, 2016; Hughes et al., 2017). Notably, caffeine increased autophagy by promoting the calcium-dependent activation of AMP-activated protein kinase (AMPK) in mammalian skeletal muscle cells (Mathew et al., 2014), and prevent skin from oxidative stress-induced senescence through the activation of autophagy. This caffeine-induced autophagy, mainly mitophagy, was mediated by A2AR/SIRT3/AMPK pathway, protecting skin from oxidative stress-induced senescence both in vitro and in vivo models (Li et al., 2018a). Therefore, caffeine also has the potential in protection of skin disease. Consistent with these studies, a recent research found that caffeine directly enhanced autophagy in concentration- and time-dependent manners in primary cultured thymocytes, which was dependent on A2AR signaling (Liu et al., 2019). Taken together, these studies from various cell models and in different tissues and organs in vivo demonstrate that caffeine enhances autophagy which is related to its therapeutic effects on diverse human diseases, including tumors, aging, liver fibrosis, and skin disease.

Using an α-Syn fibril model of PD, we recently have provided the first evidence that caffeine can attenuate abnormal α-Syn aggregation and neurotoxicity by re-establishing autophagy activity in animal models of PD (Luan et al., 2018). Specifically, chronic caffeine treatment did not affect autophagy processes in the normal mice striatum, but did selectively reverse α-Syn-induced defects in macroautophagy and CMA (Luan et al., 2018). Thus, caffeine may represent a novel pharmacological therapy for PD by targeting autophagy pathway. This study collaborates with the previous study showing that caffeine-induced autophagy protected against human prion protein (PrP) peptide (106–126)-triggered apoptosis in a SH-SY5Y neuroblastoma cell line. Therefore, autophagy enhanced by caffeine may be a valid therapeutic strategy for neurodegenerative diseases such as PD and prion (Moon et al., 2014; Corti et al., 2020).

Caffeine May Influence PD Pathology by Modulating Gut Microbiota

The gut microbiota in the human gastrointestinal (GI) tract is estimated to contain 10 times more microbial cells than human cells, and approximately 100–200-times more protein coding genes than the human genome (Qin et al., 2010). The gut microbiota critically influence various aspects of human biology, including the absorption and metabolism of nutrients, vitamins, medications, and toxic compounds; the development and differentiation of the intestinal epithelium and immune system, the maintenance of tissue homeostasis, and the prevention of pathogens invasion (Sommer and Backhed, 2013). The gut microbiota also plays an important role in gut-brain communication, and the neuroimmune system to maintain brain homeostasis, thus influencing brain function and behavior (Carabotti et al., 2015). In healthy subjects, the intestinal microbiota is generally stable over time, but compositional changes might occur following antibiotic usage or dietary modifications (Lozupone et al., 2012).

Mounting evidence suggest that the intestinal microbiota may be the triggering factor of PD pathology. Specifically, the gut microbiota encoded proteins and their metabolites can initiate accumulation of misfolding of α-Syn in the enteric nervous system through molecular mimicry and intestinal mucosal immunoinflammatory mechanisms, which thereafter could act in a prion-like fashion and spread along the gut-brain axis via vagus nerve, eventually leading to the development of PD pathology (Friedland, 2015; Klingelhoefer and Reichmann, 2015; Pellegrini et al., 2018; Ho et al., 2019; Miraglia and Colla, 2019; Cirstea et al., 2020; Zheng et al., 2020). Multiple lines of preclinical and clinical evidences support the role of gut microbiota dysfunction in various aspects of PD pathogenesis: (i) according to the widely accepted Braak Staging hypothesis about the pathogenesis of PD, α-Syn pathology may begin in the intestine and spread to the brain through the vagus nerve. This hypothesis has been supported by two lines of key clinical findings. (a) Idiopathic constipation is one of the strongest risk-factors for PD since up to 80% of PD patients develop gastrointestinal dysfunction, in particular constipation, in the early stages of PD, preceding the onset of motor symptoms by years (Schapira et al., 2017). (b) Full truncal vagotomy is associated with a decreased risk for subsequent PD, suggesting that the vagal nerve may be critically involved in the pathogenesis of PD (Svensson et al., 2015). (ii) The gut microbiota of patients with PD is altered depending on clinical motor phenotype and related to progress of PD; thus the gut microbiome may be a potential PD biomarker (Scheperjans et al., 2015a; Hopfner et al., 2017; Gerhardt and Mohajeri, 2018; Qian et al., 2018; Nuzum et al., 2020; Pietrucci et al., 2020; Ren et al., 2020). (iii) Gut microbiota is necessary to promote α-Syn pathology, neuroinflammation, neurodegeneration, and characteristic motor features in validated mice model of PD, and specific microbial metabolites are sufficient to promote PD symptoms, suggesting a casual and functional role in PD pathogenesis (Sampson et al., 2016; Sun et al., 2018; Klann et al., 2020; Koutzoumis et al., 2020). (iv) The human gut microbiota metabolizes the PD medication Levodopa (L-dopa), potentially reducing drug availability and causing side effects (Maini Rekdal et al., 2019). Collectively, these findings support that gut microbiota is critical contributor to PD pathogenesis and may represent a promising therapeutic target for the treatment of PD (Lubomski et al., 2019).

As the most important environmental and dietary factor of PD, how does long-term drinking caffeine affect the diversity of gut microbiota? Moreover, how do the human intestinal microorganism and their encoded enzymes influence the metabolism of caffeine? Caffeine is initially absorbed in the stomach and small intestine but is further fermented in the colon by gut microbiota (Scheperjans et al., 2015b). Recently, caffeine consumption is reportedly related to the colonic mucosa-associated gut microbiota (Gurwara et al., 2019); long-term coffee intake is associated with fecal microbial composition in humans, and regular consumption of coffee appears to be associated with changes in some intestinal microbiota groups in which caffeine, as the main dietary factor influencing PD development, may play a role (Gonzalez et al., 2020). Intestinal microorganisms also play a role in the metabolism of caffeine as caffeine was degraded in the gut of H. hampei, and that experimental inactivation of the gut microbiota eliminates this activity, suggesting that the detoxification of caffeine in H. hampei is mediated by the insect’s gut microbiota (Ceja-Navarro et al., 2015). Several recent studies have shed light on the relationship between coffee consumption, caffeine and gut microbiota, as well as GI function in PD (Derkinderen et al., 2014; Scheperjans et al., 2015b). Some effects of coffee on the gastrointestinal tract promote gastro-oesophageal reflux, stimulation of gallbladder contraction and an increase of colonic motor activity (Boekema et al., 1999). Coffee consumption is also inversely associated with the prevalence of self-reported constipation (Murakami et al., 2006). Notably, coffee and caffeine regulates the composition and abundance of intestinal flora under different pathologies. For example, coffee caused an increase of anti-inflammatory Bifidobacteria and a decrease of Clostridium spp. and Escherichia coli that invade the gut mucosa in PD (Khokhlova et al., 2012). Furthermore, chronic coffee consumption attenuated the increase in Firmicutes (F)-to-Bacteroidetes (B) ratio and Clostridium Cluster XI normally associated with high-fat feeding and augmented the levels of Enterobacteria (Cowan et al., 2014). Coffee or its components caffeine can also affect the gut microbiome and short-chain fatty acids (SCFAs) profile in Tsumura Suzuki obese diabetes (TSOD) mice and thereby improve hepatic inflammation. Specifically, daily intake of coffee or its components did not repair the gut dysbiosis in TSOD mice, rather, altered the percentages of six microbial genera changed in these mice, including Blautia, Coprococcus, and Prevotella, which have been implicated in inflammation or obesity (Clarke et al., 2012; Nishitsuji et al., 2018). Other studies suggest that the diversity and structure of the gut microbiota is not sensitive to caffeine, however, when the predicted metagenome functional content of the bacterial communities was analyzed, the caffeine treatments did induce a dramatic decrease of the aromatic amino acid decarboxylase gene (Scorza et al., 2019).

Consistent with these findings, our preliminary meta-genome analysis of the gut microbiota of A53T-α-Syn transgenic mice model of PD by chronic intervention of caffeine indicated that chronic caffeine treatment for one month had no significant effect on gastrointestinal function, but apparently normalized the structure and imbalance of the gut microbiota of PD model mice (unpublished data). Additional studies are warranted to study the interaction between caffeine and intestinal flora in the body, and determine whether the beneficial effects of coffee consumption on PD are mediated through the modulation of the microbiota-gut-brain axis.

Genetic Studies on the Interaction Between Caffeine and PD Risk Genes

Epidemiological investigation coupled with genetic analyses of the genetic and environmental interaction in development of PD has provided several important insights into the interaction between caffeine and several genes associated with PD pathogenesis such as NMDA-glutamate-receptor subunit, LRRK2 and estrogen in PD: (i) A recent genome-wide association and interaction study (GWAIS) uncover a complex interplay between genetic (GRIN2A, encoding an NMDA-glutamate-receptor subunit) and environmental factors (caffeine consumption) in etiology of PD, as a PD genetic modifier in inverse association with caffeine intake (Hamza et al., 2011). (ii) A study with patient-control study Swedish population has revealed an association of a single nucleotide polymorphism, GRIN2A_rs4998386, and its interaction with caffeine intake with PD (Yamada-Fowler et al., 2014). Thus, the interaction between caffeine and glutamate receptor genotypes may contribute to the protective effects of coffee drinking/caffeine intake in PD. (iii) Furthermore, a recent case control study of 812 subjects consisting of PD and healthy controls showed that caffeine intake would significantly reduce the risk of PD by 15-folds in those carrying PD gene risk variant (LRRK2 R1628P) (Kumar et al., 2015). Metabolomic analyses of the LRRK2 Cohort Consortium (LCC) samples identified caffeine, its demethylation metabolites, and trigonelline as prominent markers of resistance to PD linked to pathogenic LRRK2 mutations, more so than to idiopathic PD (Crotty et al., 2020). Exploratory analysis on potential interactions of smoking and caffeine intake with 10 genome-wide association studies of SNPs (at or near the SNCA, MAPT, LRRK2, and HLA loci) further reveal that a combined smoking and caffeine intake exposure is associated with a significant interaction with rs2896905 at SLC2A13, near LRRK2 (Gao et al., 2012). In addition, the Parkinson’s Epidemiology and Genetics Association Studies in US (PEGASUS) consortium (involving 3000 subjects of five independent well-characterized patient-control series) uncovered an association between an Adora2a variant (rs7165183 and rs5996696) and a reduced risk of PD (Popat et al., 2011), with the strongest coffee-PD association among those with homozygous carriers of the CYP1A2 polymorphisms and slow metabolizers of caffeine. These genetic studies support the protective effect of caffeine intake on PD through the interaction between caffeine and GRIN2A, LRRK2, and A2AR. These findings raise the exciting possibility of selecting patient subpopulations by these genetic polymorphisms of the GRIN2A, LRRK2, A2AR, and CYP1A1 genes.

Author Contributions

Both authors wrote and revised the manuscript. J-FC carried out the overall responsibility.

Funding

This work was supported by the National Key Research and Development Program of China (Grant No. 2016YFC1306600); Zhejiang Provincial Natural Science Foundation (Grant No. LY18H090012); and Wenzhou Science and Technology Program (Grant No. Y20190083).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Footnotes

  1. ^ https://clinicaltrials.gov/ct2/show/NCT01738178

References

Aarsland, D., Bronnick, K., Williams-Gray, C., Weintraub, D., Marder, K., Kulisevsky, J., et al. (2010). Mild cognitive impairment in Parkinson disease: a multicenter pooled analysis. Neurology 75, 1062–1069. doi: 10.1212/wnl.0b013e3181f39d0e

PubMed Abstract | CrossRef Full Text | Google Scholar

Aguiar, L. M., Nobre, H. V. Jr., Macedo, D. S., Oliveira, A. A., Freitas, R. M., Vasconcelos, S.M., et al. (2006). Neuroprotective effects of caffeine in the model of 6-hydroxydopamine lesion in rats. Pharmacol. Biochem. Behav. 84, 415–419. doi: 10.1016/j.pbb.2006.05.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Ahlskog, J. E., and Muenter, M. D. (2001). Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov. Disord. 16, 448–458. doi: 10.1002/mds.1090

PubMed Abstract | CrossRef Full Text | Google Scholar

Altman, R. D., Lang, A. E., and Postuma, R. B. (2011). Caffeine in Parkinson’s disease: a pilot open-label, dose-escalation study. Mov. Disord. 26, 2427–2431. doi: 10.1002/mds.23873

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez-Erviti, L., Rodriguez-Oroz, M. C., Cooper, J. M., Caballero, C., Ferrer, I., Obeso, J. A., et al. (2010). Chaperone-mediated autophagy markers in Parkinson disease brains. Arch. Neurol. 67, 1464–1472.

Google Scholar

Ascherio, A., Chen, H., Schwarzschild, M. A., Zhang, S. M., Colditz, G. A., and Speizer, F. E. (2003). Caffeine, postmenopausal estrogen, and risk of Parkinson’s disease. Neurology 60, 790–795. doi: 10.1212/01.wnl.0000046523.05125.87

PubMed Abstract | CrossRef Full Text | Google Scholar

Ascherio, A., Zhang, S. M., Hernan, M. A., Kawachi, I., Colditz, G. A., Speizer, F. E., et al. (2001). Prospective study of caffeine consumption and risk of Parkinson’s disease in men and women. Ann. Neurol. 50, 56–63. doi: 10.1002/ana.1052

PubMed Abstract | CrossRef Full Text | Google Scholar

Badshah, H., Ikram, M., Ali, W., Ahmad, S., Hahm, J. R., and Kim, M. O. (2019). Caffeine may abrogate LPS-induced oxidative stress and neuroinflammation by regulating Nrf2/TLR4 in adult mouse brains. Biomolecules 9:719. doi: 10.3390/biom9110719

PubMed Abstract | CrossRef Full Text | Google Scholar

Bakshi, R., Macklin, E. A., Hung, A. Y., Hayes, M. T., Hyman, B. T., Wills, A. M., et al. (2020). Associations of lower caffeine intake and plasma Urate levels with idiopathic Parkinson’s disease in the harvard biomarkers study. J. Parkinsons Dis. 10, 505–510. doi: 10.3233/jpd-191882

CrossRef Full Text | Google Scholar

Bloemberg, D., and Quadrilatero, J. (2016). Autophagic flux data in differentiated C2C12 myotubes following exposure to acetylcholine and caffeine. Data Brief. 7, 692–696. doi: 10.1016/j.dib.2016.03.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Boekema, P. J., Samsom, M., Van Berge Henegouwen, G. P., and Smout, A. J. (1999). Coffee and gastrointestinal function: facts and fiction. A review. Scand. J. Gastroenterol. Suppl. 230, 35–39. doi: 10.1080/003655299750025525

PubMed Abstract | CrossRef Full Text | Google Scholar

Boison, D., Chen, J. F., and Fredholm, B. B. (2010). Adenosine signaling and function in glial cells. Cell Death Differ. 17, 1071–1082. doi: 10.1038/cdd.2009.131

PubMed Abstract | CrossRef Full Text | Google Scholar

Brambilla, R., Cottini, L., Fumagalli, M., Ceruti, S., and Abbracchio, M. P. (2003). Blockade of A2A adenosine receptors prevents basic fibroblast growth factor-induced reactive astrogliosis in rat striatal primary astrocytes. Glia 43, 190–194. doi: 10.1002/glia.10243

PubMed Abstract | CrossRef Full Text | Google Scholar

Brothers, H. M., Marchalant, Y., and Wenk, G. L. (2010). Caffeine attenuates lipopolysaccharide-induced neuroinflammation. Neurosci. Lett. 480, 97–100. doi: 10.1016/j.neulet.2010.06.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Cadden, I. S., Partovi, N., and Yoshida, E. M. (2007). Review article: possible beneficial effects of coffee on liver disease and function. Aliment. Pharmacol. Ther. 26, 1–8. doi: 10.1111/j.1365-2036.2007.03319.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Canas, P. M., Porciuncula, L. O., Cunha, G. M., Silva, C. G., Machado, N. J., Oliveira, J. M., et al. (2009). Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by beta-amyloid peptides via p38 mitogen-activated protein kinase pathway. J. Neurosci. 29, 14741–14751. doi: 10.1523/jneurosci.3728-09.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, C., Cirrito, J. R., Lin, X., Wang, L., Verges, D. K., Dickson, A., et al. (2009). Caffeine suppresses amyloid-beta levels in plasma and brain of Alzheimer’s disease transgenic mice. J. Alzheimers Dis. 17, 681–697. doi: 10.3233/jad-2009-1071

PubMed Abstract | CrossRef Full Text | Google Scholar

Carabotti, M., Scirocco, A., Maselli, M. A., and Severi, C. (2015). The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 28, 203–209.

Google Scholar

Ceja-Navarro, J. A., Vega, F. E., Karaoz, U., Hao, Z., Jenkins, S., Lim, H. C., et al. (2015). Gut microbiota mediate caffeine detoxification in the primary insect pest of coffee. Nat. Commun. 6:7618.

Google Scholar

Cerri, S., and Blandini, F. (2019). Role of autophagy in Parkinson’s disease. Curr. Med. Chem. 26, 3702–3718.

Google Scholar

Chase, T. N., Bibbiani, F., Bara-Jimenez, W., Dimitrova, T., and Oh-Lee, J. D. (2003). Translating A2A antagonist KW6002 from animal models to parkinsonian patients. Neurology 61, S107–S111.

Google Scholar

Chaudhuri, K. R., Healy, D. G., Schapira, A. H., and National Institute for Clinical Excellence (2006). Non-motor symptoms of Parkinson’s disease: diagnosis and management. Lancet Neurol. 5, 235–245.

Google Scholar

Che, B., Wang, L., Zhang, Z., Zhang, Y., and Deng, Y. (2012). Distribution and accumulation of caffeine in rat tissues and its inhibition on semicarbazide-sensitive amine oxidase. Neurotoxicology 33, 1248–1253. doi: 10.1016/j.neuro.2012.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J. F. (2003). The adenosine A(2A) receptor as an attractive target for Parkinson’s disease treatment. Drug News Perspect. 16, 597–604. doi: 10.1358/dnp.2003.16.9.829342

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J. F. (2014). Adenosine receptor control of cognition in normal and disease. Int. Rev. Neurobiol. 119, 257–307. doi: 10.1016/b978-0-12-801022-8.00012-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J. F., Eltzschig, H. K., and Fredholm, B. B. (2013). Adenosine receptors as drug targets–what are the challenges? Nat. Rev. Drug Discov. 12, 265–286. doi: 10.1038/nrd3955

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J. F., Xu, K., Petzer, J. P., Staal, R., Xu, Y. H., Beilstein, M., et al. (2001). Neuroprotection by caffeine and A(2A) adenosine receptor inactivation in a model of Parkinson’s disease. J. Neurosci. 21:RC143.

Google Scholar

Chen, X., Lan, X., Roche, I., Liu, R., and Geiger, J. D. (2008). Caffeine protects against MPTP-induced blood-brain barrier dysfunction in mouse striatum. J. Neurochem. 107, 1147–1157.

Google Scholar

Cho, B. H., Choi, S. M., Kim, J. T., and Kim, B. C. (2018). Association of coffee consumption and non-motor symptoms in drug-naive, early-stage Parkinson’s disease. Parkinsonism Relat. Disord. 50, 42–47. doi: 10.1016/j.parkreldis.2018.02.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Cirstea, M. S., Yu, A. C., Golz, E., Sundvick, K., Kliger, D., Radisavljevic, N., et al. (2020). Microbiota composition and metabolism are associated with gut function in Parkinson’s disease. Mov. Disord. 35, 1208–1217. doi: 10.1002/mds.28052

PubMed Abstract | CrossRef Full Text | Google Scholar

Clarke, S. F., Murphy, E. F., Nilaweera, K., Ross, P. R., Shanahan, F., O’toole, P. W., et al. (2012). The gut microbiota and its relationship to diet and obesity: new insights. Gut Microbes 3, 186–202. doi: 10.4161/gmic.20168

PubMed Abstract | CrossRef Full Text | Google Scholar

Corti, O., Blomgren, K., Poletti, A., and Beart, P. M. (2020). Autophagy in neurodegeneration: new insights underpinning therapy for neurological diseases. J. Neurochem. 154, 354–371. doi: 10.1111/jnc.15002

PubMed Abstract | CrossRef Full Text | Google Scholar

Cowan, T. E., Palmnas, M. S., Yang, J., Bomhof, M. R., Ardell, K. L., Reimer, R. A., et al. (2014). Chronic coffee consumption in the diet-induced obese rat: impact on gut microbiota and serum metabolomics. J. Nutr. Biochem. 25, 489–495. doi: 10.1016/j.jnutbio.2013.12.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Crotty, G. F., Maciuca, R., Macklin, E. A., Wang, J., Montalban, M., Davis, S. S., et al. (2020). Association of caffeine and related analytes with resistance to Parkinson’s disease among LRRK2 mutation carriers: a metabolomic study. Neurology. doi: 10.1212/WNL.0000000000010863 [Epub ahead of print].

CrossRef Full Text | PubMed Abstract | Google Scholar

Cuervo, A. M., Stefanis, L., Fredenburg, R., Lansbury, P. T., and Sulzer, D. (2004). Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 305, 1292–1295. doi: 10.1126/science.1101738

PubMed Abstract | CrossRef Full Text | Google Scholar

Cunha, R. A., and Agostinho, P. M. (2010). Chronic caffeine consumption prevents memory disturbance in different animal models of memory decline. J. Alzheimers Dis. 20, Suppl 1, S95–S116.

Google Scholar

Dall’Igna, O. P., Fett, P., Gomes, M. W., Souza, D. O., Cunha, R. A., and Lara, D. R. (2007). Caffeine and adenosine A(2a) receptor antagonists prevent beta-amyloid (25-35)-induced cognitive deficits in mice. Exp. Neurol. 203, 241–245. doi: 10.1016/j.expneurol.2006.08.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Daly, J. W. (2007). Caffeine analogs: biomedical impact. Cell. Mol. Life Sci. 64, 2153–2169. doi: 10.1007/s00018-007-7051-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Dauer, W., and Przedborski, S. (2003). Parkinson’s disease: mechanisms and models. Neuron 39, 889–909.

Google Scholar

Derkinderen, P., Shannon, K. M., and Brundin, P. (2014). Gut feelings about smoking and coffee in Parkinson’s disease. Mov. Disord. 29, 976–979. doi: 10.1002/mds.25882

PubMed Abstract | CrossRef Full Text | Google Scholar

Dorsey, E. R., Constantinescu, R., Thompson, J. P., Biglan, K. M., Holloway, R. G., Kieburtz, K., et al. (2007). Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030. Neurology 68, 384–386. doi: 10.1212/01.wnl.0000247740.47667.03

PubMed Abstract | CrossRef Full Text | Google Scholar

Ebrahimi-Fakhari, D., Wahlster, L., and Mclean, P. J. (2012). Protein degradation pathways in Parkinson’s disease: curse or blessing. Acta Neuropathol. 124, 153–172. doi: 10.1007/s00401-012-1004-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Eskelinen, M. H., Ngandu, T., Tuomilehto, J., Soininen, H., and Kivipelto, M. (2009). Midlife coffee and tea drinking and the risk of late-life dementia: a population-based CAIDE study. J. Alzheimers Dis. 16, 85–91. doi: 10.3233/jad-2009-0920

PubMed Abstract | CrossRef Full Text | Google Scholar

Faivre, E., Coelho, J. E., Zornbach, K., Malik, E., Baqi, Y., Schneider, M., et al. (2018). Beneficial effect of a selective adenosine a2a receptor antagonist in the APPswe/PS1dE9 mouse model of Alzheimer’s disease. Front. Mol. Neurosci. 11:235. doi: 10.3389/fnmol.2018.00235

PubMed Abstract | CrossRef Full Text | Google Scholar

Fellner, L., Irschick, R., Schanda, K., Reindl, M., Klimaschewski, L., Poewe, W., et al. (2013). Toll-like receptor 4 is required for alpha-synuclein dependent activation of microglia and astroglia. Glia 61, 349–360. doi: 10.1002/glia.22437

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferreira, D. G., Batalha, V. L., Vicente Miranda, H., Coelho, J. E., Gomes, R., Goncalves, F. Q., et al. (2017a). Adenosine A2A receptors modulate alpha-synuclein aggregation and toxicity. Cereb. Cortex 27, 718–730.

Google Scholar

Ferreira, D. G., Temido-Ferreira, M., Vicente Miranda, H., Batalha, V. L., Coelho, J. E., Szego, E. M., et al. (2017b). alpha-synuclein interacts with PrP(C) to induce cognitive impairment through mGluR5 and NMDAR2B. Nat. Neurosci. 20, 1569–1579. doi: 10.1038/nn.4648

PubMed Abstract | CrossRef Full Text | Google Scholar

Frau, L., Costa, G., Porceddu, P. F., Khairnar, A., Castelli, M. P., Ennas, M. G., et al. (2016). Influence of caffeine on 3,4-methylenedioxymethamphetamine-induced dopaminergic neuron degeneration and neuroinflammation is age-dependent. J. Neurochem. 136, 148–162. doi: 10.1111/jnc.13377

PubMed Abstract | CrossRef Full Text | Google Scholar

Fredholm, B. B., Battig, K., Holmen, J., Nehlig, A., and Zvartau, E. E. (1999a). Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol. Rev. 51, 83–133.

Google Scholar

Fredholm, B. B., Battig, K., Holmen, J., Nehlig, A., and Zvartau, E. E. (1999b). Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol. Rev. 51, 83–133.

Google Scholar

Friedland, R. P. (2015). Mechanisms of molecular mimicry involving the microbiota in neurodegeneration. J. Alzheimers Dis. 45, 349–362. doi: 10.3233/jad-142841

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, J., Nalls, M. A., Shi, M., Joubert, B. R., Hernandez, D. G., Huang, X., et al. (2012). An exploratory analysis on gene-environment interactions for Parkinson disease. Neurobiol. Aging 33, 2528.e1–2528.e6.

Google Scholar

Garcao, P., Szabo, E. C., Wopereis, S., Castro, A. A., Tome, A. R., Prediger, R. D., et al. (2013). Functional interaction between pre-synaptic alpha6beta2-containing nicotinic and adenosine A2A receptors in the control of dopamine release in the rat striatum. Br. J. Pharmacol. 169, 1600–1611. doi: 10.1111/bph.12234

PubMed Abstract | CrossRef Full Text | Google Scholar

Gelber, R. P., Petrovitch, H., Masaki, K. H., Ross, G. W., and White, L. R. (2011). Coffee intake in midlife and risk of dementia and its neuropathologic correlates. J. Alzheimers Dis. 23, 607–615. doi: 10.3233/jad-2010-101428

PubMed Abstract | CrossRef Full Text | Google Scholar

Gerhardt, S., and Mohajeri, M. H. (2018). Changes of colonic bacterial composition in Parkinson’s disease and other neurodegenerative diseases. Nutrients 10:708. doi: 10.3390/nu10060708

PubMed Abstract | CrossRef Full Text | Google Scholar

Goetz, C. G., Poewe, W., Rascol, O., and Sampaio, C. (2005). Evidence-based medical review update: pharmacological and surgical treatments of Parkinson’s disease: 2001 to 2004. Mov. Disord. 20, 523–539. doi: 10.1002/mds.20464

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonzalez, S., Salazar, N., Ruiz-Saavedra, S., Gomez-Martin, M., De Los Reyes-Gavilan, C. G., and Gueimonde, M. (2020). Long-term coffee consumption is associated with fecal microbial composition in humans. Nutrients 12:1287. doi: 10.3390/nu12051287

PubMed Abstract | CrossRef Full Text | Google Scholar

Grosso, G., Godos, J., Galvano, F., and Giovannucci, E. L. (2017). Coffee, caffeine, and health outcomes: an umbrella review. Annu. Rev. Nutr. 37, 131–156. doi: 10.1146/annurev-nutr-071816-064941

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, Y. L., Duan, W. J., Lu, D. H., Ma, X. H., Li, X. X., Li, Z., et al. (2020). Autophagy-dependent removal of alpha-synuclein: a novel mechanism of GM1 ganglioside neuroprotection against Parkinson’s disease. Acta Pharmacol. Sin. doi: 10.1038/s41401-020-0454-y [Epub ahead of print].

CrossRef Full Text | PubMed Abstract | Google Scholar

Gurwara, S., Dai, A. N., Ajami, N., El-Serag, H. B., Graham, D. Y., and Jiao, L. (2019). Caffeine consumption and the colonic mucosa-associated gut Microbiota. Am. J. Gastroenterol. 114, S119–S120.

Google Scholar

Hameleers, P. A., Van Boxtel, M. P., Hogervorst, E., Riedel, W. J., Houx, P. J., Buntinx, F., et al. (2000). Habitual caffeine consumption and its relation to memory, attention, planning capacity and psychomotor performance across multiple age groups. Hum. Psychopharmacol. 15, 573–581. doi: 10.1002/hup.218

PubMed Abstract | CrossRef Full Text | Google Scholar

Hamza, T. H., Chen, H., Hill-Burns, E. M., Rhodes, S. L., Montimurro, J., Kay, D. M., et al. (2011). Genome-wide gene-environment study identifies glutamate receptor gene GRIN2A as a Parkinson’s disease modifier gene via interaction with coffee. PLoS Genet. 7:e1002237. doi: 10.1371/journal.pgen.1002237

PubMed Abstract | CrossRef Full Text | Google Scholar

Hauser, R. A., Cantillon, M., Pourcher, E., Micheli, F., Mok, V., Onofrj, M., et al. (2011). Preladenant in patients with Parkinson’s disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol. 10, 221–229. doi: 10.1016/s1474-4422(11)70012-6

CrossRef Full Text | Google Scholar

Higdon, J. V., and Frei, B. (2006). Coffee and health: a review of recent human research. Crit. Rev. Food Sci. Nutr. 46, 101–123. doi: 10.1080/10408390500400009

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirsch, E. C., and 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

CrossRef Full Text | Google Scholar

Hirsch, E. C., Vyas, S., and Hunot, S. (2012). Neuroinflammation in Parkinson’s disease. Parkinsonism Relat. Disord. 18(Suppl 1), S210–S212.

Google Scholar

Ho, L., Zhao, D., Ono, K., Ruan, K., Mogno, I., Tsuji, M., et al. (2019). Heterogeneity in gut microbiota drive polyphenol metabolism that influences alpha-synuclein misfolding and toxicity. J. Nutr. Biochem. 64, 170–181. doi: 10.1016/j.jnutbio.2018.10.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Hopfner, F., Kunstner, A., Muller, S. H., Kunzel, S., Zeuner, K. E., Margraf, N. G., et al. (2017). Gut microbiota in Parkinson disease in a northern German cohort. Brain Res. 1667, 41–45. doi: 10.1016/j.brainres.2017.04.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Hou, X., Watzlawik, J. O., Fiesel, F. C., and Springer, W. (2020). Autophagy in Parkinson’s disease. J. Mol. Biol. 432, 2651–2672.

Google Scholar

Hu, Q., Ren, X., Liu, Y., Li, Z., Zhang, L., Chen, X., et al. (2016). Aberrant adenosine A2A receptor signaling contributes to neurodegeneration and cognitive impairments in a mouse model of synucleinopathy. Exp. Neurol. 283, 213–223. doi: 10.1016/j.expneurol.2016.05.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Hughes, M. A., Downs, R. M., Webb, G. W., Crocker, C. L., Kinsey, S. T., and Baumgarner, B. L. (2017). Acute high-caffeine exposure increases autophagic flux and reduces protein synthesis in C2C12 skeletal myotubes. J. Muscle Res. Cell Motil. 38, 201–214. doi: 10.1007/s10974-017-9473-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Ikeda, K., Kurokawa, M., Aoyama, S., and Kuwana, Y. (2002). Neuroprotection by adenosine A2A receptor blockade in experimental models of Parkinson’s disease. J. Neurochem. 80, 262–270. doi: 10.1046/j.0022-3042.2001.00694.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Jenner, P., Mori, A., Hauser, R., Morelli, M., Fredholm, B. B., and Chen, J. F. (2009). Adenosine, adenosine A 2A antagonists, and Parkinson’s disease. Parkinsonism Relat. Disord. 15, 406–413.

Google Scholar

Kachroo, A., and Schwarzschild, M. A. (2012). Adenosine A2A receptor gene disruption protects in an alpha-synuclein model of Parkinson’s disease. Ann. Neurol. 71, 278–282. doi: 10.1002/ana.22630

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalda, A., Yu, L., Oztas, E., and Chen, J. F. (2006). Novel neuroprotection by caffeine and adenosine A(2A) receptor antagonists in animal models of Parkinson’s disease. J. Neurol. Sci. 248, 9–15. doi: 10.1016/j.jns.2006.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalia, L. V., and Kalia, S. K. (2015). alpha-Synuclein and Lewy pathology in Parkinson’s disease. Curr. Opin. Neurol. 28, 375–381.

Google Scholar

Kehagia, A. A., Barker, R. A., and Robbins, T. W. (2010). Neuropsychological and clinical heterogeneity of cognitive impairment and dementia in patients with Parkinson’s disease. Lancet Neurol. 9, 1200–1213. doi: 10.1016/s1474-4422(10)70212-x

CrossRef Full Text | Google Scholar

Khokhlova, E. V., Smeianov, V. V., Efimov, B. A., Kafarskaia, L. I., Pavlova, S. I., and Shkoporov, A. N. (2012). Anti-inflammatory properties of intestinal Bifidobacterium strains isolated from healthy infants. Microbiol. Immunol. 56, 27–39. doi: 10.1111/j.1348-0421.2011.00398.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Klann, E., Tagliamonte, M. S., Ukhanova, M., Mai, V., and Vedam-Mai, V. (2020). Gut microbiota dynamics in Parkinsonian mice. ACS Chem. Neurosci. 11, 3267–3276. doi: 10.1021/acschemneuro.0c00386

PubMed Abstract | CrossRef Full Text | Google Scholar

Klingelhoefer, L., and Reichmann, H. (2015). Pathogenesis of Parkinson disease–the gut-brain axis and environmental factors. Nat. Rev. Neurol. 11, 625–636. doi: 10.1038/nrneurol.2015.197

PubMed Abstract | CrossRef Full Text | Google Scholar

Knowlton, B. J., Mangels, J. A., and Squire, L. R. (1996). A neostriatal habit learning system in humans. Science 273, 1399–1402. doi: 10.1126/science.273.5280.1399

PubMed Abstract | CrossRef Full Text | Google Scholar

Ko, W. K. D., Camus, S. M., Li, Q., Yang, J., Mcguire, S., Pioli, E. Y., et al. (2016). An evaluation of istradefylline treatment on Parkinsonian motor and cognitive deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated macaque models. Neuropharmacology 110, 48–58. doi: 10.1016/j.neuropharm.2016.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Koutzoumis, D. N., Vergara, M., Pino, J., Buddendorff, J., Khoshbouei, H., Mandel, R. J., et al. (2020). Alterations of the gut microbiota with antibiotics protects dopamine neuron loss and improve motor deficits in a pharmacological rodent model of Parkinson’s disease. Exp. Neurol. 325, 113159. doi: 10.1016/j.expneurol.2019.113159

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, P. M., Paing, S. S., Li, H., Pavanni, R., Yuen, Y., Zhao, Y., et al. (2015). Differential effect of caffeine intake in subjects with genetic susceptibility to Parkinson’s Disease. Sci. Rep. 5:15492.

Google Scholar

Kyrozis, A., Ghika, A., Stathopoulos, P., Vassilopoulos, D., Trichopoulos, D., and Trichopoulou, A. (2013). Dietary and lifestyle variables in relation to incidence of Parkinson’s disease in Greece. Eur. J. Epidemiol. 28, 67–77. doi: 10.1007/s10654-012-9760-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Lamonaca, G., and Volta, M. (2020). Alpha-Synuclein and LRRK2 in synaptic autophagy: linking early dysfunction to late-stage pathology in Parkinson’s disease. Cells 9:1115. doi: 10.3390/cells9051115

PubMed Abstract | CrossRef Full Text | Google Scholar

Lang, A. E., and Obeso, J. A. (2004). Challenges in Parkinson’s disease: restoration of the nigrostriatal dopamine system is not enough. Lancet Neurol. 3, 309–316. doi: 10.1016/s1474-4422(04)00740-9

CrossRef Full Text | Google Scholar

Lashuel, H. A., Overk, C. R., Oueslati, A., and Masliah, E. (2013). The many faces of alpha-synuclein: from structure and toxicity to therapeutic target. Nat. Rev. Neurosci. 14, 38–48. doi: 10.1038/nrn3406

PubMed Abstract | CrossRef Full Text | Google Scholar

Laurent, C., Eddarkaoui, S., Derisbourg, M., Leboucher, A., Demeyer, D., Carrier, S., et al. (2014). Beneficial effects of caffeine in a transgenic model of Alzheimer’s disease-like tau pathology. Neurobiol. Aging 35, 2079–2090. doi: 10.1016/j.neurobiolaging.2014.03.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. J., Kim, C., and Lee, S. J. (2010). Alpha-synuclein stimulation of astrocytes: potential role for neuroinflammation and neuroprotection. Oxid. Med. Cell. Longev. 3, 283–287. doi: 10.4161/oxim.3.4.12809

PubMed Abstract | CrossRef Full Text | Google Scholar

Lees, A. J., Hardy, J., and Revesz, T. (2009). Parkinson’s disease. Lancet 373, 2055–2066.

Google Scholar

Lewis, S. J., Dove, A., Robbins, T. W., Barker, R. A., and Owen, A. M. (2003). Cognitive impairments in early Parkinson’s disease are accompanied by reductions in activity in frontostriatal neural circuitry. J. Neurosci. 23, 6351–6356. doi: 10.1523/jneurosci.23-15-06351.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W., Silva, H. B., Real, J., Wang, Y. M., Rial, D., Li, P., et al. (2015). Inactivation of adenosine A2A receptors reverses working memory deficits at early stages of Huntington’s disease models. Neurobiol. Dis. 79, 70–80. doi: 10.1016/j.nbd.2015.03.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., Chen, Y., Huang, H., Shi, M., Yang, W., Kuang, J., et al. (2017). Autophagy mediated by endoplasmic reticulum stress enhances the caffeine-induced apoptosis of hepatic stellate cells. Int. J. Mol. Med. 40, 1405–1414. doi: 10.3892/ijmm.2017.3145

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., He, Y., Chen, M., Pu, Z., Chen, L., Li, P., et al. (2016). Optogenetic activation of adenosine A2A receptor signaling in the dorsomedial striatopallidal neurons suppresses goal-directed behavior. Neuropsychopharmacology 41, 1003–1013. doi: 10.1038/npp.2015.227

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y. F., Ouyang, S. H., Tu, L. F., Wang, X., Yuan, W. L., Wang, G. E., et al. (2018a). Caffeine protects skin from oxidative stress-induced senescence through the activation of autophagy. Theranostics 8, 5713–5730. doi: 10.7150/thno.28778

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Z., Chen, X., Wang, T., Gao, Y., Li, F., Chen, L., et al. (2018b). The Corticostriatal Adenosine A2A receptor controls maintenance and retrieval of spatial working memory. Biol. Psychiatry 83, 530–541. doi: 10.1016/j.biopsych.2017.07.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Lindsay, J., Laurin, D., Verreault, R., Hebert, R., Helliwell, B., Hill, G. B., et al. (2002). Risk factors for Alzheimer’s disease: a prospective analysis from the Canadian Study of Health and Aging. Am. J. Epidemiol. 156, 445–453. doi: 10.1093/aje/kwf074

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, H. X., Yan, H. Y., Qu, W., Wen, X., Hou, L. F., Zhao, W. H., et al. (2019). Inhibition of thymocyte autophagy-associated CD4(+)T thymopoiesis is involved in asthma susceptibility in mice exposed to caffeine prenatally. Arch. Toxicol. 93, 1323–1335. doi: 10.1007/s00204-019-02418-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y. W., Yang, T., Zhao, L., Ni, Z., Yang, N., He, F., et al. (2016). Activation of Adenosine 2A receptor inhibits neutrophil apoptosis in an autophagy-dependent manner in mice with systemic inflammatory response syndrome. Sci. Rep. 6:33614.

Google Scholar

Lopez-Diego, R. S., and Weiner, H. L. (2008). Novel therapeutic strategies for multiple sclerosis–a multifaceted adversary. Nat. Rev. Drug Discov. 7, 909–925. doi: 10.1038/nrd2358

PubMed Abstract | CrossRef Full Text | Google Scholar

Lozupone, C. A., Stombaugh, J. I., Gordon, J. I., Jansson, J. K., and Knight, R. (2012). Diversity, stability and resilience of the human gut microbiota. Nature 489, 220–230. doi: 10.1038/nature11550

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, J., Wu, M., and Yue, Z. (2020). Autophagy and Parkinson’s disease. Adv. Exp. Med. Biol. 1207, 21–51.

Google Scholar

Luan, Y., Ren, X., Zheng, W., Zeng, Z., Guo, Y., Hou, Z., et al. (2018). Chronic caffeine treatment protects against alpha-synucleinopathy by reestablishing autophagy activity in the mouse striatum. Front. Neurosci. 12:301. doi: 10.3389/fnins.2018.00301

PubMed Abstract | CrossRef Full Text | Google Scholar

Lubomski, M., Davis, R. L., and Sue, C. M. (2019). The gut microbiota: A novel therapeutic target in Parkinson’s disease? Parkinsonism Relat. Disord. 66, 265–266. doi: 10.1016/j.parkreldis.2019.08.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Madeira, M. H., Boia, R., Ambrosio, A. F., and Santiago, A. R. (2017). Having a coffee break: the impact of caffeine consumption on microglia-mediated inflammation in neurodegenerative diseases. Mediators Inflamm. 2017:4761081.

Google Scholar

Maini Rekdal, V., Bess, E. N., Bisanz, J. E., Turnbaugh, P. J., and Balskus, E. P. (2019). Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism. Science 364:eaau6323. doi: 10.1126/science.aau6323

PubMed Abstract | CrossRef Full Text | Google Scholar

Majbour, N., and El-Agnaf, O. (2017). Cognitive impairment in Parkinson’s disease. Lancet Neurol. 16, 23–24.

Google Scholar

Mathew, T. S., Ferris, R. K., Downs, R. M., Kinsey, S. T., and Baumgarner, B. L. (2014). Caffeine promotes autophagy in skeletal muscle cells by increasing the calcium-dependent activation of AMP-activated protein kinase. Biochem. Biophys. Res. Commun. 453, 411–418. doi: 10.1016/j.bbrc.2014.09.094

PubMed Abstract | CrossRef Full Text | Google Scholar

Mattson, M. P. (2004). Pathways towards and away from Alzheimer’s disease. Nature 430, 631–639. doi: 10.1038/nature02621

PubMed Abstract | CrossRef Full Text | Google Scholar

McGeer, P. L., Itagaki, S., Boyes, B. E., and Mcgeer, E. G. (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 Abstract | CrossRef Full Text | Google Scholar

Mestre, T., Ferreira, J., Coelho, M. M., Rosa, M., and Sampaio, C. (2009). Therapeutic interventions for disease progression in Huntington’s disease. Cochrane Database Syst. Rev. 2009:CD006455.

Google Scholar

Mingote, S., Font, L., Farrar, A. M., Vontell, R., Worden, L. T., Stopper, C. M., et al. (2008). Nucleus accumbens adenosine A2A receptors regulate exertion of effort by acting on the ventral striatopallidal pathway. J. Neurosci. 28, 9037–9046. doi: 10.1523/jneurosci.1525-08.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Miraglia, F., and Colla, E. (2019). Microbiome, Parkinson’s disease and molecular mimicry. Cells 8:222. doi: 10.3390/cells8030222

PubMed Abstract | CrossRef Full Text | Google Scholar

Mitchell, D. C., Knight, C. A., Hockenberry, J., Teplansky, R., and Hartman, T. J. (2014). Beverage caffeine intakes in the U.S. Food Chem. Toxicol. 63, 136–142. doi: 10.1016/j.fct.2013.10.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Moon, J. H., Lee, J. H., Park, J. Y., Kim, S. W., Lee, Y. J., Kang, S. J., et al. (2014). Caffeine prevents human prion protein-mediated neurotoxicity through the induction of autophagy. Int. J. Mol. Med. 34, 553–558. doi: 10.3892/ijmm.2014.1814

PubMed Abstract | CrossRef Full Text | Google Scholar

Moors, T. E., Hoozemans, J. J., Ingrassia, A., Beccari, T., Parnetti, L., Chartier-Harlin, M. C., et al. (2017). Therapeutic potential of autophagy-enhancing agents in Parkinson’s disease. Mol. Neurodegener. 12:11.

Google Scholar

Morato, X., Lujan, R., Lopez-Cano, M., Gandia, J., Stagljar, I., Watanabe, M., et al. (2017). The Parkinson’s disease-associated GPR37 receptor interacts with striatal adenosine A2A receptor controlling its cell surface expression and function in vivo. Sci. Rep. 7:9452.

Google Scholar

Murakami, K., Okubo, H., and Sasaki, S. (2006). Dietary intake in relation to self-reported constipation among Japanese women aged 18-20 years. Eur. J. Clin. Nutr. 60, 650–657. doi: 10.1038/sj.ejcn.1602365

PubMed Abstract | CrossRef Full Text | Google Scholar

Nascimento, A. C., Erustes, A. G., Reckziegel, P., Bincoletto, C., Ureshino, R. P., Pereira, G. J. S., et al. (2020). alpha-synuclein overexpression induces lysosomal dysfunction and autophagy impairment in human neuroblastoma SH-SY5Y. Neurochem. Res. 45, 2749–2761. doi: 10.1007/s11064-020-03126-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Nehlig, A., Daval, J. L., and Debry, G. (1992). Caffeine and the central nervous system: mechanisms of action, biochemical, metabolic and psychostimulant effects. Brain Res. Brain Res. Rev. 17, 139–170. doi: 10.1016/0165-0173(92)90012-b

CrossRef Full Text | Google Scholar

Ning, Y. L., Yang, N., Chen, X., Tian, H. K., Zhao, Z. A., Zhang, X. Z., et al. (2019). Caffeine attenuates brain injury but increases mortality induced by high-intensity blast wave exposure. Toxicol. Lett. 301, 90–97. doi: 10.1016/j.toxlet.2018.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Ning, Y. L., Yang, N., Chen, X., Xiong, R. P., Zhang, X. Z., Li, P., et al. (2013). Adenosine A2A receptor deficiency alleviates blast-induced cognitive dysfunction. J. Cereb. Blood Flow Metab. 33, 1789–1798. doi: 10.1038/jcbfm.2013.127

PubMed Abstract | CrossRef Full Text | Google Scholar

Ning, Y. L., Yang, N., Chen, X., Zhao, Z. A., Zhang, X. Z., Chen, X. Y., et al. (2015). Chronic caffeine exposure attenuates blast-induced memory deficit in mice. Chin. J. Traumatol. 18, 204–211. doi: 10.1016/j.cjtee.2015.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Nishitsuji, K., Watanabe, S., Xiao, J., Nagatomo, R., Ogawa, H., Tsunematsu, T., et al. (2018). Effect of coffee or coffee components on gut microbiome and short-chain fatty acids in a mouse model of metabolic syndrome. Sci. Rep. 8:16173.

Google Scholar

Nuzum, N. D., Loughman, A., Szymlek-Gay, E. A., Hendy, A., Teo, W. P., and Macpherson, H. (2020). Gut microbiota differences between healthy older adults and individuals with Parkinson’s disease: a systematic review. Neurosci. Biobehav. Rev. 112, 227–241. doi: 10.1016/j.neubiorev.2020.02.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Olanow, C. W. (2004). The scientific basis for the current treatment of Parkinson’s disease. Annu. Rev. Med. 55, 41–60.

Google Scholar

Palacios, N., Gao, X., Mccullough, M. L., Schwarzschild, M. A., Shah, R., Gapstur, S., et al. (2012). Caffeine and risk of Parkinson’s disease in a large cohort of men and women. Mov. Disord. 27, 1276–1282. doi: 10.1002/mds.25076

PubMed Abstract | CrossRef Full Text | Google Scholar

Pellegrini, C., Antonioli, L., Colucci, R., Blandizzi, C., and Fornai, M. (2018). Interplay among gut microbiota, intestinal mucosal barrier and enteric neuro-immune system: a common path to neurodegenerative diseases? Acta Neuropathol. 136, 345–361. doi: 10.1007/s00401-018-1856-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Pietrucci, D., Teofani, A., Unida, V., Cerroni, R., Biocca, S., Stefani, A., et al. (2020). Can gut microbiota be a good predictor for Parkinson’s Disease? A machine learning approach. Brain Sci. 10:242. doi: 10.3390/brainsci10040242

PubMed Abstract | CrossRef Full Text | Google Scholar

Pitaksalee, R., Sanvarinda, Y., Sinchai, T., Sanvarinda, P., Thampithak, A., Jantaratnotai, N., et al. (2015). Autophagy inhibition by caffeine increases toxicity of methamphetamine in SH-SY5Y neuroblastoma cell line. Neurotox. Res. 27, 421–429. doi: 10.1007/s12640-014-9513-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Poehler, A. M., Xiang, W., Spitzer, P., May, V. E., Meixner, H., Rockenstein, E., et al. (2014). Autophagy modulates SNCA/alpha-synuclein release, thereby generating a hostile microenvironment. Autophagy 10, 2171–2192. doi: 10.4161/auto.36436

PubMed Abstract | CrossRef Full Text | Google Scholar

Popat, R. A., Van Den Eeden, S. K., Tanner, C. M., Kamel, F., Umbach, D. M., Marder, K., et al. (2011). Coffee, ADORA2A, and CYP1A2: the caffeine connection in Parkinson’s disease. Eur. J. Neurol. 18, 756–765. doi: 10.1111/j.1468-1331.2011.03353.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Postuma, R. B., Anang, J., Pelletier, A., Joseph, L., Moscovich, M., Grimes, D., et al. (2017). Caffeine as symptomatic treatment for Parkinson disease (Cafe-PD): a randomized trial. Neurology 89, 1795–1803. doi: 10.1212/wnl.0000000000004568

PubMed Abstract | CrossRef Full Text | Google Scholar

Postuma, R. B., and Berg, D. (2016). Advances in markers of prodromal Parkinson disease. Nat. Rev. Neurol. 12, 622–634. doi: 10.1038/nrneurol.2016.152

PubMed Abstract | CrossRef Full Text | Google Scholar

Postuma, R. B., Lang, A. E., Munhoz, R. P., Charland, K., Pelletier, A., Moscovich, M., et al. (2012). Caffeine for treatment of Parkinson disease: a randomized controlled trial. Neurology 79, 651–658.

Google Scholar

Powers, K. M., Kay, D. M., Factor, S. A., Zabetian, C. P., Higgins, D. S., Samii, A., et al. (2008). Combined effects of smoking, coffee, and NSAIDs on Parkinson’s disease risk. Mov. Disord. 23, 88–95. doi: 10.1002/mds.21782

PubMed Abstract | CrossRef Full Text | Google Scholar

Qi, H., and Li, S. (2014). Dose-response meta-analysis on coffee, tea and caffeine consumption with risk of Parkinson’s disease. Geriatr. Gerontol. Int. 14, 430–439. doi: 10.1111/ggi.12123

PubMed Abstract | CrossRef Full Text | Google Scholar

Qian, Y., Yang, X., Xu, S., Wu, C., Song, Y., Qin, N., et al. (2018). Alteration of the fecal microbiota in Chinese patients with Parkinson’s disease. Brain Behav. Immun. 70, 194–202. doi: 10.1016/j.bbi.2018.02.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, K. S., Manichanh, C., et al. (2010). A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 59–65.

Google Scholar

Qin, Y., Qiu, J., Wang, P., Liu, J., Zhao, Y., Jiang, F., et al. (2020). Impaired autophagy in microglia aggravates dopaminergic neurodegeneration by regulating NLRP3 inflammasome activation in experimental models of Parkinson’s disease. Brain Behav. Immun. doi: 10.1016/j.bbi.2020.10.010 [Epub ahead of print].

CrossRef Full Text | PubMed Abstract | Google Scholar

Rascol, O., Goetz, C., Koller, W., Poewe, W., and Sampaio, C. (2002). Treatment interventions for Parkinson’s disease: an evidence based assessment. Lancet 359, 1589–1598. doi: 10.1016/s0140-6736(02)08520-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Ray, K. (2013). Liver: Caffeine is a potent stimulator of autophagy to reduce hepatic lipid content–a coffee for NAFLD? Nat. Rev. Gastroenterol. Hepatol. 10:563. doi: 10.1038/nrgastro.2013.170

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, T. Z., Gao, Y. Y., Qiu, Y. H., Jiang, S. L., Zhang, Q. X., Zhang, J. H., et al. (2020). Gut microbiota altered in mild cognitive impairment compared with normal cognition in sporadic Parkinson’s disease. Front. Neurol. 11:137. doi: 10.3389/fneur.2020.00137

PubMed Abstract | CrossRef Full Text | Google Scholar

Richardson, P. J., Kase, H., and Jenner, P. G. (1997). Adenosine A2A receptor antagonists as new agents for the treatment of Parkinson’s disease. Trends Pharmacol. Sci. 18, 338–344. doi: 10.1016/s0165-6147(97)90660-x

CrossRef Full Text | Google Scholar

Ritchie, K., Carrière, I., Portet, F., De Mendonca, A., Dartigues, J. F., Rouaud, O., et al. (2007). The neuro-protective effects of caffeine: a prospective population study (the Three City Study). Neurology 69, 536–545. doi: 10.1212/01.wnl.0000266670.35219.0c

PubMed Abstract | CrossRef Full Text | Google Scholar

Rivero-Rios, P., Madero-Perez, J., Fernandez, B., and Hilfiker, S. (2016). Targeting the autophagy/lysosomal degradation pathway in Parkinson’s disease. Curr. Neuropharmacol. 14, 238–249. doi: 10.2174/1570159x13666151030103027

PubMed Abstract | CrossRef Full Text | Google Scholar

Ross, G. W., Abbott, R. D., Petrovitch, H., Morens, D. M., Grandinetti, A., Tung, K. H., et al. (2000). Association of coffee and caffeine intake with the risk of Parkinson disease. JAMA 283, 2674–2679. doi: 10.1001/jama.283.20.2674

PubMed Abstract | CrossRef Full Text | Google Scholar

Saaksjarvi, K., Knekt, P., Rissanen, H., Laaksonen, M. A., Reunanen, A., and Mannisto, S. (2008). Prospective study of coffee consumption and risk of Parkinson’s disease. Eur. J. Clin. Nutr. 62, 908–915.

Google Scholar

Sacino, A. N., Brooks, M., Mckinney, A. B., Thomas, M. A., Shaw, G., Golde, T. E., et al. (2014). Brain injection of alpha-synuclein induces multiple proteinopathies, gliosis, and a neuronal injury marker. J. Neurosci. 34, 12368–12378. doi: 10.1523/jneurosci.2102-14.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

Saiki, S., Sasazawa, Y., Imamichi, Y., Kawajiri, S., Fujimaki, T., Tanida, I., et al. (2011). Caffeine induces apoptosis by enhancement of autophagy via PI3K/Akt/mTOR/p70S6K inhibition. Autophagy 7, 176–187. doi: 10.4161/auto.7.2.14074

PubMed Abstract | CrossRef Full Text | Google Scholar

Sampson, T. R., Debelius, J. W., Thron, T., Janssen, S., Shastri, G. G., Ilhan, Z. E., et al. (2016). Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell 167:1469. doi: 10.1016/j.cell.2016.11.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Schapira, A. H. V., Chaudhuri, K. R., and Jenner, P. (2017). Non-motor features of Parkinson disease. Nat. Rev. Neurosci. 18, 435–450.

Google Scholar

Scheperjans, F., Aho, V., Pereira, P. A., Koskinen, K., Paulin, L., Pekkonen, E., et al. (2015a). Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov. Disord. 30, 350–358.

Google Scholar

Scheperjans, F., Pekkonen, E., Kaakkola, S., and Auvinen, P. (2015b). Linking smoking, coffee, urate, and Parkinson’s disease - A role for gut microbiota? J. Parkinsons Dis. 5, 255–262. doi: 10.3233/jpd-150557

PubMed Abstract | CrossRef Full Text | Google Scholar

Schwarzschild, M. A., Agnati, L., Fuxe, K., Chen, J. F., and Morelli, M. (2006). Targeting adenosine A2A receptors in Parkinson’s disease. Trends Neurosci. 29, 647–654. doi: 10.1016/j.tins.2006.09.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Scorza, C., Piccini, C., Martinez Busi, M., Abin Carriquiry, J. A., and Zunino, P. (2019). Alterations in the gut microbiota of rats chronically exposed to volatilized cocaine and its active adulterants caffeine and phenacetin. Neurotox. Res. 35, 111–121. doi: 10.1007/s12640-018-9936-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Senkevich, K., and Gan-Or, Z. (2020). Autophagy lysosomal pathway dysfunction in Parkinson’s disease; evidence from human genetics. Parkinsonism Relat. Disord. 73, 60–71. doi: 10.1016/j.parkreldis.2019.11.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Shoulson, I. (1998). Experimental therapeutics of neurodegenerative disorders: unmet needs. Science 282, 1072–1074. doi: 10.1126/science.282.5391.1072

PubMed Abstract | CrossRef Full Text | Google Scholar

Shoulson, I., and Chase, T. (1975). Caffeine and the antiparkinsonian response to levodopa or piribedil. Neurology 25, 722–724. doi: 10.1212/wnl.25.8.722

PubMed Abstract | CrossRef Full Text | Google Scholar

Sinchai, T., Plasen, S., Sanvarinda, Y., Jaisin, Y., Govitrapong, P., Morales, N. P., et al. (2011). Caffeine potentiates methamphetamine-induced toxicity both in vitro and in vivo. Neurosci. Lett. 502, 65–69. doi: 10.1016/j.neulet.2011.07.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Sinha, R. A., Farah, B. L., Singh, B. K., Siddique, M. M., Li, Y., Wu, Y., et al. (2014). Caffeine stimulates hepatic lipid metabolism by the autophagy-lysosomal pathway in mice. Hepatology 59, 1366–1380. doi: 10.1002/hep.26667

PubMed Abstract | CrossRef Full Text | Google Scholar

Sommer, F., and Backhed, F. (2013). The gut microbiota–masters of host development and physiology. Nat. Rev. Microbiol. 11, 227–238. doi: 10.1038/nrmicro2974

PubMed Abstract | CrossRef Full Text | Google Scholar

Sonsalla, P. K., Wong, L. Y., Harris, S. L., Richardson, J. R., Khobahy, I., Li, W., et al. (2012). Delayed caffeine treatment prevents nigral dopamine neuron loss in a progressive rat model of Parkinson’s disease. Exp. Neurol. 234, 482–487. doi: 10.1016/j.expneurol.2012.01.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, M. F., Zhu, Y. L., Zhou, Z. L., Jia, X. B., Xu, Y. D., Yang, Q., et al. (2018). Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: gut microbiota, glial reaction and TLR4/TNF-alpha signaling pathway. Brain Behav. Immun. 70, 48–60. doi: 10.1016/j.bbi.2018.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Svensson, E., Horvath-Puho, E., Thomsen, R. W., Djurhuus, J. C., Pedersen, L., Borghammer, P., et al. (2015). Vagotomy and subsequent risk of Parkinson’s disease. Ann. Neurol. 78, 522–529. doi: 10.1002/ana.24448

PubMed Abstract | CrossRef Full Text | Google Scholar

Tansey, M. G., and Goldberg, M. S. (2010). Neuroinflammation in Parkinson’s disease: its role in neuronal death and implications for therapeutic intervention. Neurobiol. Dis. 37, 510–518. doi: 10.1016/j.nbd.2009.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

van Boxtel, M. P., Schmitt, J. A., Bosma, H., and Jolles, J. (2003). The effects of habitual caffeine use on cognitive change: a longitudinal perspective. Pharmacol. Biochem. Behav. 75, 921–927. doi: 10.1016/s0091-3057(03)00171-0

CrossRef Full Text | Google Scholar

van Dam, R. M., Willett, W. C., Manson, J. E., and Hu, F. B. (2006). Coffee, caffeine, and risk of type 2 diabetes: a prospective cohort study in younger and middle-aged U.S. women. Diabetes Care 29, 398–403. doi: 10.2337/diacare.29.02.06.dc05-1512

PubMed Abstract | CrossRef Full Text | Google Scholar

van Gelder, B. M., Buijsse, B., Tijhuis, M., Kalmijn, S., Giampaoli, S., Nissinen, A., et al. (2007). Coffee consumption is inversely associated with cognitive decline in elderly European men: the FINE Study. Eur. J. Clin. Nutr. 61, 226–232. doi: 10.1038/sj.ejcn.1602495

PubMed Abstract | CrossRef Full Text | Google Scholar

Wan, S. X., Pan, X., Qian, J. J., Shu, Y., Xu, P., Zhao, J., et al. (2020). Downregulation of ATP13A2 in midbrain dopaminergic neurons is related to defective autophagy in a mouse model of Parkinson’s disease. Int. J. Clin. Exp. Pathol. 13, 1853–1858.

Google Scholar

Webb, J. L., Ravikumar, B., Atkins, J., Skepper, J. N., and Rubinsztein, D. C. (2003). Alpha-Synuclein is degraded by both autophagy and the proteasome. J. Biol. Chem. 278, 25009–25013. doi: 10.1074/jbc.m300227200

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, C. J., Augusto, E., Gomes, C. A., Singer, P., Wang, Y., Boison, D., et al. (2014). Regulation of fear responses by striatal and extrastriatal adenosine A2A receptors in forebrain. Biol. Psychiatry 75, 855–863. doi: 10.1016/j.biopsych.2013.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, C. J., Singer, P., Coelho, J., Boison, D., Feldon, J., Yee, B. K., et al. (2011). Selective inactivation of adenosine A(2A) receptors in striatal neurons enhances working memory and reversal learning. Learn. Mem. 18, 459–474. doi: 10.1101/lm.2136011

PubMed Abstract | CrossRef Full Text | Google Scholar

Wikoff, D., Welsh, B. T., Henderson, R., Brorby, G. P., Britt, J., Myers, E., et al. (2017). Systematic review of the potential adverse effects of caffeine consumption in healthy adults, pregnant women, adolescents, and children. Food Chem. Toxicol. 109, 585–648. doi: 10.1016/j.fct.2017.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Wills, A. M., Eberly, S., Tennis, M., Lang, A. E., Messing, S., Togasaki, D., et al. (2013). Caffeine consumption and risk of dyskinesia in CALM-PD. Mov. Disord. 28, 380–383. doi: 10.1002/mds.25319

PubMed Abstract | CrossRef Full Text | Google Scholar

Winkelmayer, W. C., Stampfer, M. J., Willett, W. C., and Curhan, G. C. (2005). Habitual caffeine intake and the risk of hypertension in women. JAMA 294, 2330–2335. doi: 10.1001/jama.294.18.2330

PubMed Abstract | CrossRef Full Text | Google Scholar

Winslow, A. R., and Rubinsztein, D. C. (2011). The Parkinson disease protein alpha-synuclein inhibits autophagy. Autophagy 7, 429–431. doi: 10.4161/auto.7.4.14393

PubMed Abstract | CrossRef Full Text | Google Scholar

Winter, G., Hazan, R., Bakalinsky, A. T., and Abeliovich, H. (2008). Caffeine induces macroautophagy and confers a cytocidal effect on food spoilage yeast in combination with benzoic acid. Autophagy 4, 28–36. doi: 10.4161/auto.5127

PubMed Abstract | CrossRef Full Text | Google Scholar

Wong, Y. C., and Krainc, D. (2017). alpha-synuclein toxicity in neurodegeneration: mechanism and therapeutic strategies. Nat. Med. 23, 1–13. doi: 10.1038/nm.4269

PubMed Abstract | CrossRef Full Text | Google Scholar

Xilouri, M., Brekk, O. R., and Stefanis, L. (2016). Autophagy and Alpha-Synuclein: relevance to Parkinson’s disease and related synucleopathies. Mov. Disord. 31, 178–192. doi: 10.1002/mds.26477

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, K., Xu, Y. H., Chen, J. F., and Schwarzschild, M. A. (2002). Caffeine’s neuroprotection against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity shows no tolerance to chronic caffeine administration in mice. Neurosci. Lett. 322, 13–16. doi: 10.1016/s0304-3940(02)00069-1

CrossRef Full Text | Google Scholar

Yamada-Fowler, N., Fredrikson, M., and Soderkvist, P. (2014). Caffeine interaction with glutamate receptor gene GRIN2A: Parkinson’s disease in Swedish population. PLoS One 9:e99294. doi: 10.1371/journal.pone.0099294

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, R., Zhang, J., Park, H. J., Park, E. S., Oh, S., Zheng, H., et al. (2018). Synergistic neuroprotection by coffee components eicosanoyl-5-hydroxytryptamide and caffeine in models of Parkinson’s disease and DLB. Proc. Natl. Acad. Sci. U.S.A. 115, E12053–E12062.

Google Scholar

Yasuda, T., Nakata, Y., and Mochizuki, H. (2013). alpha-Synuclein and neuronal cell death. Mol. Neurobiol. 47, 466–483.

Google Scholar

Zheng, W. X., He, R. N., Yan, Z. X., Huang, Y. W., Huang, W., Cai, Z. Y., et al. (2020). Regulation of immune-driven pathogenesis in Parkinson’s disease by gut microbiota. Brain Behav. Immun. 87, 890–897. doi: 10.1016/j.bbi.2020.01.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, S. J., Zhu, M. E., Shu, D., Du, X. P., Song, X. H., Wang, X. T., et al. (2009). Preferential enhancement of working memory in mice lacking adenosine A(2A) receptors. Brain Res. 1303, 74–83. doi: 10.1016/j.brainres.2009.09.082

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Z. D., Xie, S. P., Saw, W. T., Ho, P. G. H., Wang, H. Y., Zhou, L., et al. (2019). The therapeutic implications of tea Polyphenols against dopamine (DA) neuron degeneration in Parkinson’s Disease (PD). Cells 8:911. doi: 10.3390/cells8080911

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: caffeine, Parkinson’s disease, α-synuclein, adenosine A2A receptor, autophagy, gut microbiota

Citation: Ren X and Chen J-F (2020) Caffeine and Parkinson’s Disease: Multiple Benefits and Emerging Mechanisms. Front. Neurosci. 14:602697. doi: 10.3389/fnins.2020.602697

Received: 04 September 2020; Accepted: 25 November 2020;
Published: 17 December 2020.

Edited by:

Paula Agostinho, University of Coimbra, Portugal

Reviewed by:

Micaela Morelli, University of Cagliari, Italy
Carina Rodrigues Boeck, UFN - Universidade Franciscana, Brazil

Copyright © 2020 Ren and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xiangpeng Ren, renxpeng@mail3.sysu.edu.cn; Jiang-Fan Chen, chenjf555@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.