Appearance
This report is written by MaltSci based on the latest literature and research findings
How does Parkinson's disease develop?
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra, leading to motor dysfunctions such as bradykinesia, rigidity, and tremor. With over 10 million affected individuals globally, understanding the multifactorial etiology of PD, which includes genetic predispositions, environmental exposures, and cellular dysfunctions, is critical for identifying therapeutic targets. Genetic mutations, particularly in SNCA, LRRK2, and GBA1, account for 5-10% of cases, while the majority are sporadic, influenced by environmental toxins like pesticides and lifestyle factors. Cellular mechanisms such as mitochondrial dysfunction, oxidative stress, and neuroinflammation significantly contribute to neuronal degeneration. Neuroinflammation, driven by activated microglia, plays a dual role, exacerbating neuronal damage and promoting a vicious cycle of inflammation and degeneration. The importance of early diagnosis through advanced imaging techniques and the identification of biomarkers cannot be overstated, as timely intervention could modify disease progression. Current treatments primarily provide symptomatic relief, emphasizing the need for disease-modifying therapies that target the underlying mechanisms of neurodegeneration. Research is focused on novel strategies including gene therapy, immunomodulatory approaches, and personalized medicine, which hold promise for altering the course of PD. In conclusion, the ongoing investigation into the complex interactions between genetic, environmental, and cellular factors is essential for improving the management of Parkinson's disease and enhancing patient outcomes.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Genetic Factors in Parkinson's Disease
- 2.1 Overview of Genetic Mutations
- 2.2 Familial vs. Sporadic Cases
- 3 Environmental Influences
- 3.1 Role of Toxins and Chemicals
- 3.2 Lifestyle Factors and Their Impact
- 4 Cellular Mechanisms of Neurodegeneration
- 4.1 Mitochondrial Dysfunction
- 4.2 Protein Aggregation and Lewy Bodies
- 5 Neuroinflammation and Its Role in PD
- 5.1 Immune Response in the Central Nervous System
- 5.2 Microglial Activation and Neurodegeneration
- 6 Early Diagnosis and Therapeutic Strategies
- 6.1 Biomarkers for Early Detection
- 6.2 Current and Emerging Treatment Options
- 7 Conclusion
1 Introduction
Parkinson's disease (PD) is a progressive neurodegenerative disorder primarily characterized by the degeneration of dopaminergic neurons in the substantia nigra, leading to significant motor dysfunctions such as bradykinesia, rigidity, and tremor. The global prevalence of PD has reached alarming levels, affecting over 10 million individuals worldwide, with the burden expected to increase as populations age[1]. The complexity of PD's etiology is underscored by its multifactorial nature, involving intricate interactions between genetic predispositions, environmental exposures, and cellular dysfunctions. Understanding the pathogenesis of PD is essential not only for elucidating its underlying mechanisms but also for identifying potential therapeutic targets and developing effective interventions[2].
The significance of studying PD extends beyond the individual patient, as it poses substantial challenges to healthcare systems globally. Currently, there are no curative treatments available, and existing therapies primarily focus on alleviating symptoms rather than addressing the disease's progression[3]. As research advances, it becomes increasingly critical to uncover the molecular pathways that contribute to neuronal degeneration, which could pave the way for disease-modifying therapies[1][2].
Recent advancements in genetic research have revealed a multitude of genetic mutations associated with both familial and sporadic forms of PD. Notable genes such as SNCA (α-synuclein), LRRK2, and GBA1 have been implicated in the disease's development[3][4]. The understanding of how these genetic factors interplay with environmental influences, such as exposure to toxins and lifestyle choices, is crucial for developing a comprehensive view of PD pathogenesis[5]. Environmental factors, including pesticide exposure, have been linked to an increased risk of developing PD, further complicating the disease's etiology[1][5].
Moreover, cellular mechanisms such as mitochondrial dysfunction, oxidative stress, and neuroinflammation play pivotal roles in the neurodegenerative processes associated with PD[2][6]. The involvement of neuroinflammation, particularly the activation of microglia and the immune response in the central nervous system, has gained attention as a significant contributor to neuronal damage[2][6]. This interplay between genetic, environmental, and cellular factors creates a complex landscape that researchers are beginning to decipher.
The organization of this report will follow a structured approach to address the various dimensions of PD's development. The first section will delve into the genetic factors associated with PD, distinguishing between familial and sporadic cases and discussing the implications of identified mutations[3][4]. Next, we will explore the environmental influences, focusing on the role of toxins and lifestyle factors that may exacerbate or mitigate the risk of PD[5]. Following this, we will examine the cellular mechanisms of neurodegeneration, including mitochondrial dysfunction and the aggregation of proteins such as α-synuclein into Lewy bodies, which are hallmark features of PD[2][7].
The report will also address the critical role of neuroinflammation in PD, highlighting the immune responses within the central nervous system and the activation of microglia that contribute to the disease's progression[2][6]. Furthermore, we will discuss the importance of early diagnosis and therapeutic strategies, emphasizing biomarkers for early detection and the current landscape of treatment options, including both established and emerging therapies[1].
In conclusion, this report aims to synthesize the latest findings in the field of Parkinson's disease research, providing insights into the mechanisms underlying its development and progression. By exploring these multifaceted aspects, we hope to illuminate potential avenues for future research and clinical applications that could improve the management of this debilitating disorder.
2 Genetic Factors in Parkinson's Disease
2.1 Overview of Genetic Mutations
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons and the accumulation of alpha-synuclein aggregates. The development of PD involves a multifactorial interplay between genetic and environmental factors.
Genetic mutations play a crucial role in the etiology of Parkinson's disease. Approximately 5-10% of PD cases are attributed to monogenic forms of the disease, which are caused by specific mutations in genes such as SNCA (alpha-synuclein), LRRK2 (leucine-rich repeat kinase 2), and VPS35. These mutations can lead to autosomal dominant or recessive inheritance patterns, with high penetrance in some cases. Notably, autosomal recessive mutations in genes such as PINK1, DJ-1, and Parkin are associated with early-onset forms of PD, while mutations in SNCA and LRRK2 are linked to late-onset PD and exhibit significant variability in clinical presentation and neuropathology[8][9].
Recent advancements in genetic research have identified additional genetic contributors to PD, including rare mutations in DNAJC6, which are primarily linked to atypical PD phenotypes[8]. Genome-wide association studies have further elucidated the genetic landscape of PD, identifying 26 risk loci that contribute to the disease's susceptibility. These loci, however, typically exhibit only moderate effects on PD risk, indicating that the majority of PD cases arise from complex interactions between multiple genetic variants and environmental exposures[8].
The genetic factors associated with PD not only increase susceptibility to the disease but also interact with environmental toxins. Studies have shown that commonly used toxicants, such as rotenone and paraquat, can exacerbate the effects of genetic mutations linked to PD, particularly those affecting alpha-synuclein. This highlights the necessity of understanding the shared mechanisms between genetic predisposition and environmental triggers in the pathogenesis of PD[10].
In summary, the development of Parkinson's disease is influenced by a combination of genetic mutations and environmental factors. The genetic architecture of PD is complex, involving both monogenic and polygenic influences, which interact with environmental risk factors to modulate disease onset and progression. Understanding these interactions is essential for developing effective therapeutic strategies and improving early detection of the disease.
2.2 Familial vs. Sporadic Cases
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by the selective degeneration of dopaminergic neurons in the substantia nigra pars compacta, leading to motor and non-motor symptoms. The development of PD is influenced by both genetic and environmental factors, resulting in two main forms: familial and sporadic cases.
Familial cases of PD, although representing a small percentage of total cases, provide significant insights into the genetic underpinnings of the disease. Familial PD accounts for approximately 5-10% of all cases and is typically associated with specific Mendelian inheritance patterns. Research has identified over 18 genetic loci linked to familial forms of PD, including well-known genes such as SNCA, LRRK2, PARK2, and GBA. For instance, mutations in the SNCA gene, which encodes alpha-synuclein, have been shown to play a crucial role in the pathogenesis of familial PD. Studies have also suggested that these familial forms may exhibit autosomal dominant inheritance, as seen in some families where parkinsonism appears to be inherited through successive generations (Gasser et al. 1998; Lesage & Brice 2012) [11][12].
In contrast, sporadic cases of PD constitute the majority, with estimates indicating that they account for 90-95% of all instances. The etiology of sporadic PD remains less clear, although genetic factors are increasingly recognized as contributing elements. For example, twin studies and familial aggregation analyses suggest a hereditary component, but no specific genotypic associations have been definitively established for many sporadic cases. Environmental factors, such as exposure to toxins, have also been implicated in the development of sporadic PD. The interplay between genetic predispositions and environmental exposures likely contributes to the onset of nigral cell degeneration observed in these cases (Jenner 1999; Warner & Schapira 2003) [13][14].
Recent advancements in genome-wide association studies (GWAS) have identified several polymorphic variants that increase the risk of developing late-onset sporadic PD. Notably, heterozygous mutations in the GBA gene have been validated as significant susceptibility factors. The role of genetic factors in sporadic PD is underscored by findings that indicate genetic contributions to at least 27% of cases, with some populations showing that single genetic factors account for more than 33% of PD patients (Klein & Westenberger 2012; Karimi-Moghadam et al. 2018) [15][16].
In summary, the development of Parkinson's disease is a multifactorial process influenced by both genetic and environmental factors. Familial forms of PD are characterized by clear genetic mutations and inheritance patterns, while sporadic cases involve a complex interplay of genetic susceptibility and environmental triggers, with ongoing research aiming to elucidate the underlying mechanisms and interactions that contribute to the disease's onset and progression.
3 Environmental Influences
3.1 Role of Toxins and Chemicals
Parkinson's disease (PD) is increasingly recognized as a complex neurodegenerative disorder where environmental influences, particularly exposure to various toxins and chemicals, play a significant role in its development. While genetic factors contribute to the disease, a growing body of evidence suggests that environmental toxicants are predominant risk factors.
Recent studies have identified three major classes of environmental toxicants associated with PD: certain pesticides, solvents such as trichloroethylene, and air pollution. These substances are ubiquitous in modern environments and are known to impair mitochondrial or lysosomal function, which is crucial for neuronal health. This impairment can lead to the degeneration of dopaminergic neurons, a hallmark of PD [17].
Epidemiological studies have consistently shown associations between PD and exposure to neurotoxic pesticides and heavy metals. For instance, specific pesticides like paraquat and rotenone have been implicated in the pathogenesis of PD, as they can induce dopaminergic cell death through mechanisms involving oxidative stress and mitochondrial dysfunction [18].
Furthermore, air pollution has emerged as a significant environmental risk factor for PD. Research indicates that exposure to various pollutants, including particulate matter and nitrogen oxides, can contribute to neuroinflammation and oxidative stress, processes believed to underlie neurodegeneration [19]. The potential mechanisms through which air pollution affects PD risk include direct neuronal toxicity and systemic inflammation that may impact central nervous system function [20].
Additionally, the organochlorine pesticide dieldrin has been detected in postmortem brain tissues of PD patients, highlighting its potential role in promoting nigral cell death [21]. Although dieldrin has been banned, its persistent accumulation in the environment continues to expose humans through food sources [21].
The interaction between genetic predispositions and environmental exposures is also critical. While some genetic mutations are linked to familial forms of PD, the majority of cases are sporadic and likely result from a complex interplay of genetic and environmental factors [22]. This interplay may influence the timing and severity of neurotoxic effects, as well as the overall risk of developing PD.
The current understanding of PD emphasizes the importance of addressing modifiable risk factors, particularly environmental toxins, to develop effective prevention strategies. Improved measurement of toxicant exposure, long-term prospective studies, and increased funding for research on environmental impacts are essential steps toward reducing the burden of Parkinson's disease [17].
In conclusion, the development of Parkinson's disease is significantly influenced by environmental toxins, which can lead to mitochondrial dysfunction, oxidative stress, and neuroinflammation, ultimately contributing to the degeneration of dopaminergic neurons. Addressing these environmental factors presents a hopeful opportunity for prevention and intervention strategies in managing Parkinson's disease.
3.2 Lifestyle Factors and Their Impact
Parkinson's disease (PD) is a multifactorial neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra, leading to clinical symptoms such as resting tremor, bradykinesia, rigidity, and postural instability. The etiology of PD remains unclear, but it is widely accepted that both genetic and environmental factors contribute to its development. Recent research emphasizes the role of lifestyle factors and environmental influences, which may either increase the risk of developing PD or provide protective effects.
Age is recognized as the primary risk factor for PD, with increasing prevalence noted in older populations. However, beyond age, various lifestyle and environmental exposures have been implicated in modulating the risk for PD. Notably, smoking and caffeine consumption have been consistently associated with a reduced risk of developing PD, suggesting a potential protective effect against the disease [23].
Environmental exposures, particularly those related to occupational and residential settings, have garnered attention in understanding PD etiology. Evidence suggests that pesticide exposure is a significant risk factor for PD, as highlighted in several studies. For instance, a review identified pesticides as a notable environmental contributor to PD risk, indicating that exposure to these chemicals may impair mitochondrial function or provoke neuroinflammation, thereby increasing susceptibility to neurodegeneration [17]. Other occupational hazards, such as exposure to certain metals and solvents, have also been associated with an elevated risk of PD [24].
In addition to harmful exposures, lifestyle factors such as physical activity have been shown to influence PD risk. Regular exercise may not only reduce the likelihood of developing PD but also ameliorate symptoms in diagnosed patients. Increased physical activity has been linked to neuroprotective effects, potentially through mechanisms involving enhanced neurotrophic factor expression and improved neuronal health [25].
Furthermore, emerging evidence suggests that gut microbiota may play a crucial role in the pathogenesis of PD. The interaction between the gut microbiome and the central nervous system could influence neuroinflammatory processes and neuronal health, highlighting the importance of diet and gut health in PD risk modulation [26].
Overall, while genetic predispositions contribute to the risk of PD, lifestyle choices and environmental exposures are significant modifiable factors that can impact disease development and progression. Continued research is essential to elucidate the complex interplay between these factors and to identify potential preventive strategies that may mitigate the burden of Parkinson's disease in the population.
4 Cellular Mechanisms of Neurodegeneration
4.1 Mitochondrial Dysfunction
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra, leading to both motor and non-motor impairments. The development of PD is influenced by a multifactorial interplay of genetic, environmental, and age-related factors, with mitochondrial dysfunction emerging as a central mechanism in its pathogenesis.
Mitochondria, the cellular organelles responsible for energy production, play a critical role in maintaining neuronal health. In the context of PD, mitochondrial dysfunction can manifest through several pathways, including impaired bioenergetics, oxidative stress, altered mitochondrial dynamics, and defective mitophagy. These dysfunctions contribute to neuronal apoptosis and the overall neurodegenerative process.
Mitochondrial dysfunction in PD is often associated with increased oxidative stress, which results from an imbalance between the production of reactive oxygen species (ROS) and the ability of the cell to detoxify these harmful compounds. This oxidative stress can damage mitochondrial DNA, proteins, and lipids, further exacerbating mitochondrial impairment and neuronal injury [27][28].
Key genes associated with familial forms of PD, such as PINK1 and Parkin, are integral to mitochondrial quality control. They regulate mitochondrial dynamics, including fission and fusion processes, and promote the removal of damaged mitochondria through mitophagy [29][30]. Dysfunction in these pathways can lead to the accumulation of damaged mitochondria, which is a hallmark of PD pathology [31].
Furthermore, the interplay between mitochondrial dysfunction and neuroinflammation has been recognized as a significant contributor to PD development. Studies indicate that neuroinflammation, characterized by increased inflammatory markers in the brain, is closely linked to mitochondrial impairment, suggesting a bidirectional relationship that exacerbates neurodegeneration [32].
The pathogenesis of PD also involves age-related changes that amplify mitochondrial dysfunction, including the decline in mitochondrial biogenesis and the efficiency of mitochondrial dynamics. As individuals age, the cumulative effects of oxidative stress and mitochondrial dysfunction can lead to significant neuronal loss, particularly in the dopaminergic neurons [33][34].
In summary, the development of Parkinson's disease is intricately linked to mitochondrial dysfunction, which serves as a pivotal mechanism in the neurodegenerative process. This dysfunction not only contributes to neuronal apoptosis through oxidative stress and impaired bioenergetics but also disrupts critical cellular processes such as mitophagy and mitochondrial dynamics. Understanding these cellular mechanisms is crucial for developing targeted therapeutic strategies aimed at mitigating mitochondrial dysfunction and, consequently, the progression of Parkinson's disease [28][35].
4.2 Protein Aggregation and Lewy Bodies
Parkinson's disease (PD) is a progressive neurodegenerative disorder primarily characterized by the degeneration of dopaminergic neurons in the substantia nigra, leading to a significant loss of dopamine and the formation of intracellular protein aggregates known as Lewy bodies. The pathological hallmark of PD, Lewy bodies are predominantly composed of the protein alpha-synuclein (α-syn), which plays a critical role in the disease's pathogenesis.
The development of PD is intricately linked to the aggregation of α-syn, which is believed to initiate a cascade of neurodegenerative processes. α-Syn aggregates form Lewy bodies, and these aggregates are implicated in the disruption of cellular functions. Studies have indicated that the aggregation of α-syn is influenced by various factors, including genetic mutations, environmental toxins, and the overall cellular protein handling mechanisms. For instance, mutations in the α-syn gene, such as A30P and A53T, have been shown to alter the kinetics of α-syn aggregation, thereby affecting the onset and progression of PD (Flagmeier et al., 2016) [36].
Moreover, the accumulation of α-syn is linked to impaired protein degradation pathways, particularly the ubiquitin-proteasome system and autophagy. A decline in the activity of these systems leads to the accumulation of misfolded proteins, contributing to the formation of toxic oligomers and aggregates (Olanow et al., 2004) [37]. This accumulation can create a vicious cycle where increased protein aggregation further impairs the cell's ability to clear these aggregates, leading to neurodegeneration (Olanow & McNaught, 2011) [38].
The relationship between α-syn aggregation and mitochondrial dysfunction is also significant in the context of PD. Mitochondrial impairment can result from both genetic predispositions and environmental factors, leading to oxidative stress that exacerbates α-syn aggregation and neuronal death (Betarbet et al., 2002) [39]. Additionally, the interplay between α-syn and other proteins, such as tau and amyloid beta, has been proposed to contribute to the complexity of PD pathology, indicating that these proteins may influence each other's aggregation and toxicity (Sahoo et al., 2025) [40].
Recent research has also highlighted the importance of cellular interactions and the propagation of α-syn aggregates between neurons, suggesting that PD may exhibit prion-like characteristics (Gerez et al., 2019) [41]. This cell-to-cell transmission of α-syn aggregates can lead to the progressive spread of pathology throughout interconnected brain regions, further complicating the disease's progression.
In summary, the development of Parkinson's disease is a multifaceted process driven by the aggregation of α-synuclein, impaired protein degradation mechanisms, mitochondrial dysfunction, and intercellular propagation of toxic aggregates. Understanding these cellular mechanisms is crucial for identifying potential therapeutic targets and developing strategies for managing PD effectively.
5 Neuroinflammation and Its Role in PD
5.1 Immune Response in the Central Nervous System
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra, leading to significant motor and non-motor symptoms. The pathogenesis of PD is multifactorial, involving a combination of genetic, environmental, and inflammatory factors. A crucial aspect of PD development is neuroinflammation, which plays a significant role in both the initiation and progression of the disease.
Neuroinflammation in PD is primarily mediated by the activation of glial cells, including microglia and astrocytes, and is characterized by the presence of inflammatory cytokines and immune cell infiltration in the central nervous system (CNS). Activated microglia can release pro-inflammatory cytokines, which contribute to neuronal damage and create a self-sustaining cycle of neuroinflammation and neurodegeneration. This phenomenon has been referred to as a "vicious circle," where neuroinflammation exacerbates neuronal loss, which in turn promotes further inflammatory responses [6][42].
Recent studies have highlighted the dual role of neuroinflammation in PD. On one hand, acute neuroinflammation can serve as a protective response to injury or infection; however, when it becomes chronic, it leads to detrimental effects, including neuronal cell death [43]. This chronic inflammation is thought to be influenced by both central and peripheral immune responses. Evidence suggests that peripheral immune activation, such as the infiltration of T cells into the CNS, can prime microglia to adopt a pro-inflammatory phenotype, thus amplifying the inflammatory response within the brain [44][45].
The immune response in PD is not limited to the CNS; peripheral immune alterations have also been observed. Elevated levels of cytokines in the blood and the presence of autoantibodies indicate that the immune system's dysregulation extends beyond the brain [46]. Furthermore, interactions between the gut microbiome and the CNS, known as the gut-brain axis, have been implicated in the modulation of neuroinflammation and may play a significant role in the onset and progression of PD [47].
Understanding the interplay between neuroinflammation and the immune response is crucial for developing therapeutic strategies for PD. Current research is exploring various approaches to modulate the immune response, including immunotherapies aimed at reducing neuroinflammation, which could potentially slow the progression of the disease [48][49].
In summary, the development of Parkinson's disease is intricately linked to neuroinflammation and the immune response within the CNS. The activation of glial cells and the infiltration of peripheral immune cells contribute to a chronic inflammatory state that exacerbates neuronal degeneration. Continued research into the mechanisms of neuroinflammation and the immune response may provide insights into effective therapeutic interventions for PD.
5.2 Microglial Activation and Neurodegeneration
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra, leading to significant motor and non-motor symptoms. The development of PD is intricately linked to neuroinflammation, particularly through the activation of microglia, the resident immune cells of the central nervous system. Neuroinflammation is not merely a consequence of neuronal degeneration but is now recognized as a critical factor contributing to the pathogenesis and progression of PD.
Microglia play dual roles in PD, exhibiting both neuroprotective and neurotoxic properties. Upon activation, microglia can adopt different phenotypes, primarily categorized into M1 (pro-inflammatory) and M2 (anti-inflammatory) states. In the context of PD, M1 microglia release pro-inflammatory cytokines, reactive oxygen species (ROS), and other neurotoxic factors that exacerbate neuronal damage and contribute to neurodegeneration [50]. This pro-inflammatory response is thought to establish a self-perpetuating cycle of "inflammation-damage-reinflammation," leading to further neuronal loss [51].
Research has shown that neuroinflammation is not uniform across the stages of PD. In the prodromal and early stages of the disease, microglial activation is associated with neuroinflammation, protein aggregation, and neurodegeneration, although the exact mechanisms remain poorly understood [50]. In contrast, studies on animal models suggest that early microglial activation may not always correlate with significant neuroinflammatory processes, indicating that the relationship between microglial activity and neuronal degeneration is complex [52].
The interaction between microglia and other cell types, such as neurons and astrocytes, further complicates the neuroinflammatory landscape in PD. Activated microglia can influence neuronal health and contribute to the pathophysiology of α-synuclein aggregation, a hallmark of PD [53]. The accumulation of α-synuclein in neurons appears to trigger microglial activation, resulting in a cascade of inflammatory responses that ultimately lead to neuronal death [54].
Furthermore, the aging process significantly impacts microglial function, contributing to an altered inflammatory response that exacerbates neurodegeneration. Aging is a major risk factor for PD, and age-related changes in microglial activation states can lead to heightened inflammatory responses that promote neuronal damage [51].
Emerging therapeutic strategies focus on modulating microglial activation states to harness their neuroprotective potential while mitigating their neurotoxic effects. Targeting the inflammatory pathways and the phenotypic shifts of microglia presents a promising avenue for developing new treatments aimed at slowing the progression of PD and improving patient outcomes [55].
In conclusion, the development of Parkinson's disease is closely linked to neuroinflammation mediated by microglial activation. Understanding the complex interplay between microglia, neuronal health, and the inflammatory milieu is essential for identifying novel therapeutic targets that could alter the course of this debilitating disease.
6 Early Diagnosis and Therapeutic Strategies
6.1 Biomarkers for Early Detection
Parkinson's disease (PD) is characterized by a progressive degeneration of dopaminergic neurons primarily located in the substantia nigra, leading to motor deficits and a variety of non-motor symptoms. The exact etiology of PD remains unclear, but it is believed to arise from a complex interplay of genetic and environmental factors, along with age-related changes in the brain. As the disease progresses, it leads to a significant loss of dopamine-producing neurons, which manifests clinically when approximately 60% of these neurons are already lost at the time of diagnosis [56].
The pathogenesis of PD is multifaceted, involving mechanisms such as mitochondrial dysfunction, oxidative stress, and neuroinflammation [57][58]. Neuroinflammation has been particularly highlighted as a contributing factor to the initiation and progression of PD. Inflammatory responses in both the central nervous system and the periphery may trigger a cascade of neurodegenerative processes, although the specific triggers of this inflammation remain to be fully elucidated [58]. Recent evidence suggests that gut dysbiosis may play a role in initiating systemic inflammation, further complicating the disease's pathophysiology [58].
Early diagnosis of PD is crucial for effective management and potentially modifying the disease's course. Advanced brain imaging techniques, such as single photon emission computed tomography (SPECT) and positron emission tomography (PET), have been emphasized for their role in visualizing the integrity of striatal dopaminergic neurons in living patients. The introduction of radioligands that selectively bind to the dopamine transporter represents a significant advancement in the early diagnosis of PD [59]. Such imaging studies conducted at fixed intervals allow for monitoring the degeneration rate of dopaminergic neurons and evaluating the therapeutic effects of neuroprotective agents [59].
Biomarkers for early detection of PD are of immense importance, as they could lead to timely interventions that may alter disease progression. Recent research efforts have focused on identifying specific biomarkers that could indicate the presence of PD in early and pre-symptomatic stages [58]. The development of these biomarkers is critical, as current therapies primarily provide symptomatic relief without addressing the underlying neurodegenerative processes [60].
Current therapeutic strategies for PD are primarily symptomatic, focusing on dopamine replacement therapies, such as levodopa and dopamine receptor agonists. However, these treatments do not modify the disease's progression and often lead to complications over time [56][61]. There is a pressing need for disease-modifying therapies that can target the underlying mechanisms of neurodegeneration. Recent approaches under investigation include antioxidant therapies, immunomodulatory strategies, and novel small molecules aimed at mitigating oxidative stress and neuroinflammation [62][63].
In conclusion, the development of Parkinson's disease involves a complex interplay of neurodegenerative processes, and early diagnosis is essential for improving patient outcomes. Advancements in imaging techniques and biomarker identification are paving the way for better detection and monitoring of the disease, while ongoing research into disease-modifying therapies holds promise for altering the course of this debilitating disorder.
6.2 Current and Emerging Treatment Options
Parkinson's disease (PD) is a progressive neurodegenerative disorder primarily characterized by the selective loss of dopaminergic neurons in the substantia nigra, leading to a gradual decline in motor function. The pathogenesis of PD is complex and involves a multifaceted interplay of genetic, environmental, and age-related factors. Key mechanisms implicated in the disease include mitochondrial dysfunction, oxidative stress, neuroinflammation, and the accumulation of misfolded proteins, particularly α-synuclein, which aggregates to form Lewy bodies within neurons[57][64][65].
Current therapeutic strategies for PD predominantly focus on symptomatic relief rather than halting disease progression. Traditional treatments involve dopaminergic medications, such as levodopa, which alleviate motor symptoms but do not modify the underlying disease process. The long-term use of these therapies is often complicated by side effects and diminished efficacy over time[65][66]. Deep brain stimulation (DBS) is another intervention that provides symptomatic relief for advanced PD but similarly lacks disease-modifying effects[67].
Emerging treatment options are increasingly directed at modifying the disease course by targeting the pathogenic mechanisms associated with PD. Recent advances in molecular research have facilitated the development of disease-modifying strategies aimed at preventing α-synuclein aggregation, correcting mitochondrial dysfunction, and addressing genetic mutations such as those found in LRRK2 and GBA1[64]. Additionally, pluripotent stem cell (PSC)-derived dopaminergic neurons are being explored as a potential source for cell replacement therapy, with early-phase clinical trials demonstrating safety and functional integration of these grafts[64].
Furthermore, novel therapeutic approaches are being investigated, including the use of adenosine A(2A) receptor antagonists and glutamate AMPA receptor antagonists, which may offer symptomatic benefits without the complications associated with dopaminergic therapies[65]. Gene therapy, which encompasses techniques such as subthalamic glutamic acid decarboxylase gene therapy, is also gaining traction, with initial trials showing promise[67].
The development of biomarkers for PD is another critical area of research that could enhance early diagnosis and treatment personalization. These biomarkers could improve monitoring of disease progression and facilitate targeted interventions based on individual patient responses[60].
In summary, while current treatments for Parkinson's disease primarily focus on managing symptoms, there is a concerted effort within the scientific community to develop disease-modifying therapies that address the underlying mechanisms of the disease. These emerging strategies, combined with advances in early diagnosis and personalized medicine, hold the potential to significantly improve patient outcomes and alter the trajectory of Parkinson's disease management.
7 Conclusion
The exploration of Parkinson's disease (PD) has revealed critical insights into its multifactorial etiology, encompassing genetic, environmental, and cellular mechanisms. The findings underscore that genetic mutations, particularly in genes such as SNCA, LRRK2, and GBA1, play a significant role in both familial and sporadic forms of PD. Environmental influences, particularly exposure to neurotoxic agents and lifestyle factors, further complicate the disease's pathogenesis. Mitochondrial dysfunction, oxidative stress, and neuroinflammation emerge as pivotal cellular mechanisms driving neurodegeneration in PD. Notably, the activation of microglia and the chronic inflammatory response within the central nervous system contribute significantly to neuronal loss. The identification of biomarkers for early detection is crucial, as it could facilitate timely interventions that may alter disease progression. Current therapeutic strategies primarily focus on symptomatic relief, highlighting the urgent need for disease-modifying therapies that target the underlying neurodegenerative processes. Future research should prioritize elucidating the complex interactions between genetic predispositions, environmental exposures, and cellular dysfunctions to develop effective preventive and therapeutic strategies. Moreover, the integration of advanced diagnostic tools and the pursuit of novel therapeutic avenues will be essential in improving the management of Parkinson's disease and enhancing patient outcomes.
References
- [1] Suhas M;Sunil Kumar Kadiri. Parkinson's Disease: From Bench to Bedside-Advancements in Diagnosis and Therapeutics using Pharmacogenomic Approach.. Current gene therapy(IF=3.3). 2025. PMID:39945257. DOI: 10.2174/0115665232342211250207064205.
- [2] Cintia Roodveldt;Liliana Bernardino;Ozgur Oztop-Cakmak;Milorad Dragic;Kari E Fladmark;Sibel Ertan;Busra Aktas;Carlos Pita;Lucia Ciglar;Gaetan Garraux;Caroline Williams-Gray;Rodrigo Pacheco;Marina Romero-Ramos. The immune system in Parkinson's disease: what we know so far.. Brain : a journal of neurology(IF=11.7). 2024. PMID:38833182. DOI: 10.1093/brain/awae177.
- [3] Yashumati Ratan;Aishwarya Rajput;Ashutosh Pareek;Aaushi Pareek;Vivek Jain;Sonia Sonia;Zeba Farooqui;Ranjeet Kaur;Gurjit Singh. Advancements in Genetic and Biochemical Insights: Unraveling the Etiopathogenesis of Neurodegeneration in Parkinson's Disease.. Biomolecules(IF=4.8). 2024. PMID:38254673. DOI: 10.3390/biom14010073.
- [4] Suganya Selvaraj;Shanmughavel Piramanayagam. Impact of gene mutation in the development of Parkinson's disease.. Genes & diseases(IF=9.4). 2019. PMID:31193965. DOI: 10.1016/j.gendis.2019.01.004.
- [5] Harm J Heusinkveld;Martin van den Berg;Remco H S Westerink. In vitro dopaminergic neurotoxicity of pesticides: a link with neurodegeneration?. The veterinary quarterly(IF=5.2). 2014. PMID:25506807. DOI: 10.1080/01652176.2014.980934.
- [6] G Arena;K Sharma;G Agyeah;R Krüger;A Grünewald;J C Fitzgerald. Neurodegeneration and Neuroinflammation in Parkinson's Disease: a Self-Sustained Loop.. Current neurology and neuroscience reports(IF=5.2). 2022. PMID:35674870. DOI: 10.1007/s11910-022-01207-5.
- [7] Hao Song;Juanli Chen;Jin Huang;Peng Sun;Yanming Liu;Li Xu;Chuanfei Wei;Xin Mu;Xianjie Lu;Wei Wang;Nan Zhang;Miwei Shang;Mei Mo;Wei Zhang;Hui Zhao;Fabin Han. Epigenetic modification in Parkinson's disease.. Frontiers in cell and developmental biology(IF=4.3). 2023. PMID:37351278. DOI: 10.3389/fcell.2023.1123621.
- [8] Christina M Lill. Genetics of Parkinson's disease.. Molecular and cellular probes(IF=3.0). 2016. PMID:27818248. DOI: 10.1016/j.mcp.2016.11.001.
- [9] Andreas Puschmann. Monogenic Parkinson's disease and parkinsonism: clinical phenotypes and frequencies of known mutations.. Parkinsonism & related disorders(IF=3.4). 2013. PMID:23462481. DOI: .
- [10] Sheila M Fleming. Mechanisms of Gene-Environment Interactions in Parkinson's Disease.. Current environmental health reports(IF=9.1). 2017. PMID:28417442. DOI: 10.1007/s40572-017-0143-2.
- [11] T Gasser;B Müller-Myhsok;Z K Wszolek;R Oehlmann;D B Calne;V Bonifati;B Bereznai;E Fabrizio;P Vieregge;R D Horstmann. A susceptibility locus for Parkinson's disease maps to chromosome 2p13.. Nature genetics(IF=29.0). 1998. PMID:9500549. DOI: 10.1038/ng0398-262.
- [12] Suzanne Lesage;Alexis Brice. Role of mendelian genes in "sporadic" Parkinson's disease.. Parkinsonism & related disorders(IF=3.4). 2012. PMID:22166458. DOI: 10.1016/S1353-8020(11)70022-0.
- [13] P Jenner. Genetic susceptibility and the occurrence of Parkinson's disease.. Parkinsonism & related disorders(IF=3.4). 1999. PMID:18591137. DOI: 10.1016/s1353-8020(99)00034-6.
- [14] Thomas T Warner;Anthony H V Schapira. Genetic and environmental factors in the cause of Parkinson's disease.. Annals of neurology(IF=7.7). 2003. PMID:12666095. DOI: 10.1002/ana.10487.
- [15] Christine Klein;Ana Westenberger. Genetics of Parkinson's disease.. Cold Spring Harbor perspectives in medicine(IF=10.1). 2012. PMID:22315721. DOI: 10.1101/cshperspect.a008888.
- [16] Amin Karimi-Moghadam;Saeid Charsouei;Benjamin Bell;Mohammad Reza Jabalameli. Parkinson Disease from Mendelian Forms to Genetic Susceptibility: New Molecular Insights into the Neurodegeneration Process.. Cellular and molecular neurobiology(IF=4.8). 2018. PMID:29700661. DOI: 10.1007/s10571-018-0587-4.
- [17] E Ray Dorsey;Briana R De Miranda;Sarrah Hussain;Bastiaan R Bloem;Alexis Elbaz;Jorge Llibre-Guerra;Raymond Y Lo;Samuel M Goldman;Caroline M Tanner. Environmental toxicants and Parkinson's disease: recent evidence, risks, and prevention opportunities.. The Lancet. Neurology(IF=45.5). 2025. PMID:41109237. DOI: 10.1016/S1474-4422(25)00287-X.
- [18] Minhong Huang;Alejandra Bargues-Carot;Zainab Riaz;Hannah Wickham;Gary Zenitsky;Huajun Jin;Vellareddy Anantharam;Arthi Kanthasamy;Anumantha G Kanthasamy. Impact of Environmental Risk Factors on Mitochondrial Dysfunction, Neuroinflammation, Protein Misfolding, and Oxidative Stress in the Etiopathogenesis of Parkinson's Disease.. International journal of molecular sciences(IF=4.9). 2022. PMID:36142718. DOI: 10.3390/ijms231810808.
- [19] Hiromi Murata;Lisa M Barnhill;Jeff M Bronstein. Air Pollution and the Risk of Parkinson's Disease: A Review.. Movement disorders : official journal of the Movement Disorder Society(IF=7.6). 2022. PMID:35043999. DOI: 10.1002/mds.28922.
- [20] Natalia Palacios. Air pollution and Parkinson's disease - evidence and future directions.. Reviews on environmental health(IF=4.5). 2017. PMID:28731859. DOI: .
- [21] Anumantha G Kanthasamy;Masashi Kitazawa;Arthi Kanthasamy;Vellareddy Anantharam. Dieldrin-induced neurotoxicity: relevance to Parkinson's disease pathogenesis.. Neurotoxicology(IF=3.9). 2005. PMID:16112328. DOI: 10.1016/j.neuro.2004.07.010.
- [22] E Ray Dorsey;Bastiaan R Bloem. Parkinson's Disease Is Predominantly an Environmental Disease.. Journal of Parkinson's disease(IF=5.0). 2024. PMID:38217613. DOI: 10.3233/JPD-230357.
- [23] Caroline M Tanner. Advances in environmental epidemiology.. Movement disorders : official journal of the Movement Disorder Society(IF=7.6). 2010. PMID:20187243. DOI: 10.1002/mds.22721.
- [24] B C L Lai;S A Marion;K Teschke;J K C Tsui. Occupational and environmental risk factors for Parkinson's disease.. Parkinsonism & related disorders(IF=3.4). 2002. PMID:15177059. DOI: 10.1016/s1353-8020(01)00054-2.
- [25] Connie Marras;Colleen G Canning;Samuel M Goldman. Environment, lifestyle, and Parkinson's disease: Implications for prevention in the next decade.. Movement disorders : official journal of the Movement Disorder Society(IF=7.6). 2019. PMID:31091353. DOI: 10.1002/mds.27720.
- [26] Rafael Franco;Rafael Rivas-Santisteban;Irene Reyes-Resina;Gemma Navarro;Eva Martínez-Pinilla. Microbiota and Other Preventive Strategies and Non-genetic Risk Factors in Parkinson's Disease.. Frontiers in aging neuroscience(IF=4.5). 2020. PMID:32226375. DOI: 10.3389/fnagi.2020.00012.
- [27] Paola Imbriani;Giuseppina Martella;Paola Bonsi;Antonio Pisani. Oxidative stress and synaptic dysfunction in rodent models of Parkinson's disease.. Neurobiology of disease(IF=5.6). 2022. PMID:36007757. DOI: 10.1016/j.nbd.2022.105851.
- [28] Sadegh Moradi Vastegani;Ava Nasrolahi;Shahab Ghaderi;Rafie Belali;Masome Rashno;Maryam Farzaneh;Seyed Esmaeil Khoshnam. Mitochondrial Dysfunction and Parkinson's Disease: Pathogenesis and Therapeutic Strategies.. Neurochemical research(IF=3.8). 2023. PMID:36943668. DOI: 10.1007/s11064-023-03904-0.
- [29] Francois Mouton-Liger;Maxime Jacoupy;Jean-Christophe Corvol;Olga Corti. PINK1/Parkin-Dependent Mitochondrial Surveillance: From Pleiotropy to Parkinson's Disease.. Frontiers in molecular neuroscience(IF=3.8). 2017. PMID:28507507. DOI: 10.3389/fnmol.2017.00120.
- [30] Sandra Franco-Iborra;Miquel Vila;Celine Perier. The Parkinson Disease Mitochondrial Hypothesis: Where Are We at?. The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry(IF=3.9). 2016. PMID:25761946. DOI: 10.1177/1073858415574600.
- [31] Xiao-Le Wang;Si-Tong Feng;Ya-Ting Wang;Yu-He Yuan;Zhi-Peng Li;Nai-Hong Chen;Zhen-Zhen Wang;Yi Zhang. Mitophagy, a Form of Selective Autophagy, Plays an Essential Role in Mitochondrial Dynamics of Parkinson's Disease.. Cellular and molecular neurobiology(IF=4.8). 2022. PMID:33528716. DOI: 10.1007/s10571-021-01039-w.
- [32] Negar Ebadpour;Mahmoud Mahmoudi;Ramiar Kamal Kheder;Mohammad Abavisani;Zahra Baridjavadi;Narges Abdollahi;Seyed-Alireza Esmaeili. From mitochondrial dysfunction to neuroinflammation in Parkinson's disease: Pathogenesis and mitochondrial therapeutic approaches.. International immunopharmacology(IF=4.7). 2024. PMID:39222583. DOI: 10.1016/j.intimp.2024.113015.
- [33] Anthony H Schapira;Peter Jenner. Etiology and pathogenesis of Parkinson's disease.. Movement disorders : official journal of the Movement Disorder Society(IF=7.6). 2011. PMID:21626550. DOI: 10.1002/mds.23732.
- [34] Martin T Henrich;Wolfgang H Oertel;D James Surmeier;Fanni F Geibl. Mitochondrial dysfunction in Parkinson's disease - a key disease hallmark with therapeutic potential.. Molecular neurodegeneration(IF=17.5). 2023. PMID:37951933. DOI: 10.1186/s13024-023-00676-7.
- [35] Qing Zheng;Hanghang Liu;Yifan Gao;Guozhi Cao;Yusong Wang;Zhen Li. Ameliorating Mitochondrial Dysfunction for the Therapy of Parkinson's Disease.. Small (Weinheim an der Bergstrasse, Germany)(IF=12.1). 2024. PMID:38385823. DOI: 10.1002/smll.202311571.
- [36] Patrick Flagmeier;Georg Meisl;Michele Vendruscolo;Tuomas P J Knowles;Christopher M Dobson;Alexander K Buell;Céline Galvagnion. Mutations associated with familial Parkinson's disease alter the initiation and amplification steps of α-synuclein aggregation.. Proceedings of the National Academy of Sciences of the United States of America(IF=9.1). 2016. PMID:27573854. DOI: 10.1073/pnas.1604645113.
- [37] C Warren Olanow;Daniel P Perl;George N DeMartino;Kevin St P McNaught. Lewy-body formation is an aggresome-related process: a hypothesis.. The Lancet. Neurology(IF=45.5). 2004. PMID:15261611. DOI: 10.1016/S1474-4422(04)00827-0.
- [38] C Warren Olanow;K McNaught. Parkinson's disease, proteins, and prions: milestones.. Movement disorders : official journal of the Movement Disorder Society(IF=7.6). 2011. PMID:21626551. DOI: 10.1002/mds.23767.
- [39] Ranjita Betarbet;Todd B Sherer;Donato A Di Monte;J Timothy Greenamyre. Mechanistic approaches to Parkinson's disease pathogenesis.. Brain pathology (Zurich, Switzerland)(IF=6.2). 2002. PMID:12408237. DOI: 10.1111/j.1750-3639.2002.tb00468.x.
- [40] Tarini Ashish Sahoo;Jagdish Chand;Amarjith Thiyyar Kandy;Shanish Antony;Gomathy Subramanian. Unravelling the Proteinopathic Engagement of α-Synuclein, Tau, and Amyloid Beta in Parkinson's Disease: Mitochondrial Collapse as a Pivotal Driver of Neurodegeneration.. Neurochemical research(IF=3.8). 2025. PMID:40240583. DOI: 10.1007/s11064-025-04399-7.
- [41] Juan A Gerez;Natalia C Prymaczok;Edward Rockenstein;Uli S Herrmann;Petra Schwarz;Anthony Adame;Radoslav I Enchev;Thibault Courtheoux;Paul J Boersema;Roland Riek;Matthias Peter;Adriano Aguzzi;Eliezer Masliah;Paola Picotti. A cullin-RING ubiquitin ligase targets exogenous α-synuclein and inhibits Lewy body-like pathology.. Science translational medicine(IF=14.6). 2019. PMID:31167929. DOI: 10.1126/scitranslmed.aau6722.
- [42] Li Qian;Patrick M Flood;Jau-Shyong Hong. Neuroinflammation is a key player in Parkinson's disease and a prime target for therapy.. Journal of neural transmission (Vienna, Austria : 1996)(IF=4.0). 2010. PMID:20571837. DOI: 10.1007/s00702-010-0428-1.
- [43] Mi Eun Kim;Jun Sik Lee. Mechanisms and Emerging Regulators of Neuroinflammation: Exploring New Therapeutic Strategies for Neurological Disorders.. Current issues in molecular biology(IF=3.0). 2024. PMID:39852123. DOI: 10.3390/cimb47010008.
- [44] Natasa Kustrimovic;Franca Marino;Marco Cosentino. Peripheral Immunity, Immunoaging and Neuroinflammation in Parkinson's Disease.. Current medicinal chemistry(IF=3.5). 2019. PMID:30306855. DOI: 10.2174/0929867325666181009161048.
- [45] Bruna Araújo;Rita Caridade-Silva;Carla Soares-Guedes;Joana Martins-Macedo;Eduardo D Gomes;Susana Monteiro;Fábio G Teixeira. Neuroinflammation and Parkinson's Disease-From Neurodegeneration to Therapeutic Opportunities.. Cells(IF=5.2). 2022. PMID:36139483. DOI: 10.3390/cells11182908.
- [46] Neeraj Joshi;Sarika Singh. Updates on immunity and inflammation in Parkinson disease pathology.. Journal of neuroscience research(IF=3.4). 2018. PMID:29072332. DOI: 10.1002/jnr.24185.
- [47] Elodie Kip;Louise C Parr-Brownlie. Reducing neuroinflammation via therapeutic compounds and lifestyle to prevent or delay progression of Parkinson's disease.. Ageing research reviews(IF=12.4). 2022. PMID:35395416. DOI: 10.1016/j.arr.2022.101618.
- [48] Alhamdu Adamu;Shuo Li;Fankai Gao;Guofang Xue. The role of neuroinflammation in neurodegenerative diseases: current understanding and future therapeutic targets.. Frontiers in aging neuroscience(IF=4.5). 2024. PMID:38681666. DOI: 10.3389/fnagi.2024.1347987.
- [49] Gregory P Williams;Aubrey M Schonhoff;Alessandro Sette;Cecilia S Lindestam Arlehamn. Central and Peripheral Inflammation: Connecting the Immune Responses of Parkinson's Disease.. Journal of Parkinson's disease(IF=5.0). 2022. PMID:35754290. DOI: 10.3233/JPD-223241.
- [50] Frida Lind-Holm Mogensen;Philip Seibler;Anne Grünewald;Alessandro Michelucci. Microglial dynamics and neuroinflammation in prodromal and early Parkinson's disease.. Journal of neuroinflammation(IF=10.1). 2025. PMID:40399949. DOI: 10.1186/s12974-025-03462-y.
- [51] Yuxiang Xu;Tingting Han;Yue Wu;Haixuan Liu;Keyuan Hou;Zhaowu An;Yongjie Li;Chaoyang Zhu;Song Wang;Jianshe Wei. Role of Microglial Dysfunction in Parkinson's Disease: From Multifactorial Causes to Neurodegeneration.. Neuroscience bulletin(IF=5.8). 2025. PMID:40973880. DOI: 10.1007/s12264-025-01505-1.
- [52] Vanessa Porrini;Mariana Mota;Edoardo Parrella;Arianna Bellucci;Marina Benarese;Lara Faggi;Paolo Tonin;Pier F Spano;Marina Pizzi. Mild Inflammatory Profile without Gliosis in the c-Rel Deficient Mouse Modeling a Late-Onset Parkinsonism.. Frontiers in aging neuroscience(IF=4.5). 2017. PMID:28769786. DOI: 10.3389/fnagi.2017.00229.
- [53] Cai-Yun Liu;Xu Wang;Chang Liu;Hong-Liang Zhang. Pharmacological Targeting of Microglial Activation: New Therapeutic Approach.. Frontiers in cellular neuroscience(IF=4.0). 2019. PMID:31803024. DOI: 10.3389/fncel.2019.00514.
- [54] Haiyang Yu;Qing Chang;Tong Sun;Xin He;Lulu Wen;Jing An;Juan Feng;Yuhong Zhao. Metabolic reprogramming and polarization of microglia in Parkinson's disease: Role of inflammasome and iron.. Ageing research reviews(IF=12.4). 2023. PMID:37572760. DOI: 10.1016/j.arr.2023.102032.
- [55] Sudhakar R Subramaniam;Howard J Federoff. Targeting Microglial Activation States as a Therapeutic Avenue in Parkinson's Disease.. Frontiers in aging neuroscience(IF=4.5). 2017. PMID:28642697. DOI: 10.3389/fnagi.2017.00176.
- [56] Michael J O'Neill;Eric R Siemers. Pharmacological approaches to disease-modifying therapies in Parkinson's disease.. Expert review of neurotherapeutics(IF=3.4). 2002. PMID:19810916. DOI: 10.1586/14737175.2.6.819.
- [57] Alexander Leathem;Tamara Ortiz-Cerda;Joanne M Dennis;Paul K Witting. Evidence for Oxidative Pathways in the Pathogenesis of PD: Are Antioxidants Candidate Drugs to Ameliorate Disease Progression?. International journal of molecular sciences(IF=4.9). 2022. PMID:35805928. DOI: 10.3390/ijms23136923.
- [58] Anamaria Jurcau;Felicia Liana Andronie-Cioara;Delia Carmen Nistor-Cseppento;Nicoleta Pascalau;Marius Rus;Elisabeta Vasca;Maria Carolina Jurcau. The Involvement of Neuroinflammation in the Onset and Progression of Parkinson's Disease.. International journal of molecular sciences(IF=4.9). 2023. PMID:37834030. DOI: 10.3390/ijms241914582.
- [59] J C Stoof;A Winogrodzka;F L van Muiswinkel;E C Wolters;P Voorn;H J Groenewegen;J Booij;B Drukarch. Leads for the development of neuroprotective treatment in Parkinson's disease and brain imaging methods for estimating treatment efficacy.. European journal of pharmacology(IF=4.7). 1999. PMID:10443566. DOI: 10.1016/s0014-2999(99)00260-5.
- [60] Rumiana Tenchov;Janet M Sasso;Qiongqiong Angela Zhou. Evolving Landscape of Parkinson's Disease Research: Challenges and Perspectives.. ACS omega(IF=4.3). 2025. PMID:39866628. DOI: 10.1021/acsomega.4c09114.
- [61] C Warren Olanow;Matthew B Stern;Kapil Sethi. The scientific and clinical basis for the treatment of Parkinson disease (2009).. Neurology(IF=8.5). 2009. PMID:19470958. DOI: 10.1212/WNL.0b013e3181a1d44c.
- [62] Ting Shen;Jiali Pu;Xiaoli Si;Rong Ye;Baorong Zhang. An update on potential therapeutic strategies for Parkinson's disease based on pathogenic mechanisms.. Expert review of neurotherapeutics(IF=3.4). 2016. PMID:27138872. DOI: 10.1080/14737175.2016.1179112.
- [63] A H V Schapira. Challenges to the development of disease-modifying therapies in Parkinson's disease.. European journal of neurology(IF=3.9). 2011. PMID:21255199. DOI: 10.1111/j.1468-1331.2010.03324.x.
- [64] Tae Young Kim;Byoung Dae Lee. Current therapeutic strategies in Parkinson's disease: Future perspectives.. Molecules and cells(IF=6.5). 2025. PMID:40914459. DOI: 10.1016/j.mocell.2025.100274.
- [65] Stacy S Wu;Steven J Frucht. Treatment of Parkinson's disease : what's on the horizon?. CNS drugs(IF=7.4). 2005. PMID:16142989. DOI: 10.2165/00023210-200519090-00001.
- [66] Amaal AlDakheel;Lorraine V Kalia;Anthony E Lang. Pathogenesis-targeted, disease-modifying therapies in Parkinson disease.. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics(IF=6.9). 2014. PMID:24085420. DOI: 10.1007/s13311-013-0218-1.
- [67] Philip C Buttery;Roger A Barker. Gene and Cell-Based Therapies for Parkinson's Disease: Where Are We?. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics(IF=6.9). 2020. PMID:33128174. DOI: 10.1007/s13311-020-00940-4.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Parkinson's disease · neurodegenerative disorder · neuroinflammation · genetic factors · cellular mechanisms
© 2025 MaltSci
