Unraveling the Genetic and Environmental Connection in Parkinson’s Disease: Implications for Pathogenesis and Therapeutic Strategies

0
252

Abstract

Parkinson’s disease (PD) is a complex neurodegenerative disorder that affects millions of people worldwide and is characterized primarily by progressive motor dysfunction such as tremors, bradykinesia, and rigidity. It also triggers a variety of non-motor symptoms. The pathogenesis of PD involves a complex interaction between genetic predispositions and environmental factors. This paper explores the critical role of various genetic mutations, including those in the SNCA and LRRK2 genes, implicated in familial and sporadic forms of PD. These rare mutations offer insight into the underlying mechanisms of the disease, including alpha-synuclein misfolding and mitochondrial dysfunction. Also, environmental exposures to pesticides and heavy metals are examined for their correlation with higher PD incidence, supported by evidence of oxidative stress and neuroinflammation induced by toxins. Additionally, this paper highlights the relationships between different environmental factors, such as pesticide exposure and heavy metals, which can attack genetic vulnerabilities and result in devastating neurodegeneration. It identifies how the interaction between these genetic and environmental factors may influence the susceptibility to and severity of Parkinson’s disease. In terms of personalized medicine, this research points to the potential of gene therapies targeting specific mutations such as LRRK2, SNCA, and GBA. These customized approaches could lead to more effective treatments, offering patients tailored interventions based on their genetic and environmental risk profiles. This paper provides a foundation for future research and public health efforts to minimize ecological risks while advancing gene therapy development.

Keywords: Parkinson’s disease, genetics, environment, pathogenesis, neurodegeneration, SNCA, LRRK2, pesticides, personalized medicine

Introduction

In recent years, Parkinson’s disease (PD) research has made significant strides in understanding its genetic and environmental risk factors. However, a critical gap remains in exploring underexamined genetic pathways and their interplay with novel ecological factors. This study aims to investigate the impact of specific genetic

mutations, particularly those in the LRRK2 and GBA genes, in conjunction with emerging environmental neurotoxicants, such as pesticides and heavy metals, on the pathogenesis of Parkinson’s disease. It is hypothesized that the interaction between these genetic predispositions and environmental exposures will elucidate novel biochemical mechanisms that contribute to disease progression. By addressing these underexplored areas, this research seeks to provide innovative insights that could pave the way for new therapeutic approaches. For example, studies indicate that DNA methylation patterns may differ significantly between PD patients and healthy controls, suggesting that these modifications could be biomarkers for disease diagnosis or prognosis. Additionally, histone acetylation and deacetylation processes have been implicated in regulating genes associated with synaptic plasticity and neuronal health, further illustrating the role of epigenetics in PD pathology. Notably, the epigenetic landscape is dynamic and can be influenced by lifestyle factors, including diet and physical activity, offering potential avenues for therapeutic intervention1. By understanding the epigenetic mechanisms that underlie PD, researchers may identify novel targets for treatment that could reverse or mitigate disease progression2.

Pathogenesis of Parkinson’s Disease

PD pathogenesis involves a series of molecular and cellular mechanisms leading to neurodegeneration. A staple of PD pathology is the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta, a brain region in the basal ganglia that is crucial for motor control. Abnormal aggregates of the protein alpha-synuclein, encoded by the SNCA gene, accompany this neuronal loss3. Understanding the sequence of these pathological features is essential for developing targeted therapeutic interventions.

At the molecular level, alpha-synuclein plays a pivotal role in PD pathogenesis. Under healthy conditions, alpha-synuclein is involved in synaptic vesicle regulation, but in PD, it misfolds and aggregates to form Lewy bodies4. These aggregates disrupt normal cellular functions, notably leading to mitochondrial dysfunction. Mitochondrial dysfunction is a particularly significant pathology, resulting in reduced adenosine triphosphate (ATP) production and increased generation of reactive oxygen species (ROS)5, further damaging cellular components and contributing to neuronal death.

Cellular mechanisms in PD pathogenesis also involve impaired autophagy and proteasome function, which are crucial processes for degrading and recycling cellular waste and unwanted or damaged proteins. The accumulation of alpha-synuclein and other misfolded proteins overwhelms these degradation pathways, leading to cellular stress and apoptosis (cell death)6. Additionally, neuroinflammation plays a significant role in PD. Microglial cells, the primary immune cells in the brain, become activated in response to neuronal damage and release inflammatory cytokines7. While initially a protective measure, chronic neuroinflammation worsens neuronal injury and cell death.

The impacts of genetic predispositions and environmental factors contribute to PD pathogenesis at the cellular level. Genetic mutations like those in the LRRK2 and SNCA genes can directly affect protein aggregation, mitochondrial function, and cellular homeostasis8. Environmental toxins, such as pesticides, can induce oxidative stress and mitochondrial dysfunction, further compounding the effects of genetic vulnerabilities. These genetic and ecological detriments disrupt normal cellular processes, leading to the progressive degeneration of dopaminergic neurons.

Overall, the pathogenesis of PD is a variable process involving molecular and cellular disruptions that combine to cause neuronal death. Insights into these mechanisms are crucial for identifying potential therapeutic targets and developing strategies to halt or slow disease progression.

Dopaminergic Cellular and Synaptic Mechanisms

Understanding the cellular and synaptic mechanisms underlying Parkinson’s disease is crucial when learning how genetic predispositions and environmental exposures interact to trigger its onset. Dopamine, a neurotransmitter central to motor control and reward processing, plays a significant role in synaptic plasticity, particularly in mechanisms such as long-term potentiation (LTP) and long-term depression (LTD).

Dopaminergic Contributions to Plasticity

Dopamine is essential for regulating synaptic plasticity, which is the ability of synapses to strengthen or weaken over time in response to external stimuli and their resulting activity levels9. In the context of PD, dopamine depletion in the striatum, a brain region critical for movement regulation, disrupts these plasticity processes. LTP and LTD are two fundamental forms of synaptic plasticity. LTP refers to the long-lasting enhancement of synaptic strength, while LTD denotes a prolonged decrease in synaptic efficacy and can lead to the destruction of the synapse. Both are vital for learning, memory, and motor control.

In PD, the loss of dopaminergic neurons in the substantia nigra pars compacta leads to diminished dopamine levels in the striatum, promoting LTD over LTP10. This imbalance results in abnormal motor function, contributing to the hallmark symptoms of PD, such as bradykinesia, rigidity, and tremors11. Research has shown that dopamine is necessary for both the induction and maintenance of LTP and LTD in the corticostriatal pathways, highlighting its critical role in synaptic modulation12. Genetic predispositions play a significant role in these disruptions. Mutations in genes such as LRRK2 and SNCA directly affect dopamine synthesis, release, and receptor function, leading to altered synaptic plasticity13. Environmental exposures, such as pesticides, further compound these genetic vulnerabilities by inducing oxidative stress and mitochondrial dysfunction, which impair dopaminergic signaling and worsen synaptic deficits14.

Dopamine Therapy

Therapeutic strategies for PD often involve restoring dopamine levels to reestablish normal synaptic function. Levodopa (L-DOPA), a precursor to dopamine, is the most commonly used treatment and helps alleviate motor symptoms by replenishing brain dopamine15. However, while such treatments can temporarily improve motor function, they do not address the underlying cellular mechanisms driving neurodegeneration. The treatments can also become less effective as patients resist the drug over time.

Research indicates that genetic mutations influencing dopamine pathways alter the efficacy of these treatments. For example, patients with specific LRRK2 mutations may respond differently to dopaminergic therapies than those without such mutations16.

Additionally, environmental factors that contribute to dopamine neuron damage, such as chronic pesticide exposure, can diminish the effectiveness of dopamine replacement therapies by further disrupting synaptic integrity17.

Chronic Pain

Chronic pain is a hallmark non-motor symptom of PD, resulting from altered pain processing pathways in the brain and spinal cord. Analgesics, including opioids and non-steroidal anti-inflammatory drugs (NSAIDs), are effective at minimizing pain symptoms in PD patients. Despite their efficacy in pain management, these drugs do not modify the cellular mechanisms contributing to PD11.

Current research focuses on understanding how dopamine depletion affects pain pathways at the cellular level. Pain in PD is deeply connected to the neurodegenerative processes characteristic of the disease. Dopamine significantly influences pain pathways through the basal ganglia and associated neural circuits. In PD, the loss of dopaminergic neurons in the substantia nigra results in decreased dopamine levels, disrupting the standard processing of pain signals18.

This dopamine depletion disrupts the balance between excitatory and inhibitory neurotransmitters, such as glutamate and GABA, crucial for maintaining normal synaptic function. For example, reduced dopamine levels can increase activity in the thalamus, a key relay center for sensory information, including pain. This heightened thalamic activity enhances the transmission of pain signals to the cortex, particularly the somatosensory cortex, which processes the sensory aspects of pain, and the prefrontal cortex, which is involved in the emotional response to pain. This altered signaling contributes to the development of chronic pain in PD patients19.

Further investigations into cellular mechanisms have shown that changes in synaptic plasticity, specifically disrupted LTP and LTD, play a significant role in the persistence of chronic pain in PD. Dopamine modulates LTP and LTD at various synapses, including those in the striatum, a critical basal ganglia component. Without sufficient dopamine, these processes become dysregulated, leading to abnormal pain processing and the maintenance of chronic pain states20. These findings emphasize the need for therapeutic approaches beyond symptom management to restore normal synaptic function and neurotransmitter balance, addressing the fundamental cellular dysfunctions in PD.

Future Directions

To address the research question of how genetic predispositions and environmental exposures interact to trigger PD onset, it is essential to consider how these cellular and synaptic factors influence dopamine’s role in synaptic plasticity. Environmental factors, such as pesticide exposure, can exacerbate these effects by inducing oxidative stress and mitochondrial dysfunction, further impairing dopamine’s role in the brain21.

By examining the dopaminergic connections between genetic and environmental factors, researchers can better understand the cellular and synaptic mechanisms underlying PD. This knowledge is crucial for developing targeted therapies that not only alleviate symptoms but also address the root causes of neurodegeneration in PD. Insights into how genetic predispositions and environmental exposures disrupt synaptic plasticity and dopamine signaling can inform future research directions and potential clinical applications to mitigate PD onset and progression.

Relevant Brain Anatomy

A thorough understanding of the brain regions affected by Parkinson’s disease is essential to elucidate how genetic predispositions and environmental exposures interact to trigger its onset. Central to the pathology of PD is the substantia nigra and its projections into the striatum.

Substantia Nigra

The substantia nigra is a crucial brain region that contains DA neurons, which are integral to movement regulation. In PD, these DA neurons undergo significant degeneration, leading to the hallmark motor symptoms of the disease. The loss of these neurons is a primary pathological feature of PD and is directly influenced by genetic and environmental factors.

Genetic mutations, such as those in the LRRK2 and SNCA genes, can lead to the dysfunction and eventual death of DA neurons in the substantia nigra. These genetic alterations affect various cellular processes, including protein aggregation, mitochondrial function, and oxidative stress responses critical for neuron survival.

Striatum

The neurons in the substantia nigra project into the striatum, a brain region divided into two main sections: the caudate nucleus and the putamen. The striatum is a critical component of the basal ganglia, which coordinates movement and other functions. The projections from the substantia nigra to the striatum form the nigrostriatal pathway, which is essential for the modulation of motor activity22.

In PD, the degeneration of DA neurons in the substantia nigra results in decreased dopamine levels in the striatum. This dopamine deficiency disrupts the normal functioning of the nigrostriatal pathway, leading to impaired synaptic plasticity, particularly in processes such as LTP and LTD14. The balance between LTP and LTD is crucial for motor learning and execution, and its disruption contributes to the motor symptoms observed in PD patients.

Genetic predispositions and environmental exposures also impact the striatum’s functionality. For instance, genetic mutations affecting dopamine synthesis, release, and receptor function can alter synaptic plasticity in the striatum. Concurrently, environmental toxins can cause oxidative damage and inflammation in the striatum, further impairing its ability to maintain normal synaptic function, similar to other parts of the brain, including the substantia nigra14.

Future Directions

In the greater context of understanding PD, it is critical to consider the roles of the substantia nigra and striatum. Genetic mutations that affect DA neurons in the substantia nigra can lead to their degeneration, which is further amplified by environmental toxins. This neuronal loss disrupts the nigrostriatal pathway, resulting in decreased dopamine levels in the striatum and impaired synaptic plasticity23.

By examining the specific impacts of genetic and environmental factors on these brain regions, researchers can gain insights into the mechanisms underlying PD onset.

Understanding how these factors interact to affect the substantia nigra and striatum can progress the development of targeted therapies aimed at preserving DA neurons and maintaining normal synaptic function. This approach holds the potential to lessen PD progression and improve patient outcomes.

Oxidative Stress and Genetic Interactions

Oxidative stress is a significant driver of neurodegeneration in PD, particularly affecting dopaminergic neurons in the substantia nigra. Reactive oxygen species (ROS) accumulate due to mitochondrial dysfunction and environmental exposures like pesticides or air pollution. These ROS can damage cellular components, including proteins, lipids, and nucleic acids, leading to neuronal death24.

At the transcriptional level, oxidative stress interacts with key PD-related genes, influencing their expression. For instance, mutations in the LRRK2 (leucine-rich repeat kinase 2) gene are associated with increased oxidative stress. Studies suggest that oxidative stress upregulates LRRK2 expression, which, in turn, contributes to mitochondrial dysfunction. Furthermore, the oxidative stress response gene NRF2 is downregulated in PD patients, which impairs the cell’s antioxidant defenses, leaving neurons more vulnerable to oxidative damage25.

Post-transcriptionally, oxidative stress alters mRNA stability or modulates gene expression through microRNAs (miRNAs). For instance, miRNAs such as miR-34b/c are downregulated in PD patients, impairing mitochondrial function and increasing oxidative stress26. SNCA (α-synuclein) mRNA is another critical target, where oxidative stress can increase its expression, promoting the aggregation of α-synuclein, a hallmark of PD pathology27.

The substantia nigra, particularly the pars compacta, is heavily affected by these molecular disruptions. The dopaminergic neurons in this region are uniquely susceptible to oxidative stress due to their high metabolic activity and dopamine metabolism, which generates additional ROS. As oxidative stress accumulates, it contributes to the degeneration of these neurons, leading to the motor symptoms of PD28.

Studies have shown that oxidative stress markers, such as lipid peroxidation and DNA oxidation, are elevated in the substantia nigra of PD patients. In addition, post-mortem analyses reveal increased oxidative damage to mitochondrial DNA in this brain region, linking oxidative stress directly to neuronal death in the substantia nigra29.

Integrating molecular and genetic data on oxidative stress into understanding PD pathogenesis highlights the complex interplay between environmental factors and genetic susceptibility. These insights can lead to more targeted therapeutic approaches to modulate oxidative stress and improve mitochondrial function in PD patients.

 Parkinson’s Disease Models

Interpreting models that explore the relationships between gene mutations, environmental influences, and Parkinson’s disease involves understanding genetic and environmental factors separately and in conjunction. Animal models are crucial for this research because they allow the manipulation and observation of various aspects of PD pathology.

Genetic models focus on specific mutations that are known to be associated with PD. Alpha-synuclein models, for instance, involve genetically modified animals to overexpress human alpha-synuclein, leading to PD-like symptoms such as motor deficits and Lewy body formation. Additionally, animals with specific mutations in the alpha-synuclein gene (e.g., A53T, A30P) exhibit progressive neurodegeneration and motor impairment30. LRRK2 models include knock-in or transgenic mice with the LRRK2 G2019S mutation, which show dopaminergic neuron loss and motor deficits similar to those seen in PD patients31. LRRK2 knockout models help to reveal the effects of losing this gene’s function, highlighting compensatory mechanisms and pathways involved in PD32. PINK1 and Parkin (signaling genes involved in indicating mitochondrial damage) knockout models demonstrate mitochondrial dysfunction and increased vulnerability to stress, which mimic early-onset PD33.

Animal models that investigate environmental impacts in PD often involve toxin-induced PD. One of the most common is the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model, where administration of the neurotoxin MPTP in mice and primates causes selective dopaminergic neuron loss, replicating hallmark features of PD. The MPTP is meant to imitate the harmful compounds found in nature that cause DA neuron deterioration. Mice exposed to MPTP demonstrate DA loss similar to humans in the presence of such toxins. These models help us understand how certain environmental factors can trigger or exacerbate the disease by inducing DA neuron degeneration34. By combining genetic predispositions with ecological exposures, researchers can better understand the complex interplay that leads to PD onset. This integrated approach is crucial for developing more effective therapeutic strategies and preventive measures.

Toxin Studies

Pesticides

The interaction between genetic predispositions and environmental exposures, particularly to toxins such as pesticides, plays a crucial role in the pathogenesis of Parkinson’s disease. Research has shown that certain ecological toxins exacerbate genetic vulnerabilities by inducing oxidative stress and mitochondrial dysfunction, leading to accelerated degeneration of DA neurons in the substantia nigra. A 2017 study involving the pesticide rotenone provided compelling evidence of its detrimental effects on mitochondrial function35. Rotenone selectively inhibited mitochondrial complex I, increasing ROS production and subsequent oxidative stress. Chronic exposure to rotenone in rats resulted in the degeneration of DA neurons and the formation of alpha-synuclein aggregates, which are hallmark features of PD.

Similarly, paraquat, another widely used pesticide, has been implicated in generating oxidative stress within DA neurons. A 2009 study reported that paraquat exposure in mice significantly elevated superoxide radicals, causing oxidative damage specifically in the substantia nigra36. This effect was notably more pronounced in mice with genetic mutations affecting oxidative stress response pathways, such as those involving the

DJ-1 gene, highlighting the connection between genetic susceptibility and environmental factors.

Furthermore, epidemiological studies have shown the heightened risk of PD in individuals with genetic mutations when exposed to these environmental toxins. For example, a recent 2024 study found that individuals carrying the LRRK2 G2019S mutation had a significantly increased risk of developing PD when exposed to pesticides compared to non-carriers37. This suggests a synergistic relationship where genetic mutations and environmental toxin exposure can increase the risk of DA neuron degeneration. These findings underscore the importance of considering genetic and environmental factors to understand PD’s complex etiology and develop targeted prevention and treatment strategies.

Air Pollution

In recent epidemiological studies, air pollution has emerged as a significant environmental risk factor for PD. Several studies have investigated the association between exposure to air pollution—particulate matter (PM) and nitrogen dioxide (NO₂)—and the incidence of PD, providing quantitative evidence to support this link.

One of the most comprehensive longitudinal studies assessing the relationship between air pollution and PD was conducted by Chen et al. from 2001 to 201338. This study utilized a cohort from Ontario, Canada, and investigated long-term exposure to fine particulate matter (PM2.5, particles with diameters less than 2.5 microns). The results showed a statistically significant association between higher levels of PM2.5 exposure and an increased risk of developing PD. Specifically, for each interquartile range (IQR) increase in PM2.5 levels (3.8 µg/m³), a 4% increase in PD incidence was found (95% confidence interval, 1.01-1.08) after adjusting for various confounding variables. There were also positive associations relating PD incidence with NO₂ and ozone levels (hazard ratios ranged from 1.03 to 1.04). 

In a 2015 U.S.-based study conducted by Kioumourtzoglou et al., the researchers examined the effect of long-term exposure to PM2.5 on the development of PD39. The study followed Medicare enrollees aged 65 years or older. The results indicated that exposure to PM2.5 was significantly associated with an increased risk of PD (HR = 1.08, 95% CI: 1.04–1.12) per 1-µg/m³ increase in PM2.5.

In another influential study, Kwon et al., 2024 examined the relationship between

traffic-related air pollution/fine particulate matter and PD incidence in California40. Using a case-control design, the researchers found that individuals (average age of 68 years) exposed to higher levels of traffic-related pollutants had a significantly higher risk of PD. Specifically, they observed that exposure to PM2.5 for 10 years was associated with an odds ratio (OR) of 1.62 (95% CI: 1.22–2.15, p < 0.01) for residential exposure and 1.85 in workplaces (95% Cl 1.21, 2.85, p <0.01).

Risk Factor Genes

The LRRK2 and SNCA genes are among the most well-studied genetic factors in Parkinson’s disease, with both playing significant roles in the disease’s pathogenesis. Mutations in the LRRK2 gene, particularly the G2019S mutation, are the most common genetic cause of familial and sporadic PD41. LRRK2 encodes a kinase involved in various cellular processes, including autophagy, mitochondrial function, and inflammation. Mutations in LRRK2 are believed to enhance its kinase activity, leading to hyperphosphorylation of downstream targets, which can cause mitochondrial dysfunction and increased oxidative stress in DA neurons42. This mutation’s effect on mitochondrial function is particularly critical, as it exposes the vulnerability of these neurons to environmental toxins, such as pesticides, that also induce oxidative stress and mitochondrial damage. This interaction between LRRK2 mutations and ecological exposures underscores the gene’s central role in PD’s complex etiology.

Research suggests that individuals with specific LRRK2 mutations may experience a less favorable response to traditional dopaminergic treatments, such as Levodopa, which is commonly used to manage PD symptoms43. Although it is not known why those with the LRRK2 mutation respond less favorably, this differential response underscores the importance of LRRK2 in modulating the disease’s progression. This suggests therapeutic strategies may need to be tailored according to genetic background to maximize efficacy.

Recent research suggests alpha-synuclein may interfere with mitochondrial function and promote oxidative stress, creating a feedback loop that accelerates neurodegeneration44. Combining SNCA gene mutations and environmental factors, such as heavy metals or pesticide exposure, can strengthen alpha-synuclein aggregation and further stress cellular pathways critical for neuron survival. Exposure to heavy metals and pesticides is linked to SNCA mutations with neurodegenerative properties45. Together, the LRRK2 and SNCA genes illustrate how genetic predispositions can interact with environmental exposures to drive the progression of PD, offering insights into potential therapeutic targets.

Along with LRRK2 and SNCA, GBA is a protein-encoding gene relevant to PD incidence. Mutations in GBA that lead to a gene deficiency result in oxidative stress and are associated with neurodegenerative conditions, notably PD. The GBA1 gene provides instructions for producing an enzyme called lysosomal acid glucosylceramidase, which functions within lysosomes. Lysosomes use digestive enzymes to break down harmful substances, eliminate invading bacteria, and recycle worn-out cell components. Lysosomal acid glucosylceramidase helps break down a molecule called glucocerebroside into glucose (a sugar) and ceramide (a simpler fat molecule). Glucocerebroside, a glycolipid found in the membranes around cells, is broken down when cells die, allowing its components to be reused in forming new cells46. GBA deficiency can lead to an accumulation of glucocerebroside, contributing to increased oxidative stress. This stress, which damages cells and disrupts cellular function, is a massive contributor to PD onset.

Epigenetics

Epigenetic modifications, particularly DNA methylation, play a significant role in the pathogenesis of Parkinson’s disease by influencing gene expression without altering the underlying DNA sequence47. DNA methylation typically involves adding a methyl group to the cytosine base in DNA, which can suppress gene expression48. In PD, abnormal DNA methylation patterns have been observed in several genes associated with the disease, including those involved in mitochondrial function, oxidative stress response, and neuroinflammation49.

One of the critical environmental factors that may influence DNA methylation in PD is exposure to heavy metals, such as lead, cadmium, and mercury. These metals disrupt normal cellular processes and have been linked to increased oxidative stress and neurotoxicity50. A 2015 study suggests that heavy metals can also alter DNA methylation patterns, leading to the dysregulation of genes critical for neuronal survival. For instance, exposure to lead has been shown to induce hypomethylation (the loss of methylated cytosine bases) in the promoter regions of genes involved in antioxidant defense, reducing their expression and leaving neurons more vulnerable to oxidative damage51.

These epigenetic changes may further complicate the interaction between PD’s environmental exposures and genetic predispositions. Individuals with genetic mutations that already impair cellular defenses against oxidative stress may be particularly susceptible to the detrimental effects of altered DNA methylation induced by heavy metal exposure52. This combination of genetic and epigenetic factors could accelerate DA neuron degeneration, contributing to PD onset and progression.

Understanding the role of epigenetics in PD not only provides insight into the complex mechanisms driving the disease but also highlights potential avenues for therapeutic intervention. Targeting abnormal DNA methylation patterns, possibly through demethylating/deaminating agents or other epigenetic therapies that offer genetic variability and healing, could provide a novel approach to slowing or preventing the neurodegenerative processes in PD.

Therapeutic Approaches in Treating Parkinson’s Disease

In PD, genetic predispositions and molecular dysfunctions contribute to the progressive degeneration of dopaminergic neurons, especially in the substantia nigra. While traditional treatments like levodopa and dopamine agonists provide symptomatic relief, they do not halt the disease’s progression. Emerging therapies, such as Deep Brain Stimulation (DBS) and gene therapy, aim to target the underlying molecular dysfunctions, offering more advanced therapeutic options53.

Deep Brain Stimulation

DBS is an established treatment that involves implanting electrodes on the brain targeting specific regions, such as the subthalamic nucleus or the globus pallidus interna. These electrodes deliver electrical impulses that modulate abnormal brain activity, helping to alleviate motor symptoms like tremors, bradykinesia, and rigidity54.

Mechanism of DBS

Although the exact mechanism of how DBS alleviates PD symptoms is not fully understood, it is thought to disrupt aberrant neural circuits in PD. Dopaminergic neuron loss leads to hyperactivity in the subthalamic nucleus and other brain regions, causing motor dysfunctions. DBS reduces this hyperactivity by modulating synaptic activity, restoring more balanced signaling in the motor pathways55.

Effectiveness

DBS has been shown to improve motor function significantly in PD patients, particularly those with advanced disease who do not respond well to medications. For instance, clinical studies have demonstrated enhanced motor scores (as measured by the Unified Parkinson’s Disease Rating Scale) in patients receiving DBS. Long-term follow-up studies show these improvements can persist for up to ten years56. Limitations and Genetic Implications 

While DBS is effective in controlling motor symptoms, it does not address non-motor symptoms like cognitive decline or neuropsychiatric issues, which are often exacerbated by genetic factors like LRRK2 or SNCA mutations. Moreover, not all patients respond equally to DBS, with genetic variations potentially influencing individual outcomes.

Research suggests that PD patients with GBA mutations can experience detrimental motor impairment after DBS57. Also, patients with LRRK2 mutations may exhibit unpleasant behavioral responses to DBS, warranting further investigation on a

case-by-case basis into how genetic predispositions impact the effectiveness of this therapy.

Gene Therapy

Gene therapy offers a more direct approach to targeting the underlying genetic and molecular dysfunctions in PD. Recent advances focus on using viral vectors to deliver genetic material into specific brain regions to restore normal cellular function or slow disease progression58.

Gene Replacement Therapy

This approach involves introducing healthy copies of genes into neurons to compensate for dysfunctional or mutated genes. For example, gene therapy targeting GBA can restore the enzymatic function engaged in the production of glucocerebrosidase and glucosylceramide, potentially slowing the progression of the disease59.

Neuroprotective Gene Therapy

Specific gene therapies aim to increase the production of neuroprotective factors. For instance, viral vectors have been used to deliver the neurturin gene, which encodes a protein that promotes the survival of dopaminergic neurons. Clinical trials showed promise but involved various supplementary gene materials, making pinpointing an exact treatment difficult60.

CRISPR/Cas9 Gene Editing

Advances in gene-editing technologies like CRISPR/Cas9 can correct disease-causing mutations at the genomic level. For example, researchers are investigating

CRISPR-mediated correction of specific mutations in the LRRK2 or SNCA genes or deletion of genes such as PINK1 and PARK2 in their entirety61. While this approach is still experimental, it holds promise for permanently correcting genetic mutations associated with PD.

Challenges and Future Directions

One of the primary challenges of gene therapy is the safe and targeted delivery of genetic material. Viral vectors, such as adeno-associated viruses (AAV), have been used in clinical trials, but there is ongoing research to optimize their delivery and minimize immune responses62. Additionally, gene therapy is still in the experimental stages for many genetic mutations associated with PD, such as PINK1 and Parkin mutations, and further research is needed to determine long-term efficacy and safety.

Both DBS and gene therapy represent promising advanced treatment options for PD. While DBS primarily targets the disease’s motor symptoms by modulating abnormal brain activity, gene therapy addresses the underlying molecular and genetic disruptions contributing to PD progression. As our understanding of the genetic predispositions to PD, such as the LRRK2, SNCA, and GBA mutations, improve, these therapies will likely become more personalized, targeting specific genetic profiles for better outcomes.

Conclusion

In summary, this review underscores the intricate impacts of genetic predispositions and environmental factors in the pathogenesis of Parkinson’s disease. The evidence highlights how gene mutations like LRRK2 and SNCA can exacerbate cellular dysfunctions, particularly when combined with environmental exposures such as pesticides. This interaction accelerates the degeneration of DA neurons, leading to the hallmark symptoms of PD. Exploring cellular mechanisms, such as impaired synaptic plasticity and chronic neuroinflammation, offers critical insights into the disease’s progression.

The findings of this review have significant implications for the broader understanding of neurodegenerative diseases. By elucidating the connections between genetics, environmental factors, and epigenetic modifications, this work provides a foundation for developing more targeted and effective therapeutic strategies. These insights deepen our understanding of PD and open new avenues for research into preventive measures and treatments that could alter the course of the disease. The importance of considering genetic and environmental factors in future studies cannot be overstated, as it is essential for advancing our ability to combat Parkinson’s disease and improve patient outcomes.

Limitations

This review paper is limited because it did not systematically search for literature and needs inclusion/exclusion criteria for studies.

Acknowledgments

Dhvani Thakkar – Program Manager
Jishnu Ghose – Publication Strategist
Joanna Eckhardt – Mentor

References

  1. C. Labbé, O. Lorenzo-Betancor, O.A. Ross. Epigenetic regulation in Parkinson’s disease. Acta Neuropathol. 4(132), 515-530 (2016). []
  2. L. Lossi, C. Castagna, A. Merighi. An Overview of the Epigenetic Modifications in the Brain under Normal and Pathological Conditions. Int J Mol Sci. 7(25), (2024). []
  3. A. Kouli, K.M. Torsney, W.L. Kuan. Parkinson’s Disease: Etiology, Neuropathology, and Pathogenesis. Parkinson’s Disease: Pathogenesis and Clinical Aspects [Internet]. Brisbane (AU): Codon Publications. 1, (2018). []
  4. A. W. Willis, E. Roberts, J. C. Beck, B. Fiske, W. Ross, R. Savica, S. K. Van Den Eeden, C. M. Tanner, C. Marras on behalf of the Parkinson’s Foundation P4 Group. Incidence of Parkinson disease in North America. npj Parkinsons

    Disease. 8(170), (2022). []

  5. N. M. Kanaan, F. P. Manfredsson. Loss of functional alpha-synuclein: a toxic event in Parkinson’s disease?. Journal of Parkinson’s Disease. 2(4), 249–267 (2012). []
  6. L. Stefanis. α-Synuclein in Parkinson’s Disease. Cold Spring Harbor Perspectives in Medicine. 2(2), a009399 (2012). []
  7. C. A. Stamerra, P. Di Giosia, P. Giorgini, C. Ferri, V. N. Sukhorukov, A. Sahebkar. Mitochondrial dysfunction and cardiovascular disease: pathophysiology and emerging therapies. Oxidative Medicine and Cellular Longevity. 2022, 9530007 (2022). []
  8. R. V. Rao, D. E. Bredesen. Misfolded proteins, endoplasmic reticulum stress and neurodegeneration. Current Opinion in Cell Biology. 16(6), 653–662 (2004). []
  9. M. Colonna, O. Butovsky. Microglia function in the central nervous system during health and neurodegeneration. Annual Review of Immunology. 35, 441–468 (2017). []
  10. J. Q. Li, L. Tan, J. T. Yu. The role of the LRRK2 gene in Parkinsonism. Molecular Neurodegeneration. 9, 47 (2014). []
  11. M. Madadi Asl, A. H. Vahabie, A. Valizadeh. Dopaminergic modulation of synaptic plasticity, its role in neuropsychiatric disorders, and its computational modeling. Basic and Clinical Neuroscience. 10(1), 1–12 (2019). [] []
  12. T. Kitada, A. Pisani, D. R. Porter, H. Yamaguchi, A. Tscherter, G. Martella, P. Bonsi, C. Zhang, E. N. Pothos, J. Shen. Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice. Proceedings of the National

    Academy of Sciences. 104(27), 11441-11446 (2007). []

  13. Parkinson’s disease: challenges, progress, and promise. https://www.ninds.nih.gov/current-research/focus-disorders/parkinsons-disease-r esearch/parkinsons-disease-challenges-progress-and-promise (2015). []
  14. D. J. Surmeier, S. M. Graves, W. Shen. Dopaminergic modulation of striatal networks in health and Parkinson’s disease. Current Opinion in Neurobiology. 29, 109–117 (2014). [] [] []
  15. A. S. Vetchinova, M. R. Kapkaeva, M. V. Ivanov, K. A. Kutukova, N. M. Mudzhiri, L. E. Frumkina, A. V. Brydun, V. S. Sukhorukov, S. N. Illarioshkin. Mitochondrial dysfunction in dopaminergic neurons derived from patients with LRRK2- and SNCA-associated genetic forms of Parkinson’s disease. Current Issues in Molecular Biology. 45(10), 8395–8411 (2023). []
  16. S. Ramesh, A. S. P. M. Arachchige. Depletion of dopamine in Parkinson’s disease and relevant therapeutic options: A review of the literature. AIMS Neuroscience. 10(3), 200–231 (2023). []
  17. K. R. Gandhi, A. Saadabadi. Levodopa (L-Dopa). StatPearls [Internet]. https://www.ncbi.nlm.nih.gov/books/NBK482140/ (2024). []
  18. J. W. Lee, J. R. Cannon. LRRK2 mutations and neurotoxicant susceptibility.

    Experimental Biology and Medicine (Maywood, N.J.). 240(6), 752–759 (2015). []

  19. J. W. Lee, J. R. Cannon. LRRK2 mutations and neurotoxicant susceptibility. Experimental Biology and Medicine (Maywood, N.J.). 240(6), 752–759 (2015). []
  20. A. Kanthasamy, H. Jin, A. Charli, A. Vellareddy, A. Kanthasamy. Environmental neurotoxicant-induced dopaminergic neurodegeneration: a potential link to impaired neuroinflammatory mechanisms. Pharmacology & Therapeutics. 197, 61–82 (2019). []
  21. H. Juárez Olguín, D. Calderón Guzmán, E. Hernández García, G. Barragán Mejía. The role of dopamine and its dysfunction as a consequence of oxidative stress. Oxidative Medicine and Cellular Longevity, 9730467 (2016). []
  22. T. V. Bliss, S. F. Cooke. Long-term potentiation and long-term depression: a clinical perspective. Clinics (Sao Paulo, Brazil). 66(Suppl 1), 3–17 (2011). []
  23. M. Huang, A. Bargues-Carot, Z. Riaz, H. Wickham, G. Zenitsky, H. Jin, V. Anantharam, A. Kanthasamy, A. 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. 23(18), 10808 (2022). []
  24. V. Dias, E. Junn, M. M. Mouradian. The role of oxidative stress in Parkinson’s disease. J Parkinsons 3(4), 461-491 (2013). []
  25. E. M. Rocha, M. T. Keeney, R. Di Maio, B. R. De Miranda, J. T. Greenamyre. LRRK2 and idiopathic Parkinson’s disease. Trends Neurosci. 45(3), 224-236 (2022). []
  26. J. Konovalova, D. Gerasymchuk, I. Parkkinen, P. Chmielarz, A. Domanskyi. Interplay between MicroRNAs and Oxidative Stress in Neurodegenerative Diseases. Int J Mol Sci. 20(23), (2019). []
  27. L. Stefanis. α-Synuclein in Parkinson’s disease. Cold Spring Harb Perspect Med. 2(2), (2012). []
  28. B. G. Trist, D. J. Hare, K. L. Double. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging Cell. 18(6), (2019). []
  29. V. Dias, E. Junn, M. M. Mouradian. The role of oxidative stress in Parkinson’s disease. J Parkinsons Dis. 3(4), 461-491 (2013). []
  30. J. Sonne, V. Reddy, M. R. Beato. Neuroanatomy, Substantia Nigra. StatPearls [Internet]. https://www.ncbi.nlm.nih.gov/books/NBK536995/ (2022). []
  31. K. M. L. Cramb, D. Beccano-Kelly, S. J. Cragg, R. Wade-Martins. Impaired dopamine release in Parkinson’s disease. Brain: a Journal of Neurology. 146(8), 3117–3132 (2023). []
  32. R. Giráldez-Pérez, M. Antolín-Vallespín, M. Muñoz, A. Sánchez-Capelo. Models of α-synuclein aggregation in Parkinson’s disease. Acta Neuropathologica Communications. 2, 176 (2014). []
  33. E. E. S. Chang, P. W. Ho, H. F. Liu, S. Y. Pang, C. T. Leung, Y. Malki, Z. Y. Choi, D. B. Ramsden, S. L. Ho. LRRK2 mutant knock-in mouse models: therapeutic relevance in Parkinson’s disease. Translational Neurodegeneration. 11(1), 10 (2022). []
  34. Y. Xiong, T. M. Dawson, V. L. Dawson. Models of LRRK2-associated Parkinson’s disease. Advances in Neurobiology. 14, 163–191 (2017). []
  35. Z. Wang, S. W. Chan, H. Zhao, K. K. Miu, W. Y. Chan. Outlook of PINK1/Parkin signaling in molecular etiology of Parkinson’s disease, with insights into Pink1 knockout models. Zoological Research. 44(3), 559–576 (2023). []
  36. G. E. Meredith, D. J. Rademacher. MPTP mouse models of Parkinson’s disease: an update. Journal of Parkinson’s Disease, 1(1), 19–33 (2011). []
  37. S. Heinz, A. Freyberger, B. Lawrenz, L. Schladt, G. Schmuck, H. Ellinger-Ziegelbauer. Mechanistic investigations of the mitochondrial complex I inhibitor rotenone in the context of pharmacological and safety evaluation. Scientific Reports. 7, 45465 (2017). []
  38. S. Shin, R. T. Burnett, J. C. Kwong, P. Hystad, A. van Donkelaar, J. R. Brook, R. Copes, K. Tu, M. S. Goldberg, P. J. Villeneuve, R. V. Martin, B. J. Murray, A. S. Wilton, A. Kopp, H. Chen, Effects of ambient air pollution on incident Parkinson’s disease in Ontario, 2001 to 2013: a population-based cohort study, International Journal of Epidemiology. 47(6), 2038–2048 (2018). []
  39. M. A. Kioumourtzoglou, J. D. Schwartz, M. G. Weisskopf, et al. Long-term PM2.5 Exposure and Neurological Hospital Admissions in the Northeastern United States. Environ Health Perspect. 124(1), 23-29 (2016). []
  40. D. Kwon, K. C. Paul, Y. Yu, K. Zhang, A. D. Folle, J. Wu, J. M. Bronstein, B. Ritz.Traffic-related air pollution and Parkinson’s disease in central California. Environmental Research. 240(1), (2024). []
  41. V. Dias, E. Junn, M. M. Mouradian. The role of oxidative stress in Parkinson’s disease. Journal of Parkinson’s Disease. 3(4), 461–491 (2013). []
  42. E. G. Brown, S. M. Goldman, C. S. Coffey, A. Siderowf, T. Simuni, C. Meng, M. C. Brumm, C. Caspell-Garcia, K. Marek, C. M. Tanner, Parkinson’s Progression Markers Initiative. Occupational pesticide exposure in Parkinson’s disease related to GBA and LRRK2 variants. Journal of Parkinson’s Disease. 14(4), 737–746 (2024). []
  43. J. H. Kluss, A. Mamais, M. R. Cookson. LRRK2 links genetic and sporadic Parkinson’s disease. Biochemical Society Transactions. 47(2), 651–661 (2019). []
  44. E. M. Rocha, M. T. Keeney, R. Di Maio, B. R. De Miranda, J. T. Greenamyre. LRRK2 and idiopathic Parkinson’s disease. Trends in Neurosciences. 45(3), 224–236 (2022). []
  45. R. Saunders-Pullman, D. Raymond, S. Elango. LRRK2 Parkinson disease. GeneReviews® [Internet]. https://www.ncbi.nlm.nih.gov/books/NBK1208/ (2006). []
  46. MedlinePlus [Internet]. https://medlineplus.gov/genetics/gene/gba1/ []
  47. K. J. Lin, K. L. Lin, S. D. Chen, C. W. Liou, Y. C. Chuang, H. Y. Lin, T. K. Lin. The overcrowded crossroads: mitochondria, alpha-synuclein, and the endo-lysosomal system interaction in Parkinson’s disease. International Journal of Molecular Sciences. 20(21), 5312 (2019). []
  48. I. Ullah, L. Zhao, Y. Hai, M. Fahim, D. Alwayli, X. Wang, H. Li. “Metal elements and pesticides as risk factors for Parkinson’s disease – A review”. Toxicology Reports. 8, 607–616 (2021). []
  49. H. Song, J. Chen, J. Huang, P. Sun, Y. Liu, L. Xu, C. Wei, X. Mu, X. Lu, W. Wang, N. Zhang, M. Shang, M. Mo, W. Zhang, H. Zhao, F. & Han. Epigenetic modification in Parkinson’s disease. Frontiers in Cell and Developmental Biology. 11, 1123621 (2023). []
  50. C. M. Lanata, S. A. Chung, L. A. Criswell. DNA methylation 101: what is important to know about DNA methylation and its role in SLE risk and disease heterogeneity. Lupus Science & Medicine, 5(1), e000285 (2018). []
  51. Z. Chen, M. Rasheed, Y. Deng. The epigenetic mechanisms involved in mitochondrial dysfunction: Implication for Parkinson’s disease. Brain Pathology. 32(3), e13012 (2022). []
  52. O. M. Ijomone, O. K. Ijomone, J. D. Iroegbu, C. W. Ifenatuoha, N. F. Olung, M. Aschner. Epigenetic influence of environmentally neurotoxic metals. Neurotoxicology. 81, 51–65 (2020). []
  53. K. E. Shalaby, O. M. A. El-Agnaf. Gene-Based Therapeutics for Parkinson’s Disease. Biomedicines. 10(8), (2022). []
  54. J. K. Krauss, N. Lipsman, T. Aziz, et al. Technology of deep brain stimulation: current status and future directions. Nat Rev Neurol. 17(2), 75-87 (2021). []
  55. B. Davidson, L. Milosevic, L. Kondrataviciute, L. V. Kalia, S. K. Kalia. Neuroscience fundamentals relevant to neuromodulation: Neurobiology of deep brain stimulation in Parkinson’s disease. Neurotherapeutics. 21(3), (2024). []
  56. S. Bratsos, D. Karponis, S. N. Saleh. Efficacy and Safety of Deep Brain Stimulation in the Treatment of Parkinson’s Disease: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Cureus. 10(10), (2018). []
  57. K. Kalinderi, V. Papaliagkas, L. Fidani. Surgicogenomics: The Role of Genetics in Deep Brain Stimulation in Parkinson’s Disease Patients. Brain Sci. 14(8), (2024). []
  58. P. G. Coune, B. L. Schneider, P. Aebischer. Parkinson’s disease: gene therapies. Cold Spring Harb Perspect Med. 2(4), (2012). []
  59. I. Milenkovic, S. Blumenreich, A. H. Futerman. GBA mutations, glucosylceramide and Parkinson’s disease. Current Opinion in Neurobiology. 72, 148-154 (2022). []
  60. D. G. T. Parambi, K. S. Alharbi, R. Kumar, et al. Gene Therapy Approach with an Emphasis on Growth Factors: Theoretical and Clinical Outcomes in Neurodegenerative Diseases. Mol Neurobiol. 59(1),191-233 (2022). []
  61. J. N. Nojadeh, N. S. B. Eryilmaz, B. I. Ergüder. CRISPR/Cas9 genome editing for neurodegenerative diseases. EXCLI J. 22, 567-582 (2023). []
  62. D. Wang, P. W. L. Tai, G. Gao. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 18(5), 358-378 (2019). []

LEAVE A REPLY

Please enter your comment!
Please enter your name here