Call for Abstract
Scientific Program
37th World Congress on Neurology and Neuroscience, will be organized around the theme “NeuroInnovate: Precision Brain Health, Circuit Modulation & Advanced Neurological Therapies”
NEUROSCIENCE_2026 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in NEUROSCIENCE_2026
Submit your abstract to any of the mentioned tracks.
Register now for the conference by choosing an appropriate package suitable to you.
Rapid advances in precision neurotherapeutics are redefining how neurological disorders are treated by allowing clinicians to modulate dysfunctional neural circuits with unprecedented specificity. Current innovations combine electrophysiology, optogenetics, chemogenetics and high-resolution neural mapping to identify aberrant pathways driving disease symptoms. Targeted stimulation platforms such as adaptive deep-brain stimulation, closed-loop neurodevices and cortical modulators now personalize stimulation parameters using real-time neural data, greatly improving outcomes in conditions like epilepsy, Parkinson’s disease, depression and movement disorders. Integration of molecular signatures, computational modeling and non-invasive neuromodulatory tools such as TMS, tDCS and focused ultrasound enables multi-layered intervention strategies that adjust neural excitability, restore communication between networks and minimize off-target effects. As neuromodulation technologies converge with AI-driven analytics and neural biomarkers, therapeutic precision continues to increase, providing more durable symptom control and a deeper understanding of how circuit dysfunction evolves. This transformative shift toward individualized neural modulation represents a major step forward in therapeutic neuroscience, offering safer and more effective interventions for a broad spectrum of neurological diseases.
Artificial intelligence is reshaping neurological diagnostics by enabling faster, more sensitive and more accurate detection of complex brain disorders. Advanced machine-learning systems now analyze multimodal data—including neuroimaging, electrophysiology, cognitive metrics and genomic signatures to identify abnormalities long before clinical symptoms fully emerge. Predictive analytics platforms build personalized risk profiles by integrating longitudinal patient data, enabling clinicians to anticipate neurological decline, monitor progression and refine treatment plans with real-time guidance. AI-driven image analysis enhances the detection of microstructural changes in MRI, CT and PET scans that may escape human observation, accelerating diagnosis of diseases such as Alzheimer’s, multiple sclerosis, epilepsy and brain tumors. Automated EEG and EMG interpretation further reduces diagnostic variability and supports clinicians in acute settings. As explainable AI continues to improve transparency and regulatory acceptance, intelligent diagnostic ecosystems are emerging that combine wearable sensors, digital biomarkers and remote monitoring. These advancements are transforming neurological care from reactive management to proactive, prediction-based intervention, significantly improving clinical outcomes.
Breakthroughs in neuronal regeneration and synaptic plasticity research are laying the foundation for therapies that aim to restore lost neurological function rather than simply manage symptoms. Cellular and molecular studies have uncovered key drivers of axonal growth, dendritic repair, synapse formation and network rewiring, providing new therapeutic targets for conditions involving traumatic injury, stroke, neurodegeneration and demyelinating diseases. Advances in neurotrophic factors, extracellular matrix modulation, bioengineered scaffolds and pro-regenerative gene programs have shown promise in promoting structural and functional recovery. Modulation of plasticity through pharmacological enhancers, activity-dependent stimulation and enriched behavioral environments further strengthens neural circuit reconstruction. Cutting-edge research explores how glial cells, metabolic pathways and immune responses influence regenerative capacity. Integrating neurorehabilitation strategies with regenerative interventions is accelerating the translation of laboratory discoveries into clinical therapies. As regenerative neuroscience progresses, the prospect of restoring meaningful neurological function becomes increasingly attainable.
Understanding the molecular mechanisms underlying protein misfolding and aggregation is central to advancing therapies for major neurodegenerative disorders. Research on amyloid-beta, tau, alpha-synuclein and TDP-43 has revealed intricate pathways that trigger synaptic loss, metabolic dysfunction, oxidative stress and neuronal death. Structural biology, cryo-EM and proteomics have deepened insights into how toxic oligomers form and propagate across neural networks. Therapeutic strategies now focus on halting misfolding, blocking seeding, enhancing proteostasis and promoting clearance through autophagy and immunotherapy. Anti-amyloid and anti-tau agents, molecular chaperones, gene-silencing technologies and small-molecule stabilizers are under active development. Early detection through molecular biomarkers, including CSF profiles and PET ligands, is enabling more targeted clinical trials. These advances collectively deepen understanding of neurodegenerative cascades and guide the creation of more effective disease-modifying treatments.
High-throughput sequencing and omics technologies have revolutionized the study of neurological disorders by revealing the genetic and regulatory underpinnings of disease susceptibility and progression. Genomic profiling uncovers risk variants, rare mutations and polygenic influences that shape neurological phenotypes. Epigenomic mapping highlights how environmental exposures, lifestyle factors and disease states alter regulatory landscapes through methylation patterns, chromatin remodeling and histone modifications. Transcriptomic analyses provide snapshots of gene-expression dynamics across cell types and brain regions, helping define pathogenic pathways and therapeutic targets. Integration of multi-omics datasets through computational modeling enhances diagnostic accuracy and supports precision-driven treatment strategies. As personalized medicine advances, these molecular signatures guide patient stratification, drug-response prediction and early disease detection, transforming how complex brain disorders are understood and treated.
The interplay between the nervous and immune systems is increasingly recognized as a central contributor to neurological disease development and recovery. Microglia, astrocytes and peripheral immune cells modulate neuroinflammation, synaptic pruning, injury response and repair processes. Emerging evidence demonstrates that dysregulated immune signaling contributes to disorders such as Alzheimer’s, multiple sclerosis, Parkinson’s disease and neuropsychiatric conditions. Advances in immunotherapeutics aim to restore balanced immune activity through targeted cytokine modulation, immune checkpoint regulation, microglial reprogramming and cellular therapies. Understanding how immune pathways intersect with neural networks enables the development of innovative interventions that reduce chronic inflammation, enhance repair and protect neural integrity. As neuroimmunology continues to advance, it offers new avenues for treating conditions previously viewed through a purely neurological lens.
Modern neuropharmacology is evolving toward targeted, efficient and personalized approaches to treat complex CNS disorders. New drug classes leverage receptor-specific modulation, allosteric regulation, synaptic stabilization and neuroprotective mechanisms. Innovations in drug delivery are transforming therapeutic potential by overcoming the challenges posed by the blood–brain barrier. Nanocarriers, lipid-based vesicles, polymeric systems, intranasal routes and focused ultrasound-assisted delivery significantly improve CNS penetration. Long-acting formulations, programmable release technologies and precision dosing strategies reduce side effects and enhance therapeutic adherence. Pharmacogenomic profiling enables individualized treatment selection based on genetic and metabolic differences. Together, these advancements accelerate the development of safer and more effective neurological treatments, bringing forward a new era of personalized neuropharmacology.
The brain–gut–microbiome axis has emerged as a key regulatory system influencing neurological health, behavior and metabolic function. Microbial metabolites, immune signals and neural pathways shape processes such as neuroinflammation, neurotransmitter synthesis, stress resilience and cognitive performance. Dysbiosis has been linked to conditions ranging from depression and anxiety to Parkinson’s disease, autism spectrum disorders and multiple sclerosis. Therapeutic strategies including targeted probiotics, prebiotics, postbiotics, diet-based interventions and microbial metabolites aim to restore microbiome balance and modulate neurobiological processes. Cutting-edge research examines how metabolites influence synaptic function, glial activity and blood–brain barrier integrity. Integrating microbiome science with metabolic regulation provides new opportunities for designing individualized neurological interventions based on gut-derived biomarkers.
Advances in stem-cell science and neuroengineering are enabling the creation of transformative models and therapies for neurological disorders. Neural stem cells, induced pluripotent stem cells and brain organoids provide unprecedented insights into human neurodevelopment, disease mechanisms and personalized therapeutic responses. Organoids replicate complex neural circuits, enabling studies of genetics, infections, drug effects and degeneration. Regenerative neuroengineering integrates biomaterials, 3D scaffolds and bioelectronic interfaces to promote neural repair and functional recovery. Transplantation strategies using stem-cell–derived neurons or glia show promise for restoring connectivity in conditions such as spinal cord injury, Parkinson’s disease and stroke. As safety, differentiation control and integration improve, stem-cell–based therapies move closer to clinical reality, offering hope for regenerative treatments that rebuild damaged neural networks.
Modern neuroimaging technologies provide deep insights into brain architecture, metabolic activity and functional network dynamics. High-resolution fMRI, PET tracers, diffusion imaging and advanced spectroscopy enable early detection of disease processes, mapping of protein aggregates and precise visualization of structural abnormalities. Connectomics reveals how neural circuits reorganize in health and disease, supporting targeted interventions. Digital imaging analytics integrate AI algorithms to detect subtle functional changes, quantify biomarkers and predict progression across multiple neurological disorders. Multimodal imaging enhances diagnostic accuracy and guides therapeutic decision-making, surgical planning and response monitoring. Continued advancements in imaging resolution, portable devices and computational reconstruction are shaping the future of precision neurological care.
Research in cognitive neuroscience continues to expand understanding of how memory, attention, learning and executive function emerge from dynamic neural processes. Advances in neuromodulation, pharmacological enhancement and behavioral training aim to optimize cognition and counteract decline associated with aging, injury or disease. Memory engineering explores technologies such as targeted stimulation, sleep-based interventions and molecular modulation to strengthen or restore memory circuits. Insights into neuroplasticity guide rehabilitation strategies that harness the brain’s capacity for adaptation and reorganization. As cognitive neuroscience integrates AI analytics, neural decoding and multimodal monitoring, it continues to shape interventions that support lifelong cognitive health and functional recovery.
Understanding the neural circuitry underlying movement disorders enables the development of more targeted therapeutic strategies. Deep-brain stimulation remains a major breakthrough, with adaptive, closed-loop and personalized stimulation systems significantly improving outcomes in Parkinson’s disease, dystonia and essential tremor. Research on basal ganglia networks, cerebellar pathways and motor cortex connectivity uncovers novel targets for modulation. Pharmacological innovations and gene-based therapies complement neuromodulation, addressing underlying molecular dysfunction. Advanced imaging and computational models enhance surgical precision and optimize stimulation parameters. These combined innovations greatly expand treatment options and improve quality of life for individuals with movement disorders.
Pediatric neurology presents unique challenges due to rapidly developing neural systems and the complexity of genetic influences on early brain function. Rare neurodevelopmental syndromes often involve multi-system abnormalities, requiring multidisciplinary diagnostic and therapeutic approaches. Advances in genomic sequencing, metabolic profiling and early developmental screening enhance detection and classification of rare conditions. Targeted therapies including gene replacement, antisense oligonucleotides, metabolic modulation and neurorehabilitation are transforming care pathways. Understanding neurodevelopmental trajectories enables earlier intervention, improved prognostic prediction and more effective support strategies for affected children and families.
Stroke and traumatic brain injury remain major global health challenges, but breakthroughs in neurorepair and rehabilitation science are improving long-term outcomes. Early neuroprotection strategies aim to minimize cell death and preserve tissue. Regenerative pathways involving stem cells, growth factors and circuit reactivation support structural recovery. Rehabilitation technologies including robotics, exoskeletons, virtual reality, brain–computer interfaces and AI-guided therapy enable adaptive training environments that enhance functional restoration. Personalized rehabilitation plans, powered by digital biomarkers and neural feedback systems, optimize recovery trajectories. Combined advances in acute treatment and restorative therapies significantly elevate survival and functional independence.
Brain-computer interfaces (BCIs) and neuroprosthetics represent a major leap in restoring communication, mobility and sensory function for individuals with neurological impairment. Non-invasive, minimally invasive and implanted BCIs decode neural signals to control external devices, enabling interaction despite severe paralysis. Neuroprosthetic systems integrate with neural circuits to restore movement, vision or sensory perception. AI enhances signal interpretation, accuracy and adaptability, creating more intuitive and responsive interfaces. As materials science, neural engineering and computational neuroscience converge, intelligent neurotechnology platforms continue to expand possibilities for restoring independence and improving quality of life.
