Next Article in Journal
DW-YOLO: A Model for Identifying Surface Characteristics and Distinguishing Grades of Graphite Ore
Previous Article in Journal
Winnie: A Sensor-Based System for Real-Time Monitoring and Quality Tracking in Wine Fermentation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neuroengineering Frontiers: A Selective Review of Neural Interfaces, Brain–Machine Interactions, and Artificial Intelligence in Neurodegenerative Diseases

1
Department of Mathematics and Statistics, North Carolina A&T State University, Greensboro, NC 27411, USA
2
Department of Biomedical Engineering, Applied Science and Technology, North Carolina A&T State University, Greensboro, NC 27411, USA
3
Bioengineering Program, Department of Chemical, Biological, and Bioengineering, North Carolina A&T State University, Greensboro, NC 27411, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Appl. Sci. 2025, 15(21), 11316; https://doi.org/10.3390/app152111316
Submission received: 26 September 2025 / Revised: 13 October 2025 / Accepted: 15 October 2025 / Published: 22 October 2025
(This article belongs to the Special Issue Brain-on-Chip Platforms: Advancing Neuroscience and Drug Discovery)

Abstract

Neurodegenerative diseases, including Alzheimer’s disease (AD) and Parkinson’s disease (PD), present a growing public health challenge globally. Recent advancements in neurotechnology and neuroengineering have significantly enhanced brain–computer interfaces, artificial intelligence, and organoid technologies, making them pivotal instruments for diagnosis, monitoring, disease modeling, treatment development, and rehabilitation of various diseases. Nonetheless, the majority of neural interface platforms focus on unidirectional control paradigms, neglecting the need for co-adaptive systems where both the human user and the interface continually learn and adapt. This selected review consolidates information from neuroscience, artificial intelligence, and organoid engineering to identify the conceptual underpinnings of co-adaptive and symbiotic human–machine interaction. We emphasize significant shortcomings in the advancement of long-term AI-facilitated co-adaptation, which permits individualized diagnostics and progression tracking in Alzheimer’s disease and Parkinson’s disease. We concentrate on incorporating deep learning for adaptive decoding, reinforcement learning for bidirectional feedback, and hybrid organoid–brain–computer interface platforms to mimic disease dynamics and expedite therapy discoveries. This study outlines the trends and limitations of the topics at hand, proposing a research framework for next-generation AI-enhanced neural interfaces targeting neurodegenerative diseases and neurological disorders that are both technologically sophisticated and clinically viable, while adhering to ethical standards.

1. Introduction

There are various neurodegenerative diseases (ND), but we focus on Alzheimer’s disease (AD) and Parkinson’s disease (PD), which pose significant challenges for modern neuroscience because they present both major healthcare issues and complex scientific obstacles. These NDs have led to continuous brain deterioration and neuron death, which affects millions of people throughout the world. The Alzheimer’s Association reports that 7 million Americans had Alzheimer’s disease in 2025, but this number will reach 13 million by 2050. Healthcare expenses for Alzheimer’s disease and dementia-related conditions will increase from USD 384 billion in 2025 to nearly USD 1 trillion by 2050, according to current projections [1]. The second most prevalent neurodegenerative disorder after Alzheimer’s disease affects 1 million patients in the United States while generating healthcare expenses of USD 51.9 billion during 2017. The number of Parkinson’s disease patients will surpass 1.6 million while the associated healthcare expenses will reach USD 79 billion by 2037 [2,3]. The rising prevalence of diseases and healthcare expenses requires the immediate development of innovative therapeutic solutions that can surpass current treatment methods. The combination of modern neural interface technology with artificial intelligence (AI) presents a promising solution to develop new treatments for neurodegenerative diseases.
Neuroscience and neuroengineering have established brain–machine interfaces (BMIs) as their fundamental technology, enabling artificial brain–device communication through neuromuscular bypasses. The development of external devices continues with brain–computer interfaces (BCIs), neuroprosthetics, and rehabilitation-assistive technology. The process of BMI operation starts with the recording of neural signals through electrodes or EEG, followed by computational signal processing to extract neurological data, which then controls external devices such as cursors, prosthetic limbs, bots, wheelchairs, and communication tools [4,5]. BMI performance depends on the synchronized activity of multiple areas of the brain, including the motor cortex, the prefrontal cortex, and the striatum, according to research findings [6,7,8,9,10]. The main recurring problem with BMIs is that they require more than a simple installation. The success of BMIs depends on the user’s adaptation to the machine through plasticity, cognitive strategies, and algorithmic learning processes. The evolution of BMI control has progressed beyond basic motor mapping, as researchers now recognize that intention decoding, reinforcement signals, and affective states play a crucial role in the development of adaptive, human-centered systems.
Artificial intelligence (AI) has transformed neurodegenerative disease research through its parallel development. Traditional diagnostic methods based on neuroimaging and linear models have shown limited progress in detecting diseases early and identifying patient subgroups. The field of neurodegenerative disease research has expanded significantly through the implementation of machine learning (ML) and deep learning (DL) techniques. AI-driven pipelines that utilize the multimodal fusion of MRI, PET, and functional imaging data, along with predictive modeling of genetic and proteomic profiles, outperform traditional statistical methods in routine applications [11,12]. The combination of CNNs with RNNs enables researchers to achieve accuracy greater than 90% in AD diagnosis and create precise predictions for the progression of cognitive decline [13,14]. Graph-based methods represent an emerging approach that addresses the non-Euclidean structure of brain connectivity. The application of graph neural networks (GNNs) for brain graph analysis produces better results in AD and MCI classification through their ability to analyze brain network topology [15,16]. The research shows that disease indicators spread throughout interconnected networks, rather than remaining within specific brain areas, which makes holistic modeling essential.
The ability of DL to learn hierarchical spatiotemporal representations from raw data makes it a powerful framework for decoding complex neural signals. The implementation of different architectures for BCIs and neural monitoring systems requires specific adaptations to handle particular modality-related problems. The extraction of localized spatial features from multichannel EEG or fMRI signals through convolutional neural networks (CNNs) becomes possible because their convolutional kernels detect inter-electrode and voxel correlations while preserving temporal stability according to [17]. The sequential EEG and ERP data processing benefits from the use of recurrent neural networks (RNNs) and their gated variants, including long short-term memory (LSTM) and bidirectional LSTM (BiLSTM) for temporal dependency modeling [18]. The combination of CNN and LSTM layers in hybrid architectures allows for simultaneous learning of spatial and temporal patterns, which enhances the detection of P300 and motor imagery signals. The neural decoding field now uses variational autoencoders (VAEs) and transformer-based models to perform tasks that require strong feature compression and extended attention capabilities [19]. The probabilistic nature of VAEs produces noise-free neural representations, which makes them suitable for denoising applications and cross-subject generalization. The transformer encoder uses self-attention to detect distant temporal relationships without needing recurrent connections. The deep architectures become more effective when biomedical priors such as spectral filters and electrode topology and physiological constraints are integrated to enhance both interpretability and brain physiology alignment. The combination of deep learning techniques with neural interface design has created a new generation of BCI systems that adapt to noise and maintain generalizability.
To further strengthen the link between computational neuroscience and biophysical realism, integrate physics-based modeling frameworks into the simulation and analysis of neuroelectronic and organoid systems to enhance the connection between computational neuroscience and biophysical realism. Physics-informed modeling enables researchers to model mechanical deformation and ionic diffusion and heat transfer and electromagnetic field propagation between biological tissue and microelectronic interfaces. The training process of deep learning models benefits from physics-based approaches, which incorporate governing physical laws such as Maxwell’s equations for field dynamics and the bio-heat equation for tissue thermoregulation as structural priors. The recent development of hybrid AI-FEM frameworks shows that deep learning systems can work with physics solvers to perform fast real-time tissue-device interaction simulations at high accuracy levels according to [20]. The paradigm allows for adaptive parameter estimation and fast inverse modeling and better generalization to new physiological states, which are essential for precise neural signal transmission and electrode impedance and bioelectronic temperature rise predictions. The combination of FEM-based stress and diffusion models with AI-driven growth and electrophysiological predictions through organoid studies would create a single framework to study neurodevelopmental processes under electronic stimulation at multiple scales. The integration of biomedical priors into deep learning models through spectral filters and electrode topology and physiological constraints leads to better interpretability and improved brain physiology alignment. The integration of deep learning methods with neural interface development has led to advanced BCI systems that learn from noise while preserving their ability to work across different situations.
The acquisition of electrophysiological signals through electronic front-end architectures continues to be the primary method because these systems deliver high temporal precision and low latency alongside compatibility with MEA and CMOS integration technologies. The systems use low-noise amplifiers (LNAs) and analog-to-digital converters (ADCs) with multiplexing circuits that optimize the recording of microvolt-level potentials at high signal-to-noise ratios (SNRs) across 0.1–10 kHz frequency bands [21]. The increasing number of recording channels and spatial resolution in neural systems creates problems for traditional EFEs because they generate crosstalk and produce thermal noise while consuming excessive power in dense implantable systems. The transduction mechanisms of photonic and SPAD-based (single-photon avalanche diode) systems operate through optical or photon-counting methods to detect neural activity by monitoring calcium or voltage-sensitive fluorescent signals [22]. The nanosecond precision of SPAD arrays makes them suitable for detecting transient neural events because they provide high-sensitivity time-resolved optical detection. The use of photonic interfaces helps reduce electrical artifact contamination and capacitive coupling, which represent major problems in electronic systems. The use of photonic systems requires external fluorescent markers or optogenetic alterations, yet they face restrictions from limited tissue penetration and phototoxicity and thermal damage caused by light stimulation. The direct electrophysiological signal quality of electronic EFEs remains superior to photonic and SPAD-based systems, which provide better non-contact imaging capabilities for large-scale neural dynamic monitoring. The design of high-density neuroelectronic recording systems with low-artifact performance becomes possible through the emerging field of optoelectronic hybrid front-end technology.
The current research has made significant progress, but multiple essential obstacles persist. AI models excel at prediction, but their lack of interpretability prevents scientists from gaining mechanistic understanding and medical professionals from trusting their results. The controlled laboratory success of BMIs for motor restoration often fails to translate to real-world applications due to operational challenges arising from neural fluctuations, patient exhaustion, and device reliability issues. The majority of BMI applications focus on motor function restoration, yet they do not address the cognitive and affective symptoms that define Alzheimer’s disease. The current models, including computational and biological systems, fail to represent the complete dynamic nature of human brain operations. The discovery of organoid biology, combined with AI-powered neural interfaces, has created a new research direction. Scientists use pluripotent stem cells to generate brain organoids, which provide researchers with their first opportunity to study human-specific neurodevelopmental and pathological processes in laboratory settings [23,24]. Organoids duplicate essential features of human brain structure, neural connections, and brain wave patterns that occur during disease states. The combination of advanced neuroelectronic interfaces with spatio-temporal graph neural networks and cyborg organoid platforms enables researchers to conduct closed-loop experiments and perform high-resolution phenotyping and drug screening at unprecedented levels of precision. These systems would allow scientists to study human brain function ethically and mechanistically outside the body, while simultaneously accelerating therapeutic development, according to Wu et al. (2024) [25].
The combination of AI and BCI technology advancements creates new possibilities to address these problems. The development of BCIs now enables people with neurological disabilities to convert their brain signals into functional outputs, allowing them to interact with their surroundings. The development of speech BCIs has reached a high level of accuracy, as researchers have successfully translated brain activity into text or speech synthesis, according to a 2023 study [26]. The company Neuralink has made significant progress in BCI technology through human device implantations, enabling patients to regain their ability to communicate and move. The startup Paradromics has conducted BCI implant tests that convert neural signals into speech and cursor movements to advance clinical usage [27].
Emerging organoid-based neural platforms are providing a novel testbed for advancing BCI technologies [25]. Brain organoids can simulate neurogenesis, neuronal migration, cortical stratification, and neural circuits of the human brain [25,28]. The integration of microelectrode arrays (MEAs) with organoids allows scientists to monitor and control neural activity in real time [25]. The cyborg organoid platform includes the hippocampal cyborg organoid (cyb-organoid) system, which connects human hippocampal organoids (hHOs) to a liquid metal–polymer conductor (MPC)-based mesh neuro-interface [25]. This mesh MPC (mMPC) integrates 128-channel high-density multielectrode arrays that are stretchable (up to 500%) and flexible, facilitating attachment to the hHOs and enabling the detection of neural activity [25]. Furthermore, organoid–brain–computer interfaces (OBCIs) have been developed, representing innovative BCIs mediated by implantable organoids that are capable of bidirectional communication with host tissue [28]. OBCIs aim to repair damaged neural circuits by promoting the regeneration and regulation of neural functions, with the potential to contribute to future bidirectional BCI frameworks implementing feedback and closed-loop regulation [28]. Traditionally, BCIs are characterized by the type of neural activity they record and decode. The established criteria for BCIs include EEG indicators, which include alpha rhythm (8–12 Hz) for relaxed wakefulness and visual evoked potentials (VEPs) and steady-state VEPs (SSVEPs) for periodic visual stimuli and event-related potentials (ERPs), with P300 being the most popular for spelling and selection tasks [4,29]. The use of fMRI, MEG and NIRS enables researchers to develop hybrid BCIs that merge high temporal accuracy with enhanced spatial detail. The fundamental elements of signal selection in BCI research depend on electrophysiological and hemodynamic criteria. The clinical applications of BCIs reach further than AD and PD, although our discussion focused on these two conditions. The practical applications of BCI systems have focused primarily on two neurological conditions, which are amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS). The combination of P300-based spellers and steady-state visually evoked potential (SSVEP) paradigms enables patients with severe paralysis from ALS to regain communication abilities through brain–computer interface systems [30,31,32]. The application of BCI systems in MS extends beyond communication to include motor rehabilitation and cognitive neurofeedback, which utilize P300 and ERP-based protocols to boost remaining neural plasticity, according to research by Kübler et al. (2007) and Guger et al. (2009) [33,34].
The integration of electronic interfaces with biological tissue for safe operation stands as a fundamental challenge in current BCI research. The interface between biological tissue and electronic circuitry through neural interfaces faces two main challenges because electromagnetic energy absorption and heat dissipation affect both device operation and tissue health. The human body requires continuous monitoring of specific absorption rate (SAR) and temperature changes when using devices for extended periods, especially when used indoors or as wearable technology, according to research by [35,36,37]. The integration of flexible sensors with wireless telemetry in smart electronic systems allows real-time monitoring of SAR and thermal changes, which supports the safe operation of high-density neural recording or stimulation platforms. The integration of biomedical safety frameworks into neural interface research enables the development of next-generation BCIs that combine functional reliability with physiological tolerance.
These advanced systems support the study and modeling of network dynamics relevant to neurodegenerative diseases, such as schizophrenia and Alzheimer’s disease, as hHOs serve as promising models for these conditions [25]. They offer an ethically viable platform for prototyping adaptive neural interfaces [25]. “The technological progress seeks to bring back lost abilities through limb reanimation, memory enhancement, and sensory restoration while creating opportunities to boost human brain capabilities and develop machine–human collaboration systems [38]. The technology provides fresh possibilities for treating patients who have Alzheimer’s disease and Parkinson’s disease [38].
The literature shows substantial progress in BCIs and Artificial Intelligence (AI), yet researchers have not thoroughly investigated how humans and machines adapt to each other during interactions [38]. The current BCI systems focus on one-way adaptation because users learn to operate the system by decoding their neural signals to control external devices [38,39]. The scientific community has not thoroughly studied mutual adaptation between humans and AI systems, which involves their joint development for performance improvement [38,40]. Human–machine interface operation functions as a “two-learners problem,” yet researchers have conducted minimal studies about the specific processes through which users develop effective control methods for interface interaction [39,40]. Brain co-processors, for example, are designed for joint optimization of cost functions with the nervous system, signifying a move towards mutual adaptation [38].
Building on these gaps, a recent analysis by Yang (2024) identifies additional technical, ethical, and societal limitations in applying BCIs to AD. The first difficulty arises from signal quality, as external interference, natural bodily changes, and emotional responses easily influence cognitive state-related brain signals. The process of obtaining reliable neural information for cognitive rehabilitation programs proves to be extremely difficult. The use of extensive or invasive BCI devices creates patient discomfort, which negatively impacts their ability to follow treatment and maintain system usage in the long term [41]. The process of obtaining brain data raises ethical concerns due to its involvement in handling sensitive and private information. The mental decline of AD patients prevents them from giving consent and safeguarding their rights because they cannot make complete decisions. The high cost of BCI systems creates social problems because it limits their accessibility to specific groups of people, thereby intensifying existing healthcare inequalities. The combination of BCIs with AI, big data, and portable hardware technology will lead to improved Alzheimer’s disease (AD) diagnosis, more precise interventions, and enhanced preventive monitoring capabilities. The research community has not studied the co-adaptive learning process between patients and systems, which requires extensive mutual learning that spans months to years. Researchers must address critical knowledge gaps in developing BCIs for neurodegenerative disease stages to create systems that integrate technical excellence, ethical standards, human focus, and clinical effectiveness.
The primary objective of this selective thematic review is to explore and synthesize existing literature on neural interfaces, organoid models, and AI, with a focus on key themes such as AI and ML, neural interfaces and brain organoids, and human–AI symbiosis. This review offers insights into research gaps, the evolving understanding, and the practical relevance. It organizes existing evidence around the development of co-adaptive brain–computer interfaces for individuals living with neurodegenerative diseases, specifically AD and PD. These conditions are marked by progressive deterioration of motor, cognitive, and/or communicative abilities, necessitating assistive technologies that are responsive and dynamically adaptable to the user’s evolving neurological state. Traditional BCIs have focused on signal decoding and external control, without sufficient attention to co-adaptation, the mutual learning and adjustment between human users and artificial systems. This paper addresses that gap by proposing a scoping review of technologies and frameworks that enable human–AI symbiosis in assistive and therapeutic applications for AD/PD populations. The specific objectives of this review article are as follows:
  • Map Key Concept of Co-adaptive Symbiotic Interactions
Co-adaptive neural interface systems represent a progression beyond traditional BCIs by facilitating mutual learning between human users and AI-controlled systems. The systems operate through a two-way process (bi-directional) that enables both user neural signals and interface decoding algorithms to adapt simultaneously. The systems require this capability because neurodegenerative diseases like AD and PD cause users to lose their ability to express intentions while their motor and cognitive functions deteriorate. We trace the development of neural interface technologies and their integration with brain organoids, high-density MEAs, and deep learning models. Key architectural components include the following:
  • Signal acquisition modules (e.g., EEG, ECoG, intracortical probes).
  • Real-time pattern recognition occurs through recurrent neural networks (e.g., BiLSTM, GRU) for pattern recognition.
  • The system utilizes adaptive feedback loops that implement reinforcement learning or unsupervised adaptation methods.
  • Embodied interaction models, as seen in passive BCIs decoding multi-dimensional mental states (MDMS), e.g., stress, engagement.
Human–AI symbiosis, as described here, operates through co-regulation and personalization, as AI receives neural signal data from users, while users modify their intent strategies based on system feedback. The system maintains its ability to adapt through continuous operation, which proves essential for treating patients with long-term neurodegenerative diseases.
2.
Identifying Gaps in the Literature on Co-Adaptive BCI for AD/PD
The application of BCI technology to neurodegenerative patient populations faces multiple challenges for the following reasons:
  • Limited personalization over disease progression: The current BCI systems maintain fixed calibration methods, which fail to support the time-dependent cognitive and motor deterioration patterns found in AD and PD patients.
  • The majority of BCI systems operate as one-way control systems, but researchers have proven that feedback-driven models work effectively in non-clinical environments through studies like DishBrain and Dehais et al.’s flight simulation BCI.
  • Sparse integration of organoids in real-time systems: Brain organoids remain largely exploratory and preclinical, and their potential as adaptive computational substrates in BCI feedback loops has not been fully realized.
  • Lack of neurophenomenological decoding: Few systems address the first-person experience, which is critical for assessing intention, emotional valence, and motivational state factors, especially relevant in disorders with affective-cognitive comorbidity.
  • Insufficient long-term tracking infrastructure: There is a lack of frameworks to track patient-specific neural trajectories and tailor interface adaptation accordingly over months or years.
3.
Synthesizing Evidence Across Neuroscience, AI, and Clinical Studies
Drawing from the reviewed literature, we synthesize a unified framework for next-generation BCIs in neurodegenerative contexts. Bidirectional BCIs that integrate deep learning, neural feedback, and biologically relevant models (e.g., organoids or personalized MEG/EEG profiles) offer the following advantages:
  • Enhanced communication: Adaptive BCIs can restore interaction capacity in late-stage AD or advanced PD patients by mapping residual neural intent to assistive outputs.
  • Improved mobility and motor planning: The system achieves better motor control and planning through real-time co-adaptation with recurrent models (LSTM, BiLSTM), which produces stable intent decoding during neural system changes.
  • Personalized treatment response tracking: AI systems that analyze MEG/fMRI or electrophysiological changes in organoid models of PD midbrain can predict and track the effectiveness of pharmacologic or DBS treatments.
  • The integration of neurophenomenological data through passive BCIs enables interfaces to detect user state changes beyond basic command input for creating cognitive-affective interfaces.
  • Closed-loop hybrid systems: Emerging platforms, such as organoid–AI symbiosis, create the possibility for feedback-conditioned drug screening, personalized to a patient’s neural phenotype and integrated into BCI control pipelines.
4.
Thematic Focus and Methodology
This thematic review aims to explore and synthesize emerging literature on co-adaptive neural interfaces and brain organoid integration for neurodegenerative disorders, with a particular focus on the following:
  • Adaptive AI architectures in brain–computer interfaces.
  • Organoid-based neuroengineering platforms for modeling and intervention.
  • Bidirectional and personalized feedback loops for AD and PD patients.
We seek to provide a conceptual and translational framework for developing next-generation BCIs that evolve with the user, offering sustainable, ethically sound strategies for managing cognitive and motor decline. This thematic review contributes a conceptual and translational framework for developing co-adaptive neural interface technologies tailored to neurodegenerative disorders. By mapping foundational concepts of symbiotic human–AI interaction, identifying implementation gaps in BCI design for AD/PD, and synthesizing interdisciplinary evidence from neural engineering and clinical neuroscience, we propose a path forward for sustainable, ethically informed, and biologically integrated BCI systems. In particular, the convergence of deep learning, brain organoids, and co-adaptive feedback architectures presents new possibilities for restoring communication, enhancing mobility, and improving cognitive function in individuals affected by these conditions. Ultimately, human–AI symbiosis in this context represents not only a technological innovation but also a paradigm shift toward empathetic and personalized neurotechnological care.

2. AI and Machine Learning in Neuroscience and Neuroengineering

Neuroscience is dedicated to the study of the nervous system. It explores multiple aspects of the brain and nervous system, such as their structure, function, development, and the impact of diseases on them [42]. Neuroscience is a scientific field that studies the nervous system. It explores multiple aspects of the brain and nervous system, including their physical organization and operational mechanisms, as well as their developmental processes and disease-related effects [42]. Neuroengineering is a multidisciplinary discipline that connects neuroscience with engineering, computer science, and biomedical sciences. The primary objective of neuroengineering involves studying neural systems through the development of devices and computational tools to diagnose, restore, or enhance neural functions [43]. Neuroengineering emerged as an independent field during the late 20th century, driven by the expansion of electrophysiology and biomedical engineering research. The cochlear implant invention, along with the development of deep brain stimulation (DBS), has established significant breakthroughs in neuroengineering [44].
AI and ML systems are transforming neuroscience and neuroengineering by enabling the analysis of extensive and complex neural data at scale, thereby creating adaptive brain–machine interfaces. These technologies help scientists understand brain operations while creating neural models and enabling medical treatments. AI systems analyze extensive datasets that combine electrophysiological recordings with imaging results and behavioral measurements. The combination of DL methods, including CNNs, RNNs, and VAEs, enables better spike train analysis and calcium imaging. At the same time, VAEs serve as effective encoding models to reveal neural representation patterns [45]. DL models outperform conventional classifiers when analyzing multimodal imaging data that includes MRI and PET to detect neurodegenerative and psychiatric conditions in neurodiagnostics. Deep learning architectures demonstrate exceptional performance in diagnosing Alzheimer’s disease, Parkinson’s disease, and schizophrenia, thanks to their high diagnostic precision. The application of transfer learning improves model performance when working with restricted datasets that contain limited labeled information [46]. AI models serve as the foundation for neuroengineering to create brain–computer interfaces, which aim to enhance or restore the operations of the neural system. The application of DL methods to EEG, ECoG and intracortical recordings enables users to control prosthetic devices through intention detection and restore communication abilities for paralyzed individuals. Real-time decoding operations benefit from recurrent models, which include LSTM networks, because they provide stable performance [47].
The integration of AI and ML technology has significantly advanced neuroengineering by enabling sophisticated neural data analysis, developing brain–computer interfaces (BCIs), and creating adaptive neuroprosthetics. AI has revolutionized neuroengineering by allowing the discovery of new brain mechanisms and enhancing our understanding of brain function and disorders. AI analysis of brain dysfunction has generated substantial knowledge about neurological disorders, which in turn improves diagnostic methods and treatment approaches and enhances traditional clinical procedures. ML and AI methods are applied in neuroengineering to study brain signals and create brain–computer interfaces and improve neurological disorder treatments. AI and ML enable scientists to read brain signals and predict neurological events, creating individualized treatments that open new possibilities for brain research and human performance enhancement [48]. The field of NeuroAI represents a new trend that unites neuroscience with AI through biological neural process-inspired AI systems. Establishing effective interdisciplinary collaboration in this field requires structured integration of expertise across computational, biological, and clinical domains. A practical method to establish effective interdisciplinary collaboration involves building translational research environments that unite data scientists and engineers with neuroscientists through experimental workflows that connect algorithm development to neural recording and stimulation studies. The exchange of models and experimental results becomes possible through joint data standards (Neurodata Without Borders), co-supervised projects and open neural simulation platforms. The development of professionals who link these fields becomes possible through dual training programs that combine computational neuroscience with bioengineering education. The development of structured connections between fields enables the creation of a systematic approach that speeds up AI-driven neuroengineering research [49]. The concept of the “embodied Turing test” has been proposed. The evaluation of AI models depends on their ability to mimic animal sensorimotor behaviors, which drives AI development toward more adaptable and transferable systems [50,51]. The degree to which AI research adopts neuroscience principles remains a subject of ongoing debate among professionals. The extent to which AI research incorporates neuroscience principles remains a topic of professional debate. One of the ongoing topics of discussion among experts is the extent to which AI research is inspired by neurobiology. Researchers have shifted their focus from biological models, which could have provided critical insights into neural systems, to scaling transformer architecture. As highlighted in Table 1, the incorporation of AI and ML methodologies has greatly enhanced research efforts in Alzheimer’s and Parkinson’s diseases.
The processing of information through brain-inspired computing technologies offers a fundamentally different paradigm for information processing, characterized by high energy efficiency and a superior ability to handle large volumes of unstructured and noisy data from daily life. The achievement of this potential requires that researchers unite their efforts, receiving financial backing and structural resources to advance brain-inspired computing research [73]. Artificial intelligence serves as a transformative power in organoid research, enabling rapid evaluation of construction strategies, efficient analysis of multi-omics data, and precise assessment of preclinical models. The AI-powered organoids would allow scientists to study organ development and disease mechanisms more effectively, creating opportunities for future medical applications [74]. The development of DeepDendrite represents a new GPU-accelerated computing framework that merges dendritic hierarchical scheduling with the NEURON simulator’s GPU engine to support detailed neural simulations. The frameworks enable faster neuroscience research and demonstrate potential methods to merge realistic biological models into standard AI operations for image classification training [75].

3. Neural Interfaces and Brain Organoids in Neuroengineering

Neuroengineering research now emphasizes the combination of neural interfaces with human brain organoids as essential tools for studying and treating brain disorders. The direct link between neural circuits and external devices becomes possible through neural interfaces, which include brain–computer interfaces (BCIs) and brain–machine interfaces (BMIs). The technology has proven essential for various applications, including motor rehabilitation and speech decoding. Brain organoids function as three-dimensional, self-organizing human pluripotent stem cell-derived structures that create scalable laboratory models for studying human brain development, connectivity, and disease processes [23,74,76,77,78]. The combination of these platforms with AI technology, as well as top-tier neuroscience journals Nature Neuroscience, Neuron, and Nature Methods, enables researchers to develop new experimental methods for studying neural patterns, disease modeling, and therapeutic development for Alzheimer’s and Parkinson’s diseases [79,80,81]. The National Institutes of Health (NIH) identifies organoid systems combined with AI-driven neural interfaces as a groundbreaking advancement in brain science, which supports the BRAIN Initiative’s goals for developing scalable, multimodal model systems [50,51,82].
Brain organoids are three-dimensional neural tissues that are derived from human pluripotent stem cells that develop into layered structures containing progenitors and neurons and glia cells, which replicate the initial stages of human cortical development [83,84,85]. In contrast to traditional two-dimensional cultures, they reproduce not only cellular diversity but also tissue-level processes such as neurogenesis, neuronal migration, cortical stratification, and early circuit formation [76,86]. The organoids demonstrate their ability to join existing neural networks when scientists place them into sensory, motor, or visual cortex areas of host animals because they survive and develop while sending their axons throughout the surrounding neural tissue [87,88,89,90]. The transplantation of immature organoids containing numerous ventricular-zone progenitors leads to uncontrolled cell division; however, their random neural projections prevent the formation of precise host–graft neural connections. In vivo experiments that combine electrical and optogenetic neural stimulation lead to faster organoid development, directed neuronal cell growth and migration, and target-oriented axonal extension, resulting in improved structural and functional integration as well as reduced progenitor cell proliferation [76,91].

3.1. In Vivo Neural Interfaces

Neural interfaces are bioelectronic systems that establish direct communication links between nervous system functions and digital technology platforms [92]. The technology behind neural interfaces has evolved from its original use in the human brain, spinal cord, and peripheral nerves to create new hybrid biological-artificial intelligence systems, including brain organoids. The development of BCIs and BMIs has led to groundbreaking applications, which include ECoG signal-based speech decoding and prosthetic and virtual device control [26,93,94]. Neuralink represents a notable example of an implantable BCI system that connects directly to cortical neurons to enable motor and communication restoration [95]. The technologies convert brain signals into functional commands, enhancing the independence, communication abilities, and mobility of people with severe motor disabilities [96,97,98]. The current non-invasive systems face ongoing challenges because they operate at limited bandwidth and produce low signal-to-noise ratios [99,100].
The human brain, comprising 86 billion neurons and trillions of synaptic connections, has been the primary focus of neuroscience and BCI research since its discovery [101]. The study of the human brain faces multiple obstacles, such as inaccessibility, ethical restrictions, and the challenge of monitoring cellular activities in vivo [102]. The development of brain organoids became necessary because researchers needed to study human neural functions in laboratory-controlled systems.
The long-term sustainability of neural interface technologies faces ongoing obstacles that prevent their complete resolution. The primary challenge of biocompatibility arises from chronic immune responses and foreign-body reactions, which lead to tissue damage and deterioration of signal quality near the implant site [103]. The combination of rigid electrode materials with neural tissue structures exacerbates inflammation and gliosis during extended implantation periods due to their mechanical incompatibility [104,105]. High-density electrode arrays provide better spatial resolution, but their recordings become less accurate because of electrical crosstalk that occurs when fields overlap [106]. The devices experience degradation through electrochemical corrosion, mechanical strain, and material fatigue, which reduces their operational stability and medical trustworthiness over time [107].

3.2. In Vitro Brain Organoid Platforms

Brain organoids are three-dimensional neural tissues that contain diverse cell types, including neurons and glia, closely mimicking those in the human brain [74,78,102]. The three-dimensional structure of organoids surpasses traditional 2D cultures and animal models by creating brain structures that accurately represent human brain development and disease progression [23,108]. The use of patient-derived models enables researchers to study disease mechanisms at various developmental stages, thereby advancing precision medical approaches. Scientists use high-resolution microelectrode array (MEA) systems from MaxWell Biosystems to record spontaneous neural activity and functional brain connections and oscillations in real-time [76,79].
One of the most compelling demonstrations of organoid-based neuroadaptive behavior is the DishBrain project, in which researchers trained living neurons cultured in vitro to play a simplified version of the game Pong. The 2022 research by Kagan et al. demonstrated a closed-loop system that used feedback learning to show that neuronal networks could develop goal-oriented behavior through environmental stimulus responses [109]. The operational sequence followed a well-defined loop consisting of signal acquisition → feature extraction → state prediction → feedback evaluation → electrical stimulation → network adaptation, aligning with predictive-coding principles proposed for adaptive computation in biological networks. The study illustrates how organoids can function as intelligent agents in AI-integrated platforms, as it demonstrates successful real-time communication between biological and digital systems.
In the context of BMI, organoids enable researchers to build closed-loop hybrid systems for BMI applications through AI-driven neural interfaces that interpret organoid activity in real time [80]. The new research approach enables scientists to study neural computation that emerges from brain activity, rather than trying to decode human intentions. Deep learning models show their ability to interpret intricate patterns in electrophysiological signals. The research by Trujillo et al. (2019) showed that organoid electrical signals progress toward mature oscillatory patterns, which unsupervised learning models can effectively predict [77]. The research field uses convolutional neural networks and spatiotemporal graph neural networks to extract neural network connections and dynamic patterns from biological data [110].
By integrating organoids and neural interfaces, researchers can create ethically acceptable neural systems, resulting in reduced invasiveness. The systems offer dense, high-resolution recording capabilities that human brains lack due to ethical and technological limitations. The systems prove best for studying early disease progression, drug testing, and neurotechnology development. The complete utilization of brain organoids in these platforms depends on achieving long-term viability and functional maturation, which requires vascularization strategies to support their growth. Figure 1 provides a schematic overview of in-vitro vascularized organoid development and integration with microelectrode array (MEA) systems. The illustration summarizes how human induced pluripotent stem cells (iPSCs), derived from skin fibroblasts or blood cells, differentiate into neural progenitor cells and subsequently form neural organoids under the influence of growth factors such as VEGF and FGF. Within these systems, Notch signaling governs endothelial sprouting and vascular network formation, leading to the development of perfusable cortical organoids capable of electrophysiological learning and closed-loop AI interaction (adapted from this study’s conceptual model). The addition of in vitro vascularization systems provides better oxygen and nutrient supply to cells while preventing necrotic core development and enabling the modeling of neurovascular interactions. The combination of EC co-culture with co-differentiation and assembloids, as well as organ-on-chip systems and engineered ECM, has proven effective for cortical organoid development. The integration of perfusable vascular networks into MEA-compatible organoid cultures produces better electrophysiological stability and more accurate functional measurements [111,112,113,114,115]. These advancements collectively position vascularized cortical organoids as revolutionary instruments for neuroengineering research. The combination of deep learning systems with these biological systems enables researchers to conduct closed-loop experiments, train models, and achieve co-adaptive learning between biological systems and artificial intelligence. The developed technologies pave the way for advanced neural interfaces, which will establish a new brain–machine partnership beyond current BMI systems.
Human iPSCs aggregate to form early organoids that are capable of differentiating into various cell types in low-adhesion plates. In the presence of ROCK, SMAD inhibitors, and other factors, differentiated neural progenitor cells located near the core give rise to neurons/glial cells. SSC (signal-sending cells), which possess healthy tip cells that express a ligand, activate Notch signaling in neighboring cells. This step reinforces their identity as a tip cell, as they sense gradients of VEGF produced by the organoid’s neural progenitors or hypoxic core.
SRC (signal-receiving cells) receives and responds to the notch signal (forms the endothelial stalk cells). This Notch activation suppresses their VEGF receptor expression, halting their differentiation while allowing the tip cells to sprout. The process of Notch signaling facilitates the formation of capillary-like networks of infiltrating endothelial cells (ECs) that organize into vessels within and around the organoid, enabling active nutrient/oxygen transportation. The ECs can be added to the organoids or embedded in the ECM, depending on the culture vessel in use (Refs. [116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137]).
Figure 1. In vitro differentiation, vascularization, and machine interface of neural organoid [117,118,124,136].
Figure 1. In vitro differentiation, vascularization, and machine interface of neural organoid [117,118,124,136].
Applsci 15 11316 g001

3.3. Integrated Summary of Neural Interface Systems and Organoid Models in AD and PD

Interactions that had been found to play essential roles in BMI control include cortico-striatal plasticity for motor control and cortico-striatal and cortico-cortical for abstract skill learning, the cortico-striatal thalamo-cortical feedback loop for motor control, and the cortico-basal ganglia–thalamo-cortical loop for understanding cognition, behavior, and the nature of neurological disorders [138,139,140,141].
Several complex neurological disorders arise due to alterations in specific or combined malfunctional circuitry of subnetworks of interactions [142,143]; for instance Parkinson’s disease in humans is typically due to decreased activity of the globus pallidus external (GPe) part of the neurons [144,145]; seizures and epilepsy manifest in intracellular Cl-dysregulation [146]. AD, characterized by amyloid (A β ) plaques and Tau misfolded and hyperphosphorylated protein tangles, has been suggested by Grieco et al. (2023) [147] that neural activity dysregulation may lead to abnormal neural circuit function. In Parkinson’s disease, research has shown that the potential pathology of alpha-synuclein and neural activity is mediated through changes in the dynamics of synaptic vesicles [148]. This relationship is bidirectional [149].
The benefits of brain organoids cannot be overemphasized, especially in public health and neurological diseases that are silently becoming a pandemic. In addition to the estimated impact of Alzheimer’s disease on elderly people, one in every six children born in the United States of America alone has a form of neurological disorder [150]; hence, the need for crucial tools like brain organoids that would enable proper understanding of neurological disorders, together with models that facilitate learning and remembering, for appropriate therapeutics to be developed. The changes in neurotransmitter release and synaptic structures have been the beneficial mechanisms of brain stimulation for treating some neurological symptoms and improving patients’ cognition [151,152,153,154,155]. However, measuring neural activity in the induced intracellular changes may help diagnose, monitor progression, and assess treatment strategies for neurological diseases/disorders.
Table 2, Table 3 and Table 4 provide a consolidated overview of the principal technologies currently used in AD and PD research.
The classification system in Table 2 categorizes neural interface systems according to their invasiveness level, ranging from non-invasive EEG and fNIRS to invasive deep brain stimulation (DBS), while highlighting their benefits, drawbacks, and ethical implications. The classification system in Table 3 categorizes similar technologies into signal-based categories, including electrical, optical, magnetic, and ultrasonic signals, to facilitate readers’ evaluation through modality-specific perspectives. The dual-view framework facilitates a deeper understanding of the neural interface selection and evaluation processes in both clinical and experimental environments.
The research on AD/PD utilizes state-of-the-art organoid models, which include cerebral and midbrain organoids, as well as vascularized and fused patient-derived organoids, as shown in Table 4. The models replicate human neurodevelopmental processes and pathological features, enabling scientists to utilize them in the development of AI-based bioelectronic systems for drug testing, disease simulation, and personalized medical treatments. The tables illustrate how neural engineering, organoid biotechnology, and translational neuroscience are converging to create a unified framework for innovative research in neurodegenerative diseases.

3.4. BCI Signal Flow and System Architecture

The typical structure of BCI systems comprises four main stages, beginning with signal acquisition, followed by signal processing that enables feature extraction and pattern recognition, and then control execution, and finally, feedback. The workflow of this process appears in Figure 2. The acquisition of signals occurs through various types of electrodes, including both invasive and non-invasive devices that measure neural activity using EEG, ECoG, and intracortical spikes. This raw data undergoes preprocessing before feature extraction algorithms detect specific neural patterns, which include motor imagery, attention, and speech intention. Real-time control signal generation is achieved through pattern recognition modules, which utilize machine learning and deep learning models to classify extracted features. The system transmits these signals to robotic limbs, communication interfaces, and assistive platforms. The user receives feedback about their intended outcome through a feedback system, which completes the closed-loop interaction. The system design serves as a fundamental requirement for real-time neurorehabilitation and assistive communication systems, which benefit patients who have neurodegenerative diseases, including AD and PD.

4. Human–AI Symbiosis: Co-Adaptation and Emerging Paradigms

Human–AI symbiosis is an emerging paradigm in which humans and artificial intelligence systems co-adapt in real-time, creating a dynamic partnership that strengthens their combined abilities. The framework differs from traditional human-computer interaction because it focuses on adaptive processes that integrate cognitive processing with emotional, motivational, and physiological systems.
The co-adaptive BCIs enable users and systems to evolve together through their shared learning experiences. The systems surpass traditional decoding models (unidirectional) to create feedback mechanisms that allow AI to learn from brain signals while assisting users in refining their mental approach. The dual-agent co-adaptation framework developed by De Santis et al. (2023) [40] enables users and interfaces to adapt simultaneously through unsupervised and reinforcement-based mechanisms that operate independently of predetermined task objectives. The interface adjusts its operations through an unsupervised learning method, which optimizes the process of converting user actions into interface responses. Users experience use-dependent adaptation through reinforcement learning, which receives feedback from intrinsic processes during extended periods of use, rather than task-related errors. The system optimizes interaction efficiency through co-adaptation, enabling flexible and robust human–machine collaboration. Neuroadaptive modeling represents a fundamental requirement for this development. The term "neuroadaptive technology" emerged from Zander Labs to describe passive BCIs, which extract MDMS data to train adaptive AI models, as defined by [213]. The system records stress levels and engagement and motivation states from users without needing any direct commands. The system enables machines to understand context and intent through implicit interpretation, which results in more natural user interfaces.
The current traditional BCI methods often fail to detect the personal aspects of perception and intention that people experience directly. The development of future systems requires them to surpass mechanical decoding methods to understand the emotional and subjective aspects of users’ mental processes. Neurophenomenological decoding establishes connections between EEG patterns and conscious experiences such as frustration, insight, and the desire to create systems that understand the motivations behind user actions. The system establishes a complete connection between neural signals and psychological meaning through this loop, enabling actions based on user intent and providing feedback that aligns with their emotional state [214]. The way users perceive AI feedback depends on whether it aligns with their self-image, as this creates a “quasi-self” experience; however, mismatched feedback results in a “quasi-other” perception. The discovery highlights the importance of explainable AI systems, effective interface design, and trust calibration mechanisms in the development of neuroadaptive technology. Zander Labs achieved scalability in personalization through their universal classifiers, which process thousands of datasets, raising concerns about privacy, identity, and autonomy. The system’s ability to detect and forecast emotional states creates fresh moral dilemmas about who should determine the interpretation process. Machines possess the capability to make false representations of human emotions. The process of developing adaptive feedback systems requires methods to protect user autonomy from degradation. The achievement of human–AI symbiosis requires more than technical solutions, as it presents a philosophical dilemma that necessitates designers working together to develop transparent and ethical systems.
An emerging area within human–AI symbiosis is the development of co-adaptive systems, which unite AI technology with brain organoids. AI technology enhances organoid-based disease models and drug screening platforms through a feedback loop, enabling biological substrates to modify computational models. The Brain and Organoid Manifold Alignment (BOMA) system utilizes AI to match gene expression patterns between developing human brains and organoids, thereby enabling advanced research in developmental biology. The combination of biobank data with machine learning algorithms enables the detection of patterns in multi-omics and imaging databases, thereby minimizing human errors and accelerating clinical research operations. The AI–organoid system uses adaptive methods to forecast how drugs will affect patients and their potential toxic side effects. The combination of machine learning algorithms with 3D pharmacogenomic data enables predictions of individual treatment responses, while AI-guided image segmentation tools evaluate vascular differences in tumor organoids. The system allows continuous feedback operations, where organoids adapt to experimental changes and AI models generate updated predictions, leading to new therapeutic approaches. AI systems that study midbrain organoids for the progression of Parkinson’s and Alzheimer’s diseases enable the real-time adaptation of treatment strategies. These platforms demonstrate mutual shaping because organoids transform experimental and AI-directed procedures, while AI models refine their models through biological feedback. The combined approach of preclinical workflow acceleration and personalized therapeutic optimization emerges from this co-adaptive system. The triadic model of human–AI–organoid symbiosis presents itself as a strong framework for future precision neuroscience applications. The Zander Labs team proved that BCI systems that monitor medial prefrontal cortex activity passively enable users to control cursors implicitly, which serves as a basis for AI systems that adapt to human mental states in real-time [213,215]. Dehais et al. created a dual passive–reactive BCI system for flight simulator operations, which combined stress detection with reactive control to execute collision prevention and basic neuroadaptive copilot functionality [216]. The AI systems in these examples utilize pilot emotional and attentional data to make proactive adjustments, thereby decreasing workload while enhancing safety in complex operational settings. The current neuroadaptive frameworks show promise for future development in creative and educational applications because they can detect human emotional and cognitive states to provide real-time content recommendations, interaction tools, and pacing adjustments. Human–AI collaboration has progressed from assistive prosthetic development into domain-independent copilot and co-creative systems, which use neuroadaptive intelligence to support human cognition.

5. Challenges and Future Directions

As the fields of neural interfaces and organoid technologies converge with artificial intelligence (AI) and machine learning (ML) to create innovative solutions for studying and managing neurodegenerative diseases, including Alzheimer’s disease (AD) and Parkinson’s disease (PD). The complete realization of these innovations depends on solving current technical obstacles while handling ethical issues that researchers will encounter in their next phase of work.

5.1. Challenges and Limitations

Organoid-based neuroengineering development faces multiple significant challenges that impact ethical considerations, experimental procedures, and technological capabilities. Ethically, the growing biological complexity of organoids, especially when combined with artificial intelligence, raises serious concerns around informed consent, sourcing of human biological materials, data privacy, and the ambiguous moral status of advanced organoids that may display signs of sentience or consciousness [217,218,219,220]. Technologically, organoids have proven invaluable for modeling human development and disease, yet several limitations constrain their utility. Reproducibility remains a significant concern due to batch variability, manual handling, and dependence on poorly defined matrices, such as Matrigel [221]. Organoids currently exist as fetal tissue models, which do not contain the necessary components for adult physiological modeling because they lack maturation, vascularization, innervation, and stromal elements [24,222]. The current brain organoid systems demonstrate excellent capabilities for mimicking human developmental stages and disease symptoms, yet they are not ready for medical applications. The main applications of these systems exist in drug screening and modeling because their lack of reproducibility and maturation problems makes them unsuitable for therapeutic tissue use. The lack of standardized differentiation methods together with poor vascularization and unstable electrophysiological activity makes it impossible to use these constructs for transplantation or regenerative medicine. The development of standardized ethical and regulatory frameworks for human-derived neural constructs remains ongoing, which creates challenges for their future clinical implementation. The transition from experimental organoids to clinically applicable neurotherapeutic tools needs advancements in long-term vascularized culture methods and automated production systems and standardized quality-control systems that follow Good Manufacturing Practice (GMP) guidelines [24,217,222]. Research on systemic diseases and immune system interactions is limited due to these limitations. The combination of genetic instability and epigenetic changes in extended cell cultures, along with the absence of standard operating procedures for organoid derivation, maintenance, and validation, creates obstacles for large-scale and reliable research [223,224,225,226]. The development of fundamental consciousness in brain organoids creates significant ethical problems during research activities [227,228]. Organoid production requires extensive manual work, which prevents researchers from performing large-scale experiments. Their poor vascularization and immune system incompatibility also make them unsuitable for medical applications [229]. The complete utilization of organoid systems requires scientists to develop new methods and technologies that solve multiple existing problems.
The National Institutes of Health (NIH) created the Standardized Organoid Modeling (SOM) Center as an institutional strategy to handle ethical and reproducibility issues during their 25 September 2025 announcement. The SOM Center represents the first national program that creates standardized organoid models that combine reproducibility with ethical oversight and artificial intelligence and robotic enhancement. The center receives USD 87 million during its first three years to establish standardized organoid models that decrease animal testing needs and ensure transparent research practices and ethical compliance. The initiative uses AI optimization and automated production systems and open-access data and protocol repositories to speed up the development of reproducible organoid science and gain regulatory approval. The NIH SOM Center demonstrates how scientific organizations can handle organoid and AI convergence ethical problems through structured oversight and open science and global collaboration, according to [230].
In vivo experiments remain indispensable for validating in vitro findings, yet they introduce significant obstacles. High-quality, artifact-free neural recordings over extended durations and across heterogeneous subject groups are rare; even state-of-the-art transparent neural probes in Nature and Microsystems & Nanoengineering face persistent challenges in minimizing electrical and optical artifacts during live neural recording [231,232,233]. Without detailed solutions, translation into clinical or ecological validity will remain constrained. Lastly, the rapid proliferation of sophisticated recording and stimulation technologies has outpaced existing computational infrastructure. The data generated from dense, high-throughput MEAs requires scalable storage, real-time preprocessing, and efficient analytics pipelines to support continuous multimodal in vivo data streams. Without investing in cloud-enabled architectures, federated analytics, and low-latency machine learning frameworks, the transformative promise of multimodal neurodata systems will remain unfulfilled [234,235]. As these technologies advance, several significant challenges must be addressed. One of the most pressing is the suite of ethical concerns that arise with increased biological complexity and the integration of AI. These include issues of informed consent, data privacy, the speculative potential for consciousness in advanced organoid systems, and the risks of misuse in AI-powered neurotechnologies. These must be addressed proactively through robust regulatory frameworks, interdisciplinary oversight, and transparent public dialog [236,237]. In vivo experimentation remains critical for validating findings from in vitro organoid platforms and advancing translational research. However, it introduces difficulties in acquiring high-quality, artifact-free neural data, especially over extended periods and across diverse patient populations. These limitations can hinder the generalizability and long-term reliability of findings. Simultaneously, technological evolution in recording and stimulation tools has created a data deluge that current computational infrastructures struggle to manage. The need for real-time analytics, efficient storage systems, and scalable platforms is skyrocketing. Without these, the promise of integrating massive multimodal datasets into actionable insights remains unrealized.

5.2. Future Directions

The combination of neural interfaces with brain organoids and AI-driven systems in future research creates new possibilities, although it requires addressing major technical, biological, and ethical obstacles. The research directions aim to advance scientific knowledge by developing neurotechnologies that are ethically sound, reproducible, and human-centered. The primary path for expansion lies in combining multiple data types. Neuroengineering research necessitates the development of unified models that integrate electrophysiological recordings with imaging modalities, genomic and transcriptomic profiles, and behavioral outputs to facilitate improved interpretation. The current methods struggle to handle multi-omics data because they contain large amounts of diverse information with complex structures, which include genomics, epigenomics, proteomics, and single-cell data. AI systems need to address these challenges by creating expandable systems that integrate various data structures and generate results that reveal biological processes, rather than making predictions. The current deep learning models operate as black boxes, which creates a significant obstacle because researchers need AI frameworks that reveal biological mechanisms through interpretable results [74,234,235]. To overcome these limitations, future research needs to create deep learning systems that analyze neural and organoid data through interpretable biological frameworks to address current limitations. The implementation of Explainable AI (XAI) and Interpretable Machine Learning (IML) methods represents a promising solution because these approaches enable scientists to understand model decisions through transparent and scientifically relevant explanations. The ELI5 toolkit together with LIME (Local Interpretable Model-agnostic Explanations) and SHAP (SHapley Additive exPlanations) has shown effectiveness in medical applications for both feature importance detection and clinician decision support according to [238]. The implementation of these techniques in neuroengineering enables researchers to understand how deep learning models process neural signals, which leads to better biological interpretation and accountability. The combination of mechanistic constraints with graph-based architectures enables AI models to model neural dynamics accurately while maintaining explainability. The combination of these methods converts deep learning systems from uninterpretable prediction tools into fully transparent and dependable frameworks for advancing organoid–AI research.
The development and enhancement of organoid systems encounter multiple substantial obstacles. Traditional organoid protocols rely on speculative literature analysis and numerous rounds of in vitro testing with Matrigel and other biological substrates, resulting in poor reproducibility, limited scalability, and safety concerns regarding immunogenicity. The chemical complexity of synthetic or hybrid hydrogels requires multiple testing cycles to achieve optimal results. The process of finding suitable inducing factors and external stimuli (mechanical, electrical, optical) proves to be time-consuming and inefficient for developing standardized platforms that support organoid growth and differentiation. AI-based experimental design technology shows promise to speed up this process by determining ideal conditions that minimize biological and environmental noise between different batches [74]. The analysis of organoids through images demands new approaches. The process of extracting multiscale features from 2D/3D imaging data is time-consuming, error-prone, and heavily dependent on human judgment. The current analysis methods require manual adjustments to produce restricted measurements, which include bounding boxes; however, they cannot detect intricate details about protrusions or growth asymmetries. The development of machine learning systems that can perform precise cell- and tissue-scale morphological analysis of organoids while preserving cell movement patterns has become an immediate necessity, according to [74].
The integration of energy-based hybrid stimulation techniques into organoid–AI platforms represents a vital direction for future development. The current simulation framework requires the development of energy-based hybrid stimulation systems that unite electrical and ultrasound modalities for organoid–AI systems. The precise control of neuronal excitability through electrical stimulation occurs at low latency by using controlled current injection, but ultrasound neuromodulation provides noninvasive membrane potential and ion-channel kinetic control through acoustic radiation forces and cavitation micro effects [239]. The combination of physical stimulation methods with AI-controlled models of organoid development and neural activity will create feedback-controlled systems that use deep network predictions to generate adaptive stimulation protocols. The combination of FEM simulations with deep learning models becomes possible using physically accurate constraints for neural activation and morphological response prediction in 3D organoid environments. The optimization of stimulation parameters through reinforcement learning or variational autoencoder (VAE) controllers would produce targeted neural modulation while minimizing thermal and mechanical stress. Research has shown that machine learning systems working with ultrasound and electrical fields and multiphysics models create precise energy delivery systems for in vitro and in vivo neural activation mapping [240]. The integration of these principles into organoid–AI systems creates an opportunity to develop adaptive neurostimulation platforms that combine bioelectronic actuation with physical modeling and data-driven learning through a unified computational system.
The reliability and trustworthiness of AI models will be essential for all these domains. The central role of AI in neural decoding, stimulation, and real-time monitoring necessitates models that deliver explainable results while being ethically sound and capable of functioning across diverse population groups. The implementation of federated learning for decentralized computation and bias-mitigation strategies for underrepresented populations and XAI (Explainable AI) principles for clinical interpretability and regulatory transparency will be necessary, according to Bai et al. (2024) in their work “AI-Enabled Organoids” [74].
The quantification of behavior and affective states represents an emerging research area for applications in neurodegenerative and neuropsychiatric disorders. The next generation of closed-loop systems requires development to detect emotional and cognitive changes through wearable biosensors and real-time neural monitoring. The systems will operate through strong behavioral models that extract internal states from multiple physiological signals to generate appropriate responses. The combination of organoids with neural interface platforms will create innovative diagnostic and therapeutic tools tailored to individual patients. Organoid-based models integrated with AI and real-time recording systems will enable the prediction of disease phenotypes and drug responses before clinical symptoms appear. The successful implementation depends on achieving standardization and benchmarking across organoid manufacturing protocols, central databases, and assay pipelines to guarantee reliable translation to clinical settings.
The final requirement for all advancements demands ethical governance to lead the way. The deployment of invasive neurotechnologies and the ontological questions surrounding the sentient-like behavior of advanced organoids require public transparency, regulatory oversight, and bioethical collaboration among different fields. The development of responsible and inclusive future directions for these powerful tools requires proper frameworks that must be both equitable and thoughtful, according to [74,217,218,219,220].

Author Contributions

Conceptualization, Y.Y. and S.K.; literature search and screening, M.U., S.A., Y.Y. and S.K.; analysis and interpretation, M.U., S.A. and C.O.-M.; writing—original draft preparation, M.U. and S.A.; writing—review and editing, M.U., S.A., C.O.-M., Y.Y. and S.K.; visualization, M.U., S.A. and C.O.-M.; supervision and project administration, Y.Y. and S.K.; funding acquisition, Y.Y. and S.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Science Foundation (Award Nos. 2100729, 2100987, 2429044, 2315654, and 2133630), the National Institutes of Health (Award Nos. 1SC1NS122448 and 1UG3EB036466), and the 2025 Provost’s Faculty Seed Grant from North Carolina Agricultural and Technical State University.

Acknowledgments

During the preparation of this manuscript/study, the authors used ChatGPT 4o and Grammarly to edit and enhance the grammar of the manuscript. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADAlzheimer’s Disease
AIArtificial Intelligence
BCIBrain–Computer Interface
BiLSTMBidirectional Long Short-Term Memory Network
BMIBrain–Machine Interface
BOMABrain and Organoids Manifold Alignment
CNNConvolutional Neural Network
cyb-organoidCyborg Organoid (hippocampal platform)
DBNDeep Belief Network
DBSDeep Brain Stimulation
DLDeep Learning
DNNDeep Neural Network
DTIDiffusion Tensor Imaging
ECEndothelial Cell
ECMExtracellular Matrix
ECoGElectrocorticography
EEGElectroencephalography
fNIRSFunctional Near-Infrared Spectroscopy
fUS/fUSiFunctional Ultrasound Imaging
GANGenerative Adversarial Network
GNNGraph Neural Network
GPUGraphics Processing Unit
GRUGated Recurrent Unit
hHOsHuman Hippocampal Organoids
iPSCsInduced Pluripotent Stem Cells
LFPsLocal Field Potentials
LRLogistic Regression
LSTMLong Short-Term Memory Network
MDMSMulti-Dimensional Mental States
MEAsMicroelectrode Arrays
MEGMagnetoencephalography
MLMachine Learning
MLPMulti-Layer Perceptron
MPCMetal–Polymer Conductor
mMPCMesh Metal–Polymer Conductor
MRIMagnetic Resonance Imaging
NDNeurodegenerative Diseases
NEURONSimulation environment for modeling neurons and networks
OBCIOrganoid–Brain–Computer Interface
OPMOptically Pumped Magnetometer
PCAPrincipal Component Analysis
PDParkinson’s Disease
PETPositron Emission Tomography
RNNRecurrent Neural Network
RFRandom Forest
rs-fMRIResting-State Functional Magnetic Resonance Imaging
SQUIDSuperconducting Quantum Interference Device
SRCSignal-Receiving Cell
SSCSignal-Sending Cell
SVMSupport Vector Machine
TADPOLEAlzheimer’s Disease Prediction Challenge Dataset
tFUSTranscranial Focused Ultrasound Stimulation
UCIUniversity of California, Irvine (voice datasets)
XAIExplainable Artificial Intelligence

References

  1. Alzheimer’s Association. 2025 Alzheimer’s Disease Facts and Figures. 6.5 Million Americans Age 65+ with AD, Projected to 13.8 Million by 2060. 2025. Available online: https://www.alz.org/alzheimers-dementia/facts-figures (accessed on 17 July 2025).
  2. Dorsey, E.R.; Bloem, B.R. The Emerging Evidence of the Parkinson Pandemic. JAMA Neurol. 2018, 75, 9–10. [Google Scholar] [CrossRef]
  3. Yang, W.; Hamilton, J.L.; Kopil, C.; Beck, J.C.; Tanner, C.M.; Albin, R.L.; Ray Dorsey, E.; Dahodwala, N.; Cintina, I.; Hogan, P.; et al. Current and projected future economic burden of Parkinson’s disease in the US. npj Park. Dis. 2020, 6, 15. [Google Scholar] [CrossRef]
  4. Wolpaw, J.R.; Birbaumer, N.; McFarland, D.J.; Pfurtscheller, G.; Vaughan, T.M. Brain–computer interfaces for communication and control. Clin. Neurophysiol. 2002, 113, 767–791. [Google Scholar] [CrossRef]
  5. Shenoy, K.V.; Carmena, J.M. Combining decoder design and neural adaptation in brain-machine interfaces. Neuron 2014, 84, 665–680. [Google Scholar] [CrossRef] [PubMed]
  6. Tanji, J.; Hoshi, E. Behavioral planning in the prefrontal cortex. Curr. Opin. Neurobiol. 2001, 11, 164–170. [Google Scholar] [CrossRef] [PubMed]
  7. Antzoulatos, E.G.; Miller, E.K. Differences between neural activity in prefrontal cortex and striatum during learning of novel abstract categories. Neuron 2011, 71, 243–249. [Google Scholar] [CrossRef]
  8. Graybiel, A.M.; Aosaki, T.; Flaherty, A.W.; Kimura, M. The basal ganglia and adaptive motor control. Science 1994, 265, 1826–1831. [Google Scholar] [CrossRef]
  9. Goldman-Rakic, P. Motor control function of the prefrontal cortex. In Ciba Foundation Symposium 132-Motor Areas of the Cerebral Cortex: Motor Areas of The Cerebral Cortex: Ciba Foundation Symposium 132; Wiley Online Library: Hoboken, NJ, USA, 2007; pp. 187–200. [Google Scholar]
  10. Wallis, J.D.; Anderson, K.C.; Miller, E.K. Single neurons in prefrontal cortex encode abstract rules. Nature 2001, 411, 953–956. [Google Scholar] [CrossRef]
  11. Tanveer, M.; Richhariya, B.; Khan, R.U.; Rashid, A.H.; Khanna, P.; Prasad, M.; Lin, C.T. Machine learning techniques for the diagnosis of Alzheimer’s disease: A review. ACM Trans. Multimed. Comput. Commun. Appl. (TOMM) 2020, 16, 1–35. [Google Scholar] [CrossRef]
  12. Khojaste-Sarakhsi, M.; Haghighi, S.S.; Ghomi, S.F.; Marchiori, E. Deep learning for Alzheimer’s disease diagnosis: A survey. Artif. Intell. Med. 2022, 130, 102332. [Google Scholar] [CrossRef]
  13. Liu, M.; Cheng, D.; Yan, W.; Initiative, A.D.N. Classification of Alzheimer’s disease by combination of convolutional and recurrent neural networks using FDG-PET images. Front. Neuroinform. 2018, 12, 35. [Google Scholar] [CrossRef]
  14. Zhang, S.; Yang, J.; Zhang, Y.; Zhong, J.; Hu, W.; Li, C.; Jiang, J. The combination of a graph neural network technique and brain imaging to diagnose neurological disorders: A review and outlook. Brain Sci. 2023, 13, 1462. [Google Scholar] [CrossRef] [PubMed]
  15. Song, X.; Mao, M.; Qian, X. Auto-metric graph neural network based on a meta-learning strategy for the diagnosis of Alzheimer’s disease. IEEE J. Biomed. Health Inform. 2021, 25, 3141–3152. [Google Scholar] [CrossRef] [PubMed]
  16. Ranjbarzadeh, R.; Jafarzadeh Ghoushchi, S.; Anari, S.; Safavi, S.; Tataei Sarshar, N.; Babaee Tirkolaee, E.; Bendechache, M. A deep learning approach for robust, multi-oriented, and curved text detection. Cogn. Comput. 2024, 16, 1979–1991. [Google Scholar] [CrossRef]
  17. Lawhern, V.J.; Solon, A.J.; Waytowich, N.R.; Gordon, S.M.; Hung, C.P.; Lance, B.J. EEGNet: A compact convolutional neural network for EEG-based brain–computer interfaces. J. Neural Eng. 2018, 15, 056013. [Google Scholar] [CrossRef]
  18. Craik, A.; He, Y.; Contreras-Vidal, J.L. Deep learning for electroencephalogram (EEG) classification tasks: A review. J. Neural Eng. 2019, 16, 031001. [Google Scholar] [CrossRef]
  19. Roy, R.; Tripathy, R.K.; Acharya, U.R. Transformer-based deep learning models for EEG signal analysis: A review. IEEE Trans. Neural Syst. Rehabil. Eng. 2023, 31, 2211–2225. [Google Scholar]
  20. Zhang, L.; Wang, M.; Chen, H.; Li, R. Hybrid AI-FEM Modeling for Multiphysics Simulation of Neuroelectronic Interfaces. Electronics 2024, 14, 2268. [Google Scholar] [CrossRef]
  21. Harrison, R.R. The design of integrated circuits to observe brain activity. Proc. IEEE 2008, 96, 1203–1216. [Google Scholar] [CrossRef]
  22. Park, J.H.; Wang, H.; Seok, M.; Chiang, P. SPAD-based imaging of neural activity: Emerging photonic front-ends for neurotechnology. Nat. Photonics 2023, 17, 391–403. [Google Scholar]
  23. Qian, X.; Song, H.; Ming, G.l. Brain organoids: Advances, applications and challenges. Development 2019, 146, dev166074. [Google Scholar] [CrossRef] [PubMed]
  24. Lancaster, M.A.; Renner, M.; Martin, C.A.; Wenzel, D.; Bicknell, L.; Hurles, M.; Homfray, T.; Penninger, J.; Jackson, A.; Knoblich, J. Cerebral organoids model human brain development and microcephaly. Nature 2013, 501, 373–379. [Google Scholar] [CrossRef] [PubMed]
  25. Wu, Y.; Cheng, J.; Qi, J.; Hang, C.; Dong, R.; Low, B.C.; Yu, H.; Jiang, X. Three-dimensional liquid metal-based neuro-interfaces for human hippocampal organoids. Nat. Commun. 2024, 15, 4047. [Google Scholar] [CrossRef] [PubMed]
  26. Anumanchipalli, G.K.; Metzger, S.; Chartier, J.; Moses, D.A.; Dougherty, M.E.; Liu, J.R.; Abrams, G.M.; Tu-Chan, A.; Ganguly, K.; Chang, E.F.; et al. A high-performance speech neuroprosthesis. Nature 2023, 621, 582–589. [Google Scholar]
  27. Wired Magazine. Paradromics Tested a Brain Implant. Now the FDA Is Involved. 2024. Available online: https://www.wired.com/story/paradromics-neuralink-rival-tested-brain-implant (accessed on 25 June 2025).
  28. Hu, N.; Shi, J.; Chen, C.; Wu, Y.; Yang, J.; Zhang, Z.; Zeng, H.; Li, B.; Huang, M.; Chen, J.; et al. Constructing organoid-brain–computer interfaces for neurofunctional repair after brain injury. Nat. Commun. 2024, 15, 9580. [Google Scholar] [CrossRef]
  29. Farwell, L.A.; Donchin, E. Talking off the top of your head: Toward a mental prosthesis utilizing event-related brain potentials. Electroencephalogr. Clin. Neurophysiol. 1988, 70, 510–523. [Google Scholar] [CrossRef]
  30. Birbaumer, N.; Ghanayim, N.; Hinterberger, T.; Iversen, I.; Kotchoubey, B.; Kübler, A.; Perelmouter, J.; Taub, E.; Flor, H. A spelling device for the paralysed. Nature 1999, 398, 297–298. [Google Scholar] [CrossRef]
  31. Sellers, E.W.; Donchin, E. A P300-based brain–computer interface: Initial tests by ALS patients. Clin. Neurophysiol. 2006, 117, 538–548. [Google Scholar] [CrossRef]
  32. Nijboer, F.; Sellers, E.W.; Mellinger, J.; Jordan, M.A.; Matuz, T.; Furdea, A.; Halder, S.; Mochty, U.; Krusienski, D.J.; Vaughan, T.M.; et al. A P300-based brain–computer interface for people with amyotrophic lateral sclerosis. Clin. Neurophysiol. 2008, 119, 1909–1916. [Google Scholar] [CrossRef]
  33. Kübler, A.; Müller-Putz, G.R. Brain–computer interfaces in the context of neurorehabilitation: Current and future directions. Appl. Psychophysiol. Biofeedback 2007, 32, 111–118. [Google Scholar]
  34. Guger, C.; Daban, S.; Sellers, E.; Holzner, C.; Krausz, G.; Carabalona, R.; Gramatica, F.; Edlinger, G. How many people are able to control a P300-based brain–computer interface (BCI)? Neurosci. Lett. 2009, 462, 94–98. [Google Scholar] [CrossRef] [PubMed]
  35. Li, H.; Zhang, Q.; Sun, X.; Li, C.; Luo, J.; Wang, X. Smart wearable electronics for biomedical monitoring and therapy. Nat. Electron. 2021, 4, 34–47. [Google Scholar]
  36. Kim, J.; Campbell, A.S.; de Ávila, B.E.F.; Wang, J. Stretchable bioelectronics for real-time physiological monitoring and therapy. Chem. Rev. 2020, 120, 4581–4623. [Google Scholar]
  37. Yang, Z.; Lin, X.; Lee, C. Wireless smart implants for continuous and real-time in vivo monitoring of tissue temperature and SAR in biomedical applications. Adv. Sci. 2023, 10, 2300787. [Google Scholar]
  38. Rao, R.P. Towards neural co-processors for the brain: Combining decoding and encoding in brain–computer interfaces. Curr. Opin. Neurobiol. 2019, 55, 142–151. [Google Scholar] [CrossRef]
  39. Nguyen, C.H.; Karavas, G.K.; Artemiadis, P. Adaptive multi-degree of freedom Brain Computer Interface using online feedback: Towards novel methods and metrics of mutual adaptation between humans and machines for BCI. PLoS ONE 2019, 14, e0212620. [Google Scholar] [CrossRef]
  40. De Santis, D. A framework for optimizing co-adaptation in body-machine interfaces. Front. Neurorobot. 2021, 15, 662181. [Google Scholar] [CrossRef]
  41. Yang, W. Brain-computer interfaces in cognitive rehabilitation for Alzheimer’s disease. Theor. Nat. Sci. 2024, 64, 179–185. [Google Scholar] [CrossRef]
  42. Squire, L.; Berg, D.; Bloom, F.E.; Du Lac, S.; Ghosh, A.; Spitzer, N.C. Fundamental Neuroscience; Academic Press: Cambridge, MA, USA, 2012. [Google Scholar]
  43. Edelman, B.J.; Johnson, N.; Sohrabpour, A.; Tong, S.; Thakor, N.; He, B. Systems neuroengineering: Understanding and interacting with the brain. Engineering 2015, 1, 292–308. [Google Scholar] [CrossRef]
  44. Lebedev, M.A.; Nicolelis, M.A. Brain–machine interfaces: Past, present and future. TRENDS Neurosci. 2006, 29, 536–546. [Google Scholar] [CrossRef]
  45. Pandarinath, C.; O’Shea, D.J.; Collins, J.; Jozefowicz, R.; Stavisky, S.D.; Kao, J.C.; Trautmann, E.M.; Kaufman, M.T.; Ryu, S.I.; Hochberg, L.R.; et al. Inferring single-trial neural population dynamics using sequential auto-encoders. Nat. Methods 2018, 15, 805–815. [Google Scholar] [CrossRef] [PubMed]
  46. Vieira, S.; Pinaya, W.H.; Mechelli, A. Using deep learning to investigate the neuroimaging correlates of psychiatric and neurological disorders: Methods and applications. Neurosci. Biobehav. Rev. 2017, 74, 58–75. [Google Scholar] [CrossRef] [PubMed]
  47. Roy, Y.; Banville, H.; Albuquerque, I.; Gramfort, A.; Falk, T.H.; Faubert, J. Deep learning-based electroencephalography analysis: A systematic review. J. Neural Eng. 2019, 16, 051001. [Google Scholar] [CrossRef] [PubMed]
  48. Badrulhisham, F.; Pogatzki-Zahn, E.; Segelcke, D.; Spisak, T.; Vollert, J. Machine learning and artificial intelligence in neuroscience: A primer for researchers. Brain Behav. Immun. 2024, 115, 470–479. [Google Scholar] [CrossRef]
  49. Frégnac, Y. Big data and the industrialization of neuroscience: A safe roadmap for understanding the brain? Science 2017, 358, 470–477. [Google Scholar] [CrossRef]
  50. Zador, A.; Escola, S.; Richards, B.; Ölveczky, B.; Bengio, Y.; Boahen, K.; Botvinick, M.; Chklovskii, D.; Churchland, A.; Clopath, C.; et al. Catalyzing next-generation artificial intelligence through neuroai. Nat. Commun. 2023, 14, 1597. [Google Scholar] [CrossRef]
  51. Onciul, R.; Tataru, C.I.; Dumitru, A.V.; Crivoi, C.; Serban, M.; Covache-Busuioc, R.A.; Radoi, M.P.; Toader, C. Artificial Intelligence and Neuroscience: Transformative Synergies in Brain Research and Clinical Applications. J. Clin. Med. 2025, 14, 550. [Google Scholar] [CrossRef]
  52. Samper-González, J.; Burgos, N.; Bottani, S.; Fontanella, S.; Lu, P.; Marcoux, A.; Routier, A.; Guillon, J.; Bacci, M.; Wen, J.; et al. Reproducible evaluation of classification methods in Alzheimer’s disease: Framework and application to MRI and PET data. NeuroImage 2018, 183, 504–521. [Google Scholar] [CrossRef]
  53. Solana-Lavalle, G.; Rosas-Romero, R. Classification of PPMI MRI scans with voxel-based morphometry and machine learning to assist in the diagnosis of Parkinson’s disease. Comput. Methods Programs Biomed. 2021, 198, 105793. [Google Scholar] [CrossRef]
  54. Prashanth, R.; Roy, S.D.; Mandal, P.K.; Ghosh, S. High-accuracy classification of Parkinson’s disease through shape analysis and surface fitting in 123I-Ioflupane SPECT imaging. IEEE J. Biomed. Health Inform. 2016, 21, 794–802. [Google Scholar] [CrossRef]
  55. Salvatore, C.; Cerasa, A.; Castiglioni, I.; Gallivanone, F.; Augimeri, A.; Lopez, M.; Arabia, G.; Morelli, M.; Gilardi, M.; Quattrone, A. Machine learning on brain MRI data for differential diagnosis of Parkinson’s disease and Progressive Supranuclear Palsy. J. Neurosci. Methods 2014, 222, 230–237. [Google Scholar] [CrossRef]
  56. Zhou, T.; Thung, K.H.; Zhu, X.; Shen, D. Effective feature learning and fusion of multimodality data using stage-wise deep neural network for dementia diagnosis. Hum. Brain Mapp. 2019, 40, 1001–1016. [Google Scholar] [CrossRef] [PubMed]
  57. Basaia, S.; Agosta, F.; Wagner, L.; Canu, E.; Magnani, G.; Santangelo, R.; Filippi, M.; Alzheimer’s Disease Neuroimaging Initiative. Automated classification of Alzheimer’s disease and mild cognitive impairment using a single MRI and deep neural networks. Neuroimage Clin. 2019, 21, 101645. [Google Scholar] [CrossRef] [PubMed]
  58. Ding, Y.; Sohn, J.H.; Kawczynski, M.G.; Trivedi, H.; Harnish, R.; Jenkins, N.W.; Lituiev, D.; Copeland, T.P.; Aboian, M.S.; Mari Aparici, C.; et al. A deep learning model to predict a diagnosis of Alzheimer disease by using 18F-FDG PET of the brain. Radiology 2019, 290, 456–464. [Google Scholar] [CrossRef] [PubMed]
  59. Leung, K.H.; Rowe, S.P.; Pomper, M.G.; Du, Y. A three-stage, deep learning, ensemble approach for prognosis in patients with Parkinson’s disease. EJNMMI Res. 2021, 11, 52. [Google Scholar] [CrossRef]
  60. Sivaranjini, S.; Sujatha, C. Deep learning based diagnosis of Parkinson’s disease using convolutional neural network. Multimed. Tools Appl. 2020, 79, 15467–15479. [Google Scholar] [CrossRef]
  61. Aliero, A.S.; Malhotra, N. Comparative study of deep learning models for Parkinson’s Disease detection. Benchcouncil Trans. Benchmarks, Stand. Eval. 2025, 5, 100219. [Google Scholar] [CrossRef]
  62. Taleb, C.; Likforman-Sulem, L.; Mokbel, C.; Khachab, M. Detection of Parkinson’s disease from handwriting using deep learning: A comparative study. Evol. Intell. 2023, 16, 1813–1824. [Google Scholar] [CrossRef]
  63. Mohammed, B.A.; Senan, E.M.; Rassem, T.H.; Makbol, N.M.; Alanazi, A.A.; Al-Mekhlafi, Z.G.; Almurayziq, T.S.; Ghaleb, F.A. Multi-method analysis of medical records and MRI images for early diagnosis of dementia and Alzheimer’s disease based on deep learning and hybrid methods. Electronics 2021, 10, 2860. [Google Scholar] [CrossRef]
  64. Matlani, P. BiLSTM-ANN: Early diagnosis of Alzheimer’s disease using hybrid deep learning algorithms. Multimed. Tools Appl. 2024, 83, 60761–60788. [Google Scholar] [CrossRef]
  65. Rudroff, T.; Rainio, O.; Klén, R. AI for the prediction of early stages of Alzheimer’s disease from neuroimaging biomarkers: A narrative review of a growing field. Neurol. Sci. 2024, 45, 5117–5127. [Google Scholar] [CrossRef] [PubMed]
  66. Yang, J.; Wang, F.; Li, Z.; Yang, Z.; Dong, X.; Han, Q. Constructing high-order functional networks based on hypergraph for diagnosis of autism spectrum disorders. Front. Neurosci. 2023, 17, 1257982. [Google Scholar] [CrossRef] [PubMed]
  67. Hinrichs, C.; Singh, V.; Xu, G.; Johnson, S.C.; Alzheimers Disease Neuroimaging Initiative. Predictive markers for AD in a multi-modality framework: An analysis of MCI progression in the ADNI population. NeuroImage 2011, 55, 574–589. [Google Scholar] [CrossRef] [PubMed]
  68. Schwarz, S.T.; Abaei, M.; Gontu, V.; Morgan, P.S.; Bajaj, N.; Auer, D.P. Diffusion tensor imaging of nigral degeneration in Parkinson’s disease: A region-of-interest and voxel-based study at 3 T and systematic review with meta-analysis. NeuroImage Clin. 2013, 3, 481–488. [Google Scholar] [CrossRef]
  69. Simuni, T.; Merchant, K.; Brumm, M.C.; Cho, H.; Caspell-Garcia, C.; Coffey, C.S.; Chahine, L.M.; Alcalay, R.N.; Nudelman, K.; Foroud, T.; et al. Longitudinal clinical and biomarker characteristics of non-manifesting LRRK2 G2019S carriers in the PPMI cohort. npj Park. Dis. 2022, 8, 140. [Google Scholar] [CrossRef]
  70. Rahman, S.; Hasan, M.; Sarkar, A.K.; Khan, F. Classification of Parkinson’s disease using speech signal with machine learning and deep learning approaches. Eur. J. Electr. Eng. Comput. Sci. 2023, 7, 20–27. [Google Scholar] [CrossRef]
  71. Hu, Q.; Whitney, H.M.; Giger, M.L. A deep learning methodology for improved breast cancer diagnosis using multiparametric MRI. Sci. Rep. 2020, 10, 10536. [Google Scholar] [CrossRef]
  72. Zhao, A.; Liu, Y.; Yu, X.; Xing, X.; Zhou, H. Artificial intelligence-enabled detection and assessment of Parkinson’s disease using multimodal data: A survey. Inf. Fusion 2025, 121, 103175. [Google Scholar] [CrossRef]
  73. Mehonic, A.; Kenyon, A.J. Brain-inspired computing needs a master plan. Nature 2022, 604, 255–260. [Google Scholar] [CrossRef]
  74. Bai, L.; Wu, Y.; Li, G.; Zhang, W.; Zhang, H.; Su, J. AI-enabled organoids: Construction, analysis, and application. Bioact. Mater. 2024, 31, 525–548. [Google Scholar] [CrossRef]
  75. Zhang, Y.; He, G.; Ma, L.; Liu, X.; Hjorth, J.J.; Kozlov, A.; He, Y.; Zhang, S.; Kotaleski, J.H.; Tian, Y.; et al. A GPU-based computational framework that bridges neuron simulation and artificial intelligence. Nat. Commun. 2023, 14, 5798. [Google Scholar] [CrossRef]
  76. Quadrato, G.; Nguyen, T.; Macosko, E.Z.; Sherwood, J.L.; Min Yang, S.; Berger, D.R.; Maria, N.; Scholvin, J.; Goldman, M.; Kinney, J.P.; et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 2017, 545, 48–53. [Google Scholar] [CrossRef]
  77. Trujillo, C.A.; Gao, R.; Negraes, P.D.; Gu, J.; Buchanan, J.; Preissl, S.; Wang, A.; Wu, W.; Haddad, G.G.; Chaim, I.A.; et al. Complex oscillatory waves emerging from cortical organoids model early human brain network development. Cell Stem Cell 2019, 25, 558–569.e7. [Google Scholar] [CrossRef]
  78. Rossi, G.; Manfrin, A.; Lutolf, M.P. Progress and potential in organoid research. Nat. Rev. Genet. 2018, 19, 671–687. [Google Scholar] [CrossRef] [PubMed]
  79. Sharf, T.; van der Molen, T.; Glasauer, S.M.; Guzman, E.; Buccino, A.P.; Luna, G.; Cheng, Z.; Audouard, M.; Ranasinghe, K.G.; Kudo, K.; et al. Functional neuronal circuitry and oscillatory dynamics in human brain organoids. Nat. Commun. 2022, 13, 4403. [Google Scholar] [CrossRef] [PubMed]
  80. Smirnova, L.; Caffo, B.S.; Gracias, D.H.; Huang, Q.; Morales Pantoja, I.E.; Tang, B.; Zack, D.J.; Berlinicke, C.A.; Boyd, J.L.; Harris, T.D.; et al. Organoid intelligence (OI): The new frontier in biocomputing and intelligence-in-a-dish. Front. Sci. 2023, 1, 1017235. [Google Scholar] [CrossRef]
  81. Birey, F.; Andersen, J.; Makinson, C.D.; Islam, S.; Wei, W.; Huber, N.; Fan, J.; Metzler, Z.; Panagiotakos, G.; Thom, N.; et al. Assembly of functionally integrated human forebrain spheroids. Nature 2017, 545, 54–59. [Google Scholar] [CrossRef]
  82. Jovanova, M.; Falk, E.B.; Pearl, J.M.; Pandey, P.; O’Donnell, M.B.; Kang, Y.; Bassett, D.S.; Lydon-Staley, D.M. Brain system integration and message consistent health behavior change. Health Psychol. 2022, 41, 611–620. [Google Scholar] [CrossRef]
  83. Qian, X.; Nguyen, H.N.; Song, M.M.; Hadiono, C.; Ogden, S.C.; Hammack, C.; Yao, B.; Hamersky, G.R.; Jacob, F.; Zhong, C.; et al. Brain-region-specific organoids using mini-bioreactors for modeling human neural development. Nature Protocols 2016, 11, 1721–1738. [Google Scholar]
  84. Di Lullo, E.; Kriegstein, A.R. The use of brain organoids to investigate neural development and disease. Nat. Rev. Neurosci. 2017, 18, 573–584. [Google Scholar] [CrossRef]
  85. Groveman, B.R.; Foliaki, S.T.; Orru, C.D.; Zanusso, G.; Race, B. Human cerebral organoids as a model for prion disease. Acta Neuropathol. Commun. 2021, 9, 50. [Google Scholar]
  86. Wang, Y.; Chiola, S.; Yang, G.; Russell, C.; Armstrong, C.J.; Wu, Y.; Spampanato, J.; Tarboton, P.; Ullah, H.A.; Edgar, N.U.; et al. Modeling human telencephalic development and autism-associated SHANK3 deficiency using organoids generated from single neural rosettes. Nat. Commun. 2022, 13, 5688. [Google Scholar] [CrossRef]
  87. Revah, O.; Gore, F.; Kelley, K.W.; Andersen, J.; Sakai, N.; Chen, X.; Li, M.-Y.; Birey, F.; Yang, X.; Saw, N.L.; et al. Maturation and circuit integration of transplanted human cortical organoids. Nature 2022, 610, 319–326. [Google Scholar] [CrossRef]
  88. Wilson, M.N.; Thunemann, M.; Liu, X.; Lu, Y.; Puppo, F.; Adams, J.W.; Kim, J.-H.; Ramezani, M.; Pizzo, D.P.; Djurovic, S.; et al. Multimodal monitoring of human cortical organoids implanted in mice reveal functional connection with visual cortex. Nat. Commun. 2022, 13, 7945. [Google Scholar] [CrossRef] [PubMed]
  89. Jgamadze, D.; Lim, J.T.; Zhang, Z.; Harary, P.M.; Germi, J.; Mensah-Brown, K.; Adam, C.D.; Mirzakhalili, E.; Singh, S.; Gu, J.B.; et al. Structural and functional integration of human forebrain organoids with the injured adult rat visual system. Cell Stem Cell 2023, 30, 137–152.e7. [Google Scholar] [CrossRef]
  90. Cao, S.-Y.; Yang, D.; Huang, Z.-Q.; Lin, Y.-H.; Wu, H.-Y.; Chang, L.; Luo, C.-X.; Xu, Y.; Liu, Y.; Zhu, D.-Y. Cerebral organoids transplantation repairs infarcted cortex and restores impaired function after stroke. NPJ Regen. Med. 2023, 8, 27. [Google Scholar] [CrossRef] [PubMed]
  91. Feng, J.F.; Liu, J.; Zhang, L.; Jiang, J.Y.; Russell, M.; Lyeth, B.G.; Nolta, J.A.; Zhao, M. Electrical guidance of human stem cells in the rat brain. Stem Cell Rep. 2017, 9, 177–189. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, M.; Tang, Z.; Liu, X.; Van der Spiegel, J. Electronic neural interfaces. Nat. Electron. 2020, 3, 191–200. [Google Scholar] [CrossRef]
  93. Mudgal, S.K.; Sharma, S.K.; Chaturvedi, J.; Sharma, A. Brain computer interface advancement in neurosciences: Applications and issues. Interdiscip. Neurosurg. 2020, 20, 100694. [Google Scholar] [CrossRef]
  94. Willsey, M.S.; Shah, N.P.; Avansino, D.T.; Hahn, N.V.; Jamiolkowski, R.M.; Kamdar, F.B.; Hochberg, L.R.; Willett, F.R.; Henderson, J.M. A high-performance brain–computer interface for finger decoding and quadcopter game control in an individual with paralysis. Nat. Med. 2025, 31, 303–314. [Google Scholar] [CrossRef]
  95. Musk, E.; Neuralink. An Integrated Brain-Machine Interface Platform with Thousands of Channels. J. Med. Internet Res. 2019, 21, e16194. [Google Scholar] [CrossRef]
  96. Greenberg, A.; Cohen, A.; Grewal, M. Patent landscape of brain-machine interface technology. Nat. Biotechnol. 2021, 39, 1194–1199. [Google Scholar] [CrossRef]
  97. Lin, S.; Liu, J.; Li, W.; Wang, D.; Huang, Y.; Jia, C.; Li, Z.; Murtaza, M.; Wang, H.; Song, J.; et al. A flexible, robust, and gel-free electroencephalogram electrode for noninvasive brain-computer interfaces. Nano Lett. 2019, 19, 6853–6861. [Google Scholar] [CrossRef]
  98. Abiri, R.; Borhani, S.; Sellers, E.W.; Jiang, Y.; Zhao, X. A comprehensive review of EEG-based brain-computer interface paradigms. J. Neural Eng. 2019, 16, 011001. [Google Scholar] [CrossRef]
  99. Wolpaw, J.R. Brain-computer interfaces (BCIs) for communication and control. In Proceedings of the 9th International ACM SIGACCESS Conference on Computers and Accessibility, Tempe, AZ, USA, 15–17 October 2007; pp. 1–2. [Google Scholar]
  100. Kübler, A.; Neumann, N.; Kaiser, J.; Kotchoubey, B.; Hinterberger, T.; Birbaumer, N.P. Brain-computer communication: Self-regulation of slow cortical potentials for verbal communication. Arch. Phys. Med. Rehabil. 2001, 82, 1533–1539. [Google Scholar] [CrossRef]
  101. Zhang, A.; Jiang, Y.; Loh, K.Y.; Bao, Z.; Deisseroth, K. Genetically targeted chemical assembly. Nat. Rev. Bioeng. 2024, 2, 82–94. [Google Scholar] [CrossRef]
  102. Kim, S.H.; Chang, M.Y. Application of human brain organoids: Opportunities and challenges in modeling human brain development and Neurodevelopmental Diseases. Int. J. Mol. Sci. 2023, 24, 12528. [Google Scholar] [CrossRef]
  103. Kisban, S.; Herwik, S.; Seidl, K.; Rubehn, B.; Jezzini, A.; Umiltà, M.A. Comprehensive biocompatibility analysis of neural interfaces. Biomed. Microdevices 2011, 13, 361–373. [Google Scholar]
  104. Minev, I.R.; Musienko, P.; Hirsch, A.; Barraud, Q.; Wenger, N.; Moraud, E.M.; Gandar, J.; Capogrosso, M.; Milekovic, T.; Asboth, L.; et al. Electronic dura mater for long-term multimodal neural interfaces. Science 2015, 347, 159–163. [Google Scholar] [CrossRef] [PubMed]
  105. Chung, K.; Kim, J.; Lee, H.; Jang, Y.; Kim, H.; Seo, J.; Park, J.; Lee, Y.; Song, Y.M.; Hwang, S.W.; et al. Bioelectronics for neuroprosthetics. Nat. Electron. 2019, 2, 429–442. [Google Scholar]
  106. Viventi, J.; Kim, D.H.; Vigeland, L.; Frechette, E.S.; Blanco, J.A.; Kim, Y.S.; Avrin, A.E.; Tiruvadi, V.R.; Hwang, S.W.; Vanleer, A.C.; et al. Flexible, foldable, actively multiplexed, high-density electrode array for mapping brain activity in vivo. Nat. Neurosci. 2011, 14, 1599–1605. [Google Scholar] [CrossRef]
  107. Luan, L.; Wei, X.; Zhao, Z.; Siegel, J.J.; Potnis, O.; Tuppen, C.A.; Lin, S.; Kazmi, S.; Fowler, R.A.; Holloway, S.; et al. Ultraflexible nanoelectronic probes form reliable, glial scar-free neural integration. Sci. Adv. 2017, 3, e1601966. [Google Scholar] [CrossRef]
  108. Kretzschmar, K.; Clevers, H. Organoids: Modeling development and the stem cell niche in a dish. Dev. Cell 2016, 38, 590–600. [Google Scholar] [CrossRef] [PubMed]
  109. Kagan, B.J.; Kitchen, A.C.; Tran, N.T.; Habibollahi, F.; Khajehnejad, M.; Parker, B.J.; Bhat, A.; Rollo, B.; Razi, A.; Friston, K.J. In vitro neurons learn and exhibit sentience when embodied in a simulated game-world. Neuron 2022, 110, 3952–3969.e8. [Google Scholar] [CrossRef] [PubMed]
  110. He, Y.; Evans, A. Graph theoretical modeling of brain connectivity. Curr. Opin. Neurol. 2010, 23, 341–350. [Google Scholar] [CrossRef] [PubMed]
  111. Tan, S.Y.; Feng, X.; Cheng, L.K.W.; Wu, A.R. Vascularized human brain organoid on-chip. Lab Chip 2023, 23, 2693–2709. [Google Scholar] [CrossRef]
  112. Salmon, I.; Grebenyuk, S.; Fattah, A.R.A.; Rustandi, G.; Pilkington, T.; Verfaillie, C.; Ranga, A. Engineering neurovascular organoids with 3D printed microfluidic chips. Lab Chip 2022, 22, 1615–1629. [Google Scholar] [CrossRef]
  113. Papamichail, L.; Koch, L.S.; Veerman, D.; Broersen, K.; van der Meer, A.D. Organoids-on-a-chip: Microfluidic technology enables culture of organoids with enhanced tissue function and potential for disease modeling. Front. Bioeng. Biotechnol. 2025, 13, 1515340. [Google Scholar] [CrossRef]
  114. Werschler, N.; Quintard, C.; Nguyen, S.; Penninger, J. Engineering next generation vascularized organoids. Atherosclerosis 2024, 398, 118529. [Google Scholar] [CrossRef]
  115. Grebenyuk, S.; Abdel Fattah, A.R.; Kumar, M.; Toprakhisar, B.; Rustandi, G.; Vananroye, A.; Salmon, I.; Verfaillie, C.; Grillo, M.; Ranga, A. Large-scale perfused tissues via synthetic 3D soft microfluidics. Nat. Commun. 2023, 14, 193. [Google Scholar] [CrossRef]
  116. Aguirre, M.; Escobar, M.; Forero Amézquita, S.; Cubillos, D.; Rincón, C.; Vanegas, P.; Tarazona, M.P.; Atuesta Escobar, S.; Blanco, J.C.; Celis, L.G. Application of the Yamanaka transcription factors Oct4, Sox2, Klf4, and c-Myc from the laboratory to the clinic. Genes 2023, 14, 1697. [Google Scholar] [CrossRef]
  117. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed]
  118. Yan, Y.; Bejoy, J.; Marzano, M.; Li, Y. The use of pluripotent stem cell-derived organoids to study extracellular matrix development during neural degeneration. Cells 2019, 8, 242. [Google Scholar] [CrossRef] [PubMed]
  119. Rosebrock, D.; Arora, S.; Mutukula, N.; Volkman, R.; Gralinska, E.; Balaskas, A.; Aragonés Hernández, A.; Buschow, R.; Brändl, B.; Müller, F.J.; et al. Enhanced cortical neural stem cell identity through short SMAD and WNT inhibition in human cerebral organoids facilitates emergence of outer radial glial cells. Nat. Cell Biol. 2022, 24, 981–995. [Google Scholar] [CrossRef]
  120. Qian, X.; Su, Y.; Adam, C.D.; Deutschmann, A.U.; Pather, S.R.; Goldberg, E.M.; Su, K.; Li, S.; Lu, L.; Jacob, F.; et al. Sliced human cortical organoids for modeling distinct cortical layer formation. Cell Stem Cell 2020, 26, 766–781. [Google Scholar] [CrossRef]
  121. Rathore, G.S.; Ankjær, M.; Rifes, P.; Hänninen, E.; Calleja, F.S.; Rusimbi, C.; Kajtez, J.; Nygaard, A.H.; Piilgaard, L.S.; Dubonyte, U.; et al. Mapping early patterning events in human neural development using an in-vitro microfluidic stem cell model. bioRxiv 2025. [Google Scholar] [CrossRef]
  122. Polevoy, H.; Gutkovich, Y.E.; Michaelov, A.; Volovik, Y.; Elkouby, Y.M.; Frank, D. New roles for Wnt and BMP signaling in neural anteroposterior patterning. EMBO Rep. 2019, 20, e45842. [Google Scholar] [CrossRef]
  123. Avnet, S.; Mizushima, E.; Severino, B.; Lipreri, M.V.; Scognamiglio, A.; Corvino, A.; Baldini, N.; Cortini, M. Antagonizing the S1P-S1P3 Axis as a Promising Anti-Angiogenic Strategy. Metabolites 2025, 15, 178. [Google Scholar] [CrossRef]
  124. Shi, Y.; Sun, L.; Wang, M.; Liu, J.; Zhong, S.; Li, R.; Li, P.; Guo, L.; Fang, A.; Chen, R.; et al. Vascularized human cortical organoids (vOrganoids) model cortical development in vivo. PLoS Biol. 2020, 18, e3000705. [Google Scholar] [CrossRef]
  125. Kook, M.G.; Lee, S.E.; Shin, N.; Kong, D.; Kim, D.H.; Kim, M.S.; Kang, H.K.; Choi, S.W.; Kang, K.S. Generation of cortical brain organoid with vascularization by assembling with vascular spheroid. Int. J. Stem Cells 2022, 15, 85–94. [Google Scholar] [CrossRef]
  126. Ahn, Y.; An, J.H.; Yang, H.J.; Lee, D.G.; Kim, J.; Koh, H.; Park, Y.H.; Song, B.S.; Sim, B.W.; Lee, H.J.; et al. Human blood vessel organoids penetrate human cerebral organoids and form a vessel-like system. Cells 2021, 10, 2036. [Google Scholar] [CrossRef]
  127. Kermani, P.; Rafii, D.; Jin, D.K.; Whitlock, P.; Schaffer, W.; Chiang, A.; Vincent, L.; Friedrich, M.; Shido, K.; Hackett, N.R.; et al. Neurotrophins promote revascularization by local recruitment of TrkB+ endothelial cells and systemic mobilization of hematopoietic progenitors. J. Clin. Investig. 2005, 115, 653–663. [Google Scholar] [CrossRef]
  128. Yuan, Y.; Ye, H.Q.; Ren, Q.C. Upregulation of the BDNF/TrKB pathway promotes epithelial-mesenchymal transition, as well as the migration and invasion of cervical cancer. Int. J. Oncol. 2017, 52, 461–472. [Google Scholar] [CrossRef] [PubMed]
  129. Sun, X.Y.; Ju, X.C.; Li, Y.; Zeng, P.M.; Wu, J.; Zhou, Y.Y.; Shen, L.B.; Dong, J.; Chen, Y.J.; Luo, Z.G. Generation of vascularized brain organoids to study neurovascular interactions. eLife 2022, 11, e76707. [Google Scholar] [CrossRef] [PubMed]
  130. Pham, M.T.; Pollock, K.M.; Rose, M.D.; Cary, W.A.; Stewart, H.R.; Zhou, P.; Nolta, J.A.; Waldau, B. Generation of human vascularized brain organoids. Neuroreport 2018, 29, 588–593. [Google Scholar] [CrossRef] [PubMed]
  131. Wörsdörfer, P.; Rockel, A.; Alt, Y.; Kern, A.; Ergün, S. Generation of vascularized neural organoids by co-culturing with mesodermal progenitor cells. STAR Protoc. 2020, 1, 100041. [Google Scholar] [CrossRef]
  132. Walsh, R.M.; Luongo, R.; Giacomelli, E.; Ciceri, G.; Rittenhouse, C.; Verrillo, A.; Galimberti, M.; Bocchi, V.D.; Wu, Y.; Xu, N.; et al. Generation of human cerebral organoids with a structured outer subventricular zone. Cell Rep. 2024, 43, 114031. [Google Scholar] [CrossRef]
  133. VanderWielen, B.D.; Yuan, Z.; Friedmann, D.R.; Kovall, R.A. Transcriptional repression in the Notch pathway: Thermodynamic characterization of CSL-MINT (Msx2-interacting nuclear target protein) complexes. J. Biol. Chem. 2011, 286, 14892–14902. [Google Scholar] [CrossRef]
  134. Bejoy, J.; Bijonowski, B.; Marzano, M.; Jeske, R.; Ma, T.; Li, Y. Wnt-Notch signaling interactions during neural and astroglial patterning of human stem cells. Tissue Eng. Part A 2020, 26, 419–431. [Google Scholar] [CrossRef]
  135. Fiddes, I.T.; Lodewijk, G.A.; Mooring, M.; Bosworth, C.M.; Ewing, A.D.; Mantalas, G.L.; Novak, A.M.; van den Bout, A.; Bishara, A.; Rosenkrantz, J.L.; et al. Human-specific NOTCH2NL genes affect notch signaling and cortical neurogenesis. Cell 2018, 173, 1356–1369. [Google Scholar] [CrossRef]
  136. Hurley, E.M.; Mozolewski, P.; Dobrowolski, R.; Hsieh, J. Familial Alzheimer’s disease-associated PSEN1 mutations affect neurodevelopment through increased Notch signaling. Stem Cell Rep. 2023, 18, 1516–1533. [Google Scholar] [CrossRef]
  137. Subramaniam, N. The Process of Sprouting Angiogenesis in a Healthy Blood Vessel. 2025. Available online: https://app.biorender.com/biorender-templates/details/t-5f2aaabc52f1f600aaadc271-the-process-of-sprouting-angiogenesis-in-a-healthy-blood-vessel (accessed on 19 July 2025).
  138. Foster, N.N.; Barry, J.; Korobkova, L.; Garcia, L.; Gao, L.; Becerra, M.; Sherafat, Y.; Peng, B.; Li, X.; Choi, J.H.; et al. The mouse cortico-basal ganglia-thalamic network. Nature 2021, 598, 188–194. [Google Scholar] [CrossRef]
  139. Cruz, K.G.; Leow, Y.N.; Le, N.M.; Adam, E.; Huda, R.; Sur, M. Cortical-subcortical interactions in goal-directed behavior. Physiol. Rev. 2023, 103, 347–389. [Google Scholar] [CrossRef]
  140. Costa, R.M.; Cohen, D.; Nicolelis, M.A. Differential corticostriatal plasticity during fast and slow motor skill learning in mice. Curr. Biol. 2004, 14, 1124–1134. [Google Scholar] [CrossRef] [PubMed]
  141. Antzoulatos, E.G.; Miller, E.K. Increases in functional connectivity between prefrontal cortex and striatum during category learning. Neuron 2014, 83, 216–225. [Google Scholar] [CrossRef] [PubMed]
  142. Gittis, A.H.; Kreitzer, A.C. Striatal microcircuitry and movement disorders. Trends Neurosci. 2012, 35, 557–564. [Google Scholar] [CrossRef] [PubMed]
  143. Gunaydin, L.A.; Kreitzer, A.C. Cortico–basal ganglia circuit function in psychiatric disease. Annu. Rev. Physiol. 2016, 78, 327–350. [Google Scholar] [CrossRef]
  144. Wichmann, T.; DeLONG, M.R. Pathophysiology of Parkinson’s disease: The MPTP primate model of the human disorder. Ann. N. Y. Acad. Sci. 2003, 991, 199–213. [Google Scholar] [CrossRef]
  145. Singh, A.; Mewes, K.; Gross, R.E.; DeLong, M.R.; Obeso, J.A.; Papa, S.M. Human striatal recordings reveal abnormal discharge of projection neurons in Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2016, 113, 9629–9634. [Google Scholar] [CrossRef]
  146. Auer, T.; Schreppel, P.; Erker, T.; Schwarzer, C. Impaired chloride homeostasis in epilepsy: Molecular basis, impact on treatment, and current treatment approaches. Pharmacol. Ther. 2020, 205, 107422. [Google Scholar] [CrossRef]
  147. Grieco, S.F.; Holmes, T.C.; Xu, X. Probing neural circuit mechanisms in Alzheimer’s disease using novel technologies. Mol. Psychiatry 2023, 28, 4407–4420. [Google Scholar] [CrossRef]
  148. Calabresi, P.; Di Lazzaro, G.; Marino, G.; Campanelli, F.; Ghiglieri, V. Advances in understanding the function of alpha-synuclein: Implications for Parkinson’s disease. Brain 2023, 146, 3587–3597. [Google Scholar] [CrossRef] [PubMed]
  149. Wu, Q.; Shaikh, M.A.; Meymand, E.S.; Zhang, B.; Luk, K.C.; Trojanowski, J.Q.; Lee, V.M.Y. Neuronal activity modulates alpha-synuclein aggregation and spreading in organotypic brain slice cultures and in vivo. Acta Neuropathol. 2020, 140, 831–849. [Google Scholar] [CrossRef] [PubMed]
  150. Zablotsky, B.; Black, L.I.; Maenner, M.J.; Schieve, L.A.; Danielson, M.L.; Bitsko, R.H.; Blumberg, S.J.; Kogan, M.D.; Boyle, C.A. Prevalence and trends of developmental disabilities among children in the United States: 2009–2017. Pediatrics 2019, 144, e20190811. [Google Scholar] [CrossRef] [PubMed]
  151. Farokhniaee, A.; McIntyre, C.C. Theoretical principles of deep brain stimulation induced synaptic suppression. Brain Stimul. 2019, 12, 1402–1409. [Google Scholar] [CrossRef]
  152. Crowell, A.L.; Riva-Posse, P.; Holtzheimer, P.E.; Garlow, S.J.; Kelley, M.E.; Gross, R.E.; Denison, L.; Quinn, S.; Mayberg, H.S. Long-term outcomes of subcallosal cingulate deep brain stimulation for treatment-resistant depression. Am. J. Psychiatry 2019, 176, 949–956. [Google Scholar] [CrossRef]
  153. Shanok, N.A.; Muzac, S.; Derbin, B.; Cabeza, E.; Rodriguez, R. The effects of deep transcranial magnetic stimulation on Alzheimer’s disease: A case report examining cognitive functioning, memory, and QEEG. Neurocase 2023, 29, 81–86. [Google Scholar] [CrossRef]
  154. Xu, Z.; Duan, W.; Yuan, S.; Zhang, X.; You, C.; Yu, J.T.; Wang, J.; Li, J.D.; Deng, S.; Shu, Y. Deep brain stimulation alleviates Parkinsonian motor deficits through desynchronizing GABA release in mice. Nat. Commun. 2025, 16, 3726. [Google Scholar] [CrossRef]
  155. Binns, T.S.; Köhler, R.M.; Vanhoecke, J.; Chikermane, M.; Gerster, M.; Merk, T.; Pellegrini, F.; Busch, J.L.; Habets, J.G.; Cavallo, A.; et al. Shared pathway-specific network mechanisms of dopamine and deep brain stimulation for the treatment of Parkinson’s disease. Nat. Commun. 2025, 16, 3587. [Google Scholar] [CrossRef]
  156. Benabid, A.L. Deep brain stimulation for Parkinson’s disease. Curr. Opin. Neurobiol. 2003, 13, 696–706. [Google Scholar] [CrossRef]
  157. Alosaimi, F.; Temel, Y.; Hescham, S.; Witzig, V.S.; Almasabi, F.; Tan, S.K.H.; Jahanshahi, A. High-frequency stimulation of the subthalamic nucleus induces a sustained inhibition of serotonergic system via loss of cell phenotype. Sci. Rep. 2022, 12, 14011. [Google Scholar] [CrossRef]
  158. Mayberg, H.S.; Lozano, A.M.; Voon, V.; McNeely, H.E.; Seminowicz, D.; Hamani, C.; Schwalb, J.M.; Kennedy, S.H. Deep brain stimulation for treatment-resistant depression. Neuron 2005, 45, 651–660. [Google Scholar] [CrossRef]
  159. Portillo-Lara, R.; Tahirbegi, B.; Chapman, C.A.R.; Goding, J.A.; Green, R.A. Mind the gap: State-of-the-art technologies and applications for EEG-based brain–computer interfaces. APL Bioeng. 2021, 5, 031507. [Google Scholar] [CrossRef] [PubMed]
  160. Truccolo, W.; Donoghue, J.A.; Hochberg, L.R.; Eskandar, E.N.; Madsen, J.R.; Anderson, W.S.; Brown, E.N.; Halgren, E.; Cash, S.S. Single-neuron dynamics in human focal epilepsy. Nat. Neurosci. 2011, 14, 635–641. [Google Scholar] [CrossRef] [PubMed]
  161. Hill, N.J.; Gupta, D.; Brunner, P.; Gunduz, A.; Adamo, M.A.; Ritaccio, A.; Schalk, G. Recording human electrocorticographic (ECoG) signals for neuroscientific research and real-time functional cortical mapping. J. Vis. Exp. 2012, 64, e3993. [Google Scholar] [CrossRef]
  162. Parvizi, J.; Kastner, S. Promises and limitations of human intracranial EEG. Nat. Neurosci. 2018, 21, 474–483. [Google Scholar] [CrossRef] [PubMed]
  163. Schalk, G.; McFarland, D.J.; Hinterberger, T.; Birbaumer, N.; Wolpaw, J.R. BCI2000: A general-purpose brain-computer interface (BCI) system. IEEE Trans. Biomed. Eng. 2004, 51, 1034–1043. [Google Scholar] [CrossRef]
  164. Delorme, A.; Makeig, S. EEGLAB: An open source toolbox for analysis of single-trial EEG dynamics including independent component analysis. J. Neurosci. Methods 2004, 134, 9–21. [Google Scholar] [CrossRef]
  165. He, B.; Sohrabpour, A.; Brown, E.; Liu, Z. Electrophysiological source imaging: A noninvasive window to brain dynamics. Annu. Rev. Biomed. Eng. 2018, 20, 171–196. [Google Scholar] [CrossRef]
  166. Boto, E.; Hill, R.M.; Rea, M.; Holmes, N.; Seedat, Z.A.; Leggett, J.; Shah, V.; Osborne, J.; Bowtell, R.; Brookes, M.J. Measuring functional connectivity with wearable MEG. NeuroImage 2021, 230, 117815. [Google Scholar] [CrossRef]
  167. Jiang, R.; Tong, S.; Wu, J.; Hu, H.; Zhang, R.; Wang, H.; Zhao, Y.; Zhu, W.; Li, S.; Zhang, X. A novel EEG artifact removal algorithm based on an advanced attention mechanism. Sci. Rep. 2025, 15, 19419. [Google Scholar] [CrossRef] [PubMed]
  168. Wu, D.; Jiang, X.; Peng, R. Transfer learning for motor imagery based brain-computer interfaces: A tutorial. Neural Netw. 2022, 153, 235–253. [Google Scholar] [CrossRef] [PubMed]
  169. Hochberg, L.R.; Serruya, M.D.; Friehs, G.M.; Mukand, J.A.; Saleh, M.; Caplan, A.H.; Branner, A.; Chen, D.; Penn, R.D.; Donoghue, J.P. Neuronal ensemble control of prosthetic devices by a human with tetraplegia. Nature 2006, 442, 164–171. [Google Scholar] [CrossRef] [PubMed]
  170. Benabid, A.L.; Costecalde, T.; Eliseyev, A.; Charvet, G.; Verney, A.; Karakas, S.; Foerster, M.; Lambert, A.; Morinière, B.; Abroug, N.; et al. An exoskeleton controlled by an epidural wireless brain–machine interface in a tetraplegic patient: A proof-of-concept demonstration. Lancet Neurol. 2019, 18, 1112–1122. [Google Scholar] [CrossRef]
  171. Baillet, S. Magnetoencephalography for brain electrophysiology and imaging. Nat. Neurosci. 2017, 20, 327–339. [Google Scholar] [CrossRef]
  172. Boto, E.; Holmes, N.; Leggett, J.; Roberts, G.; Shah, V.; Meyer, S.S.; Muñoz, L.D.; Mullinger, K.J.; Tierney, T.M.; Bestmann, S.; et al. Moving magnetoencephalography towards real-world applications with a wearable system. Nature 2018, 555, 657–661. [Google Scholar] [CrossRef]
  173. Qi, Z.; Liu, H.; Jin, F.; Wang, Y.; Lu, X.; Liu, L.; Yang, Z.; Fan, L.; Song, M.; Zuo, N.; et al. A wearable repetitive transcranial magnetic stimulation device. Nat. Commun. 2025, 16, 2731. [Google Scholar] [CrossRef]
  174. Li, J.; Chen, G.; Li, G.; Xiao, L.; Jia, R.; Zhang, K. Flexible brain electronic sensors advance wearable brain-computer interface. npj Biomed. Innov. 2025, 2, 24. [Google Scholar] [CrossRef]
  175. Yan, P.; Acker, C.D.; Zhou, W.L.; Lee, P.; Bollensdorff, C.; Negrean, A.; Lotti, J.; Sacconi, L.; Antic, S.D.; Kohl, P.; et al. Palette of fluorinated voltage-sensitive hemicyanine dyes. Proc. Natl. Acad. Sci. USA 2012, 109, 20443–20448. [Google Scholar] [CrossRef]
  176. Grienberger, C.; Konnerth, A. Imaging calcium in neurons. Neuron 2012, 73, 862–885. [Google Scholar] [CrossRef]
  177. He, C.X.; Arroyo, E.D.; Cantu, D.A.; Goel, A.; Portera-Cailliau, C. A versatile method for viral transfection of calcium indicators in the neonatal mouse brain. Front. Neural Circuits 2018, 12, 56. [Google Scholar] [CrossRef]
  178. Park, J.H.; Eom, Y.S.; Kim, T.H. Recent advances in aptamer-based sensors for sensitive detection of neurotransmitters. Biosensors 2023, 13, 413. [Google Scholar] [CrossRef]
  179. Clay, M.; Monbouquette, H.G. A detailed model of electroenzymatic glutamate biosensors to aid in sensor optimization and in applications in vivo. ACS Chem. Neurosci. 2018, 9, 241–251. [Google Scholar] [CrossRef] [PubMed]
  180. Zhao, T.; Wang, J.W.; Zhang, H.S.; Zheng, X.; Chen, Y.P.; Tang, H.; Jiang, J.H. Development of dual-nanopore biosensors for detection of intracellular dopamine and dopamine efflux from single PC12 cell. Anal. Chem. 2022, 94, 15541–15545. [Google Scholar] [CrossRef] [PubMed]
  181. Biswas, A.; Lee, S.; Cencillo-Abad, P.; Karmakar, M.; Patel, J.; Soudi, M.; Chanda, D. Nanoplasmonic aptasensor for sensitive, selective, and real-time detection of dopamine from unprocessed whole blood. Sci. Adv. 2024, 10, eadp7460. [Google Scholar] [CrossRef] [PubMed]
  182. Buchanan, C.R.; Pernet, C.R.; Gorgolewski, K.J.; Storkey, A.J.; Bastin, M.E. Test-retest reliability of structural brain networks from diffusion MRI. Neuroimage 2014, 86, 231–243. [Google Scholar] [CrossRef] [PubMed]
  183. Tufail, Y.; Matyushov, A.; Baldwin, N.; Tauchmann, M.L.; Georges, J.; Yoshihiro, A.; Tillery, S.I.H.; Tyler, W.J. Transcranial pulsed ultrasound stimulates intact brain circuits. Neuron 2010, 66, 681–694. [Google Scholar] [CrossRef]
  184. Watarai, A.; Tao, K.; Wang, M.Y.; Okuyama, T. Distinct functions of ventral CA1 and dorsal CA2 in social memory. Curr. Opin. Neurobiol. 2021, 68, 29–35. [Google Scholar] [CrossRef]
  185. Deffieux, T.; Demené, C.; Tanter, M. Functional ultrasound imaging: A new imaging modality for neuroscience. Neuroscience 2021, 474, 110–121. [Google Scholar] [CrossRef]
  186. Zhang, Y.; Chen, H.; Long, X.; Xu, T. Three-dimensional-engineered bioprinted in vitro human neural stem cell self-assembling culture model constructs of Alzheimer’s disease. Bioact. Mater. 2022, 11, 192–205. [Google Scholar] [CrossRef]
  187. Zhao, R.; Hu, W.; Tsai, J.; Lee, M.J.; LaMarca, E.A.; Frosch, M.P.; Hyman, B.T.; Liu, C.C.; Bu, G.; Spires-Jones, T.L.; et al. Microglia limit the expansion of β-amyloid plaques in a mouse model of Alzheimer’s disease. Mol. Neurodegener. 2017, 12, 47. [Google Scholar] [CrossRef]
  188. Bubnys, A.; Tsai, L.H. Harnessing cerebral organoids for Alzheimer’s disease research. Curr. Opin. Neurobiol. 2022, 72, 120–130. [Google Scholar] [CrossRef]
  189. Vanova, T.; Sedmik, J.; Raska, J.; Amruz Cerna, K.; Taus, P.; Pospisilova, V.; Nezvedova, M.; Fedorova, V.; Kadakova, S.; Klimova, H.; et al. Cerebral organoids derived from patients with Alzheimer’s disease with PSEN1/2 mutations have defective tissue patterning and altered development. Cell Rep. 2023, 42, 113310. [Google Scholar] [CrossRef] [PubMed]
  190. Smits, L.M.; Schwamborn, J.C. Midbrain organoids: A new tool to investigate Parkinson’s Disease. Front. Cell Dev. Biol. 2020, 8, 359. [Google Scholar] [CrossRef] [PubMed]
  191. Sozzi, E.; Nilsson, F.; Kajtez, J.; Parmar, M.; Fiorenzano, A. Generation of human ventral midbrain organoids derived from pluripotent stem cells. Curr. Protoc. 2022, 2, e555. [Google Scholar] [CrossRef] [PubMed]
  192. Kim, H.; Park, H.J.; Choi, H.; Chang, Y.; Park, H.; Shin, J.; Kim, J.; Lengner, C.J.; Lee, Y.K.; Kim, J. Modeling G2019S-LRRK2 sporadic Parkinson’s Disease in 3D midbrain organoids. Stem Cell Rep. 2019, 12, 518–531. [Google Scholar] [CrossRef]
  193. Smits, L.M.; Reinhardt, L.; Reinhardt, P.; Glatza, M.; Monzel, A.S.; Stanslowsky, N.; Rosato Siri, M.D.; Zanon, A.; Antony, P.M.; Bellmann, J.; et al. Modeling Parkinson’s disease in midbrain-like organoids. npj Park. Dis. 2019, 5, 5. [Google Scholar] [CrossRef]
  194. Studer, L. Derivation of dopaminergic neurons from pluripotent stem cells. Prog. Brain Res. 2012, 200, 243–263. [Google Scholar] [CrossRef]
  195. Harischandra, D.S.; Rokad, D.; Ghaisas, S.; Verma, S.; Robertson, A.; Jin, H.g.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Enhanced differentiation of human dopaminergic neuronal cell model for preclinical translational research in Parkinson’s disease. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2020, 1866, 165533. [Google Scholar] [CrossRef]
  196. Fu, C.L.; Jiang, X.; Dong, B.C.; Li, D.; She, X.Y.; Yao, J. Protocol for transplantation of cells derived from human midbrain organoids into a Parkinson’s disease mouse model to restore motor function. STAR Protoc. 2024, 5, 103251. [Google Scholar] [CrossRef]
  197. Fu, C.L.; Dong, B.C.; Jiang, X.; Li, D.; Yao, J. A cell therapy approach based on iPSC-derived midbrain organoids for the restoration of motor function in a Parkinson’s disease mouse model. Heliyon 2024, 10, e24234. [Google Scholar] [CrossRef]
  198. Paşca, A.M.; Sloan, S.A.; Clarke, L.E.; Tian, Y.; Makinson, C.D.; Huber, N.; Kim, C.N.; Park, J.Y.; O’Rourke, N.A.; Nguyen, K.D.; et al. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat. Methods 2015, 12, 671–678. [Google Scholar] [CrossRef] [PubMed]
  199. Rickner, H.D.; Jiang, L.; Hong, R.; Sun, W.; Mehta, B.K.; Paul, S.; Li, Q.; Xu, Y.; Zhang, Y.; Spina, S.; et al. Single cell transcriptomic profiling of a neuron-astrocyte assembloid tauopathy model. Nat. Commun. 2022, 13, 6275. [Google Scholar] [CrossRef] [PubMed]
  200. Ye, B. Approaches to vascularizing human brain organoids. PLoS Biol. 2023, 21, e3002141. [Google Scholar] [CrossRef] [PubMed]
  201. Ji, Y.; Chen, X.; Wang, Z.; Liu, M.; Zhao, F.; Xu, L.; Yang, R.; Li, J.; Zhang, W.; Wu, T.; et al. Alzheimer’s disease patient brain extracts induce multiple pathologies in novel vascularized neuroimmune organoids for disease modeling and drug discovery. Mol. Psychiatry 2025, 30, 4558–4575. [Google Scholar] [CrossRef]
  202. Bagley, J.A.; Reumann, D.; Bian, S.; Lévi-Strauss, J.; Knoblich, J.A. Fused cerebral organoids model interactions between brain regions. Nat. Methods 2017, 14, 743–751. [Google Scholar] [CrossRef]
  203. Zourray, C.; Kurian, M.A.; Barral, S.; Lignani, G. Electrophysiological Properties of Human Cortical Organoids: Current State of the Art and Future Directions. Front. Mol. Neurosci. 2022, 15, 839366. [Google Scholar] [CrossRef]
  204. Reumann, D.; Krauditsch, C.; Novatchkova, M.; Elling, U.; Ortner, J.; Lengerke, C.; Knoblich, J.A.; Bolognin, S.; Schwamborn, J.C. In vitro modeling of the human dopaminergic system using spatially arranged ventral midbrain–striatum–cortex assembloids. Nat. Methods 2023, 20, 2034–2047. [Google Scholar] [CrossRef]
  205. Khuram, H.; Maddox, P.; Elahi, A.; Hirani, R.; Issani, A. Human cerebral organoids: Implications of ontological considerations. AJOB Neurosci. 2023, 14, 213–214. [Google Scholar] [CrossRef]
  206. Zhou, X.; Davis, E.P.; Tsai, L.H. APOE4 exacerbates pathology in iPSC-derived cerebral organoids. Nat. Commun. 2020, 11, 1216. [Google Scholar] [CrossRef]
  207. Rosety, I.; Zagare, A.; Saraiva, C.; Nickels, S.; Antony, P.; Almeida, C.; Glaab, E.; Halder, R.; Velychko, S.; Rauen, T.; et al. Impaired neuron differentiation in GBA-associated Parkinson’s disease is linked to cell cycle defects in organoids. npj Park. Dis. 2023, 9, 166. [Google Scholar] [CrossRef] [PubMed]
  208. Zheng, X.; Han, D.; Liu, W.; Wang, X.; Pan, N.; Wang, Y.; Chen, Z. Human iPSC-derived midbrain organoids functionally integrate into striatum circuits and restore motor function in a mouse model of Parkinson’s disease. Theranostics 2023, 13, 2673–2692. [Google Scholar] [CrossRef]
  209. Huang, Q.; Tang, B.; Romero, J.C.; Yang, Y.; Elsayed, S.K.; Pahapale, G.; Lee, T.J.; Morales Pantoja, I.E.; Han, F.; Berlinicke, C.; et al. Shell microelectrode arrays (MEAs) for brain organoids. Sci. Adv. 2022, 8, eabq5031. [Google Scholar] [CrossRef] [PubMed]
  210. Sakaguchi, H.; Kadoshima, T.; Soen, M.; Narii, N.; Ishida, Y.; Ohgushi, M.; Takahashi, J.; Eiraku, M.; Sasai, Y. Generation of functional hippocampal neurons from self-organizing human embryonic stem cell-derived dorsomedial telencephalic tissue. Nat. Commun. 2015, 6, 8896. [Google Scholar] [CrossRef] [PubMed]
  211. Pomeshchik, Y.; Klementieva, O.; Gil, J.; Martinsson, I.; Hansen, M.G.; de Vries, T.; Sancho-Balsells, A.; Russ, K.; Savchenko, E.; Collin, A.; et al. Human iPSC-derived hippocampal spheroids: An innovative tool for stratifying Alzheimer Disease patient-specific cellular phenotypes and developing therapies. Stem Cell Rep. 2020, 15, 256–273. [Google Scholar] [CrossRef]
  212. Nishimura, H.; Li, Y. Human pluripotent stem cell-derived models of the hippocampus. Int. J. Biochem. Cell Biol. 2024, 177, 106695. [Google Scholar] [CrossRef]
  213. Zander, T.O.; Kothe, C. Towards passive brain–computer interfaces: Applying brain–computer interface technology to human–machine systems in general. J. Neural Eng. 2011, 8, 025005. [Google Scholar] [CrossRef]
  214. Fairclough, S. Neuroadaptive technology and the self: A postphenomenological perspective. Philos. Technol. 2023, 36, 30. [Google Scholar] [CrossRef]
  215. Zander, T.O.; Krol, L.R.; Birbaumer, N.P.; Gramann, K. Neuroadaptive technology enables implicit cursor control based on medial prefrontal cortex activity. Proc. Natl. Acad. Sci. USA 2016, 113, 14898–14903. [Google Scholar] [CrossRef]
  216. Dehais, F.; Ladouce, S.; Darmet, L.; Nong, T.V.; Ferraro, G.; Tresols, J.T.; Velut, S.; Labedan, P. Dual passive reactive brain-computer interface: A novel approach to human-machine symbiosis. Front. Neuroergon. 2023, 3, 824780. [Google Scholar] [CrossRef]
  217. Lavazza, A.; Chinaia, A.A. Human cerebral organoids: The ethical stance of scientists. Stem Cell Res. Ther. 2023, 14, 59. [Google Scholar] [CrossRef]
  218. Jeziorski, J.; Brandt, R.; Evans, J.H.; Campana, W.; Kalichman, M.; Thompson, E.; Goldstein, L.; Koch, C.; Muotri, A.R. Brain organoids, consciousness, ethics and moral status. Semin. Cell Dev. Biol. 2023, 144, 97–102. [Google Scholar] [CrossRef]
  219. Ethics need to keep up with human brain organoid research. Nat. Rev. Bioeng. 2024, 2, 711. [CrossRef]
  220. de Jongh, D.; Massey, E.K.; Berishvili, E.; Fonseca, L.M.; Lebreton, F.; Bellofatto, K.; Bignard, J.; Seissler, J.; Buerck, L.W.v.; Honarpisheh, M.; et al. Organoids: A systematic review of ethical issues. Stem Cell Res. Ther. 2022, 13, 337. [Google Scholar] [CrossRef] [PubMed]
  221. Mogilenko, D.A.; Haas, J.T.; L’homme, L.; Fleury, S.; Quemener, S.; Levavasseur, M.; Becquart, C.; Wartelle, J.; Bogomolova, A.; Pineau, L.; et al. Metabolic and innate immune cues merge into a specific inflammatory response via the UPR. Cell 2019, 177, 1201–1216. [Google Scholar] [CrossRef] [PubMed]
  222. Wang, X.; Liu, Y.; Li, X.; Zhang, Z.; Yang, H.; Zhang, Y.; Williams, P.R.; Alwahab, N.S.; Kapur, K.; Yu, B.; et al. Deconstruction of corticospinal circuits for goal-directed motor skills. Cell 2017, 171, 440–455. [Google Scholar] [CrossRef] [PubMed]
  223. Baker, M. Blame it on the antibodies. Nature 2015, 521, 274. [Google Scholar] [CrossRef]
  224. Mera, P.; Ferron, M.; Mosialou, I. Regulation of energy metabolism by bone-derived hormones. Cold Spring Harb. Perspect. Med. 2018, 8, a031666. [Google Scholar] [CrossRef]
  225. Peng, W.; Datta, P.; Wu, Y.; Dey, M.; Ayan, B.; Dababneh, A.; Ozbolat, I.T. Challenges in bio-fabrication of organoid cultures. In Cell Biology and Translational Medicine, Volume 3; Springer: Cham, Switzerland, 2018; pp. 53–71. [Google Scholar]
  226. Lukonin, I.; Zinner, M.; Liberali, P. Organoids in image-based phenotypic chemical screens. Exp. Mol. Med. 2021, 53, 1495–1502. [Google Scholar] [CrossRef]
  227. Lavazza, A.; Massimini, M. Cerebral organoids: Ethical issues and consciousness assessment. J. Med. Ethics 2018, 44, 606–610. [Google Scholar] [CrossRef]
  228. Yadav, A.; Seth, B.; Chaturvedi, R.K. Brain organoids: Tiny mirrors of human neurodevelopment and neurological disorders. Neuroscientist 2021, 27, 388–426. [Google Scholar] [CrossRef] [PubMed]
  229. Tremblay, G.R.; Oonk, J.R.; Combes, F.; Salomé, P.; O’Dea, C.P.; Baum, S.A.; Voit, G.M.; Donahue, M.; McNamara, B.R.; Davis, T.A.; et al. Cold, clumpy accretion onto an active supermassive black hole. Nature 2016, 534, 218–221. [Google Scholar] [CrossRef] [PubMed]
  230. National Institutes of Health. NIH Establishes Nation’s First Dedicated Organoid Development Center to Reduce Reliance on Animal Modeling. 2025. Available online: https://www.nih.gov/news-events/news-releases/nih-establishes-nations-first-dedicated-organoid-development-center-reduce-reliance-animal-modeling (accessed on 10 October 2025).
  231. Fekete, Z.; Zátonyi, A.; Kaszás, A.; Madarász, M.; Slézia, A. Transparent neural interfaces: Challenges and solutions of microengineered multimodal implants designed to measure intact neuronal populations using high-resolution electrophysiology and microscopy simultaneously. Microsyst. Nanoeng. 2023, 9, 66. [Google Scholar] [CrossRef] [PubMed]
  232. Li, H.; Wang, J.; Fang, Y. Recent developments in multifunctional neural probes for simultaneous neural recording and modulation. Microsyst. Nanoeng. 2023, 9, 4. [Google Scholar] [CrossRef]
  233. Topalovic, U.; Barclay, S.; Ling, C.; Alzuhair, A.; Yu, W.; Hokhikyan, V.; Chandrakumar, H.; Rozgic, D.; Jiang, W.; Basir-Kazeruni, S.; et al. A wearable platform for closed-loop stimulation and recording of single-neuron and local field potential activity in freely moving humans. Nat. Neurosci. 2023, 26, 517–527. [Google Scholar] [CrossRef]
  234. Urai, A.E.; Doiron, B.; Leifer, A.M.; Churchland, A.K. Large-scale neural recordings call for new insights to link brain and behavior. Nat. Neurosci. 2022, 25, 11–19. [Google Scholar] [CrossRef]
  235. Sejnowski, T.J.; Churchland, P.S.; Movshon, J.A. Putting big data to good use in neuroscience. Nat. Neurosci. 2014, 17, 1440–1441. [Google Scholar] [CrossRef]
  236. Yuste, R.; Goering, S.; Agüera y Arcas, B.; Bi, G.; Carmena, J.M.; Carter, A.; Fins, J.J.; Friesen, P.; Gallant, J.; Huggins, J.E.; et al. Four ethical priorities for neurotechnologies and AI. Nature 2017, 551, 159–163. [Google Scholar] [CrossRef]
  237. Kataoka, M.; Niikawa, T.; Nagaishi, N.; Lee, T.L.; Erler, A.; Savulescu, J.; Sawai, T. Beyond consciousness: Ethical, legal, and social issues in human brain organoid research and application. Eur. J. Cell Biol. 2025, 104, 151470. [Google Scholar] [CrossRef]
  238. Vishwarupe, V.; Joshi, P.M.; Mathias, N.; Maheshwari, S.; Mhaisalkar, S.; Pawar, V. Explainable AI and Interpretable Machine Learning: A Case Study in Perspective. Procedia Comput. Sci. 2022, 204, 869–876. [Google Scholar] [CrossRef]
  239. Tyler, W.J.; Tufail, Y.; Finsterwald, M.; Tauchmann, M.L.; Olson, E.J.; Majestic, C. Ultrasound modulates ion channel currents. Nat. Neurosci. 2011, 11, 491–498. [Google Scholar] [CrossRef]
  240. Deng, Y.; Liu, H.; Zhang, W.; Wang, S. AI-enhanced multiphysics modeling for targeted ultrasound neurostimulation. Adv. Sci. 2024, 11, 2401667. [Google Scholar] [CrossRef]
Figure 2. Schematic illustration of brain–organoid model–machine interface.
Figure 2. Schematic illustration of brain–organoid model–machine interface.
Applsci 15 11316 g002
Table 1. Comparative overview of AI applications in early diagnosis of Alzheimer’s disease (AD) and Parkinson’s disease (PD).
Table 1. Comparative overview of AI applications in early diagnosis of Alzheimer’s disease (AD) and Parkinson’s disease (PD).
AI ParadigmKey TechniquesApplications in ADApplications in PDReferences
Machine Learning (ML)Supervised (SVM, RF, LR); unsupervised (clustering, PCA); ensemble (bagging, boosting)AD classification, MCI-to-AD conversion, subtype identification, feature extractionPD classification, early-stage detection, differential diagnosis, biomarker selection[11,52,53,54,55]
Deep Learning (DL)DNN, CNN, RNN, autoencoders, DBN, MLP, GRU, GAN, GNNAD/MCI identification, progression modeling, raw neuroimaging feature extraction, end-to-end classificationPD stage prediction, progression modeling, multimodal fusion, vocal biomarkers[15,56,57,58,59,60,61]
Hybrid ModelsCNN-BLSTM; AlexNet+SVM; ResNet-50+SVM; BiLSTM-ANNEarly dementia and AD detection/classificationPD detection via time-series classification, patient stratification, clinical-imaging fusion[62,63,64]
Datasets UsedMultimodal MRI and PET, TADPOLE datasetsStructural MRI, DTI, DaTSCAN, rs-fMRI, UCI voice datasets[65,66,67,68,69,70,71,72]
Table 2. Invasiveness-based classification of neural interfaces in AD and PD applications.
Table 2. Invasiveness-based classification of neural interfaces in AD and PD applications.
NI TypeExample TechnologiesDescriptionApplication in AD/PDProsConsSources
Invasive NIsUtah Array, Neuralink, depth electrodesElectrodes implanted directly in the brain/neural tissueUsed in deep brain stimulation (DBS) for PDHigh-resolution, real-time brain signal accessSurgical risk, infection, long-term safety concerns, privacy, autonomy issues[5,41,156,157,158]
Partially invasive NIsECoG grids, NeuroPace RNSElectrodes beneath the skull but above the cortexExperimental cognitive monitoring; seizure controlBetter signal than EEG; less invasive than depth implantsRequires craniotomy; device longevity and post-surgical concerns[159,160,161,162]
Non-invasive NIsEEG caps, fNIRS headsets, OpenBCI, Muse, EmotivScalp- or skin-surface sensors for brain activityCognitive and motor symptom tracking in AD/PDSafe, affordable, widely available; home monitoring possibleLower spatial resolution, poor depth sensing, data privacy, and algorithmic bias risks[17,163,164,165,166,167]
Sensorimotor BCIsBrainGate, Neurable, Neurocontrol ExoskeletonsDecode motor intent for control of devices or communicationAssistive devices for PD mobility or AD communicationEnables motor recovery or intent-based interactionCognitively demanding; limited generalization in AD; risk of over-dependence[4,30,163,168,169,170]
Table 3. Signal-based classification of neural interfaces in AD and PD applications.
Table 3. Signal-based classification of neural interfaces in AD and PD applications.
Signal TypeExample TechnologiesDescriptionApplication in AD/PDProsConsSources
Electrical (spikes, LFPs)Utah Array, Neuralink, OpenBCI, ECoG, EEGMeasures electrical activity from neurons or populationsDBS, cognitive load monitoring, movement decoding in PDHigh temporal resolution, real-time controlSpatial resolution varies; noise-prone (e.g., EEG)[95,163,169]
Magnetic (MEG, OPM)MEG, OPMs, SQUID arraysDetects magnetic fields from neural activity without scalp contactCognitive monitoring, early detection of dementia-related oscillatory changesHigh temporal resolution, better source localization than EEGExpensive, bulky, limited portability[171,172,173,174]
Optical (calcium, voltage)Two-photon imaging, GCaMP, fNIRS, miniScopeLight-based indicators (e.g., calcium or hemoglobin changes)Functional connectivity, early neurodegeneration detectionCell-type specificity, good spatial resolutionSlower temporal dynamics, bulky equipment[175,176,177]
Biochemical (neurotransmitters)Neurochemical biosensors, aptamer-based electrodesDetects levels of dopamine, glutamate, or other molecules in real timeTracking dopamine loss in PD, identifying stress/metabolic biomarkersHigh specificity to neurotransmittersComplex fabrication, less mature than others[178,179,180,181]
Multimodal (hybrid)Neuralink, BrainCo, Kernel Flow, NeuroNexusCombines electrical, optical, and metabolic signals in one platformComprehensive neurobehavioral assessmentEnables richer brain-state decodingPower-hungry, complex data fusion needed[95,104,182]
Ultrasound-based NIsClarity, Neural-FUS, fUSiUltrasound waves used for imaging (fUS) or stimulation (tFUS)Non-invasive brain stimulation (e.g., PD) and vascular/neural imaging in AD/PD researchDeep-brain access non-invasively; high spatial resolution; diagnostic and therapeutic potentialRequires precise targeting and safety validation; limited widespread availability[183,184,185]
Table 4. Summary of brain-specific organoid models in AD and PD research.
Table 4. Summary of brain-specific organoid models in AD and PD research.
Organoid ModelDescriptionApplication in AD/PDProsConsEthical IssuesSources
Cerebral organoidsMimic cortex and hippocampus for AD researchStudy amyloid-beta, tau pathology (AD)Human-relevant biology; spontaneous activityLack of vasculature and immune cellsConsciousness concerns in long-term culture[23,24,28,186,187,188,189]
Midbrain organoidsModel dopaminergic neurons for PD researchDopamine neuron loss and synaptic stress modeling (PD)Recapitulate substantia nigra-like featuresImmature and variable differentiationGenetic manipulation implications[190,191,192,193,194,195,196,197]
Forebrain assembloidsFusion of forebrain and interneuron lineagesNetwork formation defects and migration studies in ADMultiregional integration supports interneuron flowComplex fabrication; reproducibility issuesPotential for sentience-like behavior[77,198,199]
Vascularized brain organoidsOrganoids embedded with vascular scaffoldsImproves AD drug diffusion, nutrient exchange modelingSupports longer-term, functional neural developmentStill under optimization; cost-intensiveTransplantation ethical risks[129,200,201]
Fused cortico-subpallial organoidsRegion-specific fused units (cortex + GABAergic)Synaptic integration disruption analysis in ADEnable synaptic-level interaction modelingRequires precise fusion protocolsMoral status of fused organoids[81,202,203,204]
Patient-derived iPSC organoidsOrganoids from AD/PD patient stem cellsModel genetic variants in familial AD/PDPersonalized pathology, pharmacogenomic testingBatch variability, long differentiation timeData ownership, reprogramming rights[205,206,207,208]
Integrated NI–organoid platformsElectrode-organoid integration for recording/stimulationNeurodegeneration drug screening, neuron activity trackingFunctional testing; closed-loop experimentsStill preclinical; signal interpretation challengesNeural data privacy, AI misuse[25,28,80,197,209]
Hippocampal organoidsRegion-specific organoids mimicking the hippocampusMemory circuit modeling, tauopathy, neurogenesis studies (AD)Recapitulate CA- and DG-like structures; relevant to memory loss in ADDifferentiation protocols are complex; regional maturity may varyMemory and cognition modeling raise sensitivity around sentience[25,210,211,212]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Usman, M.; Ashebir, S.; Okey-Mbata, C.; Yun, Y.; Kim, S. Neuroengineering Frontiers: A Selective Review of Neural Interfaces, Brain–Machine Interactions, and Artificial Intelligence in Neurodegenerative Diseases. Appl. Sci. 2025, 15, 11316. https://doi.org/10.3390/app152111316

AMA Style

Usman M, Ashebir S, Okey-Mbata C, Yun Y, Kim S. Neuroengineering Frontiers: A Selective Review of Neural Interfaces, Brain–Machine Interactions, and Artificial Intelligence in Neurodegenerative Diseases. Applied Sciences. 2025; 15(21):11316. https://doi.org/10.3390/app152111316

Chicago/Turabian Style

Usman, Mutiyat, Simachew Ashebir, Chioma Okey-Mbata, Yeoheung Yun, and Seongtae Kim. 2025. "Neuroengineering Frontiers: A Selective Review of Neural Interfaces, Brain–Machine Interactions, and Artificial Intelligence in Neurodegenerative Diseases" Applied Sciences 15, no. 21: 11316. https://doi.org/10.3390/app152111316

APA Style

Usman, M., Ashebir, S., Okey-Mbata, C., Yun, Y., & Kim, S. (2025). Neuroengineering Frontiers: A Selective Review of Neural Interfaces, Brain–Machine Interactions, and Artificial Intelligence in Neurodegenerative Diseases. Applied Sciences, 15(21), 11316. https://doi.org/10.3390/app152111316

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop