Next Article in Journal
Kinematics of the Tennis Serve Using an Optoelectronic Motion Capture System: Are There Correlations between Joint Angles and Racket Velocity?
Previous Article in Journal
Research on an SICM Scanning Image Resolution Enhancement Algorithm
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancements in Cerebrospinal Fluid Biosensors: Bridging the Gap from Early Diagnosis to the Detection of Rare Diseases

by
Ghazal Hatami-Fard
* and
Salzitsa Anastasova-Ivanova
*
The Hamlyn Centre, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
*
Authors to whom correspondence should be addressed.
Sensors 2024, 24(11), 3294; https://doi.org/10.3390/s24113294
Submission received: 23 April 2024 / Revised: 13 May 2024 / Accepted: 17 May 2024 / Published: 22 May 2024

Abstract

:
Cerebrospinal fluid (CSF) is a body fluid that can be used for the diagnosis of various diseases. However, CSF collection requires an invasive and painful procedure called a lumbar puncture (LP). This procedure is applied to any patient with a known risk of central nervous system (CNS) damage or neurodegenerative disease, regardless of their age range. Hence, this can be a very painful procedure, especially in infants and elderly patients. On the other hand, the detection of disease biomarkers in CSF makes diagnoses as accurate as possible. This review aims to explore novel electrochemical biosensing platforms that have impacted biomedical science. Biosensors have emerged as techniques to accelerate the detection of known biomarkers in body fluids such as CSF. Biosensors can be designed and modified in various ways and shapes according to their ultimate applications to detect and quantify biomarkers of interest. This process can also significantly influence the detection and diagnosis of CSF. Hence, it is important to understand the role of this technology in the rapidly progressing field of biomedical science.

1. Introduction

Examination of the cerebrospinal fluid (CSF) is an important diagnostic tool for numerous cancers, immunological disorders, and neurodegenerative diseases [1,2,3]. Analysis of CSF dates back to 1764 [4], and further detection of neurodegenerative diseases from CSF gained attention in the 1990s [5,6]. Since then, investigations of CSF analysis for the early diagnosis of various neurological conditions and malignancies have gained significant importance. The investigation of the components and quality of CSF is similar to blood analysis tests for the neurological system [1]. Since the nervous system lacks lymphatic drainage, the CSF acts as the lymphatic system of the brain and nervous system [7]. Hence, it is responsible for the transport of neurotransmitters and toxins from the central nervous system (CNS). However, CSF analysis is limited to the detection of specific individual biomarkers [8]. Since CSF conditions are relatively stable, any deviations in its content from the normal range can be a warning for neurological disease. The low concentrations (trace level) of a few biomarkers limit the detection of analytes by analytical laboratory tools [5] in the early stages of diseases. Laboratory analytical tools, such as mass spectroscopy [9], ELISA [10], Western blotting [11], and HPLC [12], are well-known techniques for analyte detection. However, these techniques require time for sample preparation and pretreatment, in addition to being expensive. Hence, the most important challenges are fast detection, high accuracy, and increased sensitivity for in situ analyte detection in CSF. The enzyme-linked immunosorbent assay (ELISA) is a widely applied method in clinics and hospitals across the world for accurate detection and quantification of biological agents, mainly proteins and polypeptides [13]. It offers a wide range of detection; is linear, quantifiable, and easy to use; and provides multiple output signals with different fluorophores. However, it may not be as sensitive as chemiluminescence and film, and the cost of an imager can be a disadvantage [13]. Western blotting is a technique known for its sensitivity. It can detect as little as 0.1 nanograms of protein in a sample, making it an effective early diagnostic tool [14]. It offers a wide range of detection; is linear, quantifiable, and easy to use; and provides multiple output signals with different fluorophores. However, it may not be as sensitive as chemiluminescence and film, and the cost of an imager can be a disadvantage. High-Performance Liquid Chromatography (HPLC) is a technique used for separating, identifying, and quantifying each component in a mixture of compounds [15]. It is widely used in biochemical fields for the detection of small molecules like metabolites, drugs, and biomarkers in body fluids. HPLC offers high resolutions, sensitivity, and precision, and it allows for the analysis of a wide range of samples. However, despite its advantages, HPLC can be costly, requiring large quantities of expensive organics. Techniques such as solid-phase extraction and capillary electrophoresis can be cheaper and even quicker, especially for analysis under good manufacturing practice. Poor resolutions and inconsistent recoveries are notable drawbacks of this method when crude unprocessed samples are used.
However, the specifics of its advantages and disadvantages in detecting biomarkers in CSF are not well documented and may require further research.
This review article aims to investigate the recent improvements in non-invasive, point-of-care, CSF-sensing, and monitoring systems and highlights challenges and future perspectives.

1.1. CSF’s Formation, Mechanism, and Function in the Body

Ependymal cells present in the choroid plexus (CP), which line the floors of the lateral ventricles and the roofs of the third and fourth ventricles of the brain, are predominantly responsible for CSF production [16]. Volumes of 0.2–0.7 mL/minute of CSF are generated, mostly within the lateral ventricles of the brain. About 400–600 mL of CSF is generated approximately four times a day [17]. CSF is a colourless, acellular, low-protein fluid [1]. It contains approximately 60–80% glucose, 22–38 mg/dL protein, and less than five mononuclear cells per mm3 in healthy adults [18]. CSF is a part of the extracellular fluid that contains various small molecules and proteins. CSF also plays a crucial role in cerebral blood flow regulation [19]. Carrying essential ions, CSF maintains the healthy functioning of the central nervous system. Cerebrospinal fluid is a reason for the buoyancy of the brain and reduces its weight to 25–50 g from its actual 1000–1500 g weight [20]. It also acts as a barrier to protect the brain and spinal cord from sudden shocks and damage [21]. Furthermore, toxins produced in the brain are transported to the bloodstream via CSF. The exchange of small and large molecules between the blood and CSF is controlled by the blood–CSF barrier [22]. It is also involved in nutrient exchange between the blood and nerves and provides a crucial environment for neuronal signalling. Hence, any long-term disruption of this regulation may result in a malicious disease [23]. CSF is a crucial part of the Monro–Kellie doctrine, where raised intracranial pressure is detected by an increase in CSF pressure [24]. Figure 1 shows CSF collection and analysis procedures.

1.2. CSF Collection and Challenges

To diagnose any disorder in the central nervous system through CSF screening, a patient sample should be collected and analysed. CSF sampling has been performed via needle biopsy or lumbar puncture (LP) for many years [25]. LP is an invasive procedure that enables CSF sampling in the spinal subarachnoid space. Depending on the sampling site, the components of CSF, such as plasma proteins, may have varied analyte concentrations [26].
In the early 1890s, LP was introduced as an intracranial pressure release treatment for meningitis [27]. However, this procedure has been predominantly used as a diagnostic tool [28,29]. Lumbar puncture has been used for CSF sampling and anaesthesia for years, but it has certain risks and leads to post-procedural complications. Complications caused by or through LP can occur immediately or after the procedure. One of the most common symptoms was reported to be a long-lasting severe headache after LP. This reported morbidity could result in fatality if not treated [30]. As lumbar puncture is performed via needles of different sizes, according to research investigations, a smaller needle can significantly decrease the risk of post-lumbar puncture headaches [31]. However, as the needle penetrates the subarachnoid space, it can act as a vehicle for microorganisms to cause local infections. This invasive sampling and screening method can result in inflammation or infection of the patient’s body [32]. The risk of infection increases if the patient has skin lesions or dermatological conditions, such as psoriasis. This is mostly due to the risk of S. aureus infection in patients with psoriasis and other dermatological conditions [33]. Increased risk of haemorrhage leading to a spinal haematoma and the potential for brain herniation due to raised intracranial pressure are also known to be possible complications of this procedure [32]. The overall risk of complications after lumbar puncture can be more severe in infants [34]. Despite these risks and complications during LP, it remains the primary method for anaesthesia and CSF sampling [25,35].
Given that LP remains the chief method used to study CNS disorders, diagnostic tools have been developed to better analyse CSF [36]. Furthermore, the rapid development of novel non-invasive biomedical devices has allowed the properties of CSF to be widely studied in recent decades [37].

1.3. Changes in CSF Associated with Various Diseases

As mentioned above, the footprint of any CNS (central nervous system)-associated or infectious disease can be detected in the CSF. As the CSF is in contact with neuronal cells, choroid plexuses, and the blood–brain barrier membrane, it partially contains molecules or peptides secreted from them [17,38]. Exposure of the CSF to various fungal, bacterial, viral, and parasitic infections can also be detected. Hence, alterations in the CSF can be used as tools for early diagnosis, treatment, and intracranial pressure monitoring. The CSF components in a healthy adult under normal conditions are shown in the table below (Table 1).
Accordingly, any change in the physiological state of the body can cause these values to deviate from their normal ranges.
Infections affecting CSF quality may originate from various sources. Viruses infect the CNS to transmit their genetic material (DNA or RNA) to host cells for self-replication [49]. Viral encephalitis and meningitis can cause severe inflammatory responses that can be detected in the CSF. Inflammatory biomarkers are detectable a few hours to days after infection [50]. Decreases in CSF glucose levels have been reported [51]. Detection of viruses is possible through a polymerase chain reaction (PCR) of CSF samples [52].
Bacteria can infect the central nervous system through bacteraemia, a wound in the body, or the course of a lumbar puncture [53,54]. One of the worst types of CNS infections is bacterial meningitis, which can be fatal [55]. After infection, substantial numbers of pro-inflammatory markers appear in the CSF, and the type of infection can be identified using PCR [56]. In the case of bacterial infections, in some cases, detection of bacteria in CSF samples is possible via microscopy.
Fungal infections, including moulds, yeasts, and certain fungal strains, can also penetrate the blood–brain barrier (BBB) and infect the CNS, leading to severe conditions such as meningoencephalitis, meningitis, and cerebral abscess [57]. The white blood cell count usually increases in the CSF after fungal contamination, but the type of white blood cell (Monocytes, Eosinophils, etc.) is mostly dependent on the strain of fungal contamination. Reduced glucose levels and increased protein levels have also been reported in fungal infections of the CNS; however, the distinction of fungal CNS contamination from other categories has been a challenge due to non-specific signs [58]. In many low-income to middle-income countries, parasitic infections (e.g., Malaria, Leishmaniasis, Amebiasis, Hydatidosis, Coenurosis, and Sparganosis) are among the causes of CNS diseases [54]. Unlike fungal infections, parasitic infections of the CNS have a few symptoms, such as seizures, eosinophilia, headaches, and fever [59]. Parasitic infections in CSF samples can be detected using microscopy and PCR [54].
Any of the abovementioned means of infection can lead to severe conditions involving the central nervous system. Conditions that arise from infection sources can be acute or chronic. Severe acute or chronic infections of the CNS can be starting points for neurodegenerative conditions. Acute conditions of the CNS can usually alter the quality and content of the CSF [60].
Neurodegenerative disorders can affect the CSF content. The onset of each infection or disorder affects the CSF content differently. Initially, inflammatory markers appear in the CSF in mild cases of inflammatory infections [61]. The total protein count in the CSF increases depending on the severity of the inflammation [62]. Disorders such as Parkinson’s disease, myelopathies, and neuropathies are included in this category [63,64,65].
Peripheral neuropathy is a common neurological disorder caused by various factors such as diabetes or hypothyroidism [66]. Neuropathies are diagnosed based on various symptoms and analyses, including CSF analysis. They are characterised by increased inflammatory markers and protein counts along with the physical symptoms of the patient. Furthermore, an increase in small molecules and metabolic materials in CSF white blood cells can be detected in diseases such as globoid cell leukodystrophy and mucopolysaccharidosis [67].
Subarachnoid haemorrhage is a condition that has a direct effect on CSF, usually a few hours after an incident or trauma [68,69]. This condition is painful and can sometimes be followed by a loss of consciousness. In this case, the red blood cells found in the CSF can be detected at >1000 CFU/mm [70]. CSF screening is a well-known approach used to investigate meningitis. Meningitis is inflammation of the protective membrane of the brain and spinal cord. As mentioned above, the root cause of meningitis can be a viral or bacterial infection. It can be initiated by bacterial, fungal, viral, or protozoan microorganisms. CSF analysis is a crucial tool, particularly in the diagnosis and treatment of aseptic meningitis [71]. However, it may take days to obtain the results of CSF analysis using PCR. Analysis of the CSF in aseptic meningitis can identify the source of the infection [71]. For instance, a bacterial infection increases the turbidity of the CSF, whereas viral and fungal infections do not have a significant effect on turbidity. On the other hand, in aseptic meningitis, the protein content of the CSF increases. Viral and fungal infections usually have fewer indications for diagnosis in the CSF; however, increases in white blood cells and lymphocytes in the CSF can be warning signs. Overall, a PCR test can be conducted on a CSF sample to indicate the infection type and the organism that has caused the infection.
Detection of autoimmune disease markers in CSF can offer early treatment opportunities. In general, the early signs of autoimmune diseases such as multiple sclerosis commence with the detection of increased protein levels and lymphocytes in the CSF. Elevated protein levels are a common sign of most autoimmune diseases. In some cases, such as transverse myelitis, inflammatory markers are detectable in CSF. Autoimmune Encephalitis (AEI) is an inflammatory disease associated with the central nervous system. The findings from a CSF analysis of patients with AEI suggest that the inflammation and antibody markers of AEI do not follow certain patterns in various patients. However, anti-NMDA, GABAb, and AMPA antibodies have shown inflammatory changes in CSF samples from patients [72].
Brain and spinal cord cancers also affect CSF quality, which can be a promising tool for diagnosing and preventing metastasis [73]. Reports suggest that CSF analysis, along with MRI and CT scans, is a promising tool for detecting malignant tumours. This method can be expanded to the detection of colon, lung, and prostate cancer biomarkers [74].
Furthermore, minute changes in spinal cord injuries have sensible effects on CSF quality and biomarker contents. As a result of spinal cord tissue damage, neurofilaments (NFs), tau, neuron-specific enolases (NSEs), S100 calcium-binding protein β (S100β), and glial fibrillary acidic protein (GFAP) can eventually be released into the CSF [75]. Along with the vast number of studies [76] performed on Glial Scarring in the spinal cord, the detection of CSPGs, MAG, Nogo-A, and Omgp in CSF can also be investigated in the category of spinal cord defects.
Overall, changes in CSF biomarkers can facilitate the diagnosis and prognosis of various neurological, inflammatory, and infectious conditions. The table below (Table 2) illustrates the most important biomarkers in CSF that can be associated with the most common neurological diseases.

2. An Overview of Biosensors

Biosensors are analytical devices that combine a biological component with a physicochemical detector. The term “biosensor” is often used to cover sensor devices used in order to determine the concentrations of substances and other parameters of biological interest, even where they do not utilise a biological system directly [89]. These devices can provide fast, reliable, and accurate detection of specific biological processes and analytes. The biological components, also known as bioreceptors, can be tissues, microorganisms, organelles, cell receptors, enzymes, antibodies, nucleic acids, etc. The bioreceptor interacts with, binds with, or recognises the analyte being studied (Figure 2). The transducer or detector element works in a physicochemical manner, transforming one signal into another [90].

2.1. Electrochemical Biosensors

Electrochemical biosensors are self-contained integrated devices that provide specific quantitative or semi-quantitative analytical information using a biological recognition element (biochemical receptor) held in direct contact with an electrochemical transduction element [91]. They use biological materials as sensitive components; electrodes as conversion elements; and current, potential, and resistance as the detectable characteristics of a signal. In recent decades, electrochemical biosensors have been developed to detect many biological elements. They offer advantages such as high sensitivity, robustness, reliability, and the potential for integration on a single chip [92].

2.2. Optical Biosensors

Optical biosensors use light and biological elements to detect the presence of an analyte. They primarily use enzymes and antibodies as transducing elements. Optical biosensors allow the safe non-electrical remote sensing of materials. They usually do not require reference sensors, as a comparative signal can be generated using the same light source as the sampling sensor [93]. They have exhibited good performance in detecting biological systems and have promoted significant advances in clinical diagnostics, drug discovery, food process control, and environmental monitoring [94].

2.3. Piezoelectric Biosensors

Piezoelectric biosensors use the piezoelectric effect to measure changes in pressure, acceleration, temperature, strain, or force by converting them to electrical charges. They provide a direct method for the real-time monitoring of biointeractions at the sensor surface, resulting in simplified assay formats [95]. They are well suited for the construction of biosensors that recognise affinity interactions. Assays based on reactions between antigens and antibodies, two polynucleotide strains, aptamers, and proteins are well suited for such studies [96].

2.4. Amperometric Biosensors

Amperometric biosensors function by producing a current when a potential is applied between two electrodes. They generally have response times, dynamic ranges, and sensitivities similar to potentiometric biosensors. The simplest amperometric biosensor in common use involves the Clark oxygen electrode. They provide a direct method for the real-time monitoring of biointeractions at the sensor surface, resulting in simplified assay formats [97].

2.5. Voltametric Biosensors

Voltametric biosensors are electrochemical biosensors that measure the current (flow of electrons) arising during a reaction. Generally speaking, when using voltametric biosensors, the analyte undergoes or is involved in a redox reaction that can be followed by measuring the current in an electrochemical cell [98]. They are often used in initial analytical tests because of their characteristics such as ease of understanding (simple theory), ease of use (choice of potential and current acquisition time), and the possibility to work with very simple and low-cost equipment [98].

2.6. Development of Biosensors

Biosensor development involves several steps. The first step involves the selection of a suitable biological sensing element that can interact with the analyte of interest to generate a signal [99]. Sensing elements include materials such as tissues, microorganisms, organelles, cell receptors, enzymes, antibodies, and nucleic acids. The signal generated through the interaction of the sensing element and the analyte of interest is then transformed into a measurable and quantifiable electrical signal via the transducer [100].
Modern approaches employed in biosensors involve the use of nanomaterials, such as nanoparticles (NPs), nanowires (NWs), nanorods (NRs), carbon nanotubes (CNTs), quantum dots (QDs), and dendrimers. These nanomaterials can enhance the performance of transducers, increase sensitivity, reduce response time, improve reproducibility, and lower detection limits [90].

2.7. Applications of Biosensors

Biosensors are used in a wide range of applications. They play critical roles in a myriad of fields, including biomedical diagnosis, monitoring of treatment and disease progression, drug discovery, food control, and environmental monitoring [89]. They have proven useful in disease diagnosis in human healthcare delivery, agriculture, homeland security, food security, environmental and industrial monitoring, and bioprocessing [101]. They are also used in general healthcare monitoring, screening for diseases, the clinical analysis and diagnosis of diseases, veterinary and agricultural applications, industrial processing and monitoring, and environmental pollution control [102]. Biosensors represent a promising area of research with the potential to revolutionise the diagnosis and monitoring of various diseases. As this technology matures, it is expected to become an integral part of community-based screening programs and personalised medicine.

3. Types of Biosensors

Three generations of biosensors are available on the market. Furthermore, five types of sensitive elements are used: antibodies, nucleotides, enzymes, cells, and synthetic molecules [103]. Numerous biosensors have been constructed in many forms, including electrochemical and fluorescence-tagged nanomaterials, silica or quartz, and genetic constructs with reporter genes [104].
Depending on the type of transducer used in biosensor design, biosensors can be categorised into optical biosensors, electrochemical biosensors, or mass-based biosensors.
Comparing different types of biosensors is crucial for several reasons. Each type of biosensor, whether it is an immunosensor, enzymatic sensor, peptide-based sensor, or gene-based sensor, has unique advantages and applications [105]. By comparing these biosensors, we can understand their strengths and limitations, which can guide the selection of the most appropriate biosensor for a specific application. Furthermore, such comparisons can inspire the development of new biosensors that combine the advantages of different types, leading to improved performance and expanded applications. Therefore, comparing different biosensors is an essential step in the ongoing advancement of biosensor technology.

3.1. Immunosensors

Immunosensors are biosensors that utilise antibodies as biological recognition elements. They are popular because of their excellent detection performance, selectivity, and sensitivity. Recent advancements in immunosensors allow detection to be incorporated in the latest digital technology and miniaturised without compromising performance. They are quick, highly selective, and sensitive and possess the potential to significantly improve the diagnostic processes of pathogens and toxins [106]. They are a promising alternative to the traditional immunoassays and state-of-the-art affinity sensors used to diagnose clinically important analytes/antigens due to their high affinity, versatility, compact size, fast response time, minimum sample processing, and reproducibility [107].

3.2. Enzymatic Biosensors

Enzymatic biosensors use enzymes as biological recognition elements. They are highly specific, simple, and scalable and facilitate the fast, precise, and continuous monitoring of analytes. The high specificity of enzymes enhances their ability to detect analytes with lower concentration limits [108]. They provide a direct method for the real-time monitoring of biointeractions at the sensor surface, resulting in simplified assay formats. They generally have response times, dynamic ranges, and sensitivities similar to those of potentiometric biosensors [109].

3.3. Peptide-Based Biosensors

Peptide-based biosensors use peptides as biological recognition elements. They have been rapidly developed for the detection of disease markers with high sensitivity, rapid analysis speeds, and easy miniaturisation [110]. Their benefits mainly lie in their stability and selectivity toward a target analyte. Furthermore, they can be synthesised easily and modified with specific functional groups, thus making them suitable for the development of novel architectures for biosensing platforms as well as alternative labelling tools [111].

3.4. Gene-Based Biosensors

Gene-based biosensors, also known as DNA-based biosensors, use DNA or RNA as a biological recognition element [112]. They have advantages such as wider detection targets, longer lifetimes, and lower production costs. Additionally, ingenious DNA structures can control the signal conduction near the biosensor surface, which could significantly improve the performance of the biosensors. They can be used effectively to provide simple, fast, cost-effective, sensitive, and specific detection of certain genetic diseases, cancers, and infectious diseases [113].

4. Multiplex Biosensors

Multiplex biosensors are an emerging approach to point-of-care testing. They reduce analysis time and testing costs by detecting multiple analytes or biomarkers simultaneously. These are crucial for disease detection at an early stage. The application of inexpensive substrates such as paper has immense potential and is a matter of research interest in point-of-care testing for multiplexed analysis [114]. These biosensors possess several unique advantages, including the ability to detect multiple analytes on one instrument, examine various parameters, and offer a 2D paper network with spatiotemporal complexity [115].

4.1. Advancements in Multiplex Biosensors

Recent advancements in biotechnology and nanotechnology have facilitated the development of biosensor platforms capable of real-time detection of multiple biomarkers in clinically relevant samples [116]. Both electrical and spectroscopic multiplexed detection strategies are employed to reach limits of detection below the levels in clinical samples [116]. Some of the most promising strategies for point-of-care assays involve inexpensive materials, such as paper-based microfluidic devices, or portable and accessible detectors, such as smartphones [117].

4.2. Multiplex Biosensors in Cerebrospinal Fluid (CSF) Biosensing

In recent years, biomarkers of inflammatory responses in the central nervous system (CNS) have been identified in different body fluids, such as blood, cerebrospinal fluid (CSF), and tears [118]. Progress in micro- and nanotechnology has enabled the development of biosensing platforms capable of detecting multiple biomarkers in clinically relevant samples, such as CSF. These advancements have opened new possibilities for the diagnosis and monitoring of neurological diseases [118].
For instance, an electrochemical biosensor has been reported for the highly sensitive label-free detection of CSF miR-21, relying on target-induced redox signal amplification [119]. This biosensor was developed by covalently assembling capture stands partially complementary to miR-21 on a gold nanoparticle-coated glassy carbon electrode [120].

5. Biosensing in Body Fluids

Detection of central nervous system (CNS) disease biomarkers using biosensors in various body fluids has emerged as a promising approach in the field of neurology. These biosensors have been developed to detect key biomarkers, such as brain-derived neurotrophic factor (BDNF) and neurofilament light chain (NfL), which play important roles in the development and progression of many neurological diseases, including multiple sclerosis, Alzheimer’s disease, and Parkinson’s disease [121].

5.1. Saliva-Based Biosensors

Saliva-based biosensors are gaining attention owing to their non-invasive collection and ability to detect periodontal disease and identify systemic diseases. The human body has a unique way of indicating when something is wrong [122]. Molecules in body fluids can be helpful in the early detection of diseases by enabling health and preventing disease progression. For instance, C-reactive protein (CRP) is the most predictive biomarker of Acute Myocardial Infarction [123]. Several sensing technologies are available for stress biomarker monitoring in saliva and other fluids. Enzyme-linked immunosorbent assays, colourimetric techniques, surface plasmon resonance sensing, and molecularly imprinted polymers offer sensitive and selective cortisol detection in saliva.

5.2. Blood-Based Biosensors

Blood-based biosensors have gained attention because of their ability to detect a wide range of biomarkers for various diseases. Alzheimer’s disease (AD) is a chronic neurodegenerative disorder, and its early diagnosis plays a crucial role in its prevention and treatment [124,125]. In recent years, blood-based biomarkers for early AD diagnosis have gained attention because of their non-invasiveness and minimal adverse reactions, making them an attractive approach. Among the various sensors used to detect blood biomarkers for AD, printed sensors demonstrate great potential for AD blood biomarker detection because of their ease of miniaturisation, scalability for mass production, and compatibility with various detection methods.

5.3. Tear-Based Biosensors

Tear-based biosensors are promising tools for detecting various biomarkers associated with neurological diseases. The TearAD study aimed to use tear fluid as a potential source of AD biomarkers. In previous reports, it was demonstrated that the AD biomarkers amyloid-beta and tau are measurable in tear fluid and are associated with disease severity and neurodegeneration [126]. This study aimed to validate the previous results in a larger cohort and evaluated the diagnostic accuracy of tear biomarkers when discriminating between individuals with and without neurodegeneration, as determined by hippocampal atrophy [126].

6. CSF Sensing and Detection: Current State and Progress

Recently, alternative and prompt approaches to diagnosing diseases using CSF analysis have been introduced. So far, different analytical procedures have been used in this regard, such as MRI [127], CT [128], PCR [129], GC–mass spectrometry [130], and microscopy [40,131]. As mentioned, lumbar puncture is the chief procedure for CSF sampling, which is rather painful and carries a high infection risk for patients [132]. Hence, there is a need for quick and minimally invasive techniques for CSF sampling and subsequent analysis.
Electrochemical biosensors have emerged as a practical option for minimally invasive CSF sensing [133]. Generally, a biosensor or electrochemical sensor should be both highly sensitive and selective [10] for use as a diagnostic tool. Furthermore, biosensors can be designed and characterised according to the desired application.
Since most of the problems associated with the central nervous system are delicate and difficult to address without surgery, it is important to have biosensors with suitable designs to fulfil the technical and medical requirements. For example, CSF accumulates in the brain in hydrocephalus. The conventional treatment for hydrocephalus involves implanting a shunt in the brain ventricle via surgery [134]. However, this procedure carries a high risk of failure and infection [135]. Hence, implantable sensors have been introduced as novel monitoring tools for hydrocephalus treatment and control [136]. They have been used successfully in the detection of shunt malfunctions [137,138] and CSF flow control [139]. Implantable sensors are another option for monitoring CSF pressure and have been proven to decrease the chance of infection [140]. Another continuous flow monitoring method for the detection of hydrocephalus shunt malfunctions was proposed by Pennell et al. (2016). An implantable sensor was designed to monitor and regulate shunt malfunctions. The implantable sensor was designed to uniquely characterise the type of abnormality, either blockage or decreased flow, and regulate the flow accordingly [141]. In addition to flow control, this method is less invasive and reduces the risk of infection.
The detection of biological biomarkers in CSF is an important application of electrochemical biosensors. Over the past decade, the detection of crucial biomarkers in CSF has been gaining more attention. Numerous novel approaches for detecting biomarkers in CSF samples have been introduced. For instance, biosensors integrated with microfluidics were proposed by Senel et al. in 2020 [142] to detect decreased dopamine levels in the CSF and blood. Decreased dopamine levels are a hallmark of Parkinson’s disease. Such sensors enable the detection of important biomarkers in bodily fluids. The use of a microfluidic device was also proposed as a practical approach to decrease the sample volume [142].
CSF is in contact with the cells surrounding the brain ventricles and can be exposed to variations in extracellular pH from cells, such as neuronal cells and brain tissue. The pH of CSF in a healthy adult is approximately 7.4; hence, variations in the pH of CSF can lead to an abnormal biological condition [143]. Electrochemical pH biosensors based on ZnO microneedles have successfully enabled the real-time monitoring of mouse CSF [144] in vivo. A unique fibre-based pH sensor with increased sensitivity was designed by Booth et al. in 2021 [145].
A multi-sensing system for CSF was reported to be able to detect multiple components in CSF to predict a diagnosis of Alzheimer’s disease. The sensor was successfully calibrated for dopamine, KCl, NaCl, MgCl2, NaHCO3, NaHS, and CaCl2 detection [146].
A non-invasive method to detect lactic acid in CSF was introduced by Goh et al. (2011). This method uses low-power microwave sensors [147]. Lactate is one of the biomarkers in CSF that is very helpful in detecting infections in the central nervous system [148]. A variety of biosensors have been investigated for the measurement of lactate in blood, plasma, interstitial fluid [149], and sweat [150] on various substrates [151]. However, a more innovative approach for point-of-care lactate sensing in CSF is yet to be developed.
So far, the main application of CSF sensing remains the early-onset diagnosis of central nervous system diseases [40]. Currently, Alzheimer’s disease and dementia are the most controversial diseases in terms of early diagnosis and treatment. Hence, early detection of the disease can be crucial for treatment progress. Proteins such as β-amyloid and tau are the most common indicators of disease onset in CSF. However, the detection of these proteins in CSF in the early stages requires highly sensitive detection techniques. Very recently, with the development of a highly sensitive β-amyloid sensor, investigators have been able to detect β-amyloid [152]. The ability of the sensor to detect β-amyloid levels as low as 1 pg/mL can increase the chance of β-amyloid detection and early-onset diagnosis. This biochemically modified sensor has the potential to be used as a detection sensor rather than the previously prepared sensors for β-amyloid detection [153]. However, β-amyloid is not the only biomarker that must be detected for an accurate diagnosis of central nervous system disease [154]. Tau protein also participates in the formation of amyloid plaques in the brain. Hence, detection of total tau protein in CSF can also have an impact on early diagnosis [155]. Analytical methods have been reported to detect tau protein [156], including ELISA, xMAP, and antibody-based assays, for the analysis of total tau protein levels [157]. However, these assays require time and analytical laboratory conditions that remain controversial. Biosensors, on the other hand, have been introduced as a fast and highly sensitive approach for tau protein detection in CSF. These approaches include optical [158], electrochemical [159], and piezoelectric biosensors [160]. Electrochemical biosensors are less sensitive than optical biosensors and more sensitive than piezoelectric sensors. Among biosensors, electrochemical biosensors have been widely utilised in various aspects of life, from healthcare to technological applications. Their flexibility of design, reliability, high limit of detection, and vast range of applications for various purposes make electrochemical biosensors a unique and wise choice [161]. This feature has also had an impact on the development of diagnostic electrochemical biosensors for applications in neurodegenerative disorders. Furthermore, electrochemical biosensors are easy to fabricate and can be designed for amperometry and voltammetry.
A list of recent advances in electrochemical CSF biosensors for neurodegenerative disease-related biomarkers and their ranges of detection are listed in the table below.
The table presents a list of recent developments in the field of CSF sensing (Table 3). Currently, valuable research is underway on the development of fast and minimally invasive in vitro diagnostic biosensors. These biosensors are aimed at detecting certain biomarkers associated with disorders in order to provide correct diagnoses.
According to the literature review, various methods have been developed to measure proteins in CSF. However, most of these methods are based on point-of-care detection via antibody–antigen bonding. Hence, there is a need for a method that is capable of continuously detecting analytes to improve repeatability. In addition to their development, the validation of these sensors in clinical settings is also important [146].

7. Conclusions and Future Perspectives

The detection of biomarkers in body fluids has progressed significantly, with advancements in biosensor technology playing a pivotal role. It is evident that indications of many diseases, particularly neurodegenerative diseases, can be observed in various body fluids such as cerebrospinal fluid (CSF), saliva, blood, and tears. Accurate detection of key biomarkers associated with these disorders is crucial for early diagnosis and effective treatment.
While the main and prominent collection method for CSF remains lumbar puncture, it is important to explore options to decrease its associated complications and ease the procedure. In addition, the development of fast, low-cost diagnostic devices, such as saliva-based, blood-based, and tear-based biosensors, is highly desirable for disease detection and monitoring.
Incorporating biosensors into lumbar puncture needles or designing newer needles can offer better alternative methods to achieve these goals. However, the design of these biosensors must be approached in an interdisciplinary fashion, with engineers, neurosurgeons, and biotechnologists working together to develop robust sensors with high diagnostic sensitivity and specificity.
Furthermore, the use of multiplex biosensors capable of detecting multiple biomarkers simultaneously represents a promising area of research with the potential to revolutionise the diagnosis and monitoring of neurological diseases. As this technology matures, it is expected to become an integral part of community-based screening programs and personalised medicine.
In conclusion, the future of detecting CNS disease biomarkers in various body fluids lies in the continuous advancement of biosensor technology, interdisciplinary collaboration, and the integration of these technologies into existing medical procedures and practices.

Author Contributions

Writing and literature review, G.H.-F., Review, S.A.-I. All authors have read and agreed to the published version of the manuscript.

Funding

This Research has received funding from Welcome Trust WTiTPA (CC BY) for funding their project (PSZ088) “continuous multi-sensing in cerebrospinal fluid as a therapeutical approach”.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Vernau, W.; Vernau, K.A.; Sue Bailey, C. Cerebrospinal Fluid. Clin. Biochem. Domest. Anim. 2008, 769–819. [Google Scholar] [CrossRef]
  2. Meldolesi, J. News about the Role of Fluid and Imaging Biomarkers in Neurodegenerative Diseases. Biomedicines 2021, 9, 252. [Google Scholar] [CrossRef] [PubMed]
  3. Krause, K.; Wulf, M.; Sommer, P.; Barkovits, K.; Vorgerd, M.; Marcus, K.; Eggers, B. CSF Diagnostics: A Potentially Valuable Tool in Neurodegenerative and Inflammatory Disorders Involving Motor Neurons: A Review. Diagnostics 2021, 11, 1522. [Google Scholar] [CrossRef] [PubMed]
  4. Arribas, M. The history of cerebrospinal fluid: From classical antiquity to the late modern period. Neurosci. Hist 2017, 5, 105–113. [Google Scholar]
  5. Gastaldi, M.; Zardini, E.; Leante, R.; Ruggieri, M.; Costa, G.; Cocco, E.; De Luca, G.; Cataldo, I.; Biagioli, T.; Ballerini, C. Cerebrospinal fluid analysis and the determination of oligoclonal bands. Neurol. Sci. 2017, 38, 217–224. [Google Scholar] [CrossRef]
  6. Vecchio, D. The history of cerebrospinal fluid analysis in multiple sclerosis: A great development over the last centuries. J. Brain Disord. 2017, 1, 35–37. [Google Scholar]
  7. Ousman, S.S.; Kubes, P. Immune surveillance in the central nervous system. Nat. Neurosci. 2012, 15, 1096–1101. [Google Scholar] [CrossRef] [PubMed]
  8. Ahmed, R.M.; Paterson, R.W.; Warren, J.D.; Zetterberg, H.; Brien, J.T.; Fox, N.C.; Halliday, G.M.; Schott, J.M. Biomarkers in dementia: Clinical utility and new directions. J. Neurol. Neurosurg. Psychiatry 2014, 85, 1426. [Google Scholar] [CrossRef] [PubMed]
  9. Zhou, M.; Haque, R.U.; Dammer, E.B.; Duong, D.M.; Ping, L.; Johnson, E.C.B.; Lah, J.J.; Levey, A.I.; Seyfried, N.T. Targeted mass spectrometry to quantify brain-derived cerebrospinal fluid biomarkers in Alzheimer’s disease. Clin. Proteom. 2020, 17, 19. [Google Scholar] [CrossRef]
  10. Mikuła, E. Recent Advancements in Electrochemical Biosensors for Alzheimer’s Disease Biomarkers Detection. Curr. Med. Chem. 2021, 28, 4049–4073. [Google Scholar] [CrossRef]
  11. Collins, M.A.; An, J.; Peller, D.; Bowser, R. Total protein is an effective loading control for cerebrospinal fluid western blots. J. Neurosci. Methods 2015, 251, 72–82. [Google Scholar] [CrossRef]
  12. Batllori, M.; Molero-Luis, M.; Ormazabal, A.; Casado, M.; Sierra, C.; García-Cazorla, A.; Kurian, M.; Pope, S.; Heales, S.J.; Artuch, R. Analysis of human cerebrospinal fluid monoamines and their cofactors by HPLC. Nat. Protoc. 2017, 12, 2359–2366. [Google Scholar] [CrossRef] [PubMed]
  13. Ilhan, H.; Panhwar, S.; Boyaci, I.H.; Tamer, U. Optical Based Transducers for Biosensors. In Biosensors; CRC Press: Boca Raton, FL, USA, 2022; pp. 57–87. [Google Scholar]
  14. Wethekam, L.C.; Moore, J.K. α-tubulin regulation by 5′ introns in Saccharomyces cerevisiae. Genetics 2023, 225, iyad163. [Google Scholar] [CrossRef]
  15. Ali, A.H. High-performance liquid chromatography (HPLC): A review. Ann. Adv. Chem. 2022, 6, 010–020. [Google Scholar]
  16. Brown, P.D.; Davies, S.L.; Speake, T.; Millar, I.D. Molecular mechanisms of cerebrospinal fluid production. Neuroscience 2004, 129, 957–970. [Google Scholar] [CrossRef] [PubMed]
  17. Sakka, L.; Coll, G.; Chazal, J. Anatomy and physiology of cerebrospinal fluid. Eur. Ann. Otorhinolaryngol. Head Neck Dis. 2011, 128, 309–316. [Google Scholar] [CrossRef]
  18. Jurado, R.; Walker, H.K. Cerebrospinal fluid. In Clinical Methods: The History, Physical, and Laboratory Examinations, 3rd ed.; Butterworths: Boston, MA, USA, 1990. [Google Scholar]
  19. Simon, M.J.; Iliff, J.J. Regulation of cerebrospinal fluid (CSF) flow in neurodegenerative, neurovascular and neuroinflammatory disease. Biochim. Biophys. Acta 2016, 1862, 442–451. [Google Scholar] [CrossRef]
  20. Pollay, M. The function and structure of the cerebrospinal fluid outflow system. Cerebrospinal Fluid Res. 2010, 7, 1–20. [Google Scholar] [CrossRef]
  21. Chang, H.; Nakagawa, H. Hypothesis on the pathophysiology of syringomyelia based on simulation of cerebrospinal fluid dynamics. J. Neurol. Neurosurg. Psychiatry 2003, 74, 344–347. [Google Scholar] [CrossRef]
  22. Hosoya, K.-i.; Tachikawa, M. Roles of organic anion/cation transporters at the blood–brain and blood–cerebrospinal fluid barriers involving uremic toxins. Clin. Exp. Nephrol. 2011, 15, 478–485. [Google Scholar] [CrossRef]
  23. Ghersi-Egea, J.-F.; Strazielle, N.; Catala, M.; Silva-Vargas, V.; Doetsch, F.; Engelhardt, B. Molecular anatomy and functions of the choroidal blood-cerebrospinal fluid barrier in health and disease. Acta Neuropathol. 2018, 135, 337–361. [Google Scholar] [CrossRef] [PubMed]
  24. Bothwell, S.W.; Janigro, D.; Patabendige, A. Cerebrospinal fluid dynamics and intracranial pressure elevation in neurological diseases. Fluids Barriers CNS 2019, 16, 9. [Google Scholar] [CrossRef]
  25. Doherty, C.M.; Forbes, R.B. Diagnostic Lumbar Puncture. Ulst. Med J 2014, 83, 93–102. [Google Scholar]
  26. Tumani, H.; Huss, A.; Bachhuber, F. The cerebrospinal fluid and barriers–anatomic and physiologic considerations. Handb. Clin. Neurol. 2018, 146, 21–32. [Google Scholar]
  27. Rock, R.B.; Olin, M.; Baker, C.A.; Molitor, T.W.; Peterson, P.K. Central nervous system tuberculosis: Pathogenesis and clinical aspects. Clin. Microbiol. Rev. 2008, 21, 243–261. [Google Scholar] [CrossRef] [PubMed]
  28. Oliveira, J.P.S.; Mendes, N.T.; Martins, Á.R.; Sanvito, W.L. Cerebrospinal fluid: History, collection techniques, indications, contraindications and complications. J. Bras. Patol. Med. Lab. 2020, 56, e2822020. [Google Scholar] [CrossRef]
  29. Mollan, S.P.; Ali, F.; Hassan-Smith, G.; Botfield, H.; Friedman, D.I.; Sinclair, A.J. Evolving evidence in adult idiopathic intracranial hypertension: Pathophysiology and management. J. Neurol. Neurosurg. Psychiatry 2016, 87, 982–992. [Google Scholar] [CrossRef] [PubMed]
  30. Ahmed, S.V.; Jayawarna, C.; Jude, E. Post lumbar puncture headache: Diagnosis and management. Postgrad. Med. J. 2006, 82, 713–716. [Google Scholar] [CrossRef] [PubMed]
  31. Lavi, R.; Rowe, J.M.; Avivi, I. Lumbar Puncture: It Is Time to Change the Needle. Eur. Neurol. 2010, 64, 108–113. [Google Scholar] [CrossRef]
  32. Wright, B.L.C.; Lai, J.T.F.; Sinclair, A.J. Cerebrospinal fluid and lumbar puncture: A practical review. J. Neurol. 2012, 259, 1530–1545. [Google Scholar] [CrossRef]
  33. Tomi, N.S.; Kränke, B.; Aberer, E. Staphylococcal toxins in patients with psoriasis, atopic dermatitis, and erythroderma, and in healthy control subjects. J. Am. Acad. Dermatol. 2005, 53, 67–72. [Google Scholar] [CrossRef]
  34. Schulga, P.; Grattan, R.; Napier, C.; Babiker, M.O. How to use… lumbar puncture in children. Arch. Dis. Child Educ. Pr. Ed. 2015, 100, 264–271. [Google Scholar] [CrossRef]
  35. Brown, K.; Berkowitz, D. Chapter 171-Lumbar Puncture. In Pediatric Emergency Medicine; Baren, J.M., Rothrock, S.G., Brennan, J.A., Brown, L., Eds.; W.B. Saunders: Philadelphia, PA, USA, 2008; pp. 1208–1211. [Google Scholar] [CrossRef]
  36. Seehusen, D.A.; Reeves, M.M.; Fomin, D.A. Cerebrospinal fluid analysis. Am. Fam. Physician 2003, 68, 1103–1108. [Google Scholar]
  37. Fitzgerald, J.; Fenniri, H. Cutting Edge Methods for Non-Invasive Disease Diagnosis Using E-Tongue and E-Nose Devices. Biosensors 2017, 7, 59. [Google Scholar] [CrossRef] [PubMed]
  38. Reiber, H. Proteins in cerebrospinal fluid and blood: Barriers, CSF flow rate and source-related dynamics. Restor. Neurol. Neurosci. 2003, 21, 79–96. [Google Scholar] [PubMed]
  39. Welles, E.G.; Pugh, D.G.; Wenzel, J.G.; Sorjonen, D.C. Composition of cerebrospinal fluid in healthy adult llamas. Am. J. Vet. Res. 1994, 55, 1075–1079. [Google Scholar] [CrossRef]
  40. Hrishi, A.P.; Sethuraman, M. Cerebrospinal Fluid (CSF) Analysis and Interpretation in Neurocritical Care for Acute Neurological Conditions. Indian J. Crit. Care Med. 2019, 23, S115–S119. [Google Scholar] [CrossRef]
  41. Lumb, A.B. Chapter 4-Control of Breathing. In Nunn’s Applied Respiratory Physiology, 8th ed.; Lumb, A.B., Ed.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 51–72.e52. [Google Scholar] [CrossRef]
  42. Lee, S.C.; Lueck, C.J. Cerebrospinal fluid pressure in adults. J. Neuroophthalmol. 2014, 34, 278–283. [Google Scholar] [CrossRef] [PubMed]
  43. Erdem, H.; Ozturk-Engin, D.; Cag, Y.; Senbayrak, S.; Inan, A.; Kazak, E.; Savasci, U.; Elaldi, N.; Vahaboglu, H.; Hasbun, R. Central nervous system infections in the absence of cerebrospinal fluid pleocytosis. Int. J. Infect. Dis. 2017, 65, 107–109. [Google Scholar] [CrossRef]
  44. Martín-Ancel, A.; García-Alix, A.; Salas, S.; Del Castillo, F.; Cabañas, F.; Quero, J. Cerebrospinal fluid leucocyte counts in healthy neonates. Arch. Dis. Child Fetal Neonatal. Ed. 2006, 91, F357–F358. [Google Scholar] [CrossRef]
  45. Koenig, M.; Aldrich, E.M. Chapter 25-Cerebrovascular Disorders. In Cerebrospinal Fluid in Clinical Practice; Irani, D.N., Ed.; W.B. Saunders: Philadelphia, PA, USA, 2009; pp. 225–231. [Google Scholar] [CrossRef]
  46. Leen, W.G.; Willemsen, M.A.; Wevers, R.A.; Verbeek, M.M. Cerebrospinal fluid glucose and lactate: Age-specific reference values and implications for clinical practice. PLoS ONE 2012, 7, e42745. [Google Scholar] [CrossRef] [PubMed]
  47. Jovel, J.; O’keefe, S.; Patterson, J.; Bording-Jorgensen, M.; Wang, W.; Mason, A.L.; Warren, K.G.; Wong, G.K.-S. Cerebrospinal Fluid in a Small Cohort of Patients with Multiple Sclerosis Was Generally Free of Microbial DNA. Front. Cell. Infect. Microbiol. 2017, 6, 198. [Google Scholar] [CrossRef] [PubMed]
  48. Spector, R.; Robert Snodgrass, S.; Johanson, C.E. A balanced view of the cerebrospinal fluid composition and functions: Focus on adult humans. Exp. Neurol. 2015, 273, 57–68. [Google Scholar] [CrossRef]
  49. Koyuncu, O.O.; Hogue, I.B.; Enquist, L.W. Virus infections in the nervous system. Cell Host Microbe 2013, 13, 379–393. [Google Scholar] [CrossRef]
  50. Swanson, P.A., 2nd; McGavern, D.B. Viral diseases of the central nervous system. Curr. Opin. Virol. 2015, 11, 44–54. [Google Scholar] [CrossRef]
  51. Davis, R.; Jeffery, K.; Atkins, B.L. Hypoglycorrhachia in herpes simplex encephalitis. Clin. Infect. Dis. 2004, 38, 1506–1507. [Google Scholar] [CrossRef] [PubMed]
  52. Lévêque, N.; Legoff, J.; Mengelle, C.; Mercier-Delarue, S.; N’Guyen, Y.; Renois, F.; Tissier, F.; Simon, F.; Izopet, J.; Andréoletti, L. Virological diagnosis of central nervous system infections by use of PCR coupled with mass spectrometry analysis of cerebrospinal fluid samples. J. Clin. Microbiol. 2014, 52, 212–217. [Google Scholar] [CrossRef] [PubMed]
  53. Suthar, R.; Sankhyan, N. Bacterial Infections of the Central Nervous System. Indian J. Pediatr. 2019, 86, 60–69. [Google Scholar] [CrossRef]
  54. Benninger, F.; Steiner, I. Chapter 12-CSF in acute and chronic infectious diseases. In Handbook of Clinical Neurology; Deisenhammer, F., Teunissen, C.E., Tumani, H., Eds.; Elsevier: Amsterdam, The Netherlands, 2018; Volume 146, pp. 187–206. [Google Scholar]
  55. Prasad, K.; Karlupia, N. Prevention of bacterial meningitis: An overview of Cochrane systematic reviews. Respir. Med. 2007, 101, 2037–2043. [Google Scholar] [CrossRef]
  56. Issa, M.; Mölling, P.; Bäckman, A.; Unemo, M.; Sulaiman, N.; Olcén, P. PCR of cerebrospinal fluid for diagnosis of bacterial meningitis during meningococcal epidemics; an example from Sudan. Scand. J Infect Dis. 2003, 35, 719–723. [Google Scholar] [CrossRef]
  57. Borha, A.; Parienti, J.-J.; Emery, E.; Coskun, O.; Khouri, S.; Derlon, J.-M. Granulome cérébral à Candida albicans chez un patient immunocompétent. Cas clinique. Neurochirurgie 2009, 55, 57–62. [Google Scholar] [CrossRef] [PubMed]
  58. Góralska, K.; Blaszkowska, J.; Dzikowiec, M. Neuroinfections caused by fungi. Infection 2018, 46, 443–459. [Google Scholar] [CrossRef]
  59. Nash, T.E. Parasitic Diseases that Cause Seizures. Epilepsy Curr. 2014, 14, 29–34. [Google Scholar] [CrossRef] [PubMed]
  60. Parikh, V.; Tucci, V.; Galwankar, S. Infections of the nervous system. Int. J. Crit. Illn. Inj. Sci. 2012, 2, 82–97. [Google Scholar] [CrossRef]
  61. Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in CNS neurodegenerative diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef]
  62. Deisenhammer, F.; Zetterberg, H.; Fitzner, B.; Zettl, U.K. The Cerebrospinal Fluid in Multiple Sclerosis. Front. Immunol. 2019, 10, 438156. [Google Scholar] [CrossRef]
  63. Le, W.-D.; Rowe, D.B.; Jankovic, J.; Xie, W.; Appel, S.H. Effects of Cerebrospinal Fluid From Patients With Parkinson Disease on Dopaminergic Cells. Arch. Neurol. 1999, 56, 194–200. [Google Scholar] [CrossRef] [PubMed]
  64. Toedebusch, C.M.; Bachrach, M.D.; Garcia, V.B.; Johnson, G.C.; Katz, M.L.; Shaw, G.; Coates, J.R.; Garcia, M.L. Cerebrospinal Fluid Levels of Phosphorylated Neurofilament Heavy as a Diagnostic Marker of Canine Degenerative Myelopathy. J. Vet. Intern. Med. 2017, 31, 513–520. [Google Scholar] [CrossRef]
  65. Misra, U.K.; Kalita, J.; Nair, P.P. Diagnostic approach to peripheral neuropathy. Ann. Indian Acad. Neurol. 2008, 11, 89–97. [Google Scholar] [CrossRef]
  66. Lehmann, H.C.; Wunderlich, G.; Fink, G.R.; Sommer, C. Diagnosis of peripheral neuropathy. Neurol. Res. Pract. 2020, 2, 20. [Google Scholar] [CrossRef]
  67. Corado, C.R.; Pinkstaff, J.; Jiang, X.; Galban, E.M.; Fisher, S.J.; Scholler, O.; Russell, C.; Bagel, J.H.; PA, O.D.; Ory, D.S.; et al. Cerebrospinal fluid and serum glycosphingolipid biomarkers in canine globoid cell leukodystrophy (Krabbe Disease). Mol. Cell. Neurosci. 2020, 102, 103451. [Google Scholar] [CrossRef]
  68. McKee, A.C.; Daneshvar, D.H. The neuropathology of traumatic brain injury. Handb. Clin. Neurol. 2015, 127, 45–66. [Google Scholar] [CrossRef] [PubMed]
  69. Zanier, E.R.; Refai, D.; Zipfel, G.J.; Zoerle, T.; Longhi, L.; Esparza, T.J.; Spinner, M.L.; Bateman, R.J.; Brody, D.L.; Stocchetti, N. Neurofilament light chain levels in ventricular cerebrospinal fluid after acute aneurysmal subarachnoid haemorrhage. J. Neurol. Neurosurg. Psychiatry 2011, 82, 157–159. [Google Scholar] [CrossRef] [PubMed]
  70. Perry, J.J.; Alyahya, B.; Sivilotti, M.L.A.; Bullard, M.J.; Émond, M.; Sutherland, J.; Worster, A.; Hohl, C.; Lee, J.S.; Eisenhauer, M.A.; et al. Differentiation between traumatic tap and aneurysmal subarachnoid hemorrhage: Prospective cohort study. BMJ Br. Med. J. 2015, 350, h568. [Google Scholar] [CrossRef] [PubMed]
  71. Khoury, N.T.; Hossain, M.M.; Wootton, S.H.; Salazar, L.; Hasbun, R. Meningitis with a negative cerebrospinal fluid Gram stain in adults: Risk classification for an adverse clinical outcome. Mayo Clin. Proc. 2012, 87, 1181–1188. [Google Scholar] [CrossRef]
  72. Blinder, T.; Lewerenz, J. Cerebrospinal Fluid Findings in Patients With Autoimmune Encephalitis—A Systematic Analysis. Front. Neurol. 2019, 10, 804. [Google Scholar] [CrossRef] [PubMed]
  73. Weston, C.L.; Glantz, M.J.; Connor, J.R. Detection of cancer cells in the cerebrospinal fluid: Current methods and future directions. Fluids Barriers CNS 2011, 8, 14. [Google Scholar] [CrossRef] [PubMed]
  74. Ballester, L.Y.; Lu, G.; Zorofchian, S.; Vantaku, V.; Putluri, V.; Yan, Y.; Arevalo, O.; Zhu, P.; Riascos, R.F.; Sreekumar, A.; et al. Analysis of cerebrospinal fluid metabolites in patients with primary or metastatic central nervous system tumors. Acta Neuropathol. Commun. 2018, 6, 85. [Google Scholar] [CrossRef] [PubMed]
  75. Hulme, C.H.; Brown, S.J.; Fuller, H.R.; Riddell, J.; Osman, A.; Chowdhury, J.; Kumar, N.; Johnson, W.E.; Wright, K.T. The developing landscape of diagnostic and prognostic biomarkers for spinal cord injury in cerebrospinal fluid and blood. Spinal Cord 2017, 55, 114–125. [Google Scholar] [CrossRef]
  76. Wright, K.T.; Masri, W.E.; Osman, A.; Chowdhury, J.; Johnson, W.E.B. Concise Review: Bone Marrow for the Treatment of Spinal Cord Injury: Mechanisms and Clinical Applications. Stem Cells 2011, 29, 169–178. [Google Scholar] [CrossRef]
  77. Alcolea, D.; Pegueroles, J.; Munoz, L.; Camacho, V.; López-Mora, D.; Fernández-León, A.; Le Bastard, N.; Huyck, E.; Nadal, A.; Olmedo, V. Agreement of amyloid PET and CSF biomarkers for Alzheimer’s disease on Lumipulse. Ann. Clin. Transl. Neurol. 2019, 6, 1815–1824. [Google Scholar] [CrossRef] [PubMed]
  78. Contador, J.; Pérez-Millán, A.; Tort-Merino, A.; Balasa, M.; Falgàs, N.; Olives, J.; Castellví, M.; Borrego-Écija, S.; Bosch, B.; Fernández-Villullas, G.; et al. Longitudinal brain atrophy and CSF biomarkers in early-onset Alzheimer’s disease. NeuroImage Clin. 2021, 32, 102804. [Google Scholar] [CrossRef]
  79. Magliozzi, R.; Cross, A.H. Can CSF biomarkers predict future MS disease activity and severity? Mult. Scler. J. 2020, 26, 582–590. [Google Scholar] [CrossRef] [PubMed]
  80. Toscano, S.; Patti, F. CSF biomarkers in multiple sclerosis: Beyond neuroinflammation. Neuroimmunol. Neuroinflammation 2021, 8, 14–41. [Google Scholar] [CrossRef]
  81. Pouw, M.H.; Kwon, B.K.; Verbeek, M.M.; Vos, P.E.; van Kampen, A.; Fisher, C.G.; Street, J.; Paquette, S.J.; Dvorak, M.F.; Boyd, M.C.; et al. Structural biomarkers in the cerebrospinal fluid within 24 h after a traumatic spinal cord injury: A descriptive analysis of 16 subjects. Spinal Cord 2014, 52, 428–433. [Google Scholar] [CrossRef] [PubMed]
  82. Rath, J.; Zulehner, G.; Schober, B.; Grisold, A.; Krenn, M.; Cetin, H.; Zimprich, F. Cerebrospinal fluid analysis in Guillain–Barré syndrome: Value of albumin quotients. J. Neurol. 2021, 268, 3294–3300. [Google Scholar] [CrossRef] [PubMed]
  83. Agah, E.; Saleh, F.; Sanjari Moghaddam, H.; Saghazadeh, A.; Tafakhori, A.; Rezaei, N. CSF and blood biomarkers in amyotrophic lateral sclerosis: Protocol for a systematic review and meta-analysis. Syst. Rev. 2018, 7, 237. [Google Scholar] [CrossRef] [PubMed]
  84. Drannik, A.; Martin, J.; Peterson, R.; Ma, X.; Jiang, F.; Turnbull, J. Cerebrospinal fluid from patients with amyotrophic lateral sclerosis inhibits sonic hedgehog function. PLoS ONE 2017, 12, e0171668. [Google Scholar] [CrossRef]
  85. Ahmed, S.; Rakib, A.; Nasrin, F.; Chowdhury, R.H.; Azad, A.K.; Hasan, C.M.M. Assessment of Cerebrospinal Fluid (CSF) to Differentiate between Bacterial Meningitis and Viral Meningitis. J. Adv. Med. Pharm. Sci. 2018, 17, 1–7. [Google Scholar] [CrossRef]
  86. Stephane, G.H. Estimation of CSF Lactate as a Diagnostic Marker to Differentiate Pyogenic Meningitis from Nonpyogenic Meningitis. Ph.D Thesis, Rajiv Gandhi University of Health Sciences, Bengaluru, India, 2019. [Google Scholar]
  87. Marchegiani, F.; Matacchione, G.; Ramini, D.; Marcheselli, F.; Recchioni, R.; Casoli, T.; Mercuri, E.; Lazzarini, M.; Giorgetti, B.; Cameriere, V.; et al. Diagnostic performance of new and classic CSF biomarkers in age-related dementias. Aging 2019, 11, 2420–2429. [Google Scholar] [CrossRef]
  88. Abu-Rumeileh, S.; Steinacker, P.; Polischi, B.; Mammana, A.; Bartoletti-Stella, A.; Oeckl, P.; Baiardi, S.; Zenesini, C.; Huss, A.; Cortelli, P.; et al. CSF biomarkers of neuroinflammation in distinct forms and subtypes of neurodegenerative dementia. Alzheimer’s Res. Ther. 2019, 12, 2. [Google Scholar] [CrossRef] [PubMed]
  89. Tetyana, P.; Shumbula, P.M.; Njengele-Tetyana, Z. Biosensors: Design, development and applications. In Nanopores; IntechOpen: London, UK, 2021. [Google Scholar]
  90. Naresh, V.; Lee, N. A review on biosensors and recent development of nanostructured materials-enabled biosensors. Sensors 2021, 21, 1109. [Google Scholar] [CrossRef] [PubMed]
  91. Singh, A.; Sharma, A.; Ahmed, A.; Sundramoorthy, A.K.; Furukawa, H.; Arya, S.; Khosla, A. Recent advances in electrochemical biosensors: Applications, challenges, and future scope. Biosensors 2021, 11, 336. [Google Scholar] [CrossRef] [PubMed]
  92. Wu, J.; Liu, H.; Chen, W.; Ma, B.; Ju, H. Device integration of electrochemical biosensors. Nat. Rev. Bioeng. 2023, 1, 346–360. [Google Scholar] [CrossRef]
  93. Chen, C.; Wang, J. Optical biosensors: An exhaustive and comprehensive review. Analyst 2020, 145, 1605–1628. [Google Scholar] [CrossRef] [PubMed]
  94. Uniyal, A.; Srivastava, G.; Pal, A.; Taya, S.; Muduli, A. Recent advances in optical biosensors for sensing applications: A review. Plasmonics 2023, 18, 735–750. [Google Scholar] [CrossRef]
  95. Zhou, H.; Zhang, Y.; Qiu, Y.; Wu, H.; Qin, W.; Liao, Y.; Yu, Q.; Cheng, H. Stretchable piezoelectric energy harvesters and self-powered sensors for wearable and implantable devices. Biosens. Bioelectron. 2020, 168, 112569. [Google Scholar] [CrossRef]
  96. Deng, W.; Zhou, Y.; Libanori, A.; Chen, G.; Yang, W.; Chen, J. Piezoelectric nanogenerators for personalized healthcare. Chem. Soc. Rev. 2022, 51, 3380–3435. [Google Scholar] [CrossRef]
  97. Bollella, P. Enzyme-based amperometric biosensors: 60 years later… Quo Vadis? Anal. Chim. Acta 2022, 1234, 340517. [Google Scholar] [CrossRef]
  98. Gulaboski, R.; Mirceski, V. Application of voltammetry in biomedicine-Recent achievements in enzymatic voltammetry. Maced. J. Chem. Chem. Eng. 2020, 39, 153–166. [Google Scholar] [CrossRef]
  99. Song, L.; Zhuge, Y.; Zuo, X.; Li, M.; Wang, F. DNA walkers for biosensing development. Adv. Sci. 2022, 9, 2200327. [Google Scholar] [CrossRef]
  100. Khattab, A.M. Basics of Biological Sensors. In Handbook of Nanosensors: Materials and Technological Applications; Springer: Berlin/Heidelberg, Germany, 2023; pp. 1–43. [Google Scholar]
  101. Tripathi, A.; Bonilla-Cruz, J. Review on healthcare biosensing nanomaterials. ACS Appl. Nano Mater. 2023, 6, 5042–5074. [Google Scholar] [CrossRef]
  102. Bhatnagar, A.; Masih, J.; Kumar, R. Biosensor: Application in Environmental Management. In Harnessing Microbial Potential for Multifarious Applications; Springer: Berlin/Heidelberg, Germany, 2024; pp. 455–488. [Google Scholar]
  103. Simon, E. Biological and Medical Sensors 12. In Sensors in Science and Technology; Springer: Wiesbaden, Germany, 2022; p. 651. [Google Scholar] [CrossRef]
  104. Chaudhary, V.; Khanna, V.; Ahmed Awan, H.T.; Singh, K.; Khalid, M.; Mishra, Y.K.; Bhansali, S.; Li, C.Z.; Kaushik, A. Towards hospital-on-chip supported by 2D MXenes-based 5(th) generation intelligent biosensors. Biosens. Bioelectron. 2023, 220, 114847. [Google Scholar] [CrossRef]
  105. Paimard, G.; Ghasali, E.; Baeza, M. Screen-printed electrodes: Fabrication, modification, and biosensing applications. Chemosensors 2023, 11, 113. [Google Scholar] [CrossRef]
  106. Aydin, M.; Aydin, E.B.; Sezgintürk, M.K. Advances in immunosensor technology. Adv. Clin. Chem. 2021, 102, 1–62. [Google Scholar]
  107. Li, Z.; Zhang, J.; Huang, Y.; Zhai, J.; Liao, G.; Wang, Z.; Ning, C. Development of electroactive materials-based immunosensor towards early-stage cancer detection. Coord. Chem. Rev. 2022, 471, 214723. [Google Scholar] [CrossRef]
  108. Cavalcante, F.T.; de A. Falcão, I.R.; da S. Souza, J.E.; Rocha, T.G.; de Sousa, I.G.; Cavalcante, A.L.; de Oliveira, A.L.; de Sousa, M.C.; dos Santos, J.C. Designing of nanomaterials-based enzymatic biosensors: Synthesis, properties, and applications. Electrochem 2021, 2, 149–184. [Google Scholar] [CrossRef]
  109. Xie, Y.; Liu, T.; Chu, Z.; Jin, W. Recent advances in electrochemical enzymatic biosensors based on regular nanostructured materials. J. Electroanal. Chem. 2021, 893, 115328. [Google Scholar] [CrossRef]
  110. Wang, M.; Li, L.; Zhang, L.; Zhao, J.; Jiang, Z.; Wang, W. Peptide-derived biosensors and their applications in tumor immunology-related detection. Anal. Chem. 2021, 94, 431–441. [Google Scholar] [CrossRef]
  111. Vanova, V.; Mitrevska, K.; Milosavljevic, V.; Hynek, D.; Richtera, L.; Adam, V. Peptide-based electrochemical biosensors utilized for protein detection. Biosens. Bioelectron. 2021, 180, 113087. [Google Scholar] [CrossRef]
  112. Zhang, L.; Guo, W.; Lu, Y. Advances in cell-free biosensors: Principle, mechanism, and applications. Biotechnol. J. 2020, 15, 2000187. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, K.; Fan, Z.; Ding, Y.; Xie, M. A pH-engineering regenerative DNA tetrahedron ECL biosensor for the assay of SARS-CoV-2 RdRp gene based on CRISPR/Cas12a trans-activity. Chem. Eng. J. 2022, 429, 132472. [Google Scholar] [CrossRef] [PubMed]
  114. Glatz, R.T.; Ates, H.C.; Mohsenin, H.; Weber, W.; Dincer, C. Designing electrochemical microfluidic multiplexed biosensors for on-site applications. Anal. Bioanal. Chem. 2022, 414, 6531–6540. [Google Scholar] [CrossRef] [PubMed]
  115. Beduk, T.; Beduk, D.; Hasan, M.R.; Guler Celik, E.; Kosel, J.; Narang, J.; Salama, K.N.; Timur, S. Smartphone-based multiplexed biosensing tools for health monitoring. Biosensors 2022, 12, 583. [Google Scholar] [CrossRef] [PubMed]
  116. Cheng, H.-P.; Yang, T.-H.; Wang, J.-C.; Chuang, H.-S. Recent Trends and Innovations in Bead-Based Biosensors for Cancer Detection. Sensors 2024, 24, 2904. [Google Scholar] [CrossRef] [PubMed]
  117. Shu, T.; Hunter, H.; Zhou, Z.; Sun, Y.; Cheng, X.; Ma, J.; Su, L.; Zhang, X.; Serpe, M.J. Portable point-of-care diagnostic devices: An updated review. Anal. Methods 2021, 13, 5418–5435. [Google Scholar] [CrossRef] [PubMed]
  118. Kharati, M.; Foroutanparsa, S.; Rabiee, M.; Salarian, R.; Rabiee, N.; Rabiee, G. Early diagnosis of multiple sclerosis based on optical and electrochemical biosensors: Comprehensive perspective. Curr. Anal. Chem. 2020, 16, 557–569. [Google Scholar] [CrossRef]
  119. Wang, C.; Liu, Y.; Chen, R.; Wang, X.; Wang, Y.; Wei, J.; Zhang, K.; Zhang, C. Electrochemical biosensing of circulating microRNA-21 in cerebrospinal fluid of medulloblastoma patients through target-induced redox signal amplification. Microchim. Acta 2022, 189, 105. [Google Scholar] [CrossRef] [PubMed]
  120. Song, S.; Lee, J.U.; Jeon, M.J.; Kim, S.; Sim, S.J. Detection of multiplex exosomal miRNAs for clinically accurate diagnosis of Alzheimer’s disease using label-free plasmonic biosensor based on DNA-Assembled advanced plasmonic architecture. Biosens. Bioelectron. 2022, 199, 113864. [Google Scholar] [CrossRef]
  121. Wang, Q.; Wang, J.; Huang, Y.; Du, Y.; Zhang, Y.; Cui, Y.; Kong, D.-m. Development of the DNA-based biosensors for high performance in detection of molecular biomarkers: More rapid, sensitive, and universal. Biosens. Bioelectron. 2022, 197, 113739. [Google Scholar] [CrossRef]
  122. Malon, R.S.; Sadir, S.; Balakrishnan, M.; Córcoles, E.P. Saliva-based biosensors: Noninvasive monitoring tool for clinical diagnostics. Biomed. Res. Int. 2014, 2014, 962903. [Google Scholar] [CrossRef] [PubMed]
  123. John, R.V.; Devasiya, T.; VR, N.; Adigal, S.; Lukose, J.; Kartha, V.; Chidangil, S. Cardiovascular biomarkers in body fluids: Progress and prospects in optical sensors. Biophys. Rev. 2022, 14, 1023–1050. [Google Scholar] [CrossRef] [PubMed]
  124. Guest, F.L.; Guest, P.C.; Martins-de-Souza, D. The emergence of point-of-care blood-based biomarker testing for psychiatric disorders: Enabling personalized medicine. Biomark. Med. 2016, 10, 431–443. [Google Scholar] [CrossRef]
  125. Verber, N. Biochemical Biomarkers of Disease in a Deeply-Phenotyped Cohort of Patients with Motor Neuron Disease. Ph.D. Thesis, University of Sheffield, Sheffield, UK, 2021. [Google Scholar]
  126. Kaštelan, S.; Braš, M.; Pjevač, N.; Bakija, I.; Tomić, Z.; Pjevač Keleminić, N.; Gverović Antunica, A. Tear biomarkers and Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 13429. [Google Scholar] [CrossRef]
  127. Espinosa, P.S.; Rizvi, Z.; Sharma, P.; Hindi, F.; Filatov, A. Neurological complications of coronavirus disease (COVID-19): Encephalopathy, MRI brain and cerebrospinal fluid findings: Case 2. Cureus 2020, 12, e7930. [Google Scholar] [CrossRef]
  128. Dobrocky, T.; Mosimann, P.J.; Zibold, F.; Mordasini, P.; Raabe, A.; Ulrich, C.T.; Gralla, J.; Beck, J.; Piechowiak, E.I. Cryptogenic cerebrospinal fluid leaks in spontaneous intracranial hypotension: Role of dynamic CT myelography. Radiology 2018, 289, 766–772. [Google Scholar] [CrossRef]
  129. Ruggeri, M.; Rojas, A.; Chai, O.; Purzyc, H.; Hanael, E.; Rapoport, K.; Barnoon, I.; Konstantin, L.; Baneth, G.; Shamir, M. Detection of intraspinal Spirocerca lupi in canine cerebrospinal fluid by polymerase chain reaction. J. Comp. Pathol. 2019, 170, 105–112. [Google Scholar] [CrossRef] [PubMed]
  130. Tani, N.; Ikeda, T.; Aoki, Y.; Shida, A.; Oritani, S.; Ishikawa, T. Evaluation of screening for drug use using postmortem prolactin levels in serum and cerebrospinal fluid. Hum. Exp. Toxicol. 2019, 38, 1244–1253. [Google Scholar] [CrossRef]
  131. Emelyanov, A.; Shtam, T.; Kamyshinsky, R.; Garaeva, L.; Verlov, N.; Miliukhina, I.; Kudrevatykh, A.; Gavrilov, G.; Zabrodskaya, Y.; Pchelina, S. Cryo-electron microscopy of extracellular vesicles from cerebrospinal fluid. PLoS ONE 2020, 15, e0227949. [Google Scholar] [CrossRef]
  132. Liang, H.; Zhang, L.; Gao, A.; Li, Y.; Jiang, Z.; Hu, F.; Shao, B.; Liu, Y.; Zhang, X. Risk Factors for Infections Related to Lumbar Drainage in Spontaneous Subarachnoid Hemorrhage. Neurocrit Care 2016, 25, 243–249. [Google Scholar] [CrossRef]
  133. Hassan, Q.; Li, S.; Ferrag, C.; Kerman, K. Electrochemical biosensors for the detection and study of α-synuclein related to Parkinson’s disease—A review. Anal. Chim. Acta 2019, 1089, 32–39. [Google Scholar] [CrossRef]
  134. Wang, Z.; Zhang, Y.; Hu, F.; Ding, J.; Wang, X. Pathogenesis and pathophysiology of idiopathic normal pressure hydrocephalus. CNS Neurosci. Ther. 2020, 26, 1230–1240. [Google Scholar] [CrossRef] [PubMed]
  135. Yang, M.M.; Hader, W.; Bullivant, K.; Brindle, M.; Riva-Cambrin, J. Calgary Shunt Protocol, an adaptation of the Hydrocephalus Clinical Research Network shunt protocol, reduces shunt infections in children. J. Neurosurg. Pediatr. 2019, 23, 559–567. [Google Scholar] [CrossRef] [PubMed]
  136. Richard, K.E.; Block, F.R.; Weiser, R.R. First clinical results with a telemetric shunt-integrated ICP-sensor. Neurol. Res. 1999, 21, 117–120. [Google Scholar] [CrossRef] [PubMed]
  137. Krishnan, S.R.; Arafa, H.M.; Kwon, K.; Deng, Y.; Su, C.-J.; Reeder, J.T.; Freudman, J.; Stankiewicz, I.; Chen, H.-M.; Loza, R.; et al. Continuous, noninvasive wireless monitoring of flow of cerebrospinal fluid through shunts in patients with hydrocephalus. NPJ Digit. Med. 2020, 3, 29. [Google Scholar] [CrossRef] [PubMed]
  138. Qin, C.; Stamos, B.; Dasgupta, P.K. Inline Shunt Flow Monitor for Hydrocephalus. Anal. Chem. 2017, 89, 8170–8176. [Google Scholar] [CrossRef]
  139. Qin, C.; Olivencia-Yurvati, A.H.; Williams Jr, A.G.; Eskildsen, D.; Mallet, R.T.; Dasgupta, P.K. Inline flow sensor for ventriculoperitoneal shunts: Experimental evaluation in swine. Med. Eng. Phys. 2019, 67, 66–72. [Google Scholar] [CrossRef] [PubMed]
  140. Gamero, M.; Kim, W.S.; Hong, S.; Vorobiev, D.; Morgan, C.D.; Park, S.I. Multimodal Sensing Capabilities for the Detection of Shunt Failure. Sensors 2021, 21, 1747. [Google Scholar] [CrossRef] [PubMed]
  141. Pennell, T.; Yi, J.L.; Kaufman, B.A.; Krishnamurthy, S. Noninvasive measurement of cerebrospinal fluid flow using an ultrasonic transit time flow sensor: A preliminary study. J. Neurosurg. Pediatr. 2016, 17, 270–277. [Google Scholar] [CrossRef]
  142. Senel, M.; Dervisevic, E.; Alhassen, S.; Dervisevic, M.; Alachkar, A.; Cadarso, V.J.; Voelcker, N.H. Microfluidic Electrochemical Sensor for Cerebrospinal Fluid and Blood Dopamine Detection in a Mouse Model of Parkinson’s Disease. Anal. Chem. 2020, 92, 12347–12355. [Google Scholar] [CrossRef]
  143. Jalalvand, E.; Robertson, B.; Tostivint, H.; Löw, P.; Wallén, P.; Grillner, S. Cerebrospinal Fluid-Contacting Neurons Sense pH Changes and Motion in the Hypothalamus. J. Neurosci. 2018, 38, 7713–7724. [Google Scholar] [CrossRef] [PubMed]
  144. Mani, G.K.; Miyakoda, K.; Saito, A.; Yasoda, Y.; Kajiwara, K.; Kimura, M.; Tsuchiya, K. Microneedle pH Sensor: Direct, Label-Free, Real-Time Detection of Cerebrospinal Fluid and Bladder pH. ACS Appl. Mater. Interfaces 2017, 9, 21651–21659. [Google Scholar] [CrossRef]
  145. Booth, M.A.; Gowers, S.A.N.; Hersey, M.; Samper, I.C.; Park, S.; Anikeeva, P.; Hashemi, P.; Stevens, M.M.; Boutelle, M.G. Fiber-Based Electrochemical Biosensors for Monitoring pH and Transient Neurometabolic Lactate. Anal. Chem. 2021, 93, 6646–6655. [Google Scholar] [CrossRef] [PubMed]
  146. Schirinzi, T.; Cordella, A.; Mercuri, N.B.; D’Amico, A.; Palombi, A.; Zompanti, A.; Grasso, S.; Pennazza, G.; Santonico, M. Design of an Innovative Methodology for Cerebrospinal Fluid Analysis: Preliminary Results. Sensors 2021, 21, 3767. [Google Scholar] [CrossRef]
  147. Goh, J.H.; Mason, A.; Al-Shamma’a, A.I.; Field, M.; Shackcloth, M.; Browning, P. Non Invasive Microwave Sensor for the Detection of Lactic Acid in Cerebrospinal Fluid (CSF). J. Phys. Conf. Ser. 2011, 307, 012017. [Google Scholar] [CrossRef]
  148. Stephani, C.; Choi, A.H.K.; Moerer, O. Point-of-care detection of lactate in cerebrospinal fluid. Intensive Care Med. Exp. 2021, 9, 18. [Google Scholar] [CrossRef]
  149. Fisher, O.; Benson, R.A.; Imray, C.H. The clinical application of purine nucleosides as biomarkers of tissue Ischemia and hypoxia in humans in vivo. Biomark. Med. 2019, 13, 953–964. [Google Scholar] [CrossRef]
  150. Currano, L.J.; Sage, F.C.; Hagedon, M.; Hamilton, L.; Patrone, J.; Gerasopoulos, K. Wearable Sensor System for Detection of Lactate in Sweat. Sci. Rep. 2018, 8, 15890. [Google Scholar] [CrossRef]
  151. Yang, X.; Fu, T.; Kota, P.K.; Tjia, M.; Nguyen, C.M.; Chiao, J.-C. Lactate Sensors on Flexible Substrates. Biosensors 2016, 6, 48. [Google Scholar] [CrossRef]
  152. Abbasi, H.Y.; Tehrani, Z.; Devadoss, A.; Ali, M.M.; Moradi-Bachiller, S.; Albani, D.; Guy, O.J. Graphene based electrochemical immunosensor for the ultra-sensitive label free detection of Alzheimer’s beta amyloid peptides Aβ (1–42). Nanoscale Adv. 2021, 3, 2295–2304. [Google Scholar] [CrossRef]
  153. Nabers, A.; Ollesch, J.; Schartner, J.; Kötting, C.; Genius, J.; Hafermann, H.; Klafki, H.; Gerwert, K.; Wiltfang, J. Amyloid-β-Secondary Structure Distribution in Cerebrospinal Fluid and Blood Measured by an Immuno-Infrared-Sensor: A Biomarker Candidate for Alzheimer’s Disease. Anal. Chem. 2016, 88, 2755–2762. [Google Scholar] [CrossRef] [PubMed]
  154. Valkova, P.; Pohanka, M. Novel Trends in Electrochemical Biosensors for Early Diagnosis of Alzheimer’s Disease. Int. J. Anal. Chem. 2021, 2021, 9984876. [Google Scholar] [CrossRef] [PubMed]
  155. Karki, H.P.; Jang, Y.; Jung, J.; Oh, J. Advances in the development paradigm of biosample-based biosensors for early ultrasensitive detection of alzheimer’s disease. J. Nanobiotechnology 2021, 19, 72. [Google Scholar] [CrossRef] [PubMed]
  156. Scarano, S.; Lisi, S.; Ravelet, C.; Peyrin, E.; Minunni, M. Detecting Alzheimer’s disease biomarkers: From antibodies to new bio-mimetic receptors and their application to established and emerging bioanalytical platforms—A critical review. Anal. Chim. Acta 2016, 940, 21–37. [Google Scholar] [CrossRef] [PubMed]
  157. Ameri, M.; Shabaninejad, Z.; Movahedpour, A.; Sahebkar, A.; Mohammadi, S.; Hosseindoost, S.; Ebrahimi, M.S.; Savardashtaki, A.; Karimipour, M.; Mirzaei, H. Biosensors for detection of Tau protein as an Alzheimer’s disease marker. Int. J. Biol. Macromol. 2020, 162, 1100–1108. [Google Scholar] [CrossRef] [PubMed]
  158. Kim, S.; Wark, A.W.; Lee, H.J. Femtomolar detection of tau proteins in undiluted plasma using surface plasmon resonance. Anal. Chem. 2016, 88, 7793–7799. [Google Scholar] [CrossRef] [PubMed]
  159. Carlin, N.; Martic-Milne, S. Anti-tau antibodies based electrochemical sensor for detection of tau protein biomarkers. J. Electrochem. Soc. 2018, 165, G3018. [Google Scholar] [CrossRef]
  160. Li, D.; Scarano, S.; Lisi, S.; Palladino, P.; Minunni, M. Real-time tau protein detection by sandwich-based piezoelectric biosensing: Exploring tubulin as a mass enhancer. Sensors 2018, 18, 946. [Google Scholar] [CrossRef] [PubMed]
  161. Grieshaber, D.; MacKenzie, R.; Vörös, J.; Reimhult, E. Electrochemical Biosensors-Sensor Principles and Architectures. Sensors 2008, 8, 1400–1458. [Google Scholar] [CrossRef]
  162. Kim, H.J.; Park, D.; Yun, G.; Kim, H.; Kim, H.-G.; Lee, K.M.; Hong, I.K.; Park, K.-C.; San Lee, J.; Hwang, K.S. Screening for cerebral amyloid angiopathy based on serological biomarkers analysis using a dielectrophoretic force-driven biosensor platform. Lab A Chip 2021, 21, 4557–4565. [Google Scholar] [CrossRef]
  163. Özcan, N.; Medetalibeyoglu, H.; Akyıldırım, O.; Atar, N.; Yola, M.L. Electrochemical detection of amyloid-β protein by delaminated titanium carbide MXene/multi-walled carbon nanotubes composite with molecularly imprinted polymer. Mater. Today Commun. 2020, 23, 101097. [Google Scholar] [CrossRef]
  164. You, M.; Yang, S.; An, Y.; Zhang, F.; He, P. A novel electrochemical biosensor with molecularly imprinted polymers and aptamer-based sandwich assay for determining amyloid-β oligomer. J. Electroanal. Chem. 2020, 862, 114017. [Google Scholar] [CrossRef]
  165. Rezabakhsh, A.; Rahbarghazi, R.; Fathi, F. Surface plasmon resonance biosensors for detection of Alzheimer’s biomarkers; an effective step in early and accurate diagnosis. Biosens. Bioelectron. 2020, 167, 112511. [Google Scholar] [CrossRef] [PubMed]
  166. Sangubotla, R.; Kim, J. Fiber-optic biosensor based on the laccase immobilization on silica-functionalized fluorescent carbon dots for the detection of dopamine and multi-color imaging applications in neuroblastoma cells. Mater. Sci. Eng. C 2021, 122, 111916. [Google Scholar] [CrossRef]
  167. Ganesana, M.; Trikantzopoulos, E.; Maniar, Y.; Lee, S.T.; Venton, B.J. Development of a novel micro biosensor for in vivo monitoring of glutamate release in the brain. Biosens. Bioelectron. 2019, 130, 103–109. [Google Scholar] [CrossRef]
Figure 1. (A) Lumbar puncture is the most commonly used technique for CSF collection. As a biomarker of various central nervous system disorders, it is detectable in CSF [22]. (B) A variety of tests and techniques are available for the analysis of CSF samples, most of which are lab-based and require long processing times and special equipment. Point-of-care biosensors are the fastest approach and can be used in situ for the detection of biomarkers of interest [23]. (C) Electrochemical detection of analytes of interest can be carried out using a working electrode that can be modified for the detection of a particular analyte, after which a signal is transduced to a potentiostat for quantification of the concentration [24].
Figure 1. (A) Lumbar puncture is the most commonly used technique for CSF collection. As a biomarker of various central nervous system disorders, it is detectable in CSF [22]. (B) A variety of tests and techniques are available for the analysis of CSF samples, most of which are lab-based and require long processing times and special equipment. Point-of-care biosensors are the fastest approach and can be used in situ for the detection of biomarkers of interest [23]. (C) Electrochemical detection of analytes of interest can be carried out using a working electrode that can be modified for the detection of a particular analyte, after which a signal is transduced to a potentiostat for quantification of the concentration [24].
Sensors 24 03294 g001aSensors 24 03294 g001b
Figure 2. Electrochemical detection of analytes of interest (this figure was generated using biorender.com, accessed on 22 March 2024).
Figure 2. Electrochemical detection of analytes of interest (this figure was generated using biorender.com, accessed on 22 March 2024).
Sensors 24 03294 g002
Table 1. CSF components in a healthy adult.
Table 1. CSF components in a healthy adult.
QuantityReferences
ColourNo colour[39,40]
pH7.4[41]
Pressure50–200 mm H2O[42]
Overall cell count<5 cells per mL[43]
Red blood cells0[40]
White blood cells0–5 (lymphocytes)[44]
Protein15–40 mg/dL[45]
Glucose50–80 mg/dL[46]
Lactate1–3 mmol/L[46]
MicroorganismsNo[47]
Mg, K, Na, CaTrace [48]
Table 2. Biomarkers associated with the most important neurological diseases.
Table 2. Biomarkers associated with the most important neurological diseases.
Clinical ConditionBiomarkers in CSFReferences
Alzheimer’sAβ1-40, Aβ1-42, tTAU, pTAU, cortisol[77,78]
Multiple sclerosisOCBs (oligoclonal bands)
Increased IgA, IgM
CRTAC, Tetranectin, autotaxin-T
Immunoglobulins: Ig ϒ1, Ig heavy chain V-III, and Ig-k-chain
[79,80]
Spinal cord injuriesPro-inflammatory cytokines in CSF, NSEs, S100β, and NFH[81]
Guillain–Barre SyndromeIncreased CSF protein level[82]
Amyotrophic lateral sclerosisTotal protein concentration, IL-1β, and TNF-α[83,84]
MeningitisIncreased lactate level, CSF glucose/blood glucose < 0.4[85,86]
DementiaTau protein, Aβ1-42, NF light[87,88]
Table 3. Details of various biosensing platforms for various analytes in CSF.
Table 3. Details of various biosensing platforms for various analytes in CSF.
BiomarkerElectrochemical Sensor SubstrateRelated DiseaseLODReferences
Tau proteinBiosensor cell lines,
Sandwich-based antibody
Alzheimer’s 316 pg–100 ng
34 ng/L
[118,123]
1-42, Aβ1-40, Amyloid-βDielectrophoretic force-driven
MXene/multi-wall carbon printed nanotube (molecularly imprinted)
Molecularly imprinted polymers and aptamers
Gold-based plasmon resonance biosensors
Alzheimer’s Pg/mL
1.0 fg mL−1–100.0 fg mL−1
1.22 pg mL−1
2.4 pg/mL
[162,163,164,165]
DopamineAptamer-based biosensor
Microfluidic Au-based biosensor
Silica-functionalised fluorescent carbon dots
Alzheimer’s
Parkinson’s
Parkinson’s
µM
0.1 nM
41.2 nM
[142,164,166]
GlutamateIn vivo sensing biosensor (Pt wire-based)
Carbon–Pt microparticle-based biosensor
Brain Glutamate monitoring
Brain Glutamate
0.044 µM
0.03 µM
[167]
Serotonin Reusable aptasensor (Au-based)
Flexible WS2/Graphene/Polyimide Electrode-based biosensor
Alzheimer’s and Parkinson’s
NA
[130]
Acetylcholine EDOT-based solid-state biosensor
Amperometry biosensors
Alzheimer’s and Parkinson’s [131]
Glycated albumin His6-RAGE VC1-modified electrodes used as biosensors
Aptamer-conjugated magnetic nanoparticles used to precipitate glycated albumin
Alzheimer’s [132]
CortisolNADelirium, bacterial meningitis, Alzheimer’s diseaseNANA
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

Hatami-Fard, G.; Anastasova-Ivanova, S. Advancements in Cerebrospinal Fluid Biosensors: Bridging the Gap from Early Diagnosis to the Detection of Rare Diseases. Sensors 2024, 24, 3294. https://doi.org/10.3390/s24113294

AMA Style

Hatami-Fard G, Anastasova-Ivanova S. Advancements in Cerebrospinal Fluid Biosensors: Bridging the Gap from Early Diagnosis to the Detection of Rare Diseases. Sensors. 2024; 24(11):3294. https://doi.org/10.3390/s24113294

Chicago/Turabian Style

Hatami-Fard, Ghazal, and Salzitsa Anastasova-Ivanova. 2024. "Advancements in Cerebrospinal Fluid Biosensors: Bridging the Gap from Early Diagnosis to the Detection of Rare Diseases" Sensors 24, no. 11: 3294. https://doi.org/10.3390/s24113294

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