Biomarker Analysis Services for Down Syndrome
Drug R&D Solutions

Biomarker Analysis Services for Down Syndrome

Inquiry

Protheragen offers specialized biomarker analysis services dedicated exclusively to supporting drug discovery and preclinical development for Down Syndrome therapeutic research. Our comprehensive biomarker panel is designed to advance the understanding of Down Syndrome pathophysiology, enabling the identification and characterization of molecular targets relevant to disease mechanisms. All services are strictly research-focused and are not intended for clinical diagnostic use.

Biomarker Discovery and Identification

The foundation of effective therapeutic intervention lies in the robust discovery and identification of disease-relevant biomarkers. Protheragen's biomarker discovery services are tailored to facilitate drug development for Down Syndrome by identifying molecular targets and pathways underlying disease progression. Our approach encompasses systematic screening of candidate biomarkers, followed by rigorous validation using state-of-the-art analytical technologies. This process ensures the selection of high-confidence molecular indicators that can inform target engagement, efficacy, and mechanism-of-action studies during preclinical stages.

Multi Omics: Leveraging cutting-edge -omics technologies—including genomics, transcriptomics, proteomics, and metabolomics—Protheragen enables a comprehensive, systems-level study of biological processes relevant to Down Syndrome. Our multi-omics approach facilitates the identification of DNA, RNA, protein, and metabolite biomarkers, offering a holistic view of disease mechanisms. This strategy allows for the exploration of key disease pathways, such as those involved in neurodevelopment, synaptic function, and cellular stress responses, which are pertinent to Down Syndrome research.

Candidate Validation: Candidate biomarker validation at Protheragen involves a suite of strategies, including correlation with Down Syndrome pathophysiology, cross-platform verification, and preliminary functional screening. We prioritize candidates based on their biological relevance, reproducibility, and potential impact on therapeutic development. Criteria for promising biomarkers include strong association with disease mechanisms, robust detection in relevant biological matrices, and suitability for downstream assay development.

Biomarker Assay Development and Validation

Diverse Technological Platforms: Our custom assay development capabilities span a diverse array of technological platforms, allowing precise adaptation to specific research requirements for Down Syndrome. We design and optimize assays for a range of applications, integrating platform selection with project objectives to ensure sensitivity, specificity, and scalability.

Immunoassays: We offer a full suite of immunoassay technologies, including ELISA, chemiluminescent assays, and multiplex immunoassays, enabling sensitive and quantitative detection of protein biomarkers.

Mass Spectrometry: Our mass spectrometry services (LC-MS/MS) provide high-resolution, quantitative analysis of peptides, proteins, and metabolites, supporting biomarker discovery and validation.

Flow Cytometry: Flow cytometry platforms allow for detailed cellular phenotyping and quantification of cell surface and intracellular markers relevant to Down Syndrome research.

Molecular Diagnostics: We employ advanced molecular diagnostic techniques, such as qPCR and digital PCR, for the detection and quantification of nucleic acid biomarkers.

Histopathology And Imaging: Our histopathology and imaging services encompass immunohistochemistry and advanced imaging modalities to localize and quantify biomarker expression in tissue samples.

Rigorous Method Validation: All analytical methods undergo rigorous validation according to established research guidelines. Performance characteristics—including sensitivity, specificity, linearity, reproducibility, and robustness—are systematically evaluated. Comprehensive quality control measures are implemented throughout the workflow to ensure data integrity and reproducibility.

Biomarker Quantitative and Qualitative Analysis

Protheragen offers robust quantitative analysis capabilities, enabling precise measurement of biomarker levels across diverse sample types. Our protocols are optimized for accuracy and reproducibility, supporting the generation of reliable data for preclinical research applications.

Sample Analysis: We handle a wide range of biological sample types, including tissue, plasma, serum, cerebrospinal fluid, and cell lysates. Our analysis protocols are standardized and tailored to the unique requirements of each project, incorporating stringent quality control at every step to ensure the integrity and reliability of results.

High Throughput Capabilities: Our high-throughput analytical platforms enable multiplexed biomarker analysis, significantly increasing efficiency and conserving valuable samples. These capabilities facilitate large-scale studies and accelerate preclinical research timelines, while maintaining data quality and consistency.

Key Biomarkers for Down Syndrome Drug Development

Gene Target Biological Function Application as a Biomarker
amyloid beta precursor protein (APP) Amyloid beta precursor protein (APP) is a type I transmembrane glycoprotein that is widely expressed in many tissues, including the central nervous system. APP plays a role in neuronal growth, survival, and synaptic formation and repair. It is processed through two main proteolytic pathways: the non-amyloidogenic pathway, which precludes amyloid beta (Aβ) formation, and the amyloidogenic pathway, which generates Aβ peptides through sequential cleavage by beta- and gamma-secretases. The accumulation of Aβ peptides, particularly Aβ42, is associated with the formation of amyloid plaques in the brain, a hallmark of Alzheimer's disease pathology. Besides its role in Aβ production, APP and its cleavage products have been implicated in cell signaling, neurite outgrowth, and cell adhesion. APP and its proteolytic fragments, especially amyloid beta peptides (such as Aβ40 and Aβ42), are measured in biological fluids such as cerebrospinal fluid (CSF) and plasma as indicators of amyloidogenic processing. Altered levels of these peptides are associated with neurodegenerative conditions, most notably Alzheimer's disease. Quantification of APP-derived peptides is utilized in research and clinical investigations to assess amyloid pathology, monitor disease progression, and evaluate responses to therapeutic interventions targeting amyloid metabolism.
beta-2-microglobulin (B2M) Beta-2-microglobulin (B2M) is a low molecular weight protein that forms the light chain component of major histocompatibility complex (MHC) class I molecules, which are present on the surface of nearly all nucleated cells. B2M associates non-covalently with the alpha chain of MHC class I molecules, contributing to their structural stability and facilitating the presentation of endogenous peptide antigens to cytotoxic T lymphocytes. B2M is continuously shed from cell surfaces into the bloodstream and is filtered by the kidneys, with subsequent reabsorption and catabolism in the proximal tubules. B2M is commonly measured in serum, plasma, and urine as a biomarker. Its concentrations are used in clinical settings to assess renal function, as elevated serum levels can indicate impaired glomerular filtration or tubular dysfunction. B2M is also utilized in the context of hematological malignancies, such as multiple myeloma and certain lymphomas, where increased levels may correlate with tumor burden or disease progression. Additionally, B2M is used to monitor immune activation or dysfunction, including in conditions such as HIV infection and some autoimmune diseases.
cystathionine beta-synthase (CBS) Cystathionine beta-synthase (CBS) is a pyridoxal phosphate-dependent enzyme that catalyzes the condensation of homocysteine and serine to form cystathionine in the transsulfuration pathway. This pathway is essential for the metabolism of sulfur-containing amino acids, facilitating the conversion of homocysteine to cysteine. CBS activity helps regulate homocysteine levels, and its function is critical for maintaining cellular redox balance and proper methylation processes. Mutations in the CBS gene can lead to homocystinuria, a metabolic disorder characterized by elevated homocysteine and associated clinical manifestations. CBS has been investigated as a biomarker in several contexts. Deficient CBS activity or mutations are used in the diagnosis of classical homocystinuria, aiding in the identification and management of this inherited metabolic disorder. Additionally, alterations in CBS expression or activity have been studied in relation to cardiovascular diseases, certain cancers, and neurological disorders, where changes in homocysteine metabolism may have pathophysiological relevance. Measurement of CBS activity, gene mutations, or protein levels in biological samples can provide information relevant to disease diagnosis, prognosis, or therapeutic monitoring.
dipeptidyl peptidase 4 (DPP4) Dipeptidyl peptidase 4 (DPP4), also known as CD26, is a membrane-bound serine exopeptidase expressed on various cell types, including epithelial and endothelial cells, as well as in a soluble form in plasma. DPP4 cleaves dipeptides from the N-terminus of polypeptides, preferentially when the penultimate residue is proline or alanine. This enzymatic activity regulates the biological activity of multiple chemokines, neuropeptides, and peptide hormones such as glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and stromal cell-derived factor-1 (SDF-1/CXCL12). Through these actions, DPP4 is involved in glucose metabolism, immune regulation, signal transduction, and cell adhesion. DPP4 has been studied as a biomarker in several contexts. Its circulating levels and enzymatic activity have been investigated in metabolic disorders, including type 2 diabetes mellitus, where altered DPP4 activity is associated with impaired incretin hormone function. DPP4 expression has also been evaluated in certain cancers, such as hepatocellular carcinoma and colorectal cancer, in relation to tumor progression and prognosis. Additionally, DPP4 has been explored as a biomarker in inflammatory and cardiovascular diseases, reflecting its involvement in immune modulation and endothelial function.
dual specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) Dual specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) is a member of the DYRK family of protein kinases. It is characterized by its ability to autophosphorylate on tyrosine residues and phosphorylate exogenous substrates on serine/threonine residues. DYRK1A plays a crucial role in neurodevelopment, particularly in neuronal proliferation, differentiation, and synaptic plasticity. It is highly expressed in the brain and is involved in the regulation of gene expression, cell cycle progression, and apoptosis. The gene encoding DYRK1A is located on chromosome 21, and its dosage is implicated in the pathogenesis of Down syndrome. DYRK1A is also involved in various signaling pathways, including those regulating cell survival and neurogenesis. Alterations in DYRK1A expression or activity have been studied in the context of neurodevelopmental disorders, including Down syndrome, where gene dosage effects contribute to the phenotype. DYRK1A has also been investigated in neurodegenerative diseases such as Alzheimer's disease, as well as in certain cancers. In these contexts, DYRK1A levels or activity have been evaluated as potential indicators of disease state, progression, or response to therapy. Measurement of DYRK1A may provide information relevant to diagnosis, prognosis, or therapeutic monitoring in research and clinical studies.
epidermal growth factor receptor (EGFR) The epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein that belongs to the receptor tyrosine kinase family. EGFR is activated by binding of specific ligands, such as epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-α), leading to receptor dimerization and autophosphorylation of tyrosine residues in its intracellular domain. This activation initiates multiple downstream signaling pathways, including the RAS-RAF-MEK-ERK and PI3K-AKT cascades, which regulate cellular processes such as proliferation, differentiation, survival, and migration. EGFR is expressed in various tissues and plays a critical role in normal cellular development and tissue homeostasis. EGFR is used as a biomarker in several clinical contexts, particularly in oncology. Its overexpression, gene amplification, or activating mutations have been identified in various cancers, including non-small cell lung cancer (NSCLC), colorectal cancer, and head and neck squamous cell carcinoma. Assessment of EGFR status can inform prognosis and guide therapeutic decisions, such as the use of EGFR-targeted therapies (e.g., tyrosine kinase inhibitors and monoclonal antibodies). EGFR mutation analysis in tumor specimens is commonly employed to predict response to specific targeted treatments.
microtubule associated protein tau (MAPT) Microtubule associated protein tau (MAPT) encodes the tau protein, which is predominantly expressed in neurons of the central nervous system. Tau binds to and stabilizes microtubules, which are essential components of the neuronal cytoskeleton involved in maintaining cell shape, axonal transport, and neuronal polarity. Tau modulates microtubule dynamics through its microtubule-binding domains and is regulated by phosphorylation; hyperphosphorylation of tau reduces its affinity for microtubules and can lead to the formation of insoluble aggregates known as neurofibrillary tangles. These tangles are a pathological hallmark of several neurodegenerative diseases. MAPT and its protein product, tau, are utilized as biomarkers in neurodegenerative disease research and clinical practice. Elevated levels of total tau and phosphorylated tau in cerebrospinal fluid (CSF) are associated with neuronal injury and are measured in the context of Alzheimer's disease and other tauopathies. The presence of tau aggregates in brain tissue is also assessed post-mortem to assist in disease characterization. Measurement of tau species in CSF and, more recently, in blood, is used to support differential diagnosis and monitor disease progression.
oligodendrocyte transcription factor 2 (OLIG2) Oligodendrocyte transcription factor 2 (OLIG2) is a basic helix-loop-helix (bHLH) transcription factor that plays a critical role in the development of the central nervous system. OLIG2 is essential for the specification and differentiation of oligodendrocyte precursor cells, which give rise to oligodendrocytes—the myelinating cells of the central nervous system. It is also involved in the development of motor neurons and the maintenance of neural progenitor populations during embryogenesis. OLIG2 regulates the expression of genes required for lineage commitment, proliferation, and maturation of these neural cell types. OLIG2 is utilized as a biomarker for identifying cells of oligodendrocyte lineage in both developmental and pathological contexts. In neuropathology, OLIG2 immunohistochemistry is commonly used to distinguish oligodendroglial tumors, such as oligodendrogliomas, from other gliomas and central nervous system neoplasms. Its expression is also assessed in studies of demyelinating diseases and neural cell lineage tracing. The presence of OLIG2 in tumor samples can assist in tumor classification and diagnosis.
synuclein alpha (SNCA) Synuclein alpha (SNCA) encodes the protein alpha-synuclein, which is abundantly expressed in the central nervous system, particularly at presynaptic nerve terminals. Alpha-synuclein is involved in the regulation of synaptic vesicle trafficking and neurotransmitter release. It is believed to play a role in synaptic plasticity and neuronal differentiation. The protein is natively unfolded but can adopt different conformations depending on its environment. Alpha-synuclein is also implicated in the maintenance of neuronal integrity and may participate in the regulation of dopamine neurotransmission. Alpha-synuclein is used as a biomarker primarily in neurodegenerative diseases, particularly synucleinopathies such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Its pathological aggregation into insoluble fibrils and Lewy bodies is a characteristic feature of these disorders. Measurement of alpha-synuclein levels in cerebrospinal fluid, blood, or other tissues has been explored for diagnostic, prognostic, and disease-monitoring purposes. Altered levels or aggregation patterns of alpha-synuclein are associated with disease states, making it a candidate for biomarker studies in neurodegenerative conditions.
translocator protein (TSPO) The translocator protein (TSPO), formerly known as the peripheral benzodiazepine receptor (PBR), is an 18 kDa protein primarily located on the outer mitochondrial membrane. TSPO is involved in several biological processes, including the transport of cholesterol into mitochondria, which is a critical step in steroidogenesis. It also participates in the regulation of mitochondrial function, cell proliferation, apoptosis, and the cellular response to stress. TSPO is expressed in various tissues, with high levels in steroidogenic organs, and is upregulated in activated microglia and macrophages during neuroinflammatory and immune responses. TSPO has been widely used as a biomarker for neuroinflammation, particularly in the context of central nervous system (CNS) disorders. Its expression increases in activated microglia and astrocytes, which are associated with inflammatory processes in the brain. Positron emission tomography (PET) imaging using radioligands that bind to TSPO enables the in vivo visualization and quantification of neuroinflammatory activity in conditions such as multiple sclerosis, Alzheimer's disease, Parkinson's disease, and traumatic brain injury. TSPO expression has also been investigated as a biomarker in peripheral inflammation and certain cancers.

Partner with Protheragen to Advance Innovation

Explore Research Opportunities with Protheragen. Our biomarker research services for Down Syndrome provide a comprehensive suite of capabilities for the exploratory identification, characterization, and analysis of molecular targets relevant to therapeutic development. All biomarkers discussed are research targets only; we do not claim any as validated or mandatory for any application. Our focus is exclusively on preclinical research stages, and we maintain scientific objectivity throughout all projects.

We invite you to connect with Protheragen to discuss collaborative opportunities in exploratory biomarker research for Down Syndrome. Our team is committed to advancing scientific knowledge through objective, preclinical research partnerships.

HOW WE WORK

Make Order

Make Order

Experimental Scheme

Experimental Scheme

Implementation

Implementation

Conclusion

Conclusion
Make an Inquiry