At Protheragen, we offer specialized biomarker analysis services dedicated to supporting Fragile X Syndrome research and therapeutic development. Our comprehensive biomarker panel is designed to advance the understanding of disease pathophysiology and facilitate the discovery of novel drug targets. All our services are exclusively focused on drug discovery and preclinical development stages; we do not provide clinical diagnostic services.
The foundation of effective therapeutic intervention lies in the precise identification and characterization of disease-relevant biomarkers. Protheragen’s biomarker discovery services are tailored to accelerate drug development by systematically screening and validating molecular indicators associated with Fragile X Syndrome. Our process encompasses high-throughput screening, in-depth data analysis, and rigorous validation to ensure robust candidate selection for further preclinical investigation.
Multi Omics: Our approach leverages cutting-edge -omics technologies, including genomics, transcriptomics, proteomics, and metabolomics, to enable a comprehensive study of biological systems implicated in Fragile X Syndrome. Through integrated analysis of DNA, RNA, protein, and metabolite profiles, we identify and characterize biomarkers that reflect underlying disease mechanisms. This multi-layered strategy allows for the elucidation of critical pathways such as synaptic plasticity, neuronal development, and signaling cascades relevant to Fragile X Syndrome pathophysiology.
Candidate Validation: Candidate validation at Protheragen involves a suite of strategies designed to confirm the association of potential biomarkers with Fragile X Syndrome pathophysiology. Preliminary screening processes incorporate both in vitro and in vivo models, with candidates prioritized based on biological relevance, reproducibility, and translational potential. Criteria for promising candidates include differential expression, mechanistic linkage to disease pathways, and feasibility for downstream assay development.
Diverse Technological Platforms: We offer custom assay development capabilities utilizing a diverse range of technological platforms. Our laboratory infrastructure supports the adaptation of analytical platforms to meet specific research requirements, ensuring optimal sensitivity, specificity, and throughput for the detection and quantification of Fragile X Syndrome–associated biomarkers.
Immunoassays: Our services include the development and implementation of ELISA, chemiluminescent, and multiplex immunoassays for the sensitive and specific detection of protein biomarkers.
Mass Spectrometry: We employ advanced LC-MS/MS platforms for the precise quantification and characterization of proteins, peptides, and metabolites relevant to Fragile X Syndrome.
Flow Cytometry: Our flow cytometry solutions enable high-dimensional phenotyping and quantitation of cellular biomarkers in diverse sample types.
Molecular Diagnostics: We utilize PCR-based and next-generation sequencing methods for the detection of genetic and transcriptomic biomarkers, including gene expansions and expression profiling.
Histopathology And Imaging: Tissue-based analyses are supported by histopathology and advanced imaging techniques to localize and quantify biomarker expression in situ.
Rigorous Method Validation: All assay methods undergo rigorous validation in accordance with recognized guidelines to ensure accuracy, precision, sensitivity, and specificity. Performance characteristics are systematically evaluated, and robust quality control measures are implemented throughout the process to maintain data integrity and reproducibility.
Protheragen offers quantitative analysis capabilities across a spectrum of biomarker types, utilizing validated protocols to ensure reliable measurement of analyte concentrations. Our quantitative platforms support both absolute and relative quantification, tailored to the requirements of preclinical research.
Sample Analysis: We handle a wide array of sample types, including cell lysates, tissue homogenates, and biofluids. Our analysis protocols are optimized for each matrix, with stringent quality measures such as sample tracking, contamination control, and replicate analysis to ensure data reliability.
High Throughput Capabilities: Our high-throughput analytical platforms enable multiplexed biomarker analysis, facilitating the efficient processing of large sample cohorts. These capabilities provide significant advantages in terms of speed, data richness, and sample conservation, supporting comprehensive preclinical studies.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| bromodomain containing 4 (BRD4) | BRD4 (bromodomain containing 4) is a member of the BET (bromodomain and extraterminal domain) protein family. It functions as an epigenetic regulator by recognizing and binding to acetylated lysine residues on histone tails via its bromodomains. BRD4 plays a critical role in regulating gene transcription by recruiting transcriptional machinery, such as P-TEFb (positive transcription elongation factor b), to promoter and enhancer regions. This facilitates the transition of RNA polymerase II into productive elongation during transcription. BRD4 is involved in cell cycle progression, especially at the G1/S transition, and has been implicated in the regulation of genes associated with proliferation, inflammation, and cellular stress responses. | BRD4 expression and activity have been studied as potential biomarkers in various diseases, particularly in cancer. Elevated BRD4 levels or altered localization have been reported in certain malignancies, including hematological cancers and solid tumors, and are associated with disease progression and prognosis in some contexts. BRD4 status has also been investigated in relation to response to BET inhibitors, which are under evaluation as targeted therapies. In addition, BRD4 has been explored as a biomarker in inflammatory and fibrotic disorders, where its expression may reflect disease activity. |
| fragile X messenger ribonucleoprotein 1 (FMR1) | Fragile X messenger ribonucleoprotein 1 (FMR1) encodes the FMRP protein, which is an RNA-binding protein involved in the regulation of synaptic plasticity and neuronal development. FMRP is primarily expressed in the brain and plays a key role in the transport, stability, and translation of specific mRNAs at synapses. It is involved in the modulation of synaptic connections by regulating the local protein synthesis necessary for synaptic maturation and function. Loss or reduction of FMRP disrupts normal neural development and is associated with cognitive impairment. | FMR1 is used as a biomarker in the diagnosis of fragile X syndrome, the most common inherited form of intellectual disability and a leading monogenic cause of autism spectrum disorder. Molecular testing for FMR1 gene expansions, particularly CGG trinucleotide repeat expansions in the 5' untranslated region, can identify individuals with full mutations, premutations, or intermediate alleles. Assessment of FMR1 status is applied in clinical and carrier screening, prenatal diagnosis, and in the evaluation of individuals with unexplained intellectual disability or related neurodevelopmental conditions. |
| gamma-aminobutyric acid type A receptor subunit delta (GABRD) | The gamma-aminobutyric acid type A receptor subunit delta (GABRD) encodes the delta subunit of the GABA-A receptor, a ligand-gated chloride channel that mediates inhibitory neurotransmission in the central nervous system. GABA-A receptors are pentameric complexes composed of various subunits, and the incorporation of the delta subunit typically results in extrasynaptic receptors with high sensitivity to GABA and low desensitization. These delta-containing GABA-A receptors are primarily responsible for mediating tonic inhibitory currents, which regulate neuronal excitability and network activity. The delta subunit is most abundantly expressed in the cerebellum, thalamus, and hippocampus, contributing to the modulation of processes such as motor coordination, anxiety, and responses to neurosteroids. | Altered expression or function of GABRD has been studied in relation to several neurological and psychiatric conditions. Changes in GABRD levels have been observed in disorders such as epilepsy, particularly forms characterized by abnormal inhibitory signaling, as well as in mood disorders and autism spectrum disorders. Measurement of GABRD expression or detection of genetic variants in the GABRD gene has been explored in research settings as a potential indicator of disease state, susceptibility, or treatment response in these conditions. |
| glutamate metabotropic receptor 5 (GRM5) | Glutamate metabotropic receptor 5 (GRM5), also known as mGluR5, is a G protein-coupled receptor predominantly expressed in the central nervous system. It binds the neurotransmitter glutamate, the primary excitatory neurotransmitter in the brain. Upon activation, GRM5 stimulates phospholipase C via Gq proteins, leading to the production of inositol trisphosphate (IP3) and diacylglycerol (DAG), which in turn promote intracellular calcium release and activation of protein kinase C. GRM5 is involved in modulating synaptic plasticity, neuronal excitability, and various signaling pathways underlying learning, memory, and neurodevelopment. It also plays a role in the regulation of motor control, anxiety, and nociception. | GRM5 expression and signaling have been studied as potential biomarkers in several neurological and psychiatric conditions. Altered GRM5 levels or activity have been reported in disorders such as schizophrenia, autism spectrum disorder, fragile X syndrome, Alzheimer's disease, and Parkinson's disease. In addition, GRM5 has been investigated using molecular imaging techniques, such as positron emission tomography (PET), to assess receptor availability and distribution in vivo. These studies have explored correlations between GRM5 expression and disease states, progression, or response to therapeutic interventions. |
| glycogen synthase kinase 3 beta (GSK3B) | Glycogen synthase kinase 3 beta (GSK3B) is a serine/threonine protein kinase that plays a central role in multiple cellular processes, including glycogen metabolism, cell cycle regulation, gene expression, and apoptosis. Originally identified for its role in phosphorylating and inhibiting glycogen synthase, GSK3B is now recognized as a key regulator in signaling pathways such as Wnt/β-catenin, insulin, and neurotrophic signaling. It is involved in modulating the activity of various transcription factors and is implicated in neuronal development, synaptic plasticity, and the cellular response to stress. GSK3B activity is tightly regulated by phosphorylation and protein-protein interactions, reflecting its broad impact on cellular physiology. | GSK3B has been studied as a biomarker in several disease contexts, most notably in neurodegenerative disorders such as Alzheimer's disease, where altered expression or activity of GSK3B has been associated with tau hyperphosphorylation and neurofibrillary tangle formation. It has also been investigated in the context of psychiatric disorders, diabetes, and various cancers, where changes in GSK3B levels or phosphorylation status may correlate with disease state or progression. Measurement of GSK3B expression or activity in tissues or biofluids is used in research to explore disease mechanisms and assess potential therapeutic responses. |
| histone deacetylase 6 (HDAC6) | Histone deacetylase 6 (HDAC6) is a member of the class IIb histone deacetylase family. Unlike most HDACs, HDAC6 is predominantly localized in the cytoplasm and possesses two catalytic domains. Its primary biological function involves the deacetylation of non-histone proteins, including α-tubulin, Hsp90, and cortactin. Through these activities, HDAC6 regulates various cellular processes such as protein degradation via the aggresome-autophagy pathway, cell motility, and stress responses. HDAC6 also participates in the regulation of cytoskeletal dynamics and cellular trafficking, contributing to the maintenance of protein homeostasis and cellular adaptation to stress. | HDAC6 expression and activity levels have been investigated as potential biomarkers in several disease contexts, particularly in oncology. Altered HDAC6 expression has been reported in various cancer types, including breast, ovarian, and multiple myeloma, where its levels may correlate with disease progression or prognosis. HDAC6 has also been studied as a pharmacodynamic biomarker in the context of HDAC inhibitor therapies, where changes in its activity or substrate acetylation status may reflect therapeutic response. Additionally, HDAC6 levels have been explored in neurodegenerative disorders, as its function is implicated in protein aggregation and clearance. |
| matrix metallopeptidase 9 (MMP9) | Matrix metallopeptidase 9 (MMP9) is a member of the matrix metalloproteinase (MMP) family, which comprises zinc-dependent endopeptidases involved in the degradation of extracellular matrix (ECM) components. MMP9 specifically cleaves type IV and V collagens, elastin, and gelatin, facilitating processes such as tissue remodeling, wound healing, and angiogenesis. It is secreted as an inactive proenzyme and activated extracellularly. MMP9 activity is tightly regulated at the levels of gene expression, activation of the zymogen, and inhibition by endogenous tissue inhibitors of metalloproteinases (TIMPs). MMP9 is produced by various cell types, including neutrophils, macrophages, and epithelial cells, and is involved in normal physiological processes as well as pathological conditions characterized by ECM remodeling. | MMP9 has been studied as a biomarker in a variety of clinical contexts, particularly those involving inflammation, tissue remodeling, and cancer. Elevated levels of MMP9 have been observed in serum, plasma, or tissue samples from patients with cardiovascular diseases (such as atherosclerosis and acute coronary syndromes), inflammatory conditions (including rheumatoid arthritis and inflammatory bowel disease), and several malignancies (such as colorectal, breast, and lung cancers). In these settings, MMP9 levels have been investigated for their association with disease presence, progression, or prognosis. Additionally, MMP9 has been explored as a marker for monitoring therapeutic response and disease activity in certain conditions. |
| mechanistic target of rapamycin kinase (MTOR) | The mechanistic target of rapamycin kinase (MTOR) is a serine/threonine protein kinase that functions as a central regulator of cell growth, proliferation, metabolism, and survival. MTOR forms the catalytic core of two distinct multiprotein complexes: mTORC1 and mTORC2. mTORC1 integrates signals from nutrients, growth factors, energy status, and stress to regulate processes such as protein synthesis, autophagy, and lipid metabolism primarily via downstream effectors like S6K and 4E-BP1. mTORC2 is involved in the regulation of cytoskeletal organization and controls cell survival and metabolism through phosphorylation of AGC family kinases, including AKT. MTOR signaling is highly conserved and plays a critical role in cellular and organismal homeostasis. | MTOR expression levels, mutations, and pathway activation status have been investigated as biomarkers in various clinical contexts, particularly in oncology. Aberrant MTOR signaling has been associated with multiple cancer types, making it relevant for assessing tumor progression, therapeutic response, and prognosis. MTOR pathway components are also evaluated as pharmacodynamic biomarkers to monitor the efficacy of mTOR inhibitors in clinical trials and treatment settings. Additionally, alterations in MTOR activity have been explored as potential indicators in metabolic diseases and certain neurodegenerative disorders. |
| methyl-CpG binding protein 2 (MECP2) | Methyl-CpG binding protein 2 (MECP2) is a nuclear protein that binds specifically to methylated cytosine residues in DNA, particularly at CpG dinucleotides. MECP2 functions as a transcriptional regulator, primarily acting as a transcriptional repressor by recruiting co-repressor complexes, such as histone deacetylases, to methylated DNA regions. This activity contributes to chromatin condensation and gene silencing. MECP2 is highly expressed in the central nervous system and plays a critical role in neuronal maturation, synaptic function, and maintenance of neural networks. Mutations in the MECP2 gene are known to cause Rett syndrome, a severe neurodevelopmental disorder. | MECP2 is utilized as a molecular biomarker in the context of neurodevelopmental disorders, most notably Rett syndrome, where mutations in the MECP2 gene are frequently detected. Genetic testing for MECP2 mutations is used to support the diagnosis of Rett syndrome and related neurodevelopmental conditions. Additionally, altered MECP2 expression or mutation status has been investigated in other neurological and psychiatric disorders, as well as in certain cancers, to explore associations with disease phenotype or progression. |
| ribosomal protein S6 kinase B1 (RPS6KB1) | Ribosomal protein S6 kinase B1 (RPS6KB1), also known as p70 S6 kinase, is a serine/threonine kinase that plays a central role in the regulation of cell growth, proliferation, and metabolism. It is a downstream effector of the mechanistic target of rapamycin (mTOR) signaling pathway. Upon activation by mTOR complex 1 (mTORC1), RPS6KB1 phosphorylates several substrates, including the ribosomal protein S6, leading to increased translation of mRNAs involved in cell cycle progression and protein synthesis. RPS6KB1 is also implicated in the regulation of cell size, glucose homeostasis, and autophagy. | RPS6KB1 expression and/or activation status has been investigated as a biomarker in various cancer types, including breast, gastric, and prostate cancers. Its overexpression or increased phosphorylation has been associated with tumor progression, poor prognosis, and resistance to certain therapies. Measurement of RPS6KB1 levels or activity is used in research settings to assess mTOR pathway activation and to explore its potential as a marker for disease aggressiveness or response to targeted therapies. |
Explore Research Opportunities with Protheragen. Our biomarker research services offer advanced analytical capabilities and scientific expertise to support preclinical studies in Fragile X Syndrome. We emphasize the exploratory and research-oriented nature of our work, providing access to a wide array of molecular targets for investigation. Please note that all biomarkers discussed are considered research targets only; we do not claim any as validated or mandatory for Fragile X Syndrome research. Our focus remains on preclinical research stages, maintaining scientific objectivity and rigor throughout our collaborative efforts.
We welcome discussions about exploratory biomarker research in Fragile X Syndrome. Connect with our team at Protheragen to explore opportunities for scientific collaboration and knowledge exchange, with a shared commitment to advancing preclinical research through objective and innovative approaches.
Make Order
Experimental Scheme
Implementation
Conclusion