At Alfa Cytology, we offer specialized biomarker analysis services tailored for Kidney research and therapeutic development. Our comprehensive biomarker panel is designed to advance the understanding of Kidney disease pathophysiology and support the discovery and characterization of novel drug targets. All services are exclusively focused on supporting drug discovery through preclinical development stages; we do not provide clinical diagnostic services.
The foundation of effective therapeutic intervention lies in the precise identification and characterization of relevant biomarkers. Alfa Cytology’s biomarker discovery services are integral to the early phases of drug development, enabling the detection of molecular indicators that inform target selection, efficacy assessment, and safety profiling. Our discovery approach encompasses high-throughput screening, literature mining, and in silico prediction, followed by rigorous experimental validation to ensure the reliability of candidate biomarkers for advancing preclinical research.
Multi Omics: Leveraging cutting-edge -omics technologies, Alfa Cytology integrates genomics, transcriptomics, proteomics, and metabolomics to provide a holistic view of biological systems relevant to Kidney disease. This comprehensive approach enables the identification of DNA, RNA, protein, and metabolite biomarkers associated with key disease pathways such as cell survival, proliferation, angiogenesis, DNA repair, and metabolic regulation. By mapping multi-layered molecular networks, we elucidate mechanisms underlying Kidney disease progression and therapeutic response.
Candidate Validation: Our candidate validation strategies involve a combination of orthogonal assay systems and cross-platform confirmation to establish robust associations with Kidney pathophysiology. Preliminary screening processes utilize analytical and functional assays to prioritize candidates based on expression patterns, biological relevance, and reproducibility. Criteria for promising candidates include specificity to disease processes, detectability in relevant sample types, and potential for monitoring therapeutic effects in preclinical models.
Diverse Technological Platforms: Alfa Cytology develops and customizes biomarker assays across a range of technological platforms to meet specific research requirements. Our capabilities include the adaptation of immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology and imaging platforms, ensuring flexible and precise measurement of biomarkers relevant to Kidney research.
Immunoassays: We offer a suite of immunoassay formats, including ELISA, chemiluminescent assays, and multiplex bead-based platforms, for sensitive and specific quantification of protein biomarkers.
Mass Spectrometry: Our LC-MS/MS capabilities enable high-resolution, quantitative analysis of proteins, peptides, and metabolites, supporting both targeted and untargeted biomarker discovery.
Flow Cytometry: Flow cytometry is utilized for multiparametric analysis of cell-surface and intracellular markers, facilitating phenotypic and functional characterization of cell populations.
Molecular Diagnostics: We employ PCR, qPCR, and digital PCR platforms for the detection and quantification of DNA and RNA biomarkers, including gene mutations and expression signatures.
Histopathology And Imaging: Advanced histopathology, immunohistochemistry, and imaging techniques are used to localize and quantify biomarker expression within tissue sections, providing spatial context to molecular findings.
Rigorous Method Validation: All assay methods undergo rigorous validation in accordance with relevant regulatory and industry guidelines. Performance characteristics such as sensitivity, specificity, linearity, accuracy, precision, and reproducibility are systematically evaluated. Comprehensive quality control measures, including the use of appropriate standards and controls, are implemented to ensure data integrity and reliability throughout the analytical process.
Our quantitative analysis capabilities enable precise and reproducible measurement of biomarker levels across diverse sample types. We utilize state-of-the-art instrumentation and validated protocols to ensure accurate quantification, supporting robust data generation for preclinical Kidney research.
Sample Analysis: Alfa Cytology processes a wide range of sample types, including tissue homogenates, cell lysates, plasma, serum, urine, and other biological fluids relevant to Kidney studies. Each analysis follows standardized protocols for sample preparation, handling, and storage, with stringent quality assurance procedures to minimize variability and maintain sample integrity.
High Throughput Capabilities: Our high-throughput analytical platforms support multiplexed biomarker analysis, enabling simultaneous measurement of multiple targets within a single sample. This approach enhances efficiency, conserves valuable biological material, and accelerates data acquisition for large-scale preclinical studies.
Gene Target | Biological Function | Application as a Biomarker |
---|---|---|
AKT serine/threonine kinase 1 (AKT1) | AKT serine/threonine kinase 1 (AKT1) is a member of the AKT family of serine/threonine kinases and plays a central role in multiple cellular processes. AKT1 is activated downstream of phosphoinositide 3-kinase (PI3K) signaling in response to growth factors, hormones, and other stimuli. Upon activation, AKT1 phosphorylates a diverse array of substrates involved in the regulation of cell survival, proliferation, metabolism, and growth. Specifically, AKT1 promotes cell survival by inhibiting apoptotic pathways, facilitates glucose uptake and glycogen synthesis, and contributes to cell cycle progression. Dysregulation of AKT1 activity has been implicated in various pathological conditions, including cancer, diabetes, and cardiovascular diseases. | AKT1 has been studied as a biomarker in several contexts, particularly in oncology. Alterations in AKT1 expression levels, mutations (such as E17K), or changes in phosphorylation status have been associated with tumor development, progression, and therapeutic response in certain cancer types. Measurement of AKT1 activity or mutation status may provide information relevant to disease characterization, prognosis, and potential therapeutic strategies, especially in cancers where the PI3K/AKT pathway is frequently altered. |
BRCA2 DNA repair associated (BRCA2) | BRCA2 (Breast Cancer 2, DNA repair associated) encodes a protein that plays a critical role in the maintenance of genomic stability. BRCA2 is primarily involved in the repair of DNA double-strand breaks through homologous recombination. It facilitates the recruitment and loading of RAD51 recombinase onto single-stranded DNA at sites of damage, enabling accurate DNA repair. Loss or dysfunction of BRCA2 impairs homologous recombination, leading to increased genomic instability and susceptibility to tumorigenesis. | BRCA2 is used as a biomarker primarily in oncology, particularly in breast, ovarian, prostate, and pancreatic cancers. Germline or somatic mutations in BRCA2 are associated with increased risk for these malignancies. Detection of BRCA2 mutations can inform cancer risk assessment, guide decisions regarding genetic counseling, and influence strategies for cancer screening and prevention. In patients with cancer, BRCA2 mutation status may also aid in selecting targeted therapies, such as PARP inhibitors, which exploit deficiencies in homologous recombination repair. |
KIT proto-oncogene, receptor tyrosine kinase (KIT) | The KIT proto-oncogene, receptor tyrosine kinase (KIT), encodes a transmembrane receptor that belongs to the type III receptor tyrosine kinase family. KIT is activated by binding its ligand, stem cell factor (SCF), leading to receptor dimerization and autophosphorylation of intracellular tyrosine residues. This activation initiates multiple downstream signaling pathways, including the PI3K/AKT, RAS/MAPK, and JAK/STAT pathways, which regulate cell proliferation, differentiation, survival, and apoptosis. KIT plays essential roles in hematopoiesis, melanogenesis, gametogenesis, and the development of interstitial cells of Cajal in the gastrointestinal tract. | KIT is utilized as a biomarker in several clinical contexts, particularly in the diagnosis and classification of certain tumors. Immunohistochemical detection of KIT protein (CD117) is commonly used to identify gastrointestinal stromal tumors (GISTs), as the majority of these tumors exhibit KIT expression or activating mutations. KIT expression is also assessed in mast cell diseases such as systemic mastocytosis, and in some cases of melanoma, seminoma, and other neoplasms. The presence of KIT mutations or overexpression can provide diagnostic, prognostic, and therapeutic information, especially in guiding the use of tyrosine kinase inhibitors in relevant malignancies. |
albumin (ALB) | Albumin (ALB) is the most abundant plasma protein in humans, primarily synthesized by the liver. Its main biological functions include maintaining colloid osmotic (oncotic) pressure in the blood, which is essential for proper distribution of body fluids between blood vessels and tissues. Albumin also serves as a carrier protein, binding and transporting a variety of endogenous and exogenous substances such as hormones, fatty acids, bilirubin, drugs, and metal ions. Additionally, albumin contributes to antioxidant defense by binding free radicals and has a role in buffering blood pH. | Measurement of serum albumin levels is widely utilized in clinical practice to assess nutritional status, liver function, and kidney function. Hypoalbuminemia (reduced albumin concentration) is frequently observed in conditions such as chronic liver disease, nephrotic syndrome, malnutrition, and systemic inflammation. Conversely, elevated albumin levels are less common and may occur in cases of dehydration. Albumin is also used as a prognostic indicator in various clinical contexts, including critical illness and chronic diseases. |
carbonic anhydrase 9 (CA9) | Carbonic anhydrase 9 (CA9) is a transmembrane enzyme that catalyzes the reversible hydration of carbon dioxide to bicarbonate and protons. This reaction is important for maintaining acid-base balance in various tissues. CA9 is primarily expressed at low levels in normal tissues but is strongly induced under hypoxic conditions, largely through the activity of hypoxia-inducible factor 1-alpha (HIF-1α). By regulating pH at the cellular and extracellular levels, CA9 contributes to cellular adaptation to hypoxia and plays a role in cell proliferation, survival, and migration, particularly in the context of the tumor microenvironment. | CA9 has been utilized as a biomarker for tumor hypoxia and is frequently overexpressed in several solid tumors, including renal cell carcinoma, cervical cancer, and certain other malignancies. Its expression is associated with hypoxic regions within tumors, and detection of CA9 can assist in identifying hypoxic tumor cells. Immunohistochemical assessment of CA9 is used in diagnostic pathology, particularly in distinguishing renal cell carcinoma subtypes. Additionally, CA9 expression levels have been studied as a tool for prognosis and for monitoring response to therapy in oncology. |
epidermal growth factor receptor (EGFR) | Epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein and member of the receptor tyrosine kinase family. Upon binding to its ligands, such as epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-α), EGFR undergoes dimerization and autophosphorylation of its intracellular tyrosine kinase domain. This activation initiates multiple downstream signaling cascades, including the RAS-RAF-MEK-ERK and PI3K-AKT pathways, which regulate cellular processes such as proliferation, differentiation, survival, and migration. EGFR is expressed in various epithelial tissues and plays a critical role in normal development and tissue homeostasis. | EGFR is used as a biomarker in oncology, particularly in non-small cell lung cancer (NSCLC), colorectal cancer, and head and neck squamous cell carcinoma. Assessment of EGFR protein expression, gene amplification, or the presence of activating mutations can provide information relevant to prognosis and potential therapeutic approaches. For example, specific EGFR mutations in NSCLC are associated with sensitivity to tyrosine kinase inhibitors (TKIs). EGFR status may also inform the selection of patients for targeted therapies and can be used to monitor response to treatment. |
kinase insert domain receptor (KDR) | Kinase insert domain receptor (KDR), also known as vascular endothelial growth factor receptor 2 (VEGFR-2), is a type III receptor tyrosine kinase primarily expressed on endothelial cells. KDR functions as the main mediator of the mitogenic, angiogenic, and permeability-enhancing effects of vascular endothelial growth factor (VEGF). Upon VEGF binding, KDR undergoes dimerization and autophosphorylation, activating downstream signaling pathways such as the MAPK/ERK and PI3K/AKT cascades. These pathways regulate endothelial cell proliferation, migration, survival, and new blood vessel formation (angiogenesis), which are critical for both physiological processes such as embryonic development and wound healing, as well as pathological conditions including tumor growth and metastasis. | KDR is used as a biomarker in oncology and vascular medicine. Its expression levels can be assessed in tumor tissue to evaluate angiogenic activity, which may inform prognosis in certain cancers. KDR is also monitored to assess response to anti-angiogenic therapies, including VEGF or VEGFR-targeted agents. Additionally, KDR gene mutations and polymorphisms are studied in relation to cancer susceptibility, therapeutic response, and resistance mechanisms. In non-malignant conditions, KDR expression and circulating levels have been investigated as indicators of endothelial function and vascular injury. |
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 integrates signals from nutrients, growth factors, energy status, and cellular stress to modulate key cellular processes, including protein synthesis, autophagy, lipid biosynthesis, and cytoskeletal organization. MTOR operates as part of two distinct complexes, mTORC1 and mTORC2, each with unique protein components and downstream effects. mTORC1 primarily regulates protein translation and autophagy, while mTORC2 is involved in cytoskeletal organization and cell survival signaling. | MTOR expression levels, activity status, and genetic alterations have been investigated as biomarkers in various diseases, particularly cancer. Aberrant MTOR signaling has been associated with tumor development, progression, and therapeutic response. MTOR pathway activation, assessed by phosphorylation of MTOR or its downstream effectors, has been used to characterize tumor biology and to predict or monitor response to targeted therapies, such as mTOR inhibitors. Additionally, alterations in MTOR or related pathway components have been examined in other conditions, including metabolic and neurological disorders. |
tumor protein p53 (TP53) | Tumor protein p53 (TP53) encodes a transcription factor that plays a central role in regulating the cell cycle, DNA repair, apoptosis, and senescence. Under normal conditions, p53 is maintained at low levels, but in response to cellular stress such as DNA damage, it becomes stabilized and activated. Activated p53 binds to specific DNA sequences to regulate the expression of target genes involved in cell cycle arrest, facilitating DNA repair or, if the damage is irreparable, inducing programmed cell death (apoptosis). Through these mechanisms, p53 acts as a critical tumor suppressor, maintaining genomic stability and preventing the propagation of damaged cells. | TP53 is widely used as a biomarker in oncology due to its frequent mutation in a variety of human cancers. Assessment of TP53 mutation status, protein expression, or functional activity can provide information about tumor development, prognosis, and potential therapeutic response. Immunohistochemical detection of p53 protein accumulation in tumor tissues and molecular analysis of TP53 gene mutations are common approaches in clinical and research settings. These applications are based on the established association between TP53 alterations and tumorigenesis across multiple cancer types. |
vascular endothelial growth factor A (VEGFA) | Vascular endothelial growth factor A (VEGFA) is a key signaling protein that regulates angiogenesis, the formation of new blood vessels from pre-existing vasculature. VEGFA is produced by various cell types, including endothelial cells, macrophages, and tumor cells, in response to hypoxia and other stimuli. It binds primarily to VEGF receptor 1 (VEGFR-1) and VEGF receptor 2 (VEGFR-2) on endothelial cells, activating intracellular signaling pathways that promote endothelial cell proliferation, migration, and survival. Additionally, VEGFA increases vascular permeability and contributes to tissue repair and regeneration processes. Its activity is tightly regulated under physiological conditions, and dysregulation is associated with pathological angiogenesis in diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. | VEGFA is widely studied as a biomarker for angiogenesis-related processes and pathological conditions characterized by abnormal vascular growth. Elevated levels of VEGFA in blood, tissue, or other biological fluids have been associated with various malignancies, including colorectal, breast, and lung cancers, as well as with ocular diseases such as diabetic retinopathy and age-related macular degeneration. Measurement of VEGFA concentrations is utilized in clinical research to assess disease progression, monitor therapeutic response to anti-angiogenic treatments, and provide prognostic information in certain cancer types. |
Explore Research Opportunities with Alfa Cytology. Our biomarker research services offer advanced analytical and discovery capabilities for Kidney therapeutic development, leveraging multi-omics platforms and state-of-the-art assay technologies. Please note that all biomarkers discussed are research targets only and are not claimed as validated or mandatory markers. Our services are strictly focused on preclinical research stages, supporting exploratory studies with scientific rigor and objectivity.
We invite you to discuss your biomarker research needs with Alfa Cytology. Let’s collaborate to advance scientific knowledge and explore new frontiers in Kidney biomarker discovery. Our focus is on exploratory research and scientific partnership, without claims of biomarker validation or necessity.
Make Order
Experimental Scheme
Implementation
Conclusion