VAMP5 (Vesicle-Associated Membrane Protein 5) is a SNARE (Soluble N-ethylmaleimide-sensitive factor Attachment protein REceptor) family protein encoded by the VAMP5 gene in humans. Located on chromosome 2p11.2, it belongs to the synaptobrevin/VAMP family and plays critical roles in membrane fusion events underlying vesicle trafficking . Unlike other VAMPs, VAMP5 is uniquely associated with non-neuronal tissues, including skeletal muscle, heart, and immune cells, and participates in specialized processes such as phagocytosis, myogenesis, and extracellular vesicle (EV) communication .
VAMP5 is a 12.7 kDa protein comprising 109 amino acids (1–72 a.a.) with a hydrophobic transmembrane domain near its C-terminus. The recombinant human VAMP5 protein (produced in E. coli) includes a 37-amino acid His-tag at the N-terminus and lacks glycosylation .
Property | Value |
---|---|
Molecular Weight | 12.7 kDa |
Amino Acid Sequence | MRGSHHHHHH...RWNIRYRIC |
Domains | SNARE motif, transmembrane domain |
Post-Translational Modifications | Non-glycosylated |
VAMP5 exhibits tissue-specific expression, with highest levels in skeletal muscle, heart, and retina, as shown by RNA and protein analyses .
High-Expression Tissues | Low/No-Expression Tissues |
---|---|
Skeletal muscle | Brain (cerebellum, cerebral cortex) |
Heart | Liver |
Retina (Müller cells) | Bone marrow |
Lung | Lymph nodes |
VAMP5 regulates Fcγ receptor-mediated phagocytosis in macrophages by interacting with SNAP23. It localizes to early phagosomes and dissociates via clathrin/dynamin-dependent mechanisms to enable phagosome maturation .
During skeletal muscle differentiation, VAMP5 mRNA and protein levels increase 8–10-fold and 6-fold, respectively. It localizes to plasma membranes and intracellular vesicles, facilitating GLUT4 trafficking and myoblast fusion .
In retinal Müller cells, VAMP5 is enriched in EVs released from endfeet and microvilli. These EVs respond to ischemic conditions, suggesting roles in retinal homeostasis and disease .
Botulism: VAMP5 is a potential substrate for botulinum neurotoxins, though its role in neuronal transmission is limited .
Genetic Polymorphisms: SNPs in VAMP5 (e.g., rs10206961, rs14242) are linked to altered disease susceptibility, though replication studies show mixed results .
Developmental Defects: VAMP5 knockout mice exhibit perinatal lethality with urinary/respiratory abnormalities .
Overexpression/knockdown of VAMP5 in macrophages alters phagocytosis efficiency.
Dissociation of VAMP5 from phagosomes is clathrin-dependent and necessary for SNAP23-mediated maturation .
VAMP5 expression surges during C2C12 myoblast differentiation, correlating with muscle-specific vesicle trafficking .
Immunofluorescence confirms plasma membrane and perinuclear vesicle localization in myotubes .
Müller cell-derived EVs contain VAMP5 and tetraspanins (CD9, CD63).
Ischemia upregulates VAMP5, altering EV secretion patterns in the retina .
VAMP5 interacts with SNARE proteins, notably SNAP23, to mediate membrane fusion. Key partners include:
Recombinant human VAMP5 is produced in E. coli with the following specifications :
Parameter | Detail |
---|---|
Purity | >90% (SDS-PAGE) |
Formulation | 20 mM Tris-HCl, 100 mM NaCl, 1 mM DTT |
Stability | Stable at -20°C; avoid freeze-thaw cycles |
VAMP5 (also known as myobrevin) is a member of the VAMP/synaptobrevin family of proteins that functions as a component of the SNARE (Soluble N-ethylmaleimide-sensitive factor attachment proteins receptor) complex. This complex is fundamental for membrane fusion events and vesicular release, enabling cell-cell communication through vesicular exocytosis and extracellular vesicle (EV) formation. In the central nervous system, VAMP5 appears to play a specialized role in glial cells. Research in mouse models has identified VAMP5 as specifically expressed in Müller cells (a type of radial glial cell in the retina) and present in extracellular vesicles released by these cells . These VAMP5-positive EVs likely serve as important mediators of cellular interactions, facilitating signal exchange and providing structural and trophic support within neural tissues .
VAMP5 demonstrates distinct cell type-specific expression patterns. Based on studies in mouse retina, VAMP5 shows predominant expression in glial cells (specifically Müller cells) while being absent or less expressed in neurons . This contrasts with other VAMP family members such as VAMP1, VAMP2, and VAMP7, which are primarily localized in plexiform layers containing synaptic connections .
For human tissue analysis, researchers should employ techniques such as:
RNA sequencing and qRT-PCR to detect VAMP5 transcripts in different cell populations
Immunolabeling to visualize protein distribution across tissues and within specific cellular compartments
Cell-specific transcript analyses using immunoaffinity-purified cell populations to establish reliable cell-specific expression profiles
Importantly, VAMP5 expression can be modulated by pathological conditions. Research has demonstrated that ischemia induces upregulation of VAMP5 in Müller cells, suggesting its involvement in cellular stress responses .
For reliable detection of VAMP5 in human samples, consider the following methodological approaches:
Antibody-based detection methods:
Immunohistochemistry on tissue sections to visualize VAMP5 distribution across different cell layers
Multi-color immunofluorescence to assess colocalization with cell-specific markers (such as GLUL or RLBP1 for glial cells) and subcellular markers
Immunocytochemistry on isolated cells to examine the intracellular distribution of VAMP5
Electron microscopy with immunogold labeling for ultrastructural localization
Biochemical analysis:
Western blot for detecting and semi-quantifying VAMP5 in cell lysates and extracellular vesicle preparations
Immunoprecipitation followed by mass spectrometry to confirm antibody specificity and identify VAMP5-associated proteins
Antibody validation:
Test antibodies on lysates from cells with VAMP5 knockdown or knockout
Verify specificity through immunoprecipitation coupled with mass spectrometry
Include appropriate negative controls (isotype antibodies) and positive controls (tissues known to express VAMP5)
The punctate distribution of VAMP5 observed in Müller cells suggests its presence in vesicle-like structures, requiring careful consideration of fixation and permeabilization protocols to preserve vesicular structures while allowing antibody access .
VAMP5, as a SNARE protein, plays a crucial role in membrane fusion events associated with extracellular vesicle (EV) biogenesis and release. Research in mouse retinal Müller cells provides insights into potential mechanisms in human cells:
Subcellular localization and EV release:
Transmission electron microscopy with immunogold labeling has localized VAMP5 in multivesicular bodies within Müller cell cytoplasm
VAMP5-positive vesicle-like structures have been observed in the extracellular space adjacent to specialized cellular domains, including endfeet facing the vitreous body and apical microvilli extending into the subretinal space
This distribution pattern suggests that VAMP5 participates in polarized release of EVs from specific cellular domains
Association with EV markers:
VAMP5 colocalizes with tetraspanin proteins CD9 and CD63, established markers of extracellular vesicles
Double-immunogold labeling followed by electron microscopy has confirmed the presence of VAMP5 together with CD9 and CD63 on EV-like structures
Experimental data indicates VAMP5 is present in CD9-positive material secreted by Müller cells but not in CD63-positive material, suggesting association with specific EV subpopulations
Size characterization of VAMP5-positive EVs:
Scanning electron microscopy and nanoparticle tracking analysis have determined that VAMP5-positive EVs typically range from 30-150 nm in diameter
This size range is consistent with small extracellular vesicles including exosomes
For investigating VAMP5's role in human EV biogenesis, researchers should consider:
Live-cell imaging with fluorescently tagged VAMP5 to track vesicle formation and release
Genetic manipulation approaches (CRISPR-Cas9, siRNA) to assess how VAMP5 depletion affects EV production
Proteomic analysis of VAMP5-positive EVs from human samples to determine their molecular composition
Distinguishing VAMP5 from other VAMP family members in functional studies requires multifaceted experimental approaches:
Expression pattern analysis:
Comparative immunolabeling of multiple VAMP proteins in the same tissue sections reveals distinct cellular distribution patterns
RNA-seq and qRT-PCR data from immunoaffinity-purified cell populations can establish cell-specific expression profiles for different VAMPs
In mouse retina, VAMP5 shows predominant expression in Müller glial cells, while VAMP1, VAMP2, and VAMP7 are predominantly located in plexiform layers containing synaptic connections
Subcellular localization:
Transmission electron microscopy with immunogold labeling can reveal the precise subcellular localization of different VAMP proteins
VAMP5 has been localized to multivesicular bodies and extracellular vesicles, while VAMP2 (Synaptobrevin) is a known component of synaptic vesicles in neurons
Regulatory response:
Examination of VAMP expression under pathological conditions (e.g., ischemia) can reveal differential regulation
VAMP5 shows upregulation in response to ischemia, suggesting a specific role in cellular stress responses
Functional analysis techniques:
Selective knockdown/knockout:
Design isoform-specific siRNAs or CRISPR-Cas9 constructs targeting unique regions of VAMP5
Compare phenotypic effects with knockdown of other VAMP family members
Rescue experiments:
Express VAMP5 in cells depleted of other VAMP proteins to assess functional redundancy
Create chimeric VAMP proteins by domain swapping to identify functional domains
Protein interaction analysis:
Perform immunoprecipitation followed by mass spectrometry to identify VAMP5-specific binding partners
Use proximity labeling approaches (BioID, APEX) to map the protein interaction network of VAMP5 versus other VAMPs
Extracellular vesicle characterization:
Compare the molecular composition of EVs containing different VAMP proteins
Assess functional effects of different VAMP-containing EVs on recipient cells
Research in mouse models indicates that VAMP5 expression is dynamically regulated under pathological conditions, particularly ischemia . Understanding the regulatory mechanisms in human cells requires investigation of:
Transcriptional regulation:
Analysis of the VAMP5 promoter region to identify binding sites for transcription factors activated during stress conditions
Chromatin immunoprecipitation (ChIP) assays to confirm transcription factor binding
Reporter gene assays to validate regulatory elements in the VAMP5 promoter
Post-transcriptional regulation:
Examination of microRNA-mediated regulation, as previous studies have identified VAMP5 as a miRNA-regulated target gene in the retina
Analysis of mRNA stability under normal versus stress conditions
Assessment of alternative splicing patterns that might generate stress-specific VAMP5 isoforms
Post-translational modifications:
Mass spectrometry-based proteomic analysis to identify potential phosphorylation, ubiquitination, or other modifications of VAMP5 during stress
Site-directed mutagenesis of potential modification sites to assess their functional significance
Immunoprecipitation under different conditions to detect changes in VAMP5 interaction partners
Experimental models for studying VAMP5 regulation:
Model System | Advantages | Applications |
---|---|---|
In vitro hypoxia/ischemia | Controlled conditions, accessible for molecular manipulation | Identify immediate regulatory mechanisms, study temporal dynamics |
Animal models of ischemia | Physiological relevance, ability to study cell-cell interactions | Validate findings in complex tissue environment, assess long-term effects |
Patient-derived samples | Direct human relevance | Correlate VAMP5 levels with clinical parameters, validate biomarker potential |
To establish causality between VAMP5 upregulation and functional outcomes, researchers should:
Manipulate VAMP5 levels prior to ischemic challenge to assess impact on cell survival
Characterize EVs released under normal versus ischemic conditions to determine how their composition and function change
Evaluate the effect of VAMP5-positive EVs on recipient cells, particularly whether they confer protection against ischemic damage
Isolating and characterizing VAMP5-positive extracellular vesicles requires a systematic approach combining multiple techniques:
Isolation protocol:
Collection of conditioned media from cell cultures or biological fluids
Differential centrifugation sequence:
300g for 10 minutes to remove cells
2,000g for 20 minutes to remove cellular debris
10,000g for 30 minutes to remove large vesicles/microvesicles
100,000g for 70-120 minutes to pellet exosomes and small EVs
Additional enrichment through:
Biophysical characterization:
Nanoparticle tracking analysis (NTA) to determine concentration and size distribution (typically 30-150 nm for VAMP5-containing vesicles)
Transmission electron microscopy (TEM) or scanning electron microscopy (SEM) to visualize vesicle morphology
Dynamic light scattering (DLS) for ensemble measurement of vesicle size distribution
Molecular characterization:
Western blot to detect VAMP5 alongside canonical EV markers (CD9, CD63, CD81)
Immunogold labeling followed by TEM to confirm VAMP5 presence on individual vesicles
Proteomic analysis using mass spectrometry to characterize the complete protein content
Flow cytometry-based analysis of vesicles bound to beads to assess co-expression of VAMP5 with other markers
Critical controls:
Include flow-through fractions lacking EVs as negative controls
Use cell or tissue lysates as positive controls for VAMP5 detection
Employ markers of potential contaminants (e.g., calnexin for endoplasmic reticulum) to assess preparation purity
Research has demonstrated successful isolation of VAMP5-positive EVs from Müller cells using CD9-based immunomagnetic separation, yielding approximately 1000-fold higher particle concentrations compared to unprocessed conditioned media .
Verifying antibody specificity is crucial for reliable VAMP5 research. A comprehensive validation strategy should include:
Biochemical validation:
Immunoprecipitation followed by mass spectrometry:
Immunoprecipitate VAMP5 from tissue or cell lysates using the antibody in question
Analyze the immunoprecipitate by mass spectrometry to confirm VAMP5 as the primary protein
Compare with immunoprecipitation using non-specific IgG
Western blot analysis with appropriate controls:
Detect a band at the expected molecular weight for VAMP5 (~13-14 kDa)
Include lysates from cells overexpressing VAMP5 as positive control
Include lysates from VAMP5-knockdown cells as negative control
Perform peptide competition assays by pre-incubating the antibody with recombinant VAMP5
Imaging-based validation:
Immunofluorescence or immunohistochemistry:
Electron microscopy with immunogold labeling:
Functional validation:
Gene silencing:
Reduce VAMP5 expression using siRNA or CRISPR-Cas9
Demonstrate corresponding decrease in antibody signal
Heterologous expression:
Express human VAMP5 in cells that normally do not express it
Confirm specific detection in transfected versus non-transfected cells
Cross-reactivity testing:
Evaluate potential cross-reactivity with other VAMP family members expressed in the same tissue
Use recombinant proteins of different VAMPs to determine specificity
Previous studies have validated anti-VAMP5 antibody specificity by demonstrating strongly reduced signals in immunoblots and immunohistochemical staining of tissues from VAMP5-deficient mice . Additionally, enrichment of VAMP5 in immunoprecipitated retinal lysates analyzed by mass spectrometry provides further evidence of antibody specificity .
Investigating VAMP5's role in extracellular vesicle-mediated cell-cell communication requires a multidisciplinary approach:
Vesicle characterization and tracking:
Live-cell imaging with fluorescently tagged VAMP5 to visualize vesicle formation, transport, and release
Super-resolution microscopy to track VAMP5-positive vesicle trajectories with nanometer precision
Correlative light and electron microscopy (CLEM) to combine functional imaging with ultrastructural analysis
Single-vesicle analysis techniques to characterize the molecular composition of individual VAMP5-positive EVs
Functional assessment of EV-mediated communication:
Co-culture systems with donor cells (VAMP5-expressing) and recipient cells to evaluate EV transfer
Reporter systems in recipient cells to detect functional effects of VAMP5-positive EVs
Selective depletion of VAMP5-positive EVs using immunoaffinity approaches to assess their specific contribution to cell-cell communication
Microfluidic devices to study EV-mediated communication under controlled conditions
Molecular mechanisms:
Proximity labeling approaches (BioID, APEX) to identify proteins interacting with VAMP5 during EV biogenesis
CRISPR-Cas9 genome editing to create VAMP5 knockout or knock-in cell lines for functional studies
Domain mapping to identify regions of VAMP5 critical for its localization to EVs
Proteomics and transcriptomics of VAMP5-positive EVs to characterize their molecular cargo
Experimental design considerations:
Technique | Application | Key Considerations |
---|---|---|
Transmission electron microscopy with immunogold labeling | Ultrastructural localization of VAMP5 | Preservation of membrane structures, antibody specificity |
Nanoparticle tracking analysis | Size distribution and concentration of VAMP5-positive EVs | Sample purity, standardization of measurement conditions |
Immunoaffinity isolation | Selective enrichment of VAMP5-positive EVs | Antibody specificity, gentle elution conditions to preserve EV integrity |
Mass spectrometry | Comprehensive cargo analysis | Sample preparation, sensitivity for low-abundance proteins |
Co-culture experiments | Functional assessment of EV-mediated effects | Cell type selection, methods to distinguish donor from recipient cells |
Research has shown that VAMP5-positive EVs are released from specific domains of Müller cells, including endfeet facing the vitreous body and apical microvilli extending into the subretinal space . This suggests that VAMP5 may contribute to domain-specific EV release, potentially targeting distinct recipient cell populations.
VAMP5-positive extracellular vesicles show promise for both biomarker development and therapeutic applications:
Biomarker potential:
VAMP5's responsive regulation to ischemic conditions suggests its utility as a stress-response biomarker
The presence of VAMP5 in extracellular vesicles allows for non-invasive sampling from biological fluids
Cell type-specific expression of VAMP5 (e.g., in glial cells) enables identification of vesicles from specific cellular origins
Quantitative changes in VAMP5-positive EVs could indicate altered cellular communication in pathological states
Methodological approaches for biomarker development:
Development of sensitive assays (such as digital ELISA) to detect low levels of VAMP5 in isolated EV fractions
Multiplex analysis combining VAMP5 with other EV markers for improved specificity
Longitudinal studies correlating VAMP5-positive EV levels with disease progression
Machine learning algorithms integrating VAMP5-EV data with other clinical parameters for improved diagnostic accuracy
Therapeutic applications:
Engineered EVs containing VAMP5 could serve as delivery vehicles for therapeutic cargo
VAMP5's association with specific EV subpopulations may allow targeting of particular cell types
Modulation of endogenous VAMP5-positive EV release could enhance protective intercellular communication
Potential clinical applications:
To advance these applications, research should focus on:
Comprehensive characterization of VAMP5-positive EV cargo in human samples
Development of standardized methods for isolation and quantification
Preclinical studies validating biomarker and therapeutic potential
Clinical studies correlating VAMP5-positive EV profiles with disease states and treatment responses
Understanding the similarities and differences between VAMP5 function in model organisms and humans is crucial for translational research:
Evolutionary conservation:
VAMP5 is part of the evolutionarily conserved SNARE protein family essential for membrane fusion events
Basic mechanisms of vesicle fusion and release are likely conserved across species
Domain structure and key functional motifs of VAMP5 show high conservation between rodents and humans
Expression patterns:
In mice, VAMP5 shows cell type-specific expression in retinal Müller cells
Human expression patterns require comprehensive mapping across tissues and cell types
Comparative transcriptomics and proteomics can identify species-specific differences in expression patterns
Functional considerations:
Species differences in extracellular vesicle composition may affect VAMP5's role in intercellular communication
Regulatory mechanisms controlling VAMP5 expression may vary between species
Response to pathological conditions may show species-specific patterns
Methodological approaches for cross-species comparison:
Approach | Application | Advantages |
---|---|---|
Comparative genomics | Analysis of VAMP5 gene structure and regulatory elements | Identifies conserved and divergent regions |
Single-cell transcriptomics | Cell type-specific expression mapping | Reveals cellular specificity across species |
Proteomics | Interactome analysis of VAMP5 | Identifies species-specific protein interactions |
Functional rescue experiments | Expression of human VAMP5 in model organism knockout systems | Directly tests functional conservation |
Translational considerations:
Findings from mouse models should be validated in human cells or tissues before clinical application
Species-specific differences in VAMP5 regulation may affect biomarker development
Therapeutic strategies targeting VAMP5 or its regulatory pathways may require species-specific optimization
Studies in mouse retina have established VAMP5 as a glial-specific SNARE component involved in extracellular vesicle release . Validating these findings in human tissues is essential for translational applications, requiring careful consideration of potential species-specific differences in expression patterns, regulation, and function.
Despite recent advances in understanding VAMP5 biology, several significant questions remain unanswered:
Molecular mechanisms:
What specific cargo molecules are selectively packaged into VAMP5-positive extracellular vesicles?
How does VAMP5 contribute to the specificity of vesicle targeting and release from particular cellular domains?
What protein interactions are critical for VAMP5's function in extracellular vesicle biogenesis?
How does VAMP5 cooperate with other SNARE proteins to mediate membrane fusion events?
Cellular and physiological roles:
What is the complete expression map of VAMP5 across human cell types and tissues?
How do VAMP5-positive extracellular vesicles influence recipient cell function?
What signaling pathways regulate VAMP5 expression and localization?
How does VAMP5-mediated vesicle release contribute to normal tissue homeostasis?
Pathological implications:
How are VAMP5 expression and function altered in human diseases?
Can VAMP5-positive extracellular vesicles serve as reliable biomarkers for specific pathologies?
Does VAMP5 upregulation during stress represent a protective or detrimental response?
Could therapeutic modulation of VAMP5 or VAMP5-positive EVs provide clinical benefit?
Research priorities should include:
Comprehensive characterization of VAMP5 expression in human tissues
Functional studies of VAMP5 in human cell systems
Development of improved tools for selective isolation and analysis of VAMP5-positive EVs
Investigation of VAMP5's role in human disease models
Understanding these aspects will advance our knowledge of extracellular vesicle-mediated communication in the central nervous system and potentially reveal new diagnostic and therapeutic approaches.
Advancing VAMP5 research requires development and refinement of several technological approaches:
Imaging technologies:
Super-resolution microscopy with improved spatial and temporal resolution to track individual VAMP5-positive vesicles in living cells
Correlative light and electron microscopy (CLEM) workflows optimized for capturing rare vesicle release events
Expanded multiplexing capabilities to simultaneously visualize VAMP5 with multiple other markers
Intravital imaging approaches to observe VAMP5-mediated vesicle dynamics in intact tissues
Vesicle isolation and analysis:
Microfluidic platforms for high-throughput, standardized isolation of VAMP5-positive EVs
Single-vesicle analysis technologies to characterize the molecular content of individual vesicles
Improved methods for preserving vesicle integrity during isolation and analysis
Standardized protocols for quantitative comparison of VAMP5-positive EVs across studies
Molecular and genetic tools:
CRISPR-Cas9 systems for tissue-specific and inducible manipulation of VAMP5 expression in vivo
Genetically encoded sensors to detect and quantify VAMP5-mediated vesicle fusion events
Proximity labeling approaches with improved spatial resolution for mapping VAMP5 interactomes
Synthetic biology approaches to create designer VAMP5-positive EVs with defined cargo
Computational and analytical advances:
Machine learning algorithms for automated detection and classification of VAMP5-positive vesicles in imaging data
Integrative multi-omics approaches to correlate VAMP5 expression with global cellular responses
Predictive modeling of VAMP5-mediated vesicle trafficking and targeting
Large-scale data integration platforms to correlate VAMP5-positive EV profiles with clinical outcomes
Translational research tools:
Humanized mouse models for studying VAMP5 function in vivo
Organoid and microphysiological systems incorporating VAMP5-expressing human cells
High-sensitivity assays for detecting VAMP5 and VAMP5-positive EVs in clinical samples
Bioengineered VAMP5-positive EVs for therapeutic applications
These technological advances would address current limitations in studying the dynamic processes of extracellular vesicle formation, release, and function, ultimately accelerating our understanding of VAMP5's role in cellular communication and its potential applications in biomarker development and therapeutic strategies.
Vesicle-associated membrane protein 5 (VAMP5), also known as myobrevin, is a member of the vesicle-associated membrane protein (VAMP)/synaptobrevin family and the SNARE superfamily. These proteins play a crucial role in the docking and fusion of vesicles with cell membranes, which is essential for various cellular processes, including neurotransmitter release and hormone secretion .
The VAMP5 gene is located on chromosome 2 at the p11.2 locus. It encodes a protein that is involved in vesicle trafficking events associated with myogenesis, such as myoblast fusion and GLUT4 trafficking . The protein consists of a single transmembrane domain, a cytoplasmic domain, and a vesicle-associated domain, which are characteristic features of the VAMP family .
VAMP5 is primarily involved in the trafficking of vesicles within cells. It is a key component of the SNARE complex, which mediates the fusion of vesicles with target membranes. This process is vital for the proper functioning of various cellular activities, including the release of neurotransmitters at synapses and the secretion of hormones .
In the context of myogenesis, VAMP5 plays a significant role in the fusion of myoblasts to form multinucleated muscle fibers. This is a critical step in muscle development and regeneration. Additionally, VAMP5 is involved in the trafficking of GLUT4, a glucose transporter, to the plasma membrane in response to insulin signaling .
Mutations or dysregulation of the VAMP5 gene have been associated with various diseases. For instance, VAMP5 has been linked to infant botulism and nephronophthisis 2 . Understanding the role of VAMP5 in these conditions can provide insights into potential therapeutic targets for treating these diseases.