MIP 1a Human

Macrophage Inflammatory Protein-1 Alpha Human Recombinant (CCL3)
Shipped with Ice Packs
In Stock

Description

Structure and Biological Properties

MIP-1α is a 7.8 kDa protein composed of 70 amino acids, including four conserved cysteine residues characteristic of CC chemokines . It is synthesized as a precursor protein (92 amino acids) and processed into its mature form . Key features include:

  • Receptors: Binds to CCR1, CCR5, and CCR4 .

  • Dimerization: Forms heterodimers with MIP-1β (CCL4), enhancing antiviral activity against HIV-1 .

  • Proteoglycan Binding: Interacts with heparin and other proteoglycans, which may modulate its activity .

ParameterMIP-1αMIP-1β
ReceptorsCCR1, CCR5, CCR4CCR5, CCR2
Primary TargetsCD8⁺ T cells, macrophagesCD4⁺ T cells, monocytes
HIV SuppressionCompetes with viral entryCompetes with viral entry
Disease AssociationRA, multiple myeloma, HIVSimilar to MIP-1α
Data compiled from .

Immune Response and Inflammation

MIP-1α recruits inflammatory cells (e.g., macrophages, eosinophils, CD8⁺ T cells) to sites of infection or injury . It also induces the production of pro-inflammatory cytokines like TNF-α and IL-6 .

Disease Pathogenesis

Elevated MIP-1α levels are observed in:

  • Rheumatoid Arthritis: Promotes osteoclastogenesis and bone erosion .

  • Multiple Myeloma: Associated with tumor progression and bone resorption .

  • HIV Infection: Serves as a suppressive factor by competing with HIV for CCR5 receptors .

DiseaseMIP-1α RoleSerum Concentration (pg/mL)
Non-Small Cell Lung CancerCorrelates with tumor burden4.51–26.9 (avg. 11.3)
Multiple MyelomaLinked to bone destructionData pending validation
Healthy ControlsBaseline inflammatory marker<12 (inter-subject variability)
Data from .

Cytokine Induction and Signaling Pathways

  • TNF-α and IL-1β: Synergistically induce MIP-1α in human monocytic cells via the TLR4-IRF3 pathway and c-Jun/NF-κB signaling .

  • Oxidative Stress: Amplifies MIP-1α secretion by increasing ROS production .

  • Microglial Activation: LPS and pro-inflammatory cytokines trigger MIP-1α in human fetal microglia, contributing to neuroinflammation .

HIV Suppression

MIP-1α and MIP-1β heterodimers inhibit HIV-1 entry by blocking CCR5, a co-receptor for viral infection . This mechanism highlights their potential as therapeutic targets in HIV-related encephalitis .

Diagnostic and Therapeutic Biomarker

A validated electrochemiluminescence assay (LLOQ: 2.97 pg/mL) quantifies MIP-1α in human serum, enabling its use as a pharmacodynamic marker in clinical trials . Key applications include:

  • Monitoring Inflammatory Diseases: Tracking disease progression in multiple myeloma and rheumatoid arthritis .

  • Drug Development: Evaluating therapies targeting CCR1/CCR5 receptors in oncology and immunology .

Product Specs

Introduction
Macrophage Inflammatory Proteins (MIPs), chemokines, are crucial in immune responses. Humans possess two primary forms: MIP-1a (CCL3) and MIP-1b (CCL4). Produced by macrophages upon bacterial endotoxin stimulation, they activate granulocytes (neutrophils, eosinophils, basophils), potentially causing acute neutrophilic inflammation. Furthermore, they stimulate the production of pro-inflammatory cytokines like interleukin 1 (IL-1), IL-6, and TNF-a from fibroblasts and macrophages. Notably, the genes encoding CCL3 and CCL4 reside on human chromosome 17.
Description
Recombinant Human Macrophage Inflammatory Protein-1 alpha, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 70 amino acids with a molecular weight of 7820 Daltons. The purification of MIP-1a is achieved through proprietary chromatographic methods.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The protein solution, at a concentration of 0.55 mg/ml and without any additives, was lyophilized.
Solubility
For reconstitution, it is advised to dissolve the lyophilized Macrophage Inflammatory Protein-1a in sterile 18MΩ-cm H2O at a minimum concentration of 100µg/ml. Further dilutions can be made using other aqueous solutions.
Stability
While lyophilized MIP-1a remains stable at room temperature for up to 3 weeks, it is recommended to store it desiccated below -18°C. After reconstitution, CCL3 should be stored at 4°C for 2-7 days. For long-term storage, freezing below -18°C is advised. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined using the following methods, and is greater than 98.0%: (a) RP-HPLC analysis (b) SDS-PAGE analysis.
Biological Activity
The biological activity is assessed based on the chemo-attraction of human monocytes, using a concentration range of 1-10ng/ml. This corresponds to a Specific Activity of 100,000-1,000,000 IU/mg.
Synonyms
Small inducible cytokine A3, CCL3, Macrophage inflammatory protein 1-alpha, MIP-1-alpha, Tonsillar lymphocyte LD78 alpha protein, G0/G1 switch regulatory protein 19-1, G0S19-1 protein, SIS-beta, PAT 464.1, chemokine (C-C motif) ligand 3, MIP1A, SCYA3, G0S19-1, LD78ALPHA.
Source
Escherichia Coli.
Amino Acid Sequence
The sequence of the first five N-terminal amino acids was determined and was found to be, Ala-Ser-Leu-Ala-Ala.

Q&A

What is MIP-1α and what are its alternative nomenclatures?

MIP-1α is a chemotactic chemokine primarily secreted by macrophages that recruits inflammatory cells and participates in wound healing processes. Due to independent isolation by multiple research groups, MIP-1α has accumulated several names in scientific literature. Under the current standardized nomenclature, human MIP-1α is officially designated as chemokine (C-C motif) ligand 3 (CCL3). Previous or alternative names include LD78alpha, GOS19-1, and SCYA3 . When conducting literature searches or discussing this protein in publications, researchers should be aware of these nomenclature variations to ensure comprehensive retrieval of relevant information.

What are the primary cellular sources of MIP-1α in humans?

MIP-1α is primarily expressed by cells of the innate immune system, including macrophages, dendritic cells, and various lymphocyte populations . During inflammatory responses, these cells secrete MIP-1α to recruit additional immune cells to sites of infection or tissue damage. The regulation of MIP-1α expression varies among these cell populations, with macrophages typically producing substantial amounts following activation by pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS). Understanding the cellular sources in your experimental system is crucial for accurate interpretation of MIP-1α's role in specific pathological contexts.

What are the major biological functions of MIP-1α in human inflammatory responses?

MIP-1α serves multiple critical functions in inflammatory responses. As a proinflammatory chemokine, it primarily mediates cell adhesion and migration of monocytes to inflammatory tissues . Beyond its chemotactic properties, MIP-1α induces the synthesis and release of other inflammatory mediators, including IL-6, IL-10, and TNF-α, thus amplifying the inflammatory cascade . Additionally, MIP-1α inhibits the proliferation of hematopoietic stem cells both in vitro and in vivo, suggesting a regulatory role in hematopoiesis . Through binding to receptors CCR1, CCR4, and CCR5, MIP-1α coordinates multiple aspects of acute inflammatory responses and immune cell recruitment.

How do the two human MIP-1α genes differ functionally?

The human genome contains two MIP-1α genes resulting from duplication and subsequent mutation: CCL3/LD78a and CCL3L1/LD78b. These isoforms share approximately 94% amino acid sequence homology but display important functional differences . While CCL3/LD78a exists as a single-copy gene in humans, CCL3L1/LD78b demonstrates copy number variation within the population. The most significant functional distinction is that CCL3L1/LD78b exhibits higher binding affinity for CCR5 compared to CCL3/LD78a . This differential binding is particularly relevant in HIV-1 research, as CCR5 functions as a coreceptor for HIV-1 entry into cells, and CCL3L1 copy number variations represent genetic determinants of HIV-1 susceptibility .

What are the validated methods for measuring MIP-1α in human clinical samples?

Multiple validated methodologies exist for detecting and quantifying MIP-1α in human clinical samples. For serum or plasma measurements, electrochemiluminescence assays have been validated with a lower limit of quantitation of approximately 2.97 pg/ml . Commercial ELISA kits are also widely used, with validated performance for cell culture supernatants, serum, and various plasma preparations (EDTA, heparin, and citrate) .

For tissue expression analysis, reverse transcriptase-polymerase chain reaction (RT-PCR) can be employed to measure MIP-1α mRNA levels, using established primer sequences (forward: TGCATCACTTGCTGCTGACACG, reverse: CAACCAGTCCATAGAAGAGG) . Immunohistochemistry techniques can visualize MIP-1α protein in tissue sections using commercially available antibodies, typically following acetone fixation and appropriate blocking procedures .

Selection of the appropriate methodology should consider the specific research question, sample type availability, required sensitivity, and the need for protein versus mRNA detection.

How should researchers optimize experimental design for MIP-1α quantification in serum and plasma samples?

When quantifying MIP-1α in serum or plasma, researchers should consider several critical optimization parameters:

  • Sample type selection: Different anticoagulants (EDTA, heparin, citrate) may affect recovery rates. According to validation studies, recovery rates range from 94-98% depending on the anticoagulant used, with heparin plasma showing slightly higher recovery (98%) compared to EDTA (96%) or citrate plasma (94%) .

  • Precision considerations: Both intra-assay and inter-assay variability should be evaluated. Published data indicate coefficient of variation (CV) ranges of 5.1-8.9% for intra-assay precision and 4.9-11.6% for inter-assay precision in serum and plasma samples .

  • Linearity and recovery validation: Researchers should verify that MIP-1α measurements maintain linearity across relevant concentration ranges in their specific sample matrix.

The following table summarizes recovery rates for MIP-1α across different sample types:

Sample TypeAverage % RecoveryRange %
Cell Culture Media10088-117
Citrate Plasma9485-104
EDTA Plasma9681-109
Heparin Plasma9882-121
Serum9587-113

This data highlights the importance of consistent sample type selection throughout a study to minimize methodological variability .

What are the optimal PCR conditions and primer designs for MIP-1α mRNA quantification?

For reliable quantification of MIP-1α mRNA, researchers should implement the following optimized conditions:

  • RNA extraction: Total RNA isolation using TRIzol or similar reagents from homogenized tissue or cell pellets, followed by dissolution in DEPC-treated water .

  • Primer design: Utilize validated primer pairs for human MIP-1α:

    • Forward primer: TGCATCACTTGCTGCTGACACG

    • Reverse primer: CAACCAGTCCATAGAAGAGG

    • Probe: CTGACTACTTTGAGACGAGC

    • Expected product size: 333 bp

  • Cycling parameters: PCR amplification should include determining the linear range for accurate semi-quantification. For MIP-1α, 32 cycles typically places amplification within the linear range .

  • Internal control: Include housekeeping gene controls such as GAPDH (primers: forward CCATGGAGAAGGCTGGGG, reverse CAAAGTTGTCATGGATGACC) for normalization .

  • Visualization: Confirmation of single band products through electrophoresis and ethidium bromide staining .

For absolute quantification, consider implementing a standard curve using known copy numbers of synthetic template. For relative quantification, the ΔΔCt method with appropriate reference genes provides reliable results when primer efficiencies are similar.

How should researchers approach immunohistochemical detection of MIP-1α in brain tissue from neuroinflammatory disorders?

For optimal immunohistochemical detection of MIP-1α in brain tissue from conditions such as multiple sclerosis, researchers should implement the following protocol:

  • Tissue preparation: Prepare 5 μm thick frozen sections of brain tissue mounted on poly L-lysine (PLL)-coated glass slides. Air dry samples before fixing in acetone for 10 minutes at room temperature .

  • Blocking procedures: To prevent non-specific binding, preincubate sections with 10% normal swine serum (for polyclonal antibodies) or 2% normal rabbit serum (for monoclonal antibodies) for 10 minutes at room temperature .

  • Primary antibody application: Use mouse anti-human MIP-1α (IgG2a) at 1:10 dilution in PBS containing 1% bovine serum albumin (BSA). Incubate for 1 hour at room temperature .

  • Controls: Include isotype control sections incubated with mouse purified IgG1 (1:100 dilution) to identify any non-specific staining .

  • Cell identification: For comprehensive analysis, parallel sections should be stained with cell-specific markers including KP1 (CD68; 1:400) for macrophages/microglia, LCA (CD45; 1:50) for leukocytes, and GFAP (1:1000) for astrocytes .

  • Data interpretation: When analyzing MIP-1α expression in neuroinflammatory conditions, correlate staining patterns with lesion activity (active demyelination versus chronic lesions) and with infiltrating cell populations to establish the cellular sources of MIP-1α in the specific pathological context.

What are the key considerations when comparing MIP-1α expression levels between healthy and diseased tissues?

When comparing MIP-1α expression between healthy and diseased tissues, researchers should account for several critical factors:

  • Baseline expression variability: MIP-1α exhibits variable expression in certain tissues even under normal conditions. Establish appropriate baseline ranges using sufficient numbers of control samples.

  • Cellular composition differences: Diseased tissues often contain altered cellular compositions, particularly inflammatory cell infiltrates, which may independently contribute to changes in MIP-1α levels. Use cell-specific markers in parallel analyses to determine whether expression changes reflect altered cellular composition or differential expression within resident cells .

  • Semi-quantitative scoring: For immunohistochemical analyses, develop a rigorous semi-quantitative scoring system with blinded evaluation by multiple observers to reduce bias.

  • Normalization strategies: For mRNA expression analysis, carefully select housekeeping genes that maintain stable expression between healthy and diseased states. GAPDH at 30 PCR cycles has been validated for MIP-1α studies in brain tissue .

  • Statistical analysis: Given the typically non-normal distribution of cytokine data, consider non-parametric statistical approaches when comparing groups, and account for multiple comparisons when analyzing complex datasets.

How does MIP-1α function as a biomarker in inflammatory and infectious diseases?

MIP-1α serves as a valuable biomarker across multiple inflammatory and infectious disease contexts. Its utility stems from its position as a proximal mediator in inflammatory cascades. Elevated MIP-1α levels have demonstrated prognostic or diagnostic value in:

  • Multiple myeloma: Where increased MIP-1α correlates with disease progression and bone lesions .

  • Multiple sclerosis: Where MIP-1α expression in brain tissue reflects inflammatory activity and has been associated with lesion development .

  • HIV infection: Where MIP-1α levels correlate with viral load and disease progression. Additionally, genetic variations in MIP-1α genes, particularly CCL3L1 copy number, influence HIV-1 susceptibility due to the interaction with CCR5, which functions as an HIV-1 co-receptor .

  • Allergic asthma: Where MIP-1α participates in eosinophil recruitment and airway hyperresponsiveness .

  • Sepsis: Where MIP-1α serves as part of the cytokine profile associated with systemic inflammatory response syndrome .

For biomarker applications, researchers should establish disease-specific reference ranges, determine sensitivity and specificity parameters through ROC curve analyses, and validate findings in independent cohorts before clinical application.

What is the significance of MIP-1α gene copy number variation in human populations?

The significance of MIP-1α gene copy number variation, particularly for CCL3L1/LD78b, extends across multiple domains of human biology and disease susceptibility:

  • HIV-1 infection vulnerability: CCL3L1 copy number represents a genetic determinant of HIV-1 susceptibility due to its high-affinity binding to CCR5, the HIV-1 co-receptor. Higher CCL3L1 copy numbers have been associated with decreased susceptibility to HIV-1 infection and delayed progression to AIDS .

  • Population genetics: CCL3L1 copy number varies significantly across human populations, potentially reflecting evolutionary selective pressures related to infectious disease exposure.

  • Inflammatory disease risk: Copy number variations may influence inflammatory response magnitude, potentially modifying susceptibility to and severity of various inflammatory conditions.

When investigating CCL3L1 copy number variations, researchers should employ reliable quantification methods such as digital PCR or paralogue ratio testing rather than standard quantitative PCR, which may lack the necessary precision for copy number determination. Additionally, population-specific reference ranges should be established due to the substantial variation in normal copy number distributions across ethnic groups.

How do the binding affinities of MIP-1α to different chemokine receptors impact experimental design?

MIP-1α binds multiple chemokine receptors (CCR1, CCR4, and CCR5) with varying affinities, which creates important considerations for experimental design :

  • Receptor expression profiling: Before conducting functional experiments, researchers should characterize the receptor expression profile of their cell populations, as differential expression of CCR1, CCR4, and CCR5 will influence cellular responsiveness to MIP-1α.

  • Isoform selection: The two MIP-1α isoforms (CCL3/LD78a and CCL3L1/LD78b) display differential binding affinities, particularly for CCR5, with CCL3L1/LD78b demonstrating higher affinity . Researchers should specify which isoform is being used in experiments and consider the implications for receptor activation.

  • Receptor blockade strategies: To dissect which receptor mediates specific MIP-1α effects, implement selective receptor antagonists or receptor-specific blocking antibodies rather than assuming a particular receptor's involvement.

  • Concentration considerations: Due to differential binding affinities, dose-response experiments across wide concentration ranges are essential to capture the full spectrum of receptor-mediated effects.

  • Heterologous desensitization: In sequential stimulation experiments, consider that MIP-1α binding may desensitize cells to subsequent stimulation with other chemokines that share receptor usage through heterologous desensitization mechanisms.

How can researchers address the challenges of MIP-1α stability in experimental samples?

MIP-1α stability presents several challenges in experimental settings that researchers must address through careful sample handling:

  • Collection protocols: Standardize collection procedures, including time between collection and processing, to minimize variability introduced by differential proteolytic degradation.

  • Storage conditions: Store samples at -80°C for long-term preservation of MIP-1α. Avoid repeated freeze-thaw cycles, which significantly reduce detectable levels. If multiple analyses are planned, aliquot samples before freezing.

  • Protease inhibitors: Consider adding protease inhibitor cocktails to samples immediately upon collection, particularly for complex matrices like tissue homogenates or biological fluids with high protease content.

  • Stabilizing additives: For certain applications, carrier proteins (e.g., BSA) at low concentrations can improve MIP-1α stability without interfering with downstream assays.

  • Quality control: Include stability control samples (spiked with known MIP-1α concentrations) processed and stored identically to experimental samples to quantify any degradation effects.

  • Validation of freeze-thaw stability: For your specific sample type and storage conditions, conduct freeze-thaw stability experiments to determine the maximum allowable cycles before significant degradation occurs.

What are the optimal approaches for distinguishing between different MIP-1α isoforms in human samples?

Distinguishing between MIP-1α isoforms (CCL3/LD78a and CCL3L1/LD78b) requires specialized techniques due to their high sequence homology (94% amino acid identity) :

  • mRNA detection: Design PCR primers that target sequence divergence regions between CCL3 and CCL3L1. Typically, these differences occur in non-coding regions or at specific polymorphic sites within the coding sequence. Validation with known standards is essential to confirm specificity.

  • Protein detection: Most commercial antibodies do not discriminate between CCL3 and CCL3L1 proteins due to their high sequence similarity. For isoform-specific detection, consider:

    • Using recombinant standards of each isoform to establish differential binding patterns

    • Developing custom antibodies targeting isoform-specific epitopes

    • Employing mass spectrometry approaches that can distinguish peptide sequences unique to each isoform

  • Functional discrimination: Leverage the differential binding affinity to CCR5 between the isoforms through competitive binding assays or cell-based functional assays using CCR5-expressing reporter systems.

  • Copy number analysis: Quantitative PCR or digital PCR methods targeting CCL3L1-specific sequences can determine gene copy number, which indicates the potential for differential expression of this isoform .

How should researchers interpret contradictory MIP-1α findings across different disease studies?

When confronted with contradictory MIP-1α findings across disease studies, researchers should systematically evaluate several potential sources of discrepancy:

  • Methodological variations: Different detection methodologies (ELISA vs. electrochemiluminescence vs. bead-based multiplex assays) may yield different absolute values. Even within the same method type, inter-kit variability can be substantial. Compare precision data between studies (intra-assay CV: 1.3-8.9%; inter-assay CV: 4.1-11.6% based on published data) .

  • Sample type differences: MIP-1α levels vary between sample types (serum vs. different plasma preparations). The recovery rates range from 94-98% depending on the matrix .

  • Disease heterogeneity: Apparent contradictions may reflect genuine biological variation between different disease subtypes or stages that were not similarly distributed across study cohorts.

  • Timing of sample collection: As an inflammatory mediator, MIP-1α exhibits temporal dynamics during disease progression. Differences in sampling timepoints relative to disease onset may explain contradictory results.

  • Confounding variables: Evaluate whether studies adequately controlled for important confounders like age, sex, concurrent medications, and comorbidities, all of which can influence MIP-1α levels.

  • Statistical approaches: Review whether appropriate statistical methods were applied, including adjustment for multiple comparisons and handling of non-normally distributed data.

Resolution of contradictory findings may require meta-analysis approaches, direct replication studies with standardized protocols, or more sophisticated stratification of patient populations to account for disease heterogeneity.

What are the key considerations when using MIP-1α as a pharmacodynamic biomarker in clinical trials?

Implementing MIP-1α as a pharmacodynamic biomarker in clinical trials requires attention to several critical considerations:

  • Assay validation: Employ fully validated assays with established performance characteristics including LLOQ, precision, accuracy, and linearity appropriate for the anticipated concentration range. A validated electrochemiluminescence assay with LLOQ of 2.97 pg/ml has been successfully implemented for clinical biomarker applications .

  • Reference ranges: Establish population-specific reference ranges accounting for age, sex, and potential diurnal variations. Biological variability data should inform sampling strategies and interpretation thresholds for meaningful changes.

  • Sample handling standardization: Implement strict sample collection, processing, and storage SOPs across all trial sites to minimize pre-analytical variability. This includes standardized collection tubes, processing timeframes, centrifugation parameters, and storage conditions.

  • Timing considerations: Determine the optimal sampling timepoints based on the pharmacokinetics of the investigational product and the expected temporal relationship between drug action and MIP-1α modulation.

  • Context of interpretation: Interpret MIP-1α changes in the context of other inflammatory markers and clinical parameters, as isolated changes may be difficult to interpret, particularly for complex immunomodulatory agents.

  • Statistical analysis plan: Develop a prospective statistical analysis plan that accounts for the typically non-normal distribution of cytokine data and includes appropriate methods for handling missing data and outliers.

Product Science Overview

Introduction

Macrophage Inflammatory Protein-1 Alpha (MIP-1α), also known as CCL3, is a chemokine that plays a crucial role in the immune response. It is produced by macrophages and other cell types and is involved in the recruitment and activation of various immune cells. This article delves into the background, biological functions, and clinical significance of MIP-1α/CCL3.

Discovery and Nomenclature

MIP-1α was first discovered by Stephen D. Wolpe in 1988 . It belongs to the CC subfamily of chemokines, which are characterized by two adjacent cysteine residues near their amino terminus. The official name for MIP-1α is CCL3, and it is also known by several other names, including G0/G1 switch regulatory protein 19-1, PAT 464.1, and small-inducible cytokine A3 .

Biological Functions

MIP-1α/CCL3 is a multifunctional peptide that performs various biological functions:

  • Chemotaxis: It recruits macrophages, lymphocytes, and eosinophils to sites of inflammation via the CCR1 or CCR5 receptors .
  • Bone Resorption: It activates bone resorption cells and directly induces bone destruction, playing a significant role in bone remodeling by inducing osteoclastogenesis .
  • Inflammatory Response: It induces inflammatory responses, including superoxide production by neutrophils .
  • Antiviral Activity: MIP-1α has been shown to have antiviral activity against HSV-1 and exhibits strong HIV-suppressive activity .
Clinical Significance

MIP-1α/CCL3 is associated with various inflammatory diseases and conditions that exhibit bone resorption, such as:

  • Periodontitis: Elevated levels of MIP-1α/CCL3 are found in the biological fluids of patients with periodontitis .
  • Multiple Myeloma: It plays a role in the pathogenesis of multiple myeloma by recruiting inflammatory cells and promoting osteoclastogenesis .
  • Rheumatoid Arthritis: MIP-1α/CCL3 is involved in the inflammatory processes of rheumatoid arthritis .
  • Sjögren Syndrome: Increased concentrations of MIP-1α/CCL3 are detected in patients with Sjögren syndrome .
Recombinant Human MIP-1α/CCL3

Recombinant human MIP-1α/CCL3 is a human full-length protein expressed in Escherichia coli with high purity and low endotoxin levels . It is used in various research applications, including SDS-PAGE, functional assays, and HPLC . The recombinant protein retains its biological activity and is used to study the chemotactic and inflammatory properties of MIP-1α/CCL3 .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.