MKI67 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
The antibody is provided as a liquid solution in phosphate-buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA), and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. The delivery time may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery times.
Synonyms
Antigen identified by monoclonal antibody Ki 67 antibody; Antigen identified by monoclonal antibody Ki-67 antibody; Antigen KI-67 antibody; Antigen KI67 antibody; Antigen Ki67 antibody; KI67_HUMAN antibody; KIA antibody; Marker of proliferation Ki-67 antibody; MIB 1 antibody; MIB antibody; MKI67 antibody; PPP1R105 antibody; Proliferation marker protein Ki-67 antibody; Proliferation related Ki 67 antigen antibody; Protein phosphatase 1 regulatory subunit 105 antibody; RP11-380J17.2 antibody
Target Names
MKI67
Uniprot No.

Target Background

Function
MKI67 antibody is crucial for maintaining the dispersal of individual mitotic chromosomes within the cytoplasm following the breakdown of the nuclear envelope. It interacts with the surface of mitotic chromosomes, specifically the perichromosomal layer, encompassing a significant portion of the chromosome surface. This interaction prevents chromosome collapse into a single mass of chromatin by creating a steric and electrostatic charge barrier. The protein exhibits a high net electrical charge and acts as a surfactant, effectively dispersing chromosomes and facilitating independent chromosome movement. MKI67 binds to DNA, demonstrating a preference for supercoiled DNA and AT-rich DNA. It does not contribute to the internal structure of mitotic chromosomes. MKI67 may play a role in chromatin organization. However, it remains unclear whether this involvement is direct or an indirect consequence of its function in maintaining the dispersed state of mitotic chromosomes (Probable).
Gene References Into Functions
  1. Immunohistochemical studies on biopsy specimens from primary breast tumors have examined the expression of RAGE, EGFR, and Ki-67. PMID: 30139236
  2. High Ki-67 expression is associated with Central Giant Cell Granuloma. PMID: 30139237
  3. Our findings identified FGFR3(high)/Ki67(high) papillary pTa tumors as a subgroup with poor prognosis and emphasize the importance of histological grading, as these tumors are classified as high grade. PMID: 30154342
  4. PD-L1, Ki-67, and p53 staining individually demonstrated significant prognostic value for patients with stage II and III colorectal cancer. PMID: 28782638
  5. Research indicates that high Ki-67 expression correlates with unfavorable prognosis and advanced clinicopathological features, potentially serving as a biomarker for disease management. [Review]. PMID: 28287186
  6. High immunoexpression of Ki67, EZH2, and SMYD3, adjusted for standard clinicopathological parameters, independently predicts outcome in patients diagnosed with prostate cancer. PMID: 29174711
  7. The combination of TERT promoter/BRAFV600E mutations and Ki-67 LI emerges as a promising marker for predicting recurrence of papillary thyroid carcinoma. PMID: 28150740
  8. Dual immunostaining for p16 and Ki-67 exhibited comparable sensitivity and improved specificity in screening for high-grade cervical intraepithelial neoplasm (HGCIN) or cervical cancer when compared with human papillomavirus (hrHPV) detection. Further investigations may be beneficial to assess the efficacy of this novel biomarker, which holds potential for use in initial screening assays. PMID: 30249873
  9. Our study demonstrates the ability to accurately approximate the true Ki67 Index without detecting individual nuclei. It also statistically highlights the weaknesses of commonly adopted approaches that utilize both tumor and non-tumor regions together while compensating for the latter with higher-order approximations. PMID: 30176814
  10. Prognosis of luminal breast carcinoma can be predicted using Ki67 as a continuous variable with a standard cutoff value of 14%. It is recommended that the specimen type used to determine Ki67 be documented in the pathological report. PMID: 28865009
  11. Ki-67 and TOPO 2A expression correlated with tumor size and tumor invasiveness in somatotropinomas. PMID: 29334118
  12. This study aimed to investigate the expression of p16 and SATB1 proteins in relation to Ki-67 antigen expression and available clinicopathological data (including receptor status, staging, and grading). PMID: 29936452
  13. Data suggest that Ki-67 is a robust prognostic factor for overall survival (OS) and disease-free survival (DFS) and should be included in all pancreatic neuroendocrine tumor pathology assessments. PMID: 29351120
  14. Ki-67, a proliferation marker, is easily identifiable and provides comparable accurate information. Unlike the poor reproducibility of mitotic counts, Ki-67 demonstrates high agreement among pathologists, exhibits greater reproducibility, provides complementary value to the MBR grading system, and correlates well with other clinicopathologic parameters. PMID: 29893312
  15. High Ki-67 expression is associated with papillary thyroid carcinoma. PMID: 29855303
  16. This study demonstrated the effectiveness of p16/Ki-67 dual staining as a method for cervical cancer screening. Applying this method could potentially reduce unnecessary colposcopy referrals and misdiagnosis. PMID: 29758205
  17. In human adenocarcinoma tissues, PFKFB3 and Ki67 protein levels were found to be correlated with clinical characteristics and overall survival. PMID: 29327288
  18. In leukoplakia, the expression of survivin in association with Ki-67 reinforces the assumption that all these lesions possess malignant potential. PMID: 28346726
  19. High Ki67 expression in the index prostate cancer lesion is an independent predictor of biochemical recurrence in patients undergoing radical prostatectomy with positive surgical margins. PMID: 29506507
  20. Ki-67 expression level did not exhibit a notably significant impact on survival in patients with extensive-stage small cell lung cancer. PMID: 28589765
  21. Dual p16 and Ki-67 staining can enhance the efficiency of cervical cancer screening methods. PMID: 29895125
  22. Both the value and the level of Ki-67 expression exhibited positive correlations with the normalized iodine concentration (NIC) values (r=0.344, P=0.002 and r=0.248, P=0.026). HIF-1alpha expression demonstrated a positive correlation with the NIC values of the RC (r=0.598, P<0.001). PMID: 29103468
  23. Immunohistochemistry and immunoblot analyses revealed a decrease in the expression levels of cyclin D1, cyclin E, pRb, and Ki67 in psoriasis lesions following treatment. These levels were similar to those observed in the normal group. PMID: 29115643
  24. Studies suggest that Ki-67 functions as an organizer of the chromosome periphery region. [Review]. PMID: 28838621
  25. High Ki-67 immunohistochemical expression levels in distant metastatic lesions were independently associated with poorer overall survival outcomes following biopsy of recurrence lesions in breast cancer patients. PMID: 28425014
  26. Data indicate that there was no trend towards higher Ki-67 antigen levels in metastatic compared to primary pancreatic neuroendocrine tumors (NETs). PMID: 28984786
  27. Data suggest that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) presents an alternative proliferative marker. PMID: 29040195
  28. A high Ki-67 LI correlated significantly with a worse prognosis in gastric cancer (GC) patients. Further cumulative studies are necessary to determine the optimal cutoff value for high Ki-67 LI before its application in clinical practice. PMID: 28561880
  29. Ki67 expression in gastric carcinoma is directly correlated with tumor grade and depth of invasion. PMID: 28965621
  30. In ACTH-secreting pituitary tumors, Ki-67 was expressed in 7 out of 28 recurrent tumors and 8 out of 27 nonrecurrent tumors. No staining was observed in normal pituitary samples. Expression was predominantly localized to the nucleus of tumor cells. There was no significant difference in Ki67 expression between the nonrecurrent and recurrent groups. PMID: 29432944
  31. Adjuvant chemotherapy was 9% less likely to be recommended by a multidisciplinary board when using the current criteria compared to using a combination of the St. Gallen criteria and Ki67 and uPA/PAI-1 status (P = 0.03). These findings indicate discordance among markers in identifying recurrence risk, despite the potential independent validity of each marker. PMID: 28954632
  32. The different values of the cycling nuclear area major dimension might also be linked to the biological behavior of the three examined groups. Additionally, endometrial epithelial cells may follow a Ki-67 increase pathway, in contrast to the relatively stable pathway utilized by rapidly proliferating adenocarcinoma cells. PMID: 28737230
  33. Age-associated expression of the proliferation marker MKI67 and the immature neuron marker DCX were unrelated, suggesting that neurogenesis-associated processes are independently altered at these points in the development from stem cell to neuron. PMID: 28766905
  34. High Ki-67 expression in localized PCa is a factor associated with a poor prognosis for prostate cancer. PMID: 28648414
  35. Dual p16 and Ki-67 expression can be utilized in cervical screening of HPV-positive women. PMID: 29566392
  36. Immunohistochemistry for alpha-enolase, Ki67, and p53 was performed in pancreatic cancer and adjacent normal tissues using the corresponding primary antibodies on commercial tissue arrays. PMID: 28824297
  37. All cases of DF exhibited significantly higher Ki67 proliferation index (P = 0.0001) along with increased mitotic figures observed both on H&E staining and with anti-PHH3. PMID: 28609344
  38. We conducted immunohistochemistry for Ki67, p16INK4a, and WNT5A in human hyperplastic polyps (HPs), sessile serrated adenomas/polyps (SSA/Ps), and traditional serrated adenomas (TSAs). The distribution of Ki67 and p16INK4a positive cells in TSAs differed from that in HPs and SSA/Ps. PMID: 28627675
  39. Ki-67 expression in ureteroscopic biopsy specimens has the potential to be helpful in clinical decision-making for patients suspected of having upper urinary tract urothelial carcinoma. PMID: 28554752
  40. For patients diagnosed with ER+/HER2- breast cancer, three distinct risk patterns based on Ki67-LI levels were confirmed according to the 2015 St Gallen consensus. For patients with clearly low or high Ki67-LI, straightforward clinical decisions can be made. However, for patients with intermediate Ki67-LI, additional factors may provide valuable information. PMID: 28061893
  41. Data suggest that Ki-67 index and survivin may be useful biomarkers for rectal cancer patients undergoing preoperative chemoradiotherapy. PMID: 29491110
  42. IHC-based post-Ki67 levels may possess distinct predictive power compared to naive IHC Ki67. PMID: 28412725
  43. Ten international pathology institutions participated in a study to determine messenger RNA expression levels of ERBB2, ESR1, PGR, and MKI67 in both centrally and locally extracted RNA from formalin-fixed, paraffin-embedded breast cancer specimens using the MammaTyper(R) test. Samples were measured repeatedly on different days within the local laboratories, and reproducibility was assessed through variance comp... PMID: 28490348
  44. Our data support the use of Ki67 evaluation to estimate the prognosis of non-small-cell lung cancer (NSCLC) patients, particularly for adenocarcinoma. PMID: 26272457
  45. Co-expression of p16 and Ki-67 was strongly associated with high-risk human papillomavirus persistence, especially with HPV16/18, and could be considered as a suitable biomarker for cervical cancer screening. PMID: 27588487
  46. High expression of VEGF and Ki-67 were independent poor prognostic factors for overall survival in adenoid cystic carcinoma. PMID: 26194375
  47. Proliferative markers, such as mitotic count and Ki67 index, have limited value in predicting recurrence or metastasis in congenital mesoblastic nephromas with a cellular component. PMID: 27484189
  48. KI-67 expression correlates with SATB1 expression in non-small cell lung carcinoma. PMID: 29374696
  49. Ki-67 proliferation index may hold diagnostic value in differentiating between partial and complete hydatidiform moles. PMID: 29374747
  50. Ki-67 proliferation index (P = 0.027) was found to be an independent prognostic factor. PMID: 27049832

Show More

Hide All

Database Links

HGNC: 7107

OMIM: 176741

KEGG: hsa:4288

STRING: 9606.ENSP00000357643

UniGene: Hs.689823

Subcellular Location
Chromosome. Nucleus. Nucleus, nucleolus.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : Immunohistochemistry

Sample type: cells

Review: IHC assays were conducted to demonstrate the effects between PESV treatment and circ_0016760 on MKI67 protein expression in vivo.

Q&A

What is the MKI67 protein and why is it a valuable cellular marker?

MKI67 (also known as Ki-67) is a nuclear, non-histone protein present during all active phases of the cell cycle (G1, S, G2, and M) but absent in quiescent (G0) cells. This distinctive expression pattern makes Ki-67 an excellent marker for proliferating cell populations .

The protein contains specific structural domains:

  • A forkhead-associated (FHA) domain (amino acids 8-98 in humans)

  • Multiple phosphorylation sites (over 200 potential sites)

  • Sixteen 120-amino acid repeats in the central region

Ki-67 plays functional roles in:

  • Interaction with Hklp2 (promotes centrosome separation and spindle bipolarity)

  • Direct interaction with NIFK

  • Binding to UBF, contributing to rRNA synthesis

Human MKI67 is approximately 350-400 kDa, with splice variants of 315-345 kDa also identified. This protein serves as a critical marker in proliferation studies across multiple fields including cancer research, developmental biology, and stem cell research .

How does MKI67 gene expression change throughout the cell cycle?

MKI67 gene expression follows a distinct pattern throughout the cell cycle:

  • G0/G1 phase: Expression is minimal, with low mRNA levels

  • Late G1 and S phase: Expression begins to rise

  • G2/M phases: Maximum expression occurs

This expression pattern is regulated through:

  • Transcriptional control: Two CHR elements and one CDE site in the MKI67 promoter

  • Repressor complexes: DREAM transcriptional repressor complexes bind to CHR sites in G0/G1 cells

  • Activator complexes: B-MYB-MuvB and FOXM1-MuvB complexes bind from S phase into G2/M

The cell cycle-dependent topographical distribution of Ki-67 protein includes:

  • Perinucleolar expression at G1

  • Nuclear matrix expression at G2

  • Chromosome association during M phase

Experimental validation of this expression pattern has been conducted in synchronized human T98G, hTERT-BJ, and mouse NIH3T3 cell lines, confirming the temporal regulation of MKI67 during proliferation .

What methodological considerations are important for optimizing Ki-67 immunohistochemistry?

Optimizing Ki-67 immunohistochemistry requires careful attention to several key parameters:

Antibody selection:

  • Commonly used clones include MIB-1, SP6, and MKI67/2462

  • Monoclonal antibodies provide greater specificity compared to polyclonal alternatives

Sample preparation:

  • Fixation: Formalin fixation (4% paraformaldehyde for 10 minutes) is standard

  • Antigen retrieval: Heat-induced epitope retrieval using citrate or EDTA buffers is critical

    • Example protocol: Heating tissue sections in 10mM Tris with 1mM EDTA, pH 9.0, for 45 min at 95°C followed by cooling at room temperature for 20 minutes

Staining protocol parameters:

  • Primary antibody dilution: Typically 1:50-1:100 for IHC applications

  • Incubation time: 30 minutes at room temperature optimal for many applications

  • Detection systems: Multimer-technology based systems like ultraView Universal DAB provide enhanced sensitivity

Counterstaining:

  • Hematoxylin provides optimal nuclear contrast

  • DAPI counterstaining is preferred for immunofluorescence applications

Quality control measures:

  • Include positive controls: Tonsil tissue is recommended as an appropriate positive control

  • Validate specificity using knockout cell lines where available

What are the best practices for quantifying Ki-67 labeling index (LI)?

The Ki-67 labeling index (LI) is a critical measurement in proliferation assessment. Several methodologies exist, each with advantages and limitations:

Manual counting methods:

  • Grid counting (stereology): Considered the gold standard reference value

    • Implementation: Superimpose grid on image and count Ki-67 positive and negative nuclear profiles

    • Calculation: 100 × (Ki-67-positive nuclear profiles)/(Ki-67-positive + Ki-67-negative nuclear profiles)

    • Time requirement: Approximately 30 minutes per tissue microarray spot

Digital image analysis (DIA) approaches:

  • Automated counting following tissue digitization with platforms like Aperio ScanScope

  • Requires proper validation, calibration, and measurement error correction procedures

Hot-spot selection strategies:

  • Visual identification of areas with highest Ki-67 expression at low magnification

  • Assessment of Ki-67 LI within identified hot-spots

  • Some protocols recommend counting 2-10 square areas with subjectively highest Ki-67 LI

Validation considerations:

  • Inter-observer variation assessment is essential

  • Coefficient Error (CE) computation provides uncertainty estimation

  • Computerized interactive morphometric (CIM) assessment helps overcome selection bias

Research indicates that DIA methods may offer superior reproducibility and prognostic strength compared to subjective counting methods in some cancer types .

How does Ki-67 expression correlate with clinical outcomes in cancer research?

Ki-67 expression has significant prognostic implications across multiple cancer types:

Breast cancer:

  • Meta-analysis of 46 studies (n=12,155 patients) demonstrated:

    • Higher probability of relapse in Ki-67 positive patients (HR=1.93, 95% CI: 1.74-2.14, p<0.001)

    • Worse survival in Ki-67 positive patients (HR=1.95, 95% CI: 1.70-2.24, p<0.001)

    • Stronger prognostic effect in node-negative patients (HR=2.31, 95% CI: 1.83-2.92) compared to node-positive patients (HR=1.59, 95% CI: 1.35-1.87)

Other cancer types with established prognostic value:

  • Grade II astrocytoma

  • Oligodendroglioma

  • Colon carcinoma

  • Renal and ureter tumors

Specific applications:

  • Differentiation of high-risk patients in renal tumors

  • Distinction between malignant and benign peripheral nerve sheath tumors

  • Prediction of responsiveness to chemotherapy or endocrine therapy in breast cancer

The evidence consistently supports Ki-67 as a robust prognostic marker across diverse neoplasms, making it a valuable tool in clinical decision-making and patient stratification .

What are the technical differences between various commercially available Ki-67 antibody clones?

Different Ki-67 antibody clones have distinct characteristics that affect their performance in various applications:

Common clones and their properties:

CloneHostTypeTarget EpitopeOptimal ApplicationsCross-Reactivity
MIB-1MouseMonoclonalCentral regionIHC, ICCHuman
SP6RabbitMonoclonalC-terminus peptideIHC, WBHuman, Mouse, Rat
MKI67/2462MouseMonoclonalKi-67 antigenIHC, ICCHuman
MKI67/2465MouseMonoclonalKi-67 antigenIHC, ICCHuman
RM360RabbitMonoclonalNot specifiedIHCHuman, Mouse (predicted)

Conjugation options and considerations:

  • Unconjugated antibodies provide flexibility in secondary detection methods

  • Fluorescent conjugates (e.g., CF® dyes, Alexa Fluor®) enable direct visualization

  • Note regarding blue fluorescent dyes (CF®405S and CF®405M): Not recommended for low abundance targets due to lower fluorescence and higher non-specific background

Application-specific performance differences:

  • For detecting low abundance targets, conjugates with brighter fluorophores are preferred

  • For multiplex immunofluorescence, antibody clone selection should consider species compatibility with other primary antibodies

  • For quantitative applications, validation with knockout cell lines is recommended to confirm specificity

What is the role of MKI67 in pathological conditions beyond cancer?

Recent research has expanded the understanding of MKI67's role beyond cancer to other pathological conditions:

Pulmonary hypertension (PH):

  • MKI67 transfection experiments demonstrate its regulatory role in cell proliferation and migration in PH pathogenesis

  • Research indicates MKI67 may serve as both a diagnostic biomarker and potential therapeutic target for PH

  • Hypoxia-induced proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) is mediated by MKI67

Neurological disorders:

  • Ki-67 serves as a neuronal marker of cell cycling and proliferation

  • Analysis of Ki-67 expression patterns helps differentiate between normal and pathological neuronal proliferation

Inflammatory conditions:

  • Ki-67 expression in peripheral blood mononuclear cells can be induced by mitogens like PHA

  • This provides a model system for studying proliferative responses in inflammatory contexts

The expanding role of MKI67 across diverse pathological conditions underscores its fundamental importance in proliferative processes and opens new avenues for diagnostic and therapeutic applications beyond traditional cancer research .

How can researchers troubleshoot common issues in Ki-67 immunostaining?

Researchers frequently encounter challenges when performing Ki-67 immunostaining. Here are evidence-based solutions to common problems:

Weak or absent staining:

  • Cause: Inadequate antigen retrieval

  • Solution: Optimize heat-induced epitope retrieval (HIER) using pH 9.0 buffers; example protocol: 10mM Tris with 1mM EDTA, pH 9.0, for 45 min at 95°C

High background staining:

  • Cause: Excessive antibody concentration or non-specific binding

  • Solution: Titrate antibody dilutions (typical range: 1:50-1:100 for IHC); incorporate blocking steps with appropriate sera or BSA (0.05% BSA in PBS is effective)

Inconsistent staining intensity:

  • Cause: Variability in fixation or processing

  • Solution: Standardize fixation protocols; consider automated immunostainers for consistent application and development times

Nuclear staining heterogeneity:

  • Cause: Biological variation in proliferation or technical artifacts

  • Solution: Implement hot-spot selection strategies; consider digital image analysis with internal controls

False positives:

  • Cause: Cross-reactivity with non-target proteins

  • Solution: Validate antibody specificity using knockout cell lines; example: Ki67/MKI67 knockout HeLa cell line serves as an excellent negative control

Poor reproducibility between batches:

  • Cause: Antibody lot variation or protocol inconsistencies

  • Solution: Maintain detailed records of antibody lots; include positive control tissues (tonsil is recommended) with each batch

What are recent advances in digital image analysis for Ki-67 quantification?

Digital image analysis (DIA) has transformed Ki-67 quantification, offering improved reproducibility and objectivity:

Technological platforms:

  • Whole slide scanning systems (e.g., Aperio ScanScope XT) enable high-resolution (0.5 μm) digital capture

  • COMET™ Panel Builder platforms facilitate multiplex immunofluorescence analysis with sequential tissue staining

Validation methodologies:

  • Stereology grid count serves as reference value for DIA calibration

  • Systematic comparison with manual counting establishes measurement error correction parameters

  • Inter-observer variability assessment quantifies reproducibility improvements

Implementation approaches:

  • Hot-spot selection: Digital tools identify regions with highest Ki-67 expression

  • Cell segmentation algorithms: Distinguish positive from negative nuclei based on staining intensity

  • Automated counting: Generate Ki-67 labeling index with reduced human subjectivity

Recent innovations:

  • Integration of Ki-67 assessment with RNAscope® for simultaneous protein and mRNA detection

  • Automated sequential immunofluorescence (seqIF™) enabling multiple marker analysis on the same tissue section

  • COMET™ Panel Builder applications for creating standardized multiplex staining protocols

Research indicates DIA approaches demonstrate superior reproducibility and stronger prognostic value compared to subjective counting methods in breast cancer and other malignancies .

How does MKI67 transcriptional regulation influence cell proliferation?

MKI67 transcriptional regulation involves sophisticated mechanisms that directly impact cell proliferation:

Promoter structure and regulation:

  • Two critical CHR (Cell cycle genes Homology Region) elements in the MKI67 promoter

  • One CDE (Cell cycle-Dependent Element) site contributes to expression control

  • These elements serve as binding sites for cell cycle-specific transcriptional complexes

Key regulatory complexes:

  • DREAM repressor complexes bind to CHR sites in G0/G1 cells, downregulating expression

  • B-MYB-MuvB complexes bind from S phase into G2/M, activating transcription

  • FOXM1-MuvB complexes contribute to upregulation in G2/M phases

Intersection with tumor suppressor pathways:

  • p53 tumor suppressor indirectly downregulates MKI67 transcription

  • RB (Retinoblastoma) tumor suppressor cooperates with DREAM/MuvB-dependent transcriptional control

  • B-MYB binding to CHR elements correlates with loss of CHR-dependent MKI67 promoter activation in knockdown experiments

Expression dynamics throughout cell cycle:

  • Low expression in G0/G1 (quiescent/early G1 cells)

  • Rising expression in late G1 and through S phase

  • Maximum expression in G2 and mitosis

This regulated expression pattern ensures precise control of proliferation markers in normal cells, while dysregulation contributes to pathological conditions characterized by abnormal proliferation .

What methodological considerations are important when using Ki-67 antibodies in flow cytometry?

Flow cytometry applications for Ki-67 require specific methodological considerations:

Sample preparation protocols:

  • Fixation and permeabilization: Critical for accessing nuclear Ki-67; FlowX FoxP3 Fixation & Permeabilization Buffer Kit is specifically recommended

  • Cell concentration: Optimal at 0.2 μg/10^6 cells

  • Mitogen stimulation: PHA (5 μg/mL for 5 days) serves as positive control for proliferation induction in PBMCs

Antibody selection and titration:

  • Primary antibody concentration typically 0.2-1 μg/mL for flow cytometry

  • Secondary detection can utilize phycoerythrin-conjugated or directly labeled antibodies

  • Include isotype controls for setting quadrant markers (e.g., MAB1050)

Multi-parameter considerations:

  • Combine with lineage markers (e.g., CD3e) for cell-specific proliferation assessment

  • Include viability dyes to exclude dead cells from analysis

  • Ensure compensation controls when using multiple fluorochromes

Data analysis strategies:

  • Gating strategy should first identify intact cells, then single cells

  • Compare stimulated versus unstimulated samples to establish positive thresholds

  • For quantitative applications, consider mean fluorescence intensity rather than percent positive alone

Validation approaches:

  • Compare flow cytometry results with other proliferation assays (e.g., BrdU incorporation)

  • Include known proliferating cell populations as positive controls

  • Knockout validation confirms antibody specificity in flow cytometry applications

How do Ki-67 antibodies compare with other proliferation markers in research applications?

Researchers must understand the relative advantages of Ki-67 compared to alternative proliferation markers:

Comparison of common proliferation markers:

MarkerDetection MethodCell Cycle PhaseAdvantagesLimitations
Ki-67 (MKI67)IHC, ICC, Flow, WBAll active phases (G1, S, G2, M)Present throughout cell cycle; well-established prognostic valueDoes not distinguish between cycle phases; some background in certain applications
PCNAIHC, WBPrimarily S phaseWell-characterized; economical antibodiesLess specific; influenced by DNA repair mechanisms
BrdUIHC, FlowS phase onlyDirect measure of DNA synthesis; pulse-chase possibleRequires in vitro/in vivo labeling; DNA denaturation step
EdUIHC, FlowS phase onlyNo DNA denaturation required; compatible with other stainsRequires labeling; potential cytotoxicity
Cyclin D1IHC, WBG1 phasePhase-specific informationMore restricted expression window
Phospho-Histone H3IHC, WB, FlowM phase onlySpecific for mitosisMisses other proliferative phases

Specific research advantages of Ki-67:

  • Comprehensive cell cycle coverage (except G0)

  • Established correlation with clinical outcomes across multiple cancer types

  • Standardized methodologies and commercially validated antibodies

  • No requirement for pre-administration of synthetic nucleosides

Methodological considerations for marker selection:

  • Research questions requiring phase-specific information may benefit from more selective markers

  • Studies examining proliferation in fixed archival tissue are ideally suited to Ki-67

  • Multi-marker approaches combining Ki-67 with phase-specific markers provide more comprehensive proliferation profiles

The evidence supports Ki-67 as a robust general proliferation marker, with specialized applications where alternative or complementary markers may be advantageous based on specific research objectives .

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 2025 TheBiotek. All Rights Reserved.