MKI67 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

The MKI67 monoclonal antibody specifically binds to the Ki-67 protein, a nuclear antigen expressed during all active phases of the cell cycle (G1, S, G2, M) but absent in quiescent (G0) cells . Key characteristics include:

  • Epitope Recognition: Targets non-histone nuclear proteins (345 kDa and 395 kDa isoforms) .

  • Specificity Validation: Confirmed via knockout cell lines (e.g., HeLa cells), where staining is absent in MKI67-null cells .

  • Cross-Reactivity: Reacts with human and bovine tissues .

Cancer Prognosis

The Ki-67 labeling index (percentage of positively stained tumor nuclei) correlates with tumor aggressiveness and patient outcomes:

Non-Oncological Roles

  • Pulmonary Hypertension (PH): MKI67 regulates pulmonary artery smooth muscle cell proliferation and migration under hypoxia, suggesting diagnostic and therapeutic potential .

  • Ribosomal RNA Synthesis: Ki-67 inactivation inhibits rRNA transcription without significantly affecting cell proliferation in vivo .

Validation and Quality Control

  • Western Blot: Detects ~320 kDa bands in HeLa and MCF-7 lysates .

  • Immunofluorescence: Nuclear localization in proliferating cells (e.g., human tonsil, PBMCs) .

  • Flow Cytometry: Distinguishes PHA-activated (Ki-67+) vs. resting lymphocytes .

Research Findings and Meta-Analyses

A 2019 meta-analysis of 53 studies (7,078 gastric cancer patients) demonstrated:

Limitations and Future Directions

  • Standardization Challenges: Variability in cut-off values (e.g., 10–30% positivity) complicates cross-study comparisons .

  • Mechanistic Gaps: While Ki-67’s role in rRNA synthesis is established, its direct contribution to tumorigenesis remains unclear .

Product Specs

Buffer
PBS, pH 7.4, containing 0.02% sodium azide as a preservative and 50% Glycerol.
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary based on the purchasing method or location. For specific delivery time estimates, please consult your local distributors.
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
This antibody is required to maintain individual mitotic chromosomes dispersed in the cytoplasm following nuclear envelope disassembly. It associates with the surface of the mitotic chromosome, the perichromosomal layer, and covers a significant portion of the chromosome surface. By forming a steric and electrostatic charge barrier, it prevents chromosomes from collapsing into a single chromatin mass. The protein exhibits a high net electrical charge and acts as a surfactant, dispersing chromosomes and facilitating independent chromosome motility. It binds to DNA, with a preference for supercoiled DNA and AT-rich DNA. This antibody does not contribute to the internal structure of mitotic chromosomes. It may play a role in chromatin organization; however, it remains unclear whether this role is direct or an indirect consequence of its function in maintaining dispersed mitotic chromosomes.
Gene References Into Functions
  • RAGE, EGFR, and Ki-67 were immunohistochemically studied for their expression in biopsy specimens from primary breast tumors. PMID: 30139236
  • High Ki-67 expression is associated with Central Giant Cell Granuloma. PMID: 30139237
  • Our research identified FGFR3(high)/Ki67(high) papillary pTa tumors as a subgroup with a poor prognosis and encourages histological grading as high-grade tumors. PMID: 30154342
  • PD-L1, Ki-67, and p53 staining individually demonstrated significant prognostic value for patients with stage II and III colorectal cancer. PMID: 28782638
  • Studies indicate that high Ki-67 expression was correlated with poor prognosis and advanced clinicopathological features, suggesting it could serve as a biomarker for disease management. PMID: 28287186
  • High Ki67, EZH2, and SMYD3 immunoexpression, adjusted for standard clinicopathological parameters, independently predicts outcome in patients with prostate cancer at diagnosis. PMID: 29174711
  • The combination of the TERT promoter/BRAFV600E mutations and Ki-67 LI is a promising marker to predict recurrence of PTC. PMID: 28150740
  • p16/Ki-67 dual immunostaining demonstrated comparable sensitivity and improved specificity in screening for high-grade cervical intraepithelial neoplasm (HGCIN) or CC when compared with hrHPV detection. Further research may be beneficial to assess the efficacy of this novel biomarker, which could potentially be used as an initial screening assay. PMID: 30249873
  • Our research demonstrates that accurately approximating the true Ki67 Index is possible without detecting individual nuclei. Additionally, we statistically demonstrate the limitations of commonly adopted approaches that use both tumor and non-tumor regions together while attempting to compensate for the latter with higher-order approximations. PMID: 30176814
  • The prognosis of luminal breast carcinoma can be predicted using Ki67 as a continuous variable and a standard cut-off value of 14%. It is essential to document the specimen type used to determine Ki67 in the pathological report. PMID: 28865009
  • Ki-67 and TOPO 2A expression correlated with tumor size and tumor invasiveness in somatotropinomas. PMID: 29334118
  • This study aimed to investigate the expression of p16 and SATB1 proteins in relation to the expression of the Ki-67 antigen and available clinicopathological data (including receptor status, staging, and grading). PMID: 29936452
  • Data suggest that Ki-67 is a strong prognostic factor for overall survival (OS) and disease-free survival (DFS) and should be included in all pancreatic neuroendocrine tumor pathology. PMID: 29351120
  • Ki-67, a proliferation marker, is easily identified and provides comparable accurate information. In contrast to the poor reproducibility of mitotic counts, Ki-67 achieves high agreement between pathologists, is more reproducible, adds complementary value to the MBR grading system, and correlates well with other clinicopathologic parameters. PMID: 29893312
  • High Ki-67 expression is associated with papillary thyroid carcinoma. PMID: 29855303
  • This study demonstrated that p16/Ki-67 dual staining represents an effective method for cervical cancer screening. Implementing this method could lead to a reduction in unnecessary colposcopy referrals and misdiagnosis. PMID: 29758205
  • In human adenocarcinoma tissues, PFKFB3 and Ki67 protein levels were related to the clinical characteristics and overall survival. PMID: 29327288
  • In leukoplakia, the expression of survivin associated with that of ki-67 reinforces the assumption that all these lesions are potentially malignant. PMID: 28346726
  • High Ki67 expression in the index prostate cancer lesion is an independent predictor of biochemical recurrence in patients with a positive surgical margin following radical prostatectomy. PMID: 29506507
  • Ki-67 expression level did not have a significantly marked impact on survival in patients with extensive-stage small cell lung cancer. PMID: 28589765
  • p16/Ki-67 dual staining can enhance the efficiency of screening methods for cervical cancer. PMID: 29895125
  • Both the value and level of Ki-67 expression were positively correlated with the normalized iodine concentration (NIC) values (r=0.344, P=0.002 and r=0.248, P=0.026); HIF-1alpha expression was positively correlated with the NIC values of the RC (r=0.598, P<0.001). PMID: 29103468
  • Immunohistochemistry and immunoblot analysis revealed that the expression levels of cyclin D1, cyclin E, pRb, and Ki67 in psoriasis lesions decreased after treatment and were similar to those in the normal group. PMID: 29115643
  • Studies suggest that Ki-67 acts as an organizer of the chromosome periphery region. PMID: 28838621
  • High Ki-67 immunohistochemical expression levels in distant metastatic lesions were independently associated with poorer overall survival outcomes after biopsy of recurrence lesions in breast cancer patients. PMID: 28425014
  • Data show there was no trend toward higher Ki-67 antigen in metastatic compared to primary pancreatic neuroendocrine tumors (NETs). PMID: 28984786
  • Data suggest that Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) is an alternative proliferative marker. PMID: 29040195
  • A high Ki-67 LI correlated significantly with a worse prognosis in gastric cancer (GC) patients. Further cumulative studies are needed to determine the optimal cutoff value for high Ki-67 LI before its application in clinical practice. PMID: 28561880
  • Ki67 expression in gastric carcinoma is directly correlated with the tumor grade and depth of invasion. PMID: 28965621
  • In ACTH-secreting pituitary tumors, Ki-67 was expressed in 7 of 28 recurrent tumors and 8 of 27 nonrecurrent tumors, with no staining in normal pituitary samples. It was primarily expressed in the nucleus of the tumor cells. There was no significant difference in Ki67 expression between the nonrecurrent and recurrent groups. PMID: 29432944
  • 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). Our data demonstrate discordance among markers in identifying the risk of recurrence, even though each marker may prove to be independently valid. PMID: 28954632
  • The different values of the cycling nuclear area major dimension may also be connected with the biological behavior of the three examined groups. Furthermore, the endometrial epithelial cells may follow a Ki-67 increase pathway, rather than the relatively stable pathway that rapidly proliferating adenocarcinoma cells might utilize. PMID: 28737230
  • Age-associated expression of the proliferation and immature neuron markers MKI67 and DCX, respectively, was unrelated, suggesting that neurogenesis-associated processes are independently altered at these points in the development from stem cell to neuron. PMID: 28766905
  • High Ki-67 expression in localized PCa is a factor of poor prognosis for prostate cancer. PMID: 28648414
  • Dual p16 and Ki-67 expression can be utilized in cervical screening of HPV-positive women. PMID: 29566392
  • The expression of alpha-enolase, Ki67, and p53 in pancreatic cancer and adjacent normal tissues was evaluated by IHC using the corresponding primary antibodies on commercial tissue arrays. PMID: 28824297
  • All cases of DF exhibited significantly higher Ki67 proliferation index (P = 0.0001) along with increased mitotic figures both on H&E and with anti-PHH3. PMID: 28609344
  • Immunohistochemistry was performed for Ki67, p16INK4a, and WNT5A in human HPs (hyperplastic polyps), 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
  • Ki-67 expression in ureteroscopic biopsy specimens is potentially helpful in clinical decision-making for patients with suspected upper urinary tract urothelial carcinoma. PMID: 28554752
  • For patients with ER+/HER2- breast cancer, three distinct risk patterns by Ki67-LI levels were confirmed according to the 2015 St Gallen consensus. For patients with clearly low or high Ki67-LI, straightforward clinical decisions could be offered; however, for patients with intermediate Ki67-LI, other factors might provide valuable information. PMID: 28061893
  • Data suggest that Ki-67 index and survivin may be useful biomarkers for rectal cancer with preoperative chemoradiotherapy. PMID: 29491110
  • IHC-based post-Ki67 levels may have distinct predictive power compared to naive IHC Ki67. PMID: 28412725
  • Ten international pathology institutions participated in this study and determined 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 with the MammaTyper(R) test. Samples were measured repeatedly on different days within the local laboratories, and reproducibility was assessed by means of variance comp... PMID: 28490348
  • Our data support Ki67 evaluation to estimate non-small-cell lung cancer (NSCLC) patients' prognosis, particularly for adenocarcinoma. PMID: 26272457
  • p16/Ki-67 co-expressions strongly associated with high-risk human papillomavirus persistence, especially with HPV16/18, and could be considered a suitable biomarker for cervical cancer screening. PMID: 27588487
  • High expression of VEGF and Ki-67 were independent poor prognostic factors for overall survival in adenoid cystic carcinoma. PMID: 26194375
  • 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
  • KI-67 expression correlates with SATB1 expression in non-small cell lung carcinoma. PMID: 29374696
  • Ki-67 proliferation index (P = 0.027) proved to be an independent prognostic factor. PMID: 27049832
  • Ki-67 may have diagnostic value in distinguishing between partial and complete hydatidiform moles. PMID: 29374747

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.

Q&A

What is the molecular structure and function of Ki67/MKI67 protein in cell proliferation?

Ki67/MKI67 is a 350-400 kDa nuclear protein belonging to the molecular group of mitotic chromosome-associated proteins. It is structurally complex, containing 3256 amino acids in length with distinct domains including an FHA domain (amino acids 8-98) followed by multiple Ser/Thr phosphorylation sites and sixteen 120 amino acid repeats (amino acids 1000-2928) . The protein is contextually expressed in all cells that are not in the G₀ phase, making it an excellent proliferation marker.

Functionally, Ki67 interacts with 160 kDa Hklp2, a protein promoting centrosome separation and spindle bipolarity . It also directly interacts with NIFK and appears to bind to UBF, playing a role in rRNA synthesis . Alternative splicing produces at least two isoform variants - one 315-345 kDa variant showing deletion of amino acids 136-495, and another containing a 58 amino acid substitution for amino acids 1-513 .

Human Ki67 shares approximately 46% amino acid sequence identity with mouse Ki67 in the C-terminal region (amino acids 3120-3256), which is important when considering cross-reactivity of antibodies across species .

How do different clones of MKI67 monoclonal antibodies differ in their epitope recognition and applications?

Different monoclonal antibody clones recognize distinct epitopes across the large Ki67 protein, significantly affecting their utility in specific applications:

  • OTI5D7 (rat monoclonal): Recognizes amino acids 1160-1493 of human MKI67 and has been validated for flow cytometry (FC) and Western blot (WB) applications . This clone shows specific reactivity with human samples and is recommended at dilutions of 1:500 for WB and 1:10 for FC .

  • MAB7617 (rabbit monoclonal): Recognizes a different epitope and has been validated for multiple applications including multiplex immunofluorescence, immunocytochemistry, flow cytometry, and Simple Western assays . This antibody has demonstrated specific nuclear staining in human tonsil and various cell lines .

  • ABIN3044570 (rabbit polyclonal): Targets amino acids 2860-3256 in the C-terminal region and has been validated for Western blotting, immunohistochemistry on paraffin sections, and immunocytochemistry .

These differences in epitope recognition can affect antibody performance in several ways:

  • Fixation sensitivity: Different epitopes may be differentially affected by fixation methods

  • Application suitability: Some epitopes perform better in certain applications

  • Specificity profiles: Antibodies targeting different regions may have different cross-reactivity profiles

When selecting an MKI67 antibody, researchers should consider the specific epitope recognized, validated applications, and performance in the relevant species and tissue types .

What validation strategies should be employed to confirm MKI67 antibody specificity?

Rigorous validation is essential for ensuring reliable results with MKI67 antibodies. A comprehensive validation strategy should include:

  • Knockout/knockdown validation: The gold standard approach involves testing antibodies in Ki67 knockout cell lines. Search results demonstrate this methodology wherein the MAB7617 antibody showed specific staining in HeLa cells but no detection in Ki67 knockout HeLa cells by both immunocytochemistry and Simple Western analysis . This confirms the antibody's specificity for the intended target.

  • Positive and negative biological controls: Testing on tissues with known Ki67 expression patterns can validate antibody performance. Human tonsil serves as an excellent positive control due to its distinct proliferation zones . Similarly, comparing antibody reactivity between mitogen-stimulated lymphocytes (high Ki67) and resting lymphocytes (low Ki67) provides validation in flow cytometry applications .

  • Multiple application validation: Confirming specificity across multiple detection methods strengthens confidence in antibody performance. For example, MAB7617 demonstrated consistent specific detection of a 320 kDa band in Western blot, nuclear staining in immunofluorescence, and expected staining patterns in flow cytometry .

  • Technical controls: Always include isotype controls in flow cytometry experiments to establish proper gating parameters and differentiate specific from non-specific binding . Similarly, include secondary-only controls in immunohistochemistry and immunofluorescence applications.

  • Cross-validation with different clones: When possible, compare results using antibodies recognizing different epitopes of Ki67 to confirm staining patterns.

These validation approaches collectively provide strong evidence of antibody specificity and reliability across experimental systems .

How should sample preparation be optimized for different MKI67 detection methods?

Sample preparation is critical for successful Ki67 detection and must be tailored to the specific application:

For immunohistochemistry/immunofluorescence on FFPE tissues:

  • Deparaffinization and rehydration must be thorough, using xylene and descending ethanol series .

  • Antigen retrieval is absolutely essential due to epitope masking during fixation. High-temperature retrieval using either citrate buffer (pH 6.0) or HIER Buffer H (pH 9) has proven effective . The search results specifically mention successful retrieval using Dewax and HIER Buffer H (pH 9) on the PreTreatment Module for tissue sections .

  • Blocking should be performed to reduce background staining, typically using BSA-containing buffers .

  • Primary antibody incubation conditions vary by protocol - from rapid protocols (2-4 minutes at 37°C) to standard protocols (overnight at 4°C or several hours at room temperature) .

  • Signal detection systems should be optimized based on the application, with fluorescent secondary antibodies (e.g., Alexa Fluor 555) for immunofluorescence .

For flow cytometry:

  • Fixation and permeabilization are critical steps since Ki67 is a nuclear antigen. Protocols using FlowX FoxP3 Fixation & Permeabilization Buffer Kit have proven effective .

  • Cell stimulation protocols for positive controls (e.g., treating PBMCs with 5 μg/mL PHA for 5 days) provide excellent benchmarks for antibody performance .

  • Appropriate gating strategies should be established using isotype controls to accurately identify positive populations .

For Western blotting:

  • Protein extraction must preserve the high molecular weight Ki67 protein (320 kDa).

  • Sample preparation requires appropriate reducing conditions .

  • Gel selection is critical, with separation systems capable of resolving high molecular weight proteins (e.g., 66-440 kDa separation system) .

  • Loading controls such as GAPDH should be included for normalization .

These optimized protocols significantly enhance detection sensitivity and specificity across applications .

What are the optimal antibody dilutions and incubation conditions for different applications?

Optimal antibody conditions vary by application, detection method, and specific antibody clone:

For Western Blotting:

  • The OTI5D7 clone is recommended at 1:500 dilution

  • The MAB7617 clone has been successfully used at 20 μg/mL for Simple Western applications, detecting Ki67 at approximately 320 kDa

  • Reducing conditions are recommended for all Western blotting applications

For Flow Cytometry:

  • The OTI5D7 clone is recommended at 1:10 dilution

  • For the MAB7617 clone, successful staining of PHA-stimulated PBMCs has been achieved followed by appropriate secondary antibody detection

  • Fixation and permeabilization are essential for accessing the nuclear Ki67 antigen

For Immunohistochemistry/Immunofluorescence:

  • For multiplex immunofluorescence using the MAB7617 clone, 10 μg/mL at 37°C for 4 minutes has proven effective on FFPE human tonsil sections

  • For immunocytochemistry with MAB7617, 1 μg/mL for 3 hours at room temperature provided specific nuclear staining

  • Secondary antibody dilutions typically range from 1:100 to 1:500 depending on the detection system

For seqIF™ staining on COMET™:

  • 0.15 μg/mL at 37°C for 2 minutes has been effective using the MAB7617 antibody

Incubation temperature significantly affects staining efficiency, with rapid protocols at 37°C requiring much shorter incubation times (2-4 minutes) compared to room temperature protocols (several hours) . Regardless of application, antibody titration experiments should be performed to determine optimal conditions for each specific experimental system .

How can multiplex immunofluorescence protocols with MKI67 and other markers be optimized?

Optimizing multiplex immunofluorescence requires careful consideration of multiple factors:

  • Antibody selection and panel design:

    • Choose antibodies from different host species when possible to minimize cross-reactivity

    • The search results demonstrate successful multiplex staining using Rabbit Anti-Human Ki67/MKI67 (MAB7617) with appropriate secondary antibodies

    • For panels including T-cell markers, successful co-staining has been demonstrated with CD3e and Ki67 in PBMCs

  • Antigen retrieval optimization:

    • A universal retrieval condition must be identified that preserves epitopes for all target antigens

    • The search results specifically mention using Dewax and HIER Buffer H (pH 9) for effective antigen retrieval prior to multiplex staining

    • Optimization experiments should test different pH conditions and retrieval durations

  • Signal detection strategy:

    • Sequential detection using fluorophores with minimal spectral overlap is recommended

    • Alexa Fluor Plus 555 has been successfully used for Ki67 detection in multiplex panels

    • DAPI counterstaining provides effective nuclear context for Ki67 assessment

  • Automated platforms:

    • The COMET™ platform has been validated for multiplex immunofluorescence including Ki67

    • Such platforms provide standardized conditions, improving reproducibility

    • The search results describe specific protocols using PreTreatment Module (PT Module) with successful results

  • Incubation parameters:

    • Rapid protocols at 37°C (2-4 minutes) have proven effective for multiplex staining

    • Secondary antibody incubation at 1:100 dilution at 37°C for 2 minutes provided optimal results

  • Image acquisition and analysis:

    • Appropriate exposure settings for each fluorophore are critical

    • Nuclear localization of Ki67 provides a clear separation from membrane or cytoplasmic markers

These optimized approaches enable simultaneous detection of proliferation status alongside lineage markers, signaling pathway activation, or microenvironment components .

What strategies can improve the detection of MKI67 in challenging samples with low expression levels?

Detecting Ki67 in samples with low expression levels requires specialized approaches:

  • Signal amplification methods:

    • Tyramide signal amplification (TSA) can significantly enhance detection sensitivity

    • Polymer-based detection systems provide greater sensitivity than traditional avidin-biotin methods

    • Extended substrate development times may increase sensitivity in chromogenic detection

  • Optimized antibody selection:

    • Choose antibodies with demonstrated high sensitivity

    • Clone MAB7617 has demonstrated detection capability in both highly proliferative tissues (tonsil) and cell lines with variable Ki67 expression

    • Consider using concentrated antibody formulations rather than prediluted versions

  • Enhanced antigen retrieval:

    • Extended high-temperature antigen retrieval may improve epitope accessibility

    • The pH 9 buffer system (HIER Buffer H) mentioned in the search results often provides superior results for nuclear antigens compared to citrate buffer (pH 6)

    • Optimization of retrieval duration can significantly impact detection sensitivity

  • Sample preparation considerations:

    • Minimize fixation time to reduce epitope masking

    • Process samples rapidly to preserve antigen integrity

    • For flow cytometry applications, optimize permeabilization conditions for nuclear access

  • Detection system sensitivity:

    • For fluorescence applications, select bright fluorophores and high-quality filter sets

    • For chromogenic IHC, consider amplification steps and high-sensitivity substrates

    • In Western blotting, enhanced chemiluminescence systems can improve detection of low abundance proteins

  • Automated platforms:

    • Platforms like COMET™ provide standardized conditions that may improve sensitivity

    • Consistent temperature control and precise timing can enhance staining quality

  • Instrument settings:

    • For flow cytometry, optimized voltage settings and careful compensation are essential

    • For imaging, extended exposure times and appropriate gain settings can reveal weak signals

These approaches collectively enhance the likelihood of detecting Ki67 in samples with naturally low proliferation rates or suboptimal preservation .

What are the standard approaches for quantifying Ki67 positivity in different experimental systems?

Standardized quantification approaches vary by experimental system:

For flow cytometry:

  • Gates should be established using appropriate isotype controls (such as MAB1050 mentioned in the search results)

  • Proliferating populations can be identified by comparing untreated versus stimulated samples (e.g., PHA-treated PBMCs show significantly increased Ki67 positivity)

  • Multiparameter analysis allows correlation of Ki67 with lineage markers (e.g., CD3e) to assess proliferation within specific cell subsets

  • Results are typically reported as percentage of Ki67-positive cells within defined populations

For immunohistochemistry/immunofluorescence:

  • Ki67 labeling index (percentage of positive nuclei) is the standard metric

  • Manual counting typically involves assessment of at least 500-1000 cells across multiple high-power fields

  • Automated image analysis provides more objective quantification and can analyze larger sample areas

  • Nuclear algorithms must be optimized to distinguish positive from negative nuclei based on staining intensity thresholds

  • The search results demonstrate clear nuclear localization of Ki67 in human tonsil and cell line samples

For Western blotting:

  • Ki67 appears as a high molecular weight band at approximately 320 kDa

  • Normalization to loading controls like GAPDH is essential for relative quantification

  • Densitometric analysis should account for potential non-linearity at high expression levels

For Simple Western™:

  • The search results demonstrate specific detection of Ki67 at 320 kDa using automated capillary-based Western systems

  • This approach provides more quantitative results compared to traditional Western blotting

  • The specificity has been validated using knockout cell lines

In all cases, appropriate positive and negative controls are essential for validating quantification methods and ensuring accurate interpretation of results .

How should researchers interpret Ki67 staining patterns in heterogeneous tissue samples?

Interpreting Ki67 staining in heterogeneous tissues requires careful consideration of multiple factors:

  • Normal tissue architecture and expected proliferation zones:

    • Human tonsil, mentioned in the search results, shows characteristic proliferation zones in germinal centers

    • Epithelial tissues typically show proliferation restricted to basal and parabasal layers

    • Skin tissue shows specific proliferation patterns as demonstrated in reconstructed human epidermis

    • Understand the expected distribution pattern before interpreting abnormal proliferation

  • Cell-type specific considerations:

    • Nuclear Ki67 staining should be evaluated in the context of cell morphology and distribution

    • In lymphoid tissues, distinguish between lymphocyte populations and stromal cells

    • In complex tissues, consider co-staining with lineage markers to identify proliferating cell types

    • Flow cytometry results demonstrate how combining Ki67 with CD3e enables assessment of T-cell specific proliferation

  • Quantification approaches for heterogeneous samples:

    • Consider region-specific analysis rather than whole-section averages

    • In tumors, analyze both tumor center and invasive margins separately

    • Report both the percentage of positive cells and their distribution pattern

    • Note areas of necrosis or poor fixation that may affect interpretation

  • Technical considerations affecting interpretation:

    • Edge artifacts may show increased staining and should be excluded from analysis

    • Ensure consistent nuclear counterstaining (like DAPI) to identify all nuclei for accurate denominator

    • Distinguish between specific nuclear staining and nonspecific background

    • The search results demonstrate clear nuclear localization of specific Ki67 staining

  • Comparative analysis:

    • Always compare with appropriate control tissues processed simultaneously

    • Consider internal controls within the same section (e.g., normal adjacent tissue)

    • In longitudinal studies, ensure consistent staining and quantification methods

These interpretative approaches help extract meaningful biological insights from complex tissue staining patterns .

What are the main sources of technical variability in Ki67 assessment and how can they be minimized?

Multiple factors contribute to technical variability in Ki67 assessment:

  • Pre-analytical variables:

    • Fixation conditions significantly impact Ki67 immunoreactivity

    • Cold ischemia time before fixation affects antigen preservation

    • Storage duration of paraffin blocks or slides may reduce antigenicity

    • Standardization approaches: Implement strict protocols for tissue handling; document fixation time; process samples consistently

  • Antibody selection and validation:

    • Different clones may recognize distinct epitopes with varying sensitivities

    • Lot-to-lot variability can affect staining intensity

    • The search results demonstrate how validation using knockout cell lines provides confidence in antibody specificity

    • Standardization approaches: Validate new antibody lots against previous standards; include consistent positive controls; employ knockout validation where possible

  • Staining protocol variables:

    • Antigen retrieval methods significantly affect staining intensity and pattern

    • Antibody dilution and incubation conditions directly impact sensitivity

    • Detection systems vary in amplification capabilities

    • Standardization approaches: Use automated staining platforms like COMET™ mentioned in the search results ; implement detailed SOPs; include protocol controls

  • Analysis methods:

    • Manual counting introduces observer variability

    • Different thresholds for "positivity" affect reported percentages

    • Sampling approach (hotspot vs. average) influences results

    • Standardization approaches: Implement digital image analysis; establish clear scoring guidelines; conduct inter-observer reproducibility assessments

  • Reporting practices:

    • Inconsistent metrics (mean vs. median; percentage vs. H-score)

    • Variable cutoffs for categorical classification

    • Differing statistical approaches

    • Standardization approaches: Follow field-specific reporting guidelines; provide detailed methodological documentation; report continuous data when possible

Technical validation studies should include repeatability assessments (same sample, same conditions, different times) and reproducibility assessments (same sample, different operators/labs) to quantify and minimize these sources of variability .

How do cell cycle dynamics affect the interpretation of Ki67 positivity in proliferation studies?

Understanding cell cycle dynamics is crucial for accurate interpretation of Ki67 data:

  • Expression pattern throughout the cell cycle:

    • Ki67 expression begins in early G1 phase, increases throughout S and G2 phases, peaks during mitosis, and rapidly declines after cell division

    • This means Ki67 positivity indicates cells in active cell cycle but doesn't distinguish between cycle phases

    • Expression intensity varies, with highest levels during mitosis, which must be considered when setting positivity thresholds

    • The search results demonstrate nuclear localization patterns characteristic of this cell cycle-dependent expression

  • Implications for proliferation assessment:

    • Ki67 positivity represents a snapshot of cycling cells at a single timepoint

    • It cannot provide information about cell cycle duration or division rate

    • A tissue with long cell cycle duration may show similar Ki67 index to one with shorter cycles despite different actual proliferation rates

    • For dynamic studies, consider combining Ki67 with additional markers like BrdU/EdU (S-phase) or phospho-histone H3 (M-phase)

  • Cell cycle perturbations affecting interpretation:

    • Cell cycle arrest in G1, S, or G2 phases can maintain Ki67 positivity without actual division

    • Treatments that induce cell cycle arrest may show paradoxical Ki67 patterns

    • Extended G0/G1 transition states may show variable Ki67 expression

    • Flow cytometry results in the search data show how mitogen stimulation dramatically increases Ki67 positivity in PBMCs

  • Technical considerations:

    • Different fixation methods may preferentially preserve Ki67 epitopes in certain cell cycle phases

    • The epitope recognized by specific antibody clones may exhibit cell cycle-dependent accessibility

    • Synchronization of cell populations in experimental systems can help calibrate interpretation

  • Emerging analytical approaches:

    • Multiparameter analysis combining Ki67 with DNA content measurement provides cell cycle phase information

    • Mathematical modeling can help infer proliferation dynamics from static Ki67 measurements

    • Single-cell tracking technologies combined with endpoint Ki67 analysis offer more comprehensive interpretation

These dynamics must be considered when comparing Ki67 indices across different tissue types, experimental conditions, or treatment timepoints .

How can MKI67 antibodies be effectively integrated into multiplex imaging technologies?

Integration of Ki67 antibodies into advanced multiplex imaging platforms offers powerful insights into proliferation within complex tissue contexts:

  • Multiplex immunofluorescence platforms:

    • The search results specifically demonstrate successful application of Ki67 antibodies in multiplex protocols using the COMET™ platform

    • Ki67 (MAB7617) has been successfully incorporated into panels detecting multiple markers simultaneously in tissues like human tonsil

    • Nuclear localization of Ki67 provides excellent spatial separation from membrane or cytoplasmic markers

    • Detection using Alexa Fluor Plus 555 secondary antibodies provides strong signal with minimal bleed-through

  • Optimized multiplex protocol parameters:

    • Antigen retrieval using pH 9 HIER Buffer H provides excellent epitope recovery for multiplex panels

    • Short, high-temperature incubations (37°C for 2-4 minutes) yield excellent staining with minimal background

    • Sequential application of antibodies minimizes cross-reactivity issues

    • DAPI counterstaining provides nuclear context for accurate interpretation

  • Advanced spatial analysis approaches:

    • Quantifying proliferating cells relative to tissue compartments or pathological features

    • Measuring distances between proliferating cells and other cell types (e.g., immune cells)

    • Creating proliferation maps across tissue landscapes

    • Correlating Ki67 positivity with expression of signaling pathway components

  • Integration with emerging technologies:

    • Mass cytometry (CyTOF) allows incorporation of Ki67 into high-parameter panels

    • Imaging mass cytometry enables subcellular resolution with 40+ parameters

    • Digital spatial profiling platforms can correlate Ki67 protein expression with spatial transcriptomics

    • Light-sheet microscopy permits 3D assessment of proliferation patterns across intact specimens

  • Data analysis considerations:

    • Machine learning algorithms for cell classification in multiplexed images

    • Spatial statistics for analyzing cell-cell interactions involving proliferating cells

    • 3D reconstruction for volumetric assessment of proliferation zones

These integrated approaches provide unprecedented contextual information about proliferation dynamics within the tissue microenvironment .

What emerging technologies are improving the quantification and interpretation of MKI67 data?

Several cutting-edge technologies are transforming Ki67 assessment:

  • Digital pathology and artificial intelligence:

    • Automated image analysis algorithms specifically designed for nuclear Ki67 quantification

    • Deep learning approaches that can identify Ki67-positive nuclei with human-level accuracy

    • Whole-slide imaging enabling comprehensive assessment rather than field selection

    • These approaches eliminate observer variability and enable analysis of larger tissue areas

  • Single-cell technologies:

    • Mass cytometry allowing simultaneous assessment of Ki67 with 40+ other markers

    • Single-cell RNA-seq correlation with Ki67 protein expression

    • Imaging mass cytometry providing spatial context at subcellular resolution

    • These methods reveal heterogeneity within seemingly uniform populations

  • Live-cell imaging approaches:

    • Fluorescent Ki67 reporter systems for live tracking of proliferation dynamics

    • Correlation of fixed-cell Ki67 staining with prior live-cell behaviors

    • Real-time assessment of treatment effects on proliferation

    • These techniques capture temporal dynamics missing from snapshot analyses

  • Computational biology integration:

    • Mathematical modeling to infer cell cycle parameters from Ki67 indices

    • Integration of Ki67 data with multi-omics datasets

    • Network analysis relating proliferation to signaling pathway activities

    • These approaches extract deeper biological insights from Ki67 measurements

  • Standardization initiatives:

    • Digital reference standards for Ki67 scoring

    • Automated staining platforms like COMET™ mentioned in the search results

    • International scoring guidelines for specific disease contexts

    • These efforts improve reproducibility across laboratories and studies

  • Spatial biology integration:

    • GeoMx Digital Spatial Profiler correlating Ki67 with spatial transcriptomics

    • CODEX multiplexed imaging for high-parameter spatial analysis

    • 3D tissue clearing with Ki67 immunolabeling for volumetric assessment

    • These technologies provide unprecedented spatial context for proliferation assessment

These emerging technologies are rapidly transforming Ki67 from a simple proliferation marker to a sophisticated tool for understanding complex cellular behaviors in health and disease .

How can MKI67 antibodies be used to assess treatment responses in preclinical and clinical research?

Ki67 antibodies serve as powerful tools for evaluating treatment efficacy in both preclinical models and clinical studies:

  • Preclinical applications:

    • Cell line studies: The search results demonstrate the utility of Ki67 antibodies in assessing proliferation in cancer cell lines like HeLa and MCF-7

    • Flow cytometric analysis allows quantitative assessment of treatment effects on proliferation, as shown in the PHA stimulation model

    • Multiplexed imaging enables simultaneous assessment of proliferation, apoptosis, and signaling pathway modulation

    • The robustness of Ki67 as a proliferation biomarker is underscored by validation with knockout cell lines

  • Time-course studies:

    • Serial sampling allows tracking of proliferation dynamics throughout treatment

    • Early changes in Ki67 can precede morphological or volume changes

    • Multiple timepoints help distinguish cytostatic from cytotoxic effects

    • Optimal assessment timing varies by treatment mechanism and must be experimentally determined

  • Combination therapy evaluation:

    • Ki67 assessment helps identify synergistic vs. additive effects on proliferation

    • Multiplex approaches allow correlation of proliferation changes with drug target engagement

    • Heterogeneity in Ki67 response can identify resistant cell populations

    • Such analyses guide rational combination strategies

  • Translational applications:

    • Parallel assessment in preclinical models and patient samples enables predictive biomarker development

    • Window-of-opportunity clinical trials frequently use Ki67 as a primary endpoint

    • Standardized scoring methods facilitate cross-study comparisons

    • Correlation of Ki67 changes with clinical outcomes helps validate surrogate endpoints

  • Methodological considerations:

    • Consistent protocols are essential for longitudinal comparisons

    • Automated platforms like COMET™ improve reproducibility for clinical applications

    • Digital image analysis reduces subjective interpretation

    • Proper controls must include vehicle-treated samples and on-slide positive controls

  • Emerging applications:

    • Ex vivo drug sensitivity testing with Ki67 readouts

    • Patient-derived organoid models with Ki67 assessment

    • Circulating tumor cell proliferation analysis

    • Spatial mapping of treatment-resistant proliferating regions

These approaches collectively enable more informed decision-making in treatment development and selection, with Ki67 serving as a robust and clinically relevant biomarker of antiproliferative efficacy .

What are the limitations of MKI67 as a proliferation marker and what complementary approaches can address these limitations?

Despite its utility, Ki67 has several limitations that can be addressed through complementary approaches:

  • Fundamental biological limitations:

    • Ki67 marks all non-G0 cells, not just actively dividing cells

    • Cannot distinguish between cell cycle phases (G1, S, G2, M)

    • Does not indicate cell cycle speed or provide information about division history

    • Complementary approach: Combine with phase-specific markers like phospho-histone H3 (M-phase), cyclins, or BrdU/EdU incorporation (S-phase)

  • Technical limitations:

    • Epitope sensitivity to fixation conditions

    • Antibody clone variability in performance

    • Subjective interpretation of staining intensity

    • The search results demonstrate rigorous validation approaches including knockout cell lines to address specificity concerns

    • Complementary approach: Implement standardized protocols; use automated staining platforms like COMET™ ; employ digital image analysis

  • Interpretative challenges:

    • Threshold setting for "positivity" varies across studies

    • Proliferation heterogeneity within samples complicates scoring

    • Limited prognostic value as a standalone marker in some contexts

    • Complementary approach: Establish consensus scoring guidelines; integrate with other prognostic markers; implement machine learning for pattern recognition

  • Alternative proliferation assessment methods:

    • DNA synthesis measurement: BrdU/EdU incorporation provides direct evidence of S-phase entry

    • Cell cycle reporters: FUCCI system enables live tracking of cell cycle progression

    • Mitotic counting: Direct enumeration of mitotic figures indicates actual division events

    • Gene expression signatures: Proliferation-associated transcriptional programs provide complementary assessment

  • Emerging technologies:

    • Mass cytometry allows simultaneous assessment of Ki67 with numerous other markers

    • Live-cell imaging techniques enable dynamic assessment of proliferation

    • Single-cell RNA-seq reveals proliferation states at transcriptional level

    • Computational modeling approaches can infer proliferation dynamics from static measurements

  • Application-specific considerations:

    • Slow-cycling tissues require additional markers beyond Ki67

    • Quiescent stem cells may rapidly enter cell cycle but show negative Ki67 at most timepoints

    • Some treatments may uncouple Ki67 expression from actual proliferation

    • Complementary approach: Design marker panels tailored to specific tissue types and research questions

By recognizing these limitations and implementing complementary approaches, researchers can develop more robust and informative proliferation assessment strategies appropriate to their specific research contexts .

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.