Next Article in Journal
The Influence of N-Acetyl-selenomethionine on Two RONS-Generating Cancer Cell Lines Compared to N-Acetyl-methionine
Previous Article in Journal
Transcription Factors Sox2 and Sox3 Directly Regulate the Expression of Genes Involved in the Onset of Oligodendrocyte Differentiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Novel Chromogens for Immunohistochemistry in Spatial Biology

1
Diagnostic BioSystems, Pleasanton, CA 94588, USA
2
Key Biomedical Services, Ojai, CA 93023, USA
*
Author to whom correspondence should be addressed.
Cells 2024, 13(11), 936; https://doi.org/10.3390/cells13110936
Submission received: 17 April 2024 / Revised: 16 May 2024 / Accepted: 26 May 2024 / Published: 29 May 2024
(This article belongs to the Section Cell Microenvironment)

Abstract

:
Spatial relations between tumor cells and host-infiltrating cells are increasingly important in both basic science and clinical research. In this study, we have tested the feasibility of using standard methods of immunohistochemistry (IHC) in a multiplex staining system using a newly developed set of chromogenic substrates for the peroxidase and alkaline phosphatase enzymes. Using this approach, we have developed a set of chromogens characterized by (1) providing fine cellular detail, (2) non-overlapping spectral profiles, (3) an absence of interactions between chromogens, (4) stability when stored, and (5) compatibility with current standard immunohistochemistry practices. When viewed microscopically under brightfield illumination, the chromogens yielded the following colors: red, black, blue, yellow, brown, and green. By selecting compatible color combinations, we have shown feasibility for four-color multiplex staining. Depending on the particular type of analysis being performed, visual analysis, without the aid of computer-assisted image analysis, was sufficient to differentiate up to four different markers.

1. Introduction

There is a substantial body of literature describing the intra-tumor heterogeneity of both tumor and host cells as well as their interactions. Extensive efforts have been made to analyze the various cell types, locations, and associations within the tumor architecture. Such methods have focused primarily on single-cell measurements by flow cytometry or nucleic acid sequencing [1,2]. However, such methods based on dissociated cells fail to take into account the spatial relationships between the various cell types.
Discovering the molecular mechanisms affecting cell growth and development is critical in understanding disease processes, and this understanding can ultimately lead to better treatments. For example, in tumors there is an intimate relationship between the tumor cells and their surroundings. This can be understood to reflect a critical connection between the tumor cells and their environment. For example, some types of immunotherapy rely upon the treatment’s ability to interfere with certain interactions between tumor cells and immune cells [3]. It is now apparent that the characterization of the tumor microenvironment and the cell populations residing therein is critical to our understanding of these diseases.
Spatial immunohistochemistry (IHC) has now become a useful tool for analyzing morphological patterns [4,5] and cellular microenvironment [6,7]. In particular, methods utilizing immunofluorescence have proven extremely valuable in identifying and quantifying host-infiltrating immune cells [8,9]. One of the advantages of utilizing fluorochromes in this context is their narrow bandwidth, enabling multiple labels to be simultaneously visualized within a single tissue. However, despite these advantages, light microscopy still remains the method of choice for most pathologists as the morphological information provided by this method is critical for a primary diagnosis. Pathologists have been evaluating cells in tissue sections using classical antibody- and morphology-based staining methods for nearly 100 years. In clinical practice, these evaluations were frequently performed using chromogenic IHC methods that allowed for the assessment of one or two proteins within a single tissue slide. While conventional IHC methods using chromogenic labels can capture spatial organization, these methods are limited by the availability of robust chromogens that can be combined together within a single stain to produce multiplex staining images.
Multispectral imaging has promise for overcoming some of these limitations. Multispectral imaging can be used with both fluorescence and brightfield microscopy methods, although fewer chromogens are available for brightfield applications, and their absorbance spectra are relatively broad compared to most fluorochromes. Recent studies were able to overcome some of these limitations by the use of a new family of chromogens with narrow spectral absorption and appropriately matched brightfield illumination channels [10]. Further developments of brightfield multispectral imaging may allow the integration of this important technology into general pathology [11,12]. However, at present an objective, measurable, and reproducible means of evaluating spatial heterogeneity at the single-cell level by brightfield IHC remains a challenging goal.

2. Materials and Methods

2.1. Sample Preparation

All tissue specimens were obtained from surgical samples as paraffin blocks. Tissues had been formalin-fixed and paraffin-embedded (FFPE) using standard histological methods. Tissue sections were prepared at 4 µ, adhered to positively charged microscope slides, and stored at room temperature until the time of staining. At the time of staining, tissue sections were deparaffinized through graded solutions of xylene and alcohol and then rehydrated in deionized water. After deparaffinization, in order to firmly attach the tissues to the microscope slide during the staining procedure, the slides and the tissues were treated with an adherence-promoting reagent (Tissue Glue, Diagnostic BioSystems, Pleasanton, CA USA) according to the manufacturer’s instructions. Deparaffinized tissue sections were then subjected to antigen retrieval by submerging slides in a solution of Tris-EDTA, pH 9.0, and heating in a pressure cooker at 15 psi for 15 min. Slides in the antigen-retrieval solution were allowed to cool at room temperature until the pressure was relieved. Slides were then placed into a Tris-buffered wash solution and cooled at room temperature for five minutes.

2.2. Antibodies

Antibodies were screened from tonsils as single stains to determine optimal staining concentrations. All antibodies were obtained from Diagnostic BioSystems, Pleasanton, CA, USA, and are listed in Table 1.

2.3. Reagents

Table 2 lists reagents used. All reagents were obtained from Diagnostic BioSystems.

2.4. Chromogens

Table 3 lists all chromogens used. All chromogens were obtained from Diagnostic BioSystems.

2.5. Immunohistochemistry

Sequential multiplex staining was used because of its flexibility for using any antibody or enzyme system. The following steps were employed using a manual method at room temperature:
StepName
1Primary Antibody
2Rinse
3Polymer (enzyme-conjugated secondary antibody)
4Rinse
5Substrate/Chromogen
6Rinse
7Elution Buffer

2.6. Peroxidase Method

Incubation time for primary antibodies and polymers was 20 min. Incubation time for substrate/chromogens for horseradish peroxidase (HRP) was 5 min.

2.7. Alkaline Phosphatase Method

Incubation time for primary antibodies and polymers was 20 min. Incubation time for substrate/chromogens for alkaline phosphatase (AP) polymers was 10 min.

2.8. Elution Buffer

The elution buffer was composed of 0.25% sodium dodecyl sulfate (SDS) in 0.1 M glycine at pH 2.0 [13]. Previous experiments had shown that an incubation time of 10 min at 50 °C was sufficient to remove all primary antibodies and secondary polymers from the stained slides.

2.9. Multiplex Staining

After a single sequence of staining and elution, slides were then re-stained with a different primary antibody using the same sequence as previously described but instead using a different colored chromogen. This sequence was repeated multiple times until up to four different chromogens had been applied to the tissues, each chromogen detecting a different antigen based on the specificity of the primary antibody.

2.10. Microscopic Evaluation

Slides were mounted using an ImmunoHisto-Sealer (Diagnostic BioSystems, Part No. K076) followed by a permanent mounting medium, according to the manufacturer’s instructions. Mounted slides were viewed using brightfield microscopy at 100× and 400× magnification. Slides were evaluated for specific staining, background staining, and chromogen mixing as indicated by the mixed colors. Specific staining was graded on a scale of 0 to 3 in 0.5 grade increments, with 0 indicating no staining, 1 indicating light staining, 2 indicating moderate staining, and 3 indicating strong staining.

3. Results

Antibodies were tested on tonsil tissue as single stains and double stains to determine the optimal concentrations and to evaluate each chromogen for sensitivity, background, and fine cellular detail. Representative images for some of the chromogens are shown in Figure 1. Immunohistochemical staining with pan-cytokeratin on tonsil tissue is shown in Figure 1A–D, depicting chromogens for green-HRP, blue-HRP, yellow-HRP, and red-AP, respectively. These figures show the expected specific staining of surface and crypt epithelium with minimal background staining of non-epithelial elements. Therefore, these chromogens can be used to identify cells of epithelial origin, including most carcinomas. Figure 1E,F show staining results for B-lymphocytes in tonsil tissue with chromogens for yellow-HRP and red-AP. A typical staining pattern shows an abundance of B-lymphocytes in germinal centers and mantle zones of lymphoid follicles, with a lesser distribution of positive cells within the interfollicular spaces. Figure 1H,I show a combination of pan-cytokeratin and B-lymphocyte markers. In Figure 1H, pan-cytokeratin is stained with yellow-HRP and B-lymphocytes are stained with blue-HRP. In Figure 1I, pan-cytokeratin is stained with green-HRP and B-lymphocytes are stained with yellow-HRP.
Next, chromogens were evaluated for their ability to withstand the elution buffer without fading. The single stains were incubated in elution buffer for 10 min at 50 °C. Photographic images were compared both before and after treatment with the elution buffer. Certain chromogens showed a partial loss of signal after incubation with the elution buffer as shown below in Table 4.
Because certain chromogens showed a partial loss of staining in the elution buffer, these chromogens were either omitted or used as final stains in a multi-stain sequence. Chromogens showing a partial loss of signal included PermaRed-HRP, PermaGreen-HRP, and PermaBlack-HRP.
Using the information gathered from these initial experiments, the optimal panels were constructed. Table 5 shows representative panels, and their corresponding microscopic images are shown in Figure 2, Figure 3 and Figure 4.
Figure 2 shows four-color immunohistochemistry staining on colorectal carcinoma. Tumor cells are stained with yellow-HRP, with pan-cytokeratin (AE1/AE3) showing a moderately to well-differentiated adenocarcinoma. Staining for Ki67 using green-HRP shows a high Ki67 index, indicating aggressive growth potential. The stained nuclei show a basal orientation in some areas of the tumor and a loss of polarity in other areas. There is a moderate background of inflammatory cells in the connective tissue between areas of carcinoma. Macrophages (CD163) are stained brown with DAB, and B-lymphocytes (CD20) are stained with blue-HRP. Box A in Figure 2 shows an area of the tumor containing a mixture of both macrophages and lymphocytes. The larger Box A shows this same area at a higher magnification.
Figure 3 depicts another moderately differentiated carcinoma showing adenocarcinoma morphology. In this example, the carcinoma cells are stained with pan-cytokeratin (AE1/AE3) using red-AP chromogen. Ki67 is stained using green-HRP and shows a moderate to high Ki67 index, indicating high growth potential (Box C). Nuclei also lost their polarization. Compared to the colorectal carcinoma in Figure 2, this carcinoma displays a substantially greater number of inflammatory cells. Macrophages (CD163) are stained brown with DAB and B-lymphocytes (CD20) are stained blue with blue-HRP. Furthermore, in this area of the tumor there appears to be some separation between the B-cells (Box A) and the macrophages (Box B). The larger boxes, A, B, and C, show the same areas at a higher magnification.
Figure 4 depicts an example of another colorectal carcinoma. The staining with red-AP for pan-cytokeratin (AE1/AE3) shows a moderately to well-differentiated adenocarcinoma. The tubular structures are widely dispersed throughout an extensive stroma (Box A). The Ki67 labeled with green-HRP shows a lower Ki67 index compared to the two previous examples of colorectal carcinoma (Figure 2 and Figure 3). There is a moderate scattering of inflammatory cells composed of macrophages (CD68, brown) and B-lymphocytes (CD79b, blue). Occasional tertiary lymphoid structures are observed and stained positive for B-lymphocytes (Box B). The larger boxes, A and B, show the same areas at a higher magnification.

4. Discussion

A thorough understanding of the various technologies strengths, weaknesses, and limitations is crucial in answering specific questions regarding tumor heterogeneity and the microenvironment. No single technology can provide all the answers to all the questions. Several unique technologies have been introduced to address some of these questions [14,15,16]. Morrison et al. [15] introduced the concept of invisible chromogens that have narrowly defined absorbance bands in the ultraviolet or near-infrared spectra, making these chromogens invisible within the visible spectrum. By selecting appropriate filters, the chromogens and conventional stains could be separately imaged. Other investigations have shown the development of new chromogens based on tyramine chemistry with narrow absorbance bands, making them ideal candidates for spectral imaging [16].
Here, we present a new technology based on classical IHC chromogenic methods that can be easily incorporated into most pathology laboratories without modification of existing equipment or practices. This method is based on various chromogenic substrates that provide excellent color separation. Multiplex staining is achieved by sequential IHC with antibody elution performed between each sequential step. Using this approach, we have demonstrated the feasibility of identifying up to four different markers with excellent color differentiation. Furthermore, the time required to achieve these results is within a few hours, thus making this method feasible within the typical laboratory’s daily workflow.
Depending on the specific antibody panel chosen for multiplex staining, the complexity of the information obtained can be overwhelming such that mere visual interpretation by the microscopist may be insufficient to extract all relevant information and interpret all expression patterns. Such situations can certainly benefit from computer-aided image analysis using suitable algorithms [17,18]. In such situations, spectrally distinct chromogens are critical for accurate results. In this study, we have shown the feasibility of a new method for creating multiplex IHC stains that takes advantage of existing methods and extends those methods by the introduction of a new set of chromogenic substrates. The implementation of spatial technologies is increasingly important to identify and locate specific cell types and patterns in the context of the tumor microenvironment, and this may well lead to new insights into previously unrecognized biological processes.

5. Conclusions

In this study, we have described a method for multiplex staining in tissue samples using standard immunohistochemical methods. This method relies on the development of a new set of chromogens that can be used individually or in combination with existing chromogens to generate multiplex stains. When used in multiplex stains, these chromogens show fine cellular detail, display low background, and retain their original colors. Chromogens exhibiting sufficient color separation from each other can be combined in such a way that simple visual interpretation of the resulting images is possible. Using this method, we have successfully demonstrated four-color immunohistochemistry. Depending on the types of staining patterns generated, visual analysis without the assistance of computer-aided image analysis may be possible.

Author Contributions

Conceptualization, B.G. and G.Y.; methodology, M.K.; writing—original draft preparation, M.K.; project administration, B.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

IRB review was not required for this study as there was no interaction or intervention with human subjects or access to identifiable private information.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in the study are included in the article. Further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors were employed by, or received funding from, Diagnostic BioSystems, Inc.

References

  1. Lee, J.; Hyeon, D.Y.; Hwang, D. Single-Cell Multiomics: Technologies and Data Analysis Methods. Exp. Mol. Med. 2020, 52, 1428–1442. [Google Scholar] [CrossRef] [PubMed]
  2. Stuart, T.; Satija, R. Integrative Single-Cell Analysis. Nat. Rev. Genet. 2019, 20, 257–272. [Google Scholar] [CrossRef] [PubMed]
  3. Topalian, S.L.; Taube, J.M.; Pardoll, D.M. Neoadjuvant checkpoint blockade for cancer immunotherapy. Science 2020, 367, eaax0182. [Google Scholar] [CrossRef] [PubMed]
  4. Hickey, J.W.; Neumann, E.K.; Radtke, A.J.; Camarillo, J.M.; Beuschel, R.T.; Albanese, A.; McDonough, E.; Hatler, J.; Wiblin, A.E.; Fisher, J.; et al. Spatial Mapping of Protein Composition and Tissue Organization: A Primer for Multiplexed Antibody-Based Imaging. Nat. Methods 2022, 19, 284–295. [Google Scholar] [CrossRef] [PubMed]
  5. Bolognesi, M.M.; Manzoni, M.; Scalia, C.R.; Zannella, S.; Bosisio, F.M.; Faretta, M.; Cattoretti, G. Multiplex Staining by Sequential Immunostaining and Antibody Removal on Routine Tissue Sections. J. Histochem. Cytochem. 2017, 65, 431–444. [Google Scholar] [CrossRef] [PubMed]
  6. Allam, M.; Cai, S.; Coskun, A.F. Multiplex Bioimaging of Single-Cell Spatial Profiles for Precision Cancer Diagnostics and Therapeutics. NPJ Precis. Oncol. 2020, 4, 11. [Google Scholar] [CrossRef] [PubMed]
  7. Fl Guillot, A.; Kohlhepp, M.S.; Bruneau, A.; Heymann, F.; Tacke, F. Deciphering the Immune Microenvironment on A Single Archival Formalin-Fixed Paraffin Embedded Tissue Section by An Immediately Implementable Multiplex Fluorescence Immunostaining Protocol. Cancers 2020, 12, 2449. [Google Scholar] [CrossRef] [PubMed]
  8. Buchwalow, I.B.; Minin, E.A.; Boecker, W. A multicolor fluorescence immunostaining technique for simultaneous antigen targeting. Acta Histochem. 2005, 107, 143–148. [Google Scholar] [CrossRef] [PubMed]
  9. Lin, J.-R.; Fallahi-Sichani, M.; Sorger, P.K. Highly Multiplexed Imaging of Single Cells Using a High-Throughput Cyclic Immunofluorescence Method. Nat. Commun. 2015, 6, 8390. [Google Scholar] [CrossRef] [PubMed]
  10. Lorsakul, A.; Bredno, J.; Ochs, R.L.; Morrison, L.; Day, W. Validation of multiplex immunohistochemistry assays using automated image analysis. In Medical Imaging 2018: Digital Pathology; SPIE: Bellingham, WA, USA, 2018; Volume 10581, p. 105810J. [Google Scholar]
  11. Morrison, L.E.; Lefever, M.R.; Behman, L.J.; Leibold, T.; Roberts, E.A.; Horchner, U.B.; Bauer, D.R. Brightfield multiplex immunohistochemistry with multispectral imaging. Lab. Investig. 2020, 100, 1124–1136. [Google Scholar] [CrossRef] [PubMed]
  12. Fiore, C.; Bailey, D.; Conlon, N.; Wu, X.; Martin, N.; Fiorentino, M.; Finn, S.; Fal, K.; Andersson, S.W.; Andren, O.; et al. Utility of multispectral imaging in automated quantitative scoring of immunohistochemistry. J. Clin. Pathol. 2012, 65, 496–502. [Google Scholar] [CrossRef]
  13. Pirici, D.; Mogoanta, L.; Kumar-Singh, S.; Piricin, I.; Margaritescu, C.; Simionescu, C.; Radu Stanescu, R. Antibody Elution Method for Multiple Immunohistochemistry on Primary Antibodies Raised in the Same Species and of the Same Subtype. J. Histochem. Cytochem. 2009, 57, 567–575. [Google Scholar] [CrossRef] [PubMed]
  14. Roberts, E.A.; Morrison, L.E.; Behman, L.J.; Draganova-Tacheva, R.; O’Neill, R.; Solomides, C.C. Chromogenic immunohistochemical quadruplex provides accurate diagnostic differentiation of non-small cell lung cancer. Ann. Diagn. Pathol. 2020, 45, 151454. [Google Scholar] [CrossRef] [PubMed]
  15. Morrison, L.E.; Lefever, M.R.; Lewis, H.N.; Kapadia, J.M.; Bauer, D.R. Conventional histological and cytological staining with simultaneous immunohistochemistry enabled by invisible chromogens. Lab. Investig. 2022, 102, 545–553. [Google Scholar] [CrossRef] [PubMed]
  16. Day, W.A.; Lefever, M.R.; Ochs, R.L.; Pedata, A.; Behman, L.J.; Ashworth-Sharpe, J.; Johnson, D.D.; May, E.J.; Grille, J.G.; Roberts, E.A.; et al. Covalently deposited dyes: A new chromogen paradigm that facilitates analysis of multiple biomarkers in situ. Lab. Investig. 2017, 97, 873–885. [Google Scholar] [CrossRef] [PubMed]
  17. Blom, S.; Paavolainen, L.; Bychkov, D.; Turkki, R.; Mäki-Teeri, P.; Hemmes, A.; Välimäki, K.; Lundin, J.; Kallioniemi, O.; Pellinen, T. Systems Pathology by Multiplexed Immunohistochemistry and Whole-Slide Digital Image Analysis. Sci. Rep. 2017, 7, 15580. [Google Scholar] [CrossRef] [PubMed]
  18. De Smet, F.; Antoranz Martinez, A.; Bosisio, F.M. Next-Generation Pathology by Multiplexed Immunohistochemistry. Trends Biochem. Sci. 2021, 46, 80–82. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Optimization of chromogens on tonsil tissue. Tonsil tissue stained with chromogens (100×). (A) Pan-cytokeratin (AE1/AE3) with green-HRP, (B) pan-cytokeratin (AE1/AE3) with blue-HRP, (C) pan-cytokeratin (AE1/AE3) with yellow-HRP, (D) pan-cytokeratin (AE1/AE3) with red-AP, (E) B-cell (CD20) with yellow-HRP, (F) B-cell (CD20) with red-AP, (G) macrophage (CD163) with DAB, (H) pan-cytokeratin (AE1/AE3) with yellow-HRP and B-cell (CD20) with blue-HRP, and (I) pan-cytokeratin (AE1/AE3) with green-HRP and B-cell (CD20) with yellow-HRP.
Figure 1. Optimization of chromogens on tonsil tissue. Tonsil tissue stained with chromogens (100×). (A) Pan-cytokeratin (AE1/AE3) with green-HRP, (B) pan-cytokeratin (AE1/AE3) with blue-HRP, (C) pan-cytokeratin (AE1/AE3) with yellow-HRP, (D) pan-cytokeratin (AE1/AE3) with red-AP, (E) B-cell (CD20) with yellow-HRP, (F) B-cell (CD20) with red-AP, (G) macrophage (CD163) with DAB, (H) pan-cytokeratin (AE1/AE3) with yellow-HRP and B-cell (CD20) with blue-HRP, and (I) pan-cytokeratin (AE1/AE3) with green-HRP and B-cell (CD20) with yellow-HRP.
Cells 13 00936 g001
Figure 2. Four-plex stain on colorectal carcinoma using chromogens for HRP. Colorectal carcinoma (100×). Pan-cytokeratin (AE1/AE3) stained with yellow-HRP. Macrophage (CD163) stained with DAB. B-cell (CD20) stained with blue-HRP. Proliferation marker (Ki67) stained with green-HRP. Box A shows an area of the tumor at its original location (smaller Box A) and a higher magnification of the selected area (larger Box A).
Figure 2. Four-plex stain on colorectal carcinoma using chromogens for HRP. Colorectal carcinoma (100×). Pan-cytokeratin (AE1/AE3) stained with yellow-HRP. Macrophage (CD163) stained with DAB. B-cell (CD20) stained with blue-HRP. Proliferation marker (Ki67) stained with green-HRP. Box A shows an area of the tumor at its original location (smaller Box A) and a higher magnification of the selected area (larger Box A).
Cells 13 00936 g002
Figure 3. Four-plex stain on colorectal carcinoma using chromogens for AP and HRP. Colorectal carcinoma (100×). Pan-cytokeratin (AE1/AE3) stained with red-AP. Macrophage (CD163) stained with DAB. B-cell (CD20) stained with blue-HRP. Proliferation marker (Ki67) stained with green-HRP. Box A shows an area of predominantly B-cells (CD20). Box B shows an area of predominantly macrophages (CD163). Box C shows an area of predominantly tumor cells (AE1/AE3) that are also positive for the proliferation marker (Ki67). Small boxes A, B, and C show the tumor areas in their original locations, and larger boxes A, B, and C show a higher magnification of the same corresponding areas.
Figure 3. Four-plex stain on colorectal carcinoma using chromogens for AP and HRP. Colorectal carcinoma (100×). Pan-cytokeratin (AE1/AE3) stained with red-AP. Macrophage (CD163) stained with DAB. B-cell (CD20) stained with blue-HRP. Proliferation marker (Ki67) stained with green-HRP. Box A shows an area of predominantly B-cells (CD20). Box B shows an area of predominantly macrophages (CD163). Box C shows an area of predominantly tumor cells (AE1/AE3) that are also positive for the proliferation marker (Ki67). Small boxes A, B, and C show the tumor areas in their original locations, and larger boxes A, B, and C show a higher magnification of the same corresponding areas.
Cells 13 00936 g003
Figure 4. Four-plex stain of colorectal carcinoma showing inflammatory cells. Colorectal carcinoma (100×) showing an area of tumor stroma with an extensive inflammatory cell infiltrate. Tumor cells are stained with red-AP for pan-cytokeratin (AE1/AE3) and green-HRP for the proliferation marker (Ki67). Macrophages (CD68) are stained with DAB and B-cells (CD79a) are stained with blue-HRP. The small boxes A and B show the tumor areas at their original locations and the large boxes A and B show the same corresponding areas at a higher magnification.
Figure 4. Four-plex stain of colorectal carcinoma showing inflammatory cells. Colorectal carcinoma (100×) showing an area of tumor stroma with an extensive inflammatory cell infiltrate. Tumor cells are stained with red-AP for pan-cytokeratin (AE1/AE3) and green-HRP for the proliferation marker (Ki67). Macrophages (CD68) are stained with DAB and B-cells (CD79a) are stained with blue-HRP. The small boxes A and B show the tumor areas at their original locations and the large boxes A and B show the same corresponding areas at a higher magnification.
Cells 13 00936 g004
Table 1. Antibody list for multiplex staining.
Table 1. Antibody list for multiplex staining.
AntibodySpeciesClonePart No.
CD3 (epsilon chain)RabbitSP7RMAB005
CD4RabbitEP204RMAB053
CD5RabbitSP19RMAB11
CD8Mouse144BMob117
CD20MouseL26Mob004
CD68MousePGM1Mob094
CD79aMouseHM57Mob118
CD163Mouse10D6Mob480
Ki67RabbitSP6RMAB004
PD-1 (CD279)MouseEH33Mob573
PD-L1 (CD274)Mouse405-9A11Mob572
Cytokeratin HMWMouse34BE12Mob059
Pan-CytokeratinMouseAE1/AE3Mob190
Table 2. Reagent list.
Table 2. Reagent list.
NamePart No.
Tissue GlueK096
Wash BufferK005
UnoVue Anti-mouse HRPMUHRP-100
UnoVue Anti-rabbit HRPRUHRP-100
UnoVue Anti-mouse APMUAP-100
UnoVue Anti-rabbit APRUAP-100
Antigen Retrieval pH 9.0K043
Primary Antibody DiluentK004
Table 3. List of chromogens.
Table 3. List of chromogens.
ChromogenPart NumberEnzyme System
PermaRed-HRPK075Peroxidase
PermaBlue-HRPK063Peroxidase
PermaGreen-HRPK074Peroxidase
PermaYellow-HRPK060Peroxidase
PermaBlack-HRPK062Peroxidase
Stable DABK047Peroxidase
PermaRed-APK049Alkaline Phosphatase
Table 4. Effects of the elution buffer on chromogens.
Table 4. Effects of the elution buffer on chromogens.
ChromogenEffect of the Elution Buffer On Chromogen Staining
PermaRed-HRPSlight loss of staining
PermaBlue-HRPNo effect
PermaGreen HRPModerate loss of staining
PermaYellow-HRPNo effect
PermaBlack-HRPSlight loss of staining
Stable DABNo effect
PermaRed-APNo effect
Table 5. Optimal panels for four-color immunohistochemistry.
Table 5. Optimal panels for four-color immunohistochemistry.
SequencePrimary AntibodyChromogenTissueImage
1Pan-cytokeratin (AE1/AE3)Yellow-HRP
2Macrophage (CD163)DABColorectalFigure 2
3B-cell (CD20)Blue-HRPCarcinoma
4Proliferation marker (Ki67)Green-HRP
1Pan-cytokeratin (AE1/AE3)AP-Red
2Macrophage (CD163)DABColorectalFigure 3
3B-cell (CD20)Blue-HRPCarcinoma
4Proliferation marker (Ki67)Green-HRP
1Pan-cytokeratin (AE1/AE3)AP-Red
2Macrophage (CD68)DABColorectalFigure 4
3B-cell (CD79a)Blue-HRPCarcinoma
4Proliferation marker (Ki67)Green-HRP
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gupta, B.; Yang, G.; Key, M. Novel Chromogens for Immunohistochemistry in Spatial Biology. Cells 2024, 13, 936. https://doi.org/10.3390/cells13110936

AMA Style

Gupta B, Yang G, Key M. Novel Chromogens for Immunohistochemistry in Spatial Biology. Cells. 2024; 13(11):936. https://doi.org/10.3390/cells13110936

Chicago/Turabian Style

Gupta, Bipin, George Yang, and Marc Key. 2024. "Novel Chromogens for Immunohistochemistry in Spatial Biology" Cells 13, no. 11: 936. https://doi.org/10.3390/cells13110936

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop