Loading

Journal of Veterinary Medicine and Research

Applications of Flow Cytometry in Veterinary Research and Small Animal Clinical Practice

Review Article | Open Access

  • 1. Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, USA
  • 2. Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, USA
+ Show More - Show Less
Corresponding Authors
Pinchuk L, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, PO Box 6100, Mississippi State, 39762-6100, United States of Ameria
Abstract

Flow cytometry is a rapid, versatile and powerful technique that allows for qualitative and quantitative assessment of multiple parameters of individuals cells and particles. With the flexibility of flow cytometry, and the reliability of results, this technology has become an essential veterinary research instrument with important clinical diagnostic capabilities, particularly in the fields of immunology and hematology. This methodology has played a key role in the discovery of immunological cells and multiple aspects of canine immune function. Flow cytometric techniques have been instrumental in the identification of regulatory T lymphocytes and roles they play in autoimmune diseases, as well as characterizing and measuring cellular products, such as cytokines and other proteins. Flow cytometry has also been used in the pharmacodynamic monitoring of immunosuppressive therapies in dogs. Besides immunological cells, flow cytometry allows for the evaluation of erythrocytes, specifically the quantitation of reticulocytes, detection of anti-erythrocyte antibodies, and diagnose of erythrocyte parasites. Similar techniques can also be applied to canine platelets to diagnose immature reticulated platelets and anti-platelet antibodies. Flow cytometry can also be used to assess various aspects of platelet function. Flow cytometry is most often applied in veterinary oncology to diagnose and stage lymphoid neoplasia and leukemia, and can also be used to assess neoplastic and non-neoplastic marrow conditions. This article will cover some of the most common and popular uses of flow cytometry in small animals, and discuss the influences that this technology has on veterinary research and small animal clinical medicine.

Keywords


•    Flow cytometry
•    Regulatory T cells
•    Erythrocytes
•    Neoplasia
•    Platelets

Citation

Thomason J, Archer T, Mackin A, Stokes J, Pinchuk L (2014) Applications of Flow Cytometry in Veterinary Research and Small Animal Clinical Practice. J Vet Med Res 1(1): 1004

ABBREVIATIONS

FSC: Forward-angle scatter; SSC: Side-angle scatter; FITC: Fluorescein isothiocyanate; PE: Phycoerythrin; APC: Allophycocyanin; CD: Cluster of Differentiation; NK: Natural Killer; IL: Interleukin; Tregs: Regulatory T cells; FOXP3: Forkhead box P3; DCs: Dendritic cells; IFN-γ: Interferon gamma; PBMCs: Peripheral blood mononucleated cells; TLRs: Toll-like receptors; ROS; Reactive oxygen species; CFSE; Carboxyfluorescein succinimidyl ester; IMTP: Immune-mediated thrombocytopenia; GP: Glycoproteins; IMHA: Immune-mediated hemolytic anemia; COX: Cyclooxygenase; IMPA: Immune-mediated polyarthritis

INTRODUCTION

Flow cytometry, a laser-based biophysical technology employed in cell counting, cell sorting and cellular marker detection, is routinely used in veterinary medicine. There have been significant advancements in flow cytometric technology over the past 30 years. Flow cytometers are highly complex and expensive instruments that have traditionally required a dedicated, highly trained professional to operate but, with technological advancements, flow cytometric analysis is becoming simpler, and can be applied to an ever widening variety of applications. Initially, flow cytometry was used as a cell counter and research tool; however, with changes and improvements to instrument design, flow cytometry has not only become an extremely effective and popular research methodology, but has also become an integral component to clinical veterinary medicine. In fact, flow cytometric techniques that were once believed to solely be used for research assessment, are currently being used by veterinarians to diagnose and monitor therapy in clinic patients on an individual basis. In many veterinary clinical pathology laboratories, the incorporation of flow cytometry into hematology analyzers has replaced manual counting of hematologic cells, allowed for assessment of cellular maturity, and provided additional diagnostic modalities. User-friendly versions of these laser-based flow cytometry hematology analyzers have also recently attained common usage in general veterinary practice. Today, flow cytometers remain essential research instruments with important clinical diagnostic capabilities but, with the flexibly of these units, and the reliability of results, the use of flow cytometry in veterinary medicine will continue to grow and be applied to a vast number of both research and clinical applications. Due to the seemingly endless number on possible uses of flow cytometry, a discussion on all uses of this methodology is beyond the scope of this article, therefore, in this article, the authors have selected some of the most common and popular uses of flow cytometry in small animals, particularly dogs, and will discuss the influences of flow cytometry on veterinary research and clinical medicine. Laser-based flow cytometry hematology analyzers will not be discussed.

FLOW CYTOMETRY PRINCIPLES

Flow cytometers are complex instruments that consist of optics, lasers, electronic networks, advanced software, and most often a fluidics system. The detailed operations of a flow cytometer are beyond the scope of this article. However, the basic concept of flow cytometry is that cells or particles flow through a laser to identify or measure the characteristics of that particular cell or particle. The sample of interest is suspended in fluid, and a pressurized line directs the cells to flow into a central core that forces the cells to form in a single file line. As the sample continues to flow, the aligned cells will pass through the laser, thereby analyzing one cell or particle at a time. [1-3]

As individual cells pass through the laser beam, two types of light scatter occur: forward-angle scatter (FSC) and side-angle scatter (SSC). The light that passes through each individual cell in a forward direction is the forward-angle scatter and represents the refractive index of the cell, which roughly correlates to size. The light that scatters in a 90-degree angle is the side-angle scatter, and represents the internal complexity of the cell. The unique light scatter properties of a particular cell type can be used to distinguish and identify subpopulations of cells, making this technique an integral part in the laser-based clinical hematology analyzers. [2,3]

To enhance the capabilities of these instruments, fluorochromes can be conjugated to antibodies that target specific antigens on a cell. These fluorescently labeled antibodies are added to the sample and incubated, and then weakly bound non-specific and unbound antibodies are washed off before the labeled sample is injected into the flow cytometer. As the fluorescently labeled cell passes through the laser, the fluorochromes will emit a light of a specific wavelength which will be collected by a detector, converted to a digital output and transferred to a computer for analysis. By incorporating multiple fluorochrome dyes that emit light at different wavelengths, such as fluorescein isothiocyanate (FITC), phycoerythrin (PE), and allophycocyanin (APC), multiple antigens can be identified on the same sample at the same time. The intensity of the emitted light is directly proportional to the quantity of bound antibody. Unfortunately, when compared to humans, there is limited availability of antibodies specific to dogs. However, depending on the antigen of interest, there may be appropriate cross-reactivity with antibodies from other species to allow flow cytometric analysis. An additional benefit of some flow cytometers is the ability to sort subpopulations of fluorescent-labeled cells that can be collected and further analyzed [2,3].

EVALUATION OF THE CANINE IMMUNE SYSTEM BY FLOW CYTOMETRY

Single, two and three parameter flow cytometry

Flow cytometry has played a key role in discovery of immunological cells, and has helped facilitate evaluation of canine immune function. Canine peripheral blood leukocyte subsets, in particular CD4+ and CD8+ T cells, CD21+ B cells, CD5lo natural killer (NK) cells and CD14+ monocytes/macrophages have been stained, identified with species-specific antibodies, and sorted by flow cytometry. [4-13] Additionally, using three color flow cytometry analysis, novel canine cell populations, in particular CD3+ CD4+ CD8+ double positive T cells with features of activated T cells, have been phenotypically identified in the peripheral blood of dogs. [14]

The discovery of a population of suppressive CD4+ T cells characterized by high constitutive expression of the IL-2Rα chain (CD25) led to remarkable progress in elucidating the phenomenon of cell-mediated suppression and its importance in self-tolerance and regulation of adaptive immune responses. [15- 17] In classical suppression assays, adult mice that expressed CD4+ CD25+ T cells were significantly less likely to develop organspecific autoimmune disease than mice that expressed CD4+ CD25- T cells. [18] The identification of these cell populations lead to the discovery of a key mechanism of peripheral tolerance and a critical component of the host immune system, regulatory T cells (Tregs), which are a small population of T lymphocytes that possess immunomodulatory properties and aid in the suppression of autoimmune responses. Using flow cytometry, CD4+ T cells with the highest CD25 expression were shown to be enriched for FOXP3, a transcription factor that is highly expressed in Tregs in humans. [19,20] Recently, using similar flow cytometric techniques, CD4+ CD25+ FOXP3+ regulatory T cells were described in dogs (Weaver et al., unpublished observation, 2013). [19-25] With analytical three-color flow cytometry and fluorescence-activated cell sorting, canine Tregs have been characterized phenotypically similar to humans, providing direct evidence of Treg regulatory function in dogs, which can also serve as a model for human diseases. [26-28]

Dendritic cells (DCs), the most potent antigen-presenting cells, represent a heterogeneous group of bone-marrow derived cells, and serve as sentinels of the immune system, capable of capturing and processing antigens. Canine DCs have been identified, via flow cytometry, in the peripheral blood [10], gastrointestinal tract [29] and skin [30], with panels of caninespecific and cross-reactive antihuman monoclonal antibodies. Current protocols for isolation and generation of canine monocyte- and bone marrow derived DCs involve extensive flow cytometric analysis of multiple phenotypical and functional changes in DCs upon maturation. [6,7,31-36]

The use of flow cytometry to identify the phenotype and function of immune cells is not limited to peripheral blood. Microglial cells are the main immune effector cell of the brain, and respond to diseases within the central nervous system. Once activated, microglial cells are capable of several immunological functions, including phagocytosis, modulation of T-cell response, and production of cytokines. The extensive immune phenotyping of the surface markers expression in canine microglial cells have been identified by single color flow cytometry analysis. [37,38]

Along with the ability to label and categorize individual cells via fluorescent antibodies, flow cytometry is also capable of identifying and measuring cellular products such as cytokines, proteins, and other factors. Cytokines are important mediators of immune responses, and are produced by a variety of activated lymphocytes. The advent of intracellular cytokine assessment by flow cytometry through staining of permeabilized cells with fluorochrome-conjugated anti-cytokine antibodies allows cytokine levels in a large numbers of cells to be rapidly examined. Several studies have described flow cytometric assays for the measurement of cytokines involved in T cell activation and polarization. Requiring only a small volume of blood, a rapid whole-blood flow cytometric assay has been developed to measure interferon-γ (IFN-γ) and interleukin-4 (IL-4) produced from canine CD4+ and CD8+ lymphocytes. [39]. In our laboratory, we have evaluated the effects of in vitro and in vivo cyclosporine exposure on a panel of cytokines, including IL-2, IL-4 and IFN-γ, produced by activated T cells to ascertain their potential as markers of immunosuppression in dogs. [12,13] Interferon γ, predominantly produced by NK and Natural Killer T cells, is a fundamental component of the innate immune response, and is vital to the immunological response to viral and intracellular bacterial infections and tumor control. In canine peripheral blood mononucleated cells (PBMC) assessed by one or two color surface expression and intracellular staining flow cytometry approaches, Huang and others recently discovered CD5lo cells that closely resembled human NK cells by expressing NK-specific cell markers and producing high levels of IFN-γ. [8] Finally, in addition to cytokine detection, flow cytometry is capable of determining the purity of samples, for example via the detection of intracellular staining of CD79α in canine B-cells after being magnetically separated. [11]

Multiparameter and polychromatic flow cytometry

Multiparameter flow cytometry is a powerful analytical tool that enables the rapid measurement of multiple physical and chemical characteristics of individual cells. Recently, a multiparametric 4-color flow cytometry assay was used to evaluate the changes related to the maturation of lymphocyte subsets as in young dogs. [40] Multiparameter flow cytomery approaches have also successfully been applied for the separation and phenotypic characterization of several canine DC populations. Purified and sorted by a 4-color flow cytometer, canine CD11c+ / HLA-DR+ /CD14- /DM5- cell populations were determined to have the same functional and morphological properties as human myeloid DCs. [10]

The development of polychromatic cytometry (greater than 4 colors) has contributed to significant progress in the field of human immunology. Recently, 12 color panels were developed to simultaneously investigate the direct activation of multiple DC subsets, monocytes, and NKs, and their relationships within the PBMC environment after stimulation with a live-influenza virus or toll-like receptor (TLR) agonists. [41] Currently, the complex polychromatic cytometry approach has not been widely utilized in clinical veterinary medicine. However, a recent report using 7-color multiparameter flow cytometry provided a comprehensive approach to characterize canine lymphocyte phenotypes expressing multiple markers in healthy research dogs. [42]

Functional assays in flow cytometry

The function of immune cells, in particular neutrophils, macrophages, monocytes and DCs, can be evaluated by multiple functional flow cytometry methods. Phagocytosis is an important early step in triggering adaptive immune responses, which require processing of bacterial pathogens and presentation of their antigens to CD4+ and CD8+ T cells. [43] Several reports characterized phagocytosis in canine neutrophils, PBMC and peripheral blood-derived monocytes/macrophages by using FITC-latex beads [9,44] and Salmonella enterica serotype Kentucky labeled with GFP, a plasmid with green fluorescence (Harris et al., unpublished observation, 2010). Additionally, flow cytometry has been employed to evaluate the active uptake of FITC-labelled dextran by canine monocyte-derived DCs. [32,33] Finally, the phagocytic properties of canine microglial cells were measured using flow cytometry by using heat-killed and lyophilized FITC-labelled Staphylococcus aureus as an antigen. [37,38]

Flow cytometry is uniquely suited for the measurement of the oxidative burst and superoxide production in neutrophils and monocyte/macrophages. Compared to more conventional techniques, the major advantages of flow cytometry for the assessment of these cells are that only a very small volume of blood is required for analysis and that the results are objective and quantifiable. Neutrophils produce reactive oxygen species (ROS) and generate superoxide anion essential to their bactericidal function. Apoptosis is the process of programmed cell death in response to a stress that leads to characteristic cell changes and death. The mitochondria play a vital role in one of the major apoptosis signaling pathways, and mitochondrial dysfunction is part of the pathogenesis of multiple viral, bacterial and parasite infections. Recently, using flow cytometry, it was determined that the oxidative stress and acceleration of apoptosis associated with chronic renal disease in dogs decreases neutrophil production of superoxide. [45]. An additional study compared the status of apoptosis of peripheral blood leukocytes in dogs with demodicosis and healthy dogs, and found that in dogs with demodecosis a significantly greater percentage of cells exhibited externalization of phosphatidylserine, a hallmark sign of depolarized mitochondrial membrane potentials and early apoptosis. [46]. Extrinsic apoptosis can be evaluated by the assessment of the cellular surface expression of CD95 (Fas) and CD178 (Fas ligand). Recently, CD95 and CD178 molecules were shown to induce apoptosis in CD4+ and CD8+ T cells isolated from peripheral blood and spleen of dogs naturally infected with Leishmania [5]. Analysis of the functional ability of canine microglial cells to produce ROS has been performed by using dihydrorhodamine and flow cytometric analysis. [37]

Cell proliferation analyses are crucial for evaluating cell growth and differentiation, and are typically based on average DNA content or on cellular metabolism parameters. Carboxyfluorescein succinimidyl ester (CFSE) is a novel cell tracing fluorescent dye used to examine the proliferative activity of cells by labeling a parent generation and determining the inheritance by the daughter generation. A recent report used flow cytometry to assess the effects of synthetic agonists on TLR 9 and the regulation of canine lymphocyte proliferation by CFSE division. [47] Also, canine CD4+ CD8+ double positive T cells express high proliferation potential based on the CFSE division technique [14]. Proliferative response of T cells in dogs have been also assessed with monoclonal antibodies to IL-2R (CD25) to detect the percentage of CD4+ CD25low activated T cells. [48-50]

Flow Cytometric Analysis of Erythrocytes

Some of the major applications of flow cytometry for erythrocytes include the quantitation of reticulocytes, detection of anti-erythrocyte antibodies, and the detection and diagnose of erythrocyte parasites.

Reticulocytes are immature red blood cells (RBCs) that contain a reticular (mesh-like) network of residual ribosomal RNA, and quantifying reticulocytes serves as a reliable measure of marrow red cell production. Although several types of fluorochromes are capable of labeling reticulocytes, thiazole orange is the most common label used to detect canine reticulocytes via flow cytometry. Thiazole orange will passively enter the reticulocyte, bind to residual RNA, and fluoresce when exposed to laser light of the appropriate wavelength. When compared to traditional manual reticulocyte counts, the thiazole orange technique correlates well as long as reticulocyte percentages are greater than 2% of all RBCs. However, as the percentage of reticulocytes decreases, so does the accuracy of the test, suggesting that flow cytometric assessment of immature erythrocytes is best performed in patients with a normal or elevated reticulocyte count. [51]

Immune-mediated hemolytic anemia (IMHA) in dogs is mediated by the presence of antibody bound to the surface of the RBC. While the traditional means of detecting these antibodies is the direct antiglobulin (Coombs’) test, flow cytometry has often been used as a simple and rapid alternative means of detecting anti-RBC antibodies, of quantitating complement involvement, and of determining the precise immunoglobulin sub-type (IgG or IgM) involved. [52-54]

Flow cytometric techniques have also been developed for detecting and quantifying RBC parasites in dogs. Parasitemia with low numbers of organisms such as Babesia can be difficult to detect by the standard method of examination of stained blood smears and, although very sensitive polymerase chain reaction (PCR) based methods have now been developed for organism detection, such methods are so sensitive that even a few parasitized RBCs can lead to a strong positive result. Flow cytometry offers a practical ‘middle ground’, in that even a small number of parasitized cells can be detected, but the precise number of infected cells can be accurately and rapidly counted. Stains that bind to protozoal nucleotides, particularly hydroethidine, have been successfully used to measure RBC parasitemia in dogs affected with Babesia gibsoni and Babesia canis. [55,56] One potential drawback with using a non-specific nucleotide label rather than an organism-specific antibody label for detecting the presence of RBC organisms is that a positive flow cytometry result may not necessarily be detecting the target organism, but may instead be detecting other sources of nucleotide, such as the residual RNA within reticulocytes [57] or other RBC organisms such as the hemotropic mycoplasma [58].

Morpological and Functional Evaluation of Canine Platelets

Flow cytometry is capable of evaluating several aspects of platelets, including quantitation of immature (reticulated) platelets, detection of anti-platelet antibodies, and the detection of activated platelets. Flow cytometric assessment of platelets can be performed on multiple sample types, including washed platelets, gel-filtered platelets, platelet-rich plasma, and whole blood samples; however, whole blood has become the preferred sample type due to minimal sample processing and reduced risk of artifactual in vitro platelet activation. The use of whole blood not only allows for the assessment of platelet function in near physiologic conditions, where platelets can interact with other blood cells, but can also allow for the assessment of platelet function with the use of a small sample volume. Furthermore, unlike many other tests of platelet function, flow cytometry is capable of evaluating platelet function in thrombocytopenic and anemic patients.

Newly formed platelets that have recently been released into circulation are known as reticulated platelets, and still contain some residual mRNA. [59]. The percentage of circulating reticulated platelets can reflect megakaryocyte proliferation, and an elevated percentage of these new platelets usually indicate accelerated platelet production, while a lower percentage suggests poor megakaryocyte function. [59] Reticulated platelet numbers can help determine whether thrombocytopenia is caused by decreased platelet production or platelet consumption/ destruction. As with reticulocytes, thiazole orange can be used to detect the residual mRNA in reticulated platelets. However, at least in humans, this fluorochrome is not specific for residual platelet RNA, and approximately half of the thiazole orange staining is associated with uptake in dense granules. [60]

Immune-mediated thrombocytopenia (IMTP) is associated with binding of anti-platelet antibodies to the platelet surface membrane, leading to destruction of the platelet by phagocytic cells. Blood samples from dogs with suspected IMTP can be combined with a fluorescently labelled species-specific antiIgG and/or anti-IgM antibody and analyzed via flow cytometry in combination with platelet-specific labels. [61] According to Kristensen and others, approximately 67% of dogs with suspected IMTP were positive anti-platelet antibodies, however, it may be difficult to distinguish between primary and secondary immune mediated thrombocytopenia. [62,63] Ideally, flow cytometry should be performed within 24 hours of sample collection, and the assay also has limited sensitivity and specificity, reducing the clinical effectiveness of this diagnostic test.

Flow cytometry can measure the activation state and reactivity of circulating platelets, diagnose inherited and acquired platelet disorders, evaluate leukocyte-platelet aggregation and procoagulant changes to the platelet surface, and facilitate the assessment and monitoring of antiplatelet therapy. Unlike platelet aggregometry, where platelet activation is necessary, flow cytometry can evaluate surface protein expression both before and after platelet activation. Samples are typically labeled with two fluorescently conjugated monoclonal antibodies. The first antibody is usually specific for glycoproteins Ib, IIb, and IIIa (ie. anti-CD41 or anti-CD61), molecules common to all platelets, and serves to identify the platelet. Some samples can be left unlabeled, however, and in these instances identification of the platelet is based on characteristic forward and side scatter. The second fluorescently conjugated antibody is specific to the epitope or receptor of interest. In an attempt to stabilize the platelet membrane, the sample can be fixed before or after the addition of antibodies, although this process may adversely affect the binding of some antibodies. Flow cytometry techniques have been developed to identify activated platelets as well as microparticles released from activated platelets. [64-67]

There are several markers that indicate the presence of an activated platelet, including glycoprotein IIb/IIIa (GP IIb/IIIa), P-selectin, and leukocyte-platelet aggregates. The primary mediator of platelet aggregation involves a conformational change of the surface GPIIb/IIIa complex, enabling the binding of fibrinogen to stabilize the platelet plug [68]. With inactivated platelets, this receptor is in a resting conformation and, upon activation, there is a conformation change, allowing this receptor to bind fibrinogen. The monoclonal antibody PAC-1 only binds to the GP IIb/IIIa receptor in an activated conformation: therefore, a PAC-1 negative platelet is considered to be inactivated or resting, while a PAC-1 positive platelet is considered to be activated [69]. One of the molecules released from platelet α-granules during activation is P-selectin. Platelet P-selectin, CD62, is a cell adhesion molecule that mediates platelet and leukocyte aggregation and generates procoagulant microparticles that contain active tissue factor and enhance fibrin deposition. AntiCD62 monoclonal antibody binds to P-selectin expressed on the platelet surface only after activation and granule release, and a P-selectin negative platelet should therefore be considered to be inactivated or resting. [63,66,67] Platelet P-selectin has been shown to be expressed in higher concentrations in dogs with primary immune-mediated hemolytic anemia (IMHA) [64], and provides evidence that platelet activation could contribute to the high rate of thromboembolic complications seen in IMHA patients. The ability of P-selectin to mediate platelet and leukocyte aggregation can also be detected with flow cytometry, and leukocyte platelet aggregates are a sensitive marker of in vivo platelet activation. [67]

In resting platelets, the aminophospholipid phosphatidylserine is located in the inner leaflet of the platelet plasma membrane but, upon activation, platelet phosphatidylserine is exposed on platelet surface membranes and binds to prothrombin, accelerating the conversion of prothrombin to thrombin, and expediting the formation of a blood clot [70]. Flow cytometric detection of annexin V, a protein that preferentially binds to phosphatidylserine on the surface of platelets, can also detect the presence of activated platelets. The inherited inability of platelets to express phosphatidylserine is known as Scott Syndrome, and has been described in humans and dogs [71]. Flow cytometry can also be used to diagnose several other heritable platelet disorders, such as Glanzmann thrombasthenia (deficiencies in the GPIIb/ IIIa complex) and storage pool defects. [72,73]

Since mature platelets do not contain a nucleus and are therefore incapable of mRNA synthesis, and because COX1 is usually constitutively expressed by cells whereas COX2 expression is more typically induced, it was long believed that COX-1 was the only COX isoform expressed in circulating platelets, and that COX induction could happen only at the level of the bone marrow. However, a flow cytometry technique utilizing permeabilization methods has been described that has enabled identification and quantification of the expression of both COX-1 and COX-2 in circulating human and canine platelets. [74,75]

Despite the significant benefits of flow cytometry for evaluating platelets, there are several disadvantages and limitations to this methodology. Ideally, to avoid ex vivo platelet activation, samples should be processed within hours of collection, reducing the clinical practicability of this methodology. Additionally, while flow cytometry is excellent at determining the expression of proteins and receptors on the surface of activated platelets, the technique does not evaluate the platelet’s actual ability to form a platelet plug. Finally, activation of platelets prior to collection leads to binding to the vascular endothelium or loss of surface receptors/proteins, preventing these platelets from being appropriately assessed by flow cytometry.

Immunophenotyping in Canine Neoplasia

In human medicine, flow cytometry has become an essential component of the assessment, diagnose, and staging of cancer. Flow cytometry is primarily used for immunophenotyping, which is the use of a panel of antibodies to identify cell lineage, for example B or T cell lymphoma. Antibodies are directed toward molecules expressed by hematopoietic lineages and lymphoid subsets. The range of immunophenotyping panels used to assess canine neoplasia is still constrained because of associated expense and a limited availability of species-specific antibodies. In veterinary oncology, although flow cytometry is most often applied to hemolymphic neoplasia [76-78], particularly lymphoma, this methodology has also been used for the assessment of other neoplastic conditions such as osteosarcoma and transitional cell carcinoma. Immunophenotyping can also play a pivotal role in the assessment of canine leukemia by defining the cell lineage, stage of maturation, and prognostic information. [76,79]

In dogs with lymphoma, flow cytometry is not intended to replace conventional diagnostic techniques such as cytology or histopathology, but instead is used to assist in the classification and staging of specific tumor types. Based on flow cytometric studies in dogs, there is predominately one type of B-cell lymphoma, while T-cell lymphomas will typically have a variety of phenotypes. Large cell lymphoma, which is similar to an immunoblastic subtype characterized by the World Health Organization, is the most common form lymphoma in dogs, and originates from a B-cell lineage. On the cell surface, most B-cell lymphomas will express CD1, CD21, CD45RA, CD79a, CD90, and MHC class II, while most T-cell lymphomas are characterized by CD3, CD4, CD8, and αβ-TCR antigens. The presence of hypercalcemia in T-cell lymphoma is most frequently associated with CD4 and αβ-TCR antigens. [76,80]

One advantage of flow cytometry is that the technique can rapidly evaluate a large number of different cell types within a sample, and use multiple simultaneous labels to identify specific cells within a heterogeneous cell population. This is particularly invaluable when applied to bone marrow samples in patients with lymphoreticular neoplasia or ‘pre-leukemic’ leukemia, where a particular malignant cell type within the marrow can be identified and characterized from within a soup of multiple different cells. [78, 81-83]. In patients without neoplasia, similar methodology can be used to quantify the distribution of particular cell types within the marrow, including cells that are typically very difficult to count, such as stem cells [84-86], and cell sorting techniques can be used to isolate purified populations of specific cells [87].

Pharmacodynamic Monitoring in Dogs

One exciting new clinical application of flow cytometry in dogs is the pharmacodynamic monitoring of immunosuppressive drugs such as cyclosporine. Cyclosporine is a powerful immunosuppressive agent which inhibits T cell production of cytokines such as IL-2 and IFN-γ, and is being used in an increased frequency in veterinary medicine. In dogs, a microemulsion formulation of cyclosporine has FDA approval for the treatment of canine atopic dermatitis, and this formulation is also used off label as an immunosuppressive agent for the treatment of other conditions such as inflammatory bowel disease, anal furunculosis, IMHA, IMTP, and immune-mediated polyarthritis [88]. When treating life-threatening conditions such as IMHA or IMTP, getting the treatment right as soon as possible can make a huge difference in patient survival. Unfortunately, the best way to use cyclosporine in these conditions is unknown. Patients are usually started at high cyclosporine doses, with therapy adjusted based on pharmacokinetic monitoring of cyclosporine blood concentrations. In our experience, however, dogs can have variable cyclosporine blood concentrations at the same oral drug dose and, even at comparable drug blood concentrations, some dogs will respond positively to medical management while others will not. Pharmacokinetic monitoring alone, therefore, does not necessarily lead to acceptable treatment outcomes.

In human medicine, pharmacodynamic monitoring is now used to supplement pharmacokinetic monitoring, with the goal of improving treatment outcomes in clinical patients receiving cyclosporine. [89,90] Pharmacodynamic monitoring is the use of an assay to measure the biological effects of a drug on a specific target cell or tissue. Cyclosporine is a mainstay drug in the field of human transplantation medicine [91,92] and, because effective immunosuppression is essential in transplant patients, a range of pharmacodynamic assays have been developed to evaluate the effects of cyclosporine on T cell function. Such assays have now also been adapted for use in dogs.

Our research group has over the past few years worked to develop pharmacodynamic assays for the monitoring of cyclosporine therapy in dogs. Initially, we performed an in vitro study using flow cytometry to measure the effects of incubation with cyclosporine on activated T cell expression of both surface markers (CD25 and CD95) and intracellular cytokines (IL2, IL-4, and IFN-γ) in order to identify candidate markers for subsequent pharmacodymamic studies [12]. Dogs were then given oral cyclosporine at various doses, and flow cytometry was used to evaluate T cell expression of IL-2, IL-4, and IFN-γ. Lower cyclosporine doses were found to suppress IFN-γ expression, and higher drug doses suppressed both IL-2 and IFN-γ [13]. While our study demonstrated that flow cytometry could be utilized to explore new markers for use in pharmacodynamic assays of cyclosporine therapy in dogs, we also encountered some of the limitations common seen with flow cytometry as a routine diagnostic tool, in that the technique was too labor intense and time sensitive for routine clinical use. However, we were able to use the information gained from our flow cytometric assay to then develop and refine a more robust quantitative reverse transcription polymerase chain reaction assay to measure activated T cell expression of IL-2 and IFN-γ for routine use in clinical patients.

DISCUSSION AND CONCLUSION

Flow cytometry is a laser-based technology that uses light scatter and fluorescence properties to identify, count and assess function in selected subpopulations of cells. Some of the most common applications in small animal medicine include inpractice hematology analyzers, precise assessment of cells within the immune system, detection of antibodies on RBCs and platelets, identification of erythrocyte parasites, assessment of aspects of platelet function, characterization of bone marrow cell types, immunophenotyping of neoplastic cells, and pharmacodynamic monitoring. As technology continues to grow and become more user-friendly, the potential benefits and uses of flow cytometry in small animal veterinary medicine are practically endless.

REFERENCES

1. Herzenberg LA, Parks D, Sahaf B, Perez O, Roederer M, Herzenberg LA,. The history and future of the fluorescence activated cell sorter and flow cytometry: a view from Stanford. Clin Chem. 2002; 48: 1819- 1827.

2. Tarrant JM. The role of flow cytometry in companion animal diagnostic medicine. Vet J. 2005; 170: 278-288.

3. Wilkerson MJ. Principles and applications of flow cytometry and cell sorting in companion animal medicine. Vet Clin North Am Small Anim Pract. 2012; 42: 53-71.

4. Otani I, Ohta K, Ishikawa A, Yamada T, Ishinazaka T, Ohtaki T, Tsumagari S. Flow cytometric analysis of canine umbilical cord blood lymphocytes. J Vet Med Sci. 2008; 70: 285-287.

5. Silva KL, Melo LM, Perosso J, Oliveira BB, Santos PS, Eugênio Fde R, Lima VM. CD95 (FAS) and CD178 (FASL) induce the apoptosis of CD4+ and CD8+ cells isolated from the peripheral blood and spleen of dogs naturally infected with Leishmania spp. Vet Parasitol. 2013; 197: 470- 476.

6. Bonnefont-Rebeix C, Marchal T, Bernaud J, Pin JJ, Leroux C, Lebecque S, Chabanne L. Toll-like receptor 3 (TLR3): a new marker of canine monocytes-derived dendritic cells (cMo-DC). Vet Immunol Immunopathol. 2007; 118: 134-139.

7. Bund D, Buhmann R, Gökmen F, Kremser A, Dreyssig J, Kolb HJ, Schmetzer HM. Canine-DCs using different serum-free methods as an approach to provide an animal-model for immunotherapeutic strategies. Cell Immunol. 2010; 263: 88-98.

8. Huang YC, Hung SW, Jan TR, Liao KW, Cheng CH, Wang YS, Chu RM. CD5-low expression lymphocytes in canine peripheral blood show characteristics of natural killer cells. J Leukoc Biol. 2008; 84: 1501- 1510.

9. Goto-Koshino Y, Ohno K, Nakajima M, Mochizuki H, Kanemoto H, Tsujimoto H. A rapid and simple method to obtain canine peripheral blood-derived macrophages. J Vet Med Sci. 2011; 73: 773-778. 

10. Mielcarek M, Kucera KA, Nash R, Torok-Storb B, McKenna HJ. Identification and characterization of canine dendritic cells generated in vivo. Biol Blood Marrow Transplant. 2007; 13: 1286-1293.

11. Kawarai S, Sato K, Horiguchi A, Kurata K, Kiuchi A, Tsujimoto H, Sakaguchi M. Potential immunological adjuvant of `K’-type CpGoligodeoxynucleotides enhanced the cell proliferation and IL-6 mRNA transcription in canine B cells. J Vet Med Sci. 2011; 73: 177-184.

12. Fellman CL, Stokes JV, Archer TM, Pinchuk LM, Lunsford KV, Mackin AJ,. Cyclosporine A affects the in vitro expression of T cell activationrelated molecules and cytokines in dogs. Vet Immunol Immunopathol. 2011; 140: 175-180.

13. Archer TM, Fellman CL, Stokes JV, Pinchuk LM, Lunsford KV, Pruett SB, Langston VC. Pharmacodynamic monitoring of canine T-cell cytokine responses to oral cyclosporine. J Vet Intern Med. 2011; 25: 1391-1397.

14. Bismarck D, Schütze N, Moore P, Büttner M, Alber G, Buttlar Hv,. Canine CD4+CD8+ double positive T cells in peripheral blood have features of activated T cells. Vet Immunol Immunopathol. 2012; 149: 157-166.

15. Asano M, Toda M, Sakaguchi N, Sakaguchi S. Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med. 1996; 184: 387-396.

16. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic selftolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of selftolerance causes various autoimmune diseases. J Immunol. 1995; 155: 1151-1164.

17. Sakaguchi S, Toda M, Asano M, Itoh M, Morse SS, Sakaguchi N,. T cellmediated maintenance of natural self-tolerance: its breakdown as a possible cause of various autoimmune diseases. J Autoimmun. 1996; 9: 211-220.

18. Thornton AM, Shevach EM. Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J Immunol. 2000; 164: 183-190.

19. Pinheiro D, Singh Y, Grant CR, Appleton RC, Sacchini F, Walker KR, Chadbourne AH. Phenotypic and functional characterization of a CD4(+) CD25(high) FOXP3(high) regulatory T-cell population in the dog. Immunology. 2011; 132: 111-122.

20. Biller BJ, Elmslie RE, Burnett RC, Avery AC, Dow SW. Use of FoxP3 expression to identify regulatory T cells in healthy dogs and dogs with cancer. Vet Immunol Immunopathol. 2007; 116: 69-78.

21. Tominaga M, Horiuchi Y, Ichikawa M, Yamashita M, Okano K, Jikumaru Y, Nariai Y. Flow cytometric analysis of peripheral blood and tumorinfiltrating regulatory T cells in dogs with oral malignant melanoma. J Vet Diagn Invest. 2010; 22: 438-441.

22. Abrams VK, Hwang B, Lesnikova M, Gass MJ, Wayner E, CastillaLlorente C, Georges GE. A novel monoclonal antibody specific for canine CD25 (P4A10): selection and evaluation of canine Tregs. Vet Immunol Immunopathol. 2010; 135: 257-265.

23. Knueppel A, Lange S, Sekora A, Altmann S, Freund M, Junghanss C,. Phenotypic and functional characterization of freshly isolated and expanded canine regulatory T cells. Exp Anim. 2011; 60: 471-479.

24. Knueppel A, Lange S, Altmann S, Sekora A, Knuebel G, Vogel H, et al. Upfront denileukin diftitox as in vivo regulatory T-cell depletion in order to enhance vaccination effects in a canine allogeneic hematopoietic stem cell transplantation model. Vet Immunol Immunopathol. 2012; 145: 233-240.

25. Junginger J, Schwittlick U, Lemensieck F, Nolte I, Hewicker-Trautwein M. Immunohistochemical investigation of Foxp3 expression in the intestine in healthy and diseased dogs. Vet Res. 2012; 43: 23.

26. Gordon I, Paoloni M, Mazcko C, Khanna C. The Comparative Oncology Trials Consortium: using spontaneously occurring cancers in dogs to inform the cancer drug development pathway. PLoS Med. 2009; 6: e1000161.

27. Wilbe M, Sundberg K, Hansen IR, Strandberg E, Nachreiner RF, Hedhammar A, Kennedy LJ. Increased genetic risk or protection for canine autoimmune lymphocytic thyroiditis in Giant Schnauzers depends on DLA class II genotype. Tissue Antigens. 2010; 75: 712-719.

28. Bauer N1, Eralp O, Moritz A. Reference intervals and method optimization for variables reflecting hypocoagulatory and hypercoagulatory states in dogs using the STA Compact automated analyzer. J Vet Diagn Invest. 2009; 21: 803-814.

29. Junginger J, Lemensieck F, Moore PF, Schwittlick U, Nolte I, HewickerTrautwein M,. Canine gut dendritic cells in the steady state and in inflammatory bowel disease. Innate Immun. 2014; 20: 145-160.

30. Ricklin ME, Roosje P, Summerfield A. Characterization of canine dendritic cells in healthy, atopic, and non-allergic inflamed skin. J Clin Immunol. 2010; 30: 845-854. 31.Qeska V, Baumgärtner W, Beineke A. Species-specific properties and translational aspects of canine dendritic cells. Vet Immunol Immunopathol. 2013; 151: 181-192.

32. Wang YS, Chi KH, Liao KW, Liu CC, Cheng CL, Lin YC, Cheng CH. Characterization of canine monocyte-derived dendritic cells with phenotypic and functional differentiation. Can J Vet Res. 2007; 71: 165-174.

33. Wang YS, Chi KH, Chu RM. Cytokine profiles of canine monocytederived dendritic cells as a function of lipopolysaccharide- or tumor necrosis factor-alpha-induced maturation. Vet Immunol Immunopathol. 2007; 118: 186-198.

34. Wijewardana V, Sugiura K, Oichi T, Fujimoto M, Akazawa T, Hatoya S, et al. Generation of canine dendritic cells from peripheral blood monocytes without using purified cytokines. Vet Immunol Immunopathol. 2006; 114: 37-48.

35. Wijewardana V, Sugiura K, Yahata M, Akazawa T, Wijesekera DP, Imamoto S, Hatoya S. Production of canine soluble CD40 ligand to induce maturation of monocyte derived dendritic cells for cancer immunotherapy. Vet Immunol Immunopathol. 2013; 156: 121-127.

36. Bonnefont-Rebeix C, de Carvalho CM, Bernaud J, Chabanne L, Marchal T, Rigal D,. CD86 molecule is a specific marker for canine monocytederived dendritic cells. Vet Immunol Immunopathol. 2006; 109: 167- 176.

37. Stein VM, Baumgärtner W, Kreienbrock L, Zurbriggen A, Vandevelde M, Tipold A,. Canine microglial cells: stereotypy in immunophenotype and specificity in function? Vet Immunol Immunopathol. 2006; 113: 277-287.

38. Ensinger EM, Boekhoff TM, Carlson R, Beineke A, Rohn K, Tipold A, Stein VM. Regional topographical differences of canine microglial immunophenotype and function in the healthy spinal cord. J Neuroimmunol. 2010; 227: 144-152.

39. Papadogiannakis EI, Kontos VI, Tamamidou M, Roumeliotou A. Determination of intracellular cytokines IFN-gamma and IL-4 in canine T lymphocytes by flow cytometry following whole-blood culture. Can J Vet Res. 2009; 73: 137-143.

40. Villaescusa A, García-Sancho M, Rodríguez-Franco F, Sainz A. Early-life longitudinal survey of peripheral blood lymphocyte subsets in Beagle dogs. Vet Immunol Immunopathol. 2012; 149: 126-131.

41. Gondois-Rey F, Granjeaud S, Kieu Sle T, Herrera D, Hirsch I, Olive D,. Multiparametric cytometry for exploration of complex cellular dynamics. Cytometry A. 2012; 81: 332-342.

42. Platt R, Ng T2, Glover S3, Roof M4, Kimura K, Roth JA. Canine peripheral blood lymphocyte phenotyping by 7-color multiparameter flow cytometry. Anal Quant Cytol Histol. 2013; 35: 197-204.

43. Geissmann F, Auffray C, Palframan R, Wirrig C, Ciocca A, Campisi L, et al. Blood monocytes: distinct subsets, how they relate to dendritic cells, and their possible roles in the regulation of T-cell responses. Immunol Cell Biol. 2008; 86: 398-408.

44. Kim YJ, Kang JH, Yang MP. Zinc increases the phagocytic capacity of canine peripheral blood phagocytes in vitro. Vet Res Commun. 2009; 33: 251-261.

45. Silva AC, de Almeida BF, Soeiro CS, Ferreira WL, de Lima VM, Ciarlini PC,. Oxidative stress, superoxide production, and apoptosis of neutrophils in dogs with chronic kidney disease. Can J Vet Res. 2013; 77: 136-141.

46. Singh SK, Dimri U, Sharma MC, Swarup D, Sharma B, Pandey HO, Kumari P. The role of apoptosis in immunosuppression of dogs with demodicosis. Vet Immunol Immunopathol. 2011; 144: 487-492.

47. Im Hof M, Williamson L, Summerfield A, Balmer V, Dutoit V, Kandimalla ER, et al. Effect of synthetic agonists of toll-like receptor 9 on canine lymphocyte proliferation and cytokine production in vitro. Vet Immunol Immunopathol. 2008; 124: 120-131.

48. Masuda K, Yasuda N. The antibody against human CD25, ACT-, recognizes canine T-lymphocytes in the G2/M and G0/G1 phases of the cell cycle during proliferation. J Vet Med Sci. 2008; 70: 1285-1287.

49. Mizuno T, Suzuki R, Umeki S, Okuda M. Crossreactivity of antibodies to canine CD25 and Foxp3 and identification of canine CD4+CD25 +Foxp3+ cells in canine peripheral blood. J Vet Med Sci. 2009; 71: 1561-1568.

50. Fujimura M, Masuda K, Hayashiya M, Okayama T. Flow cytometric analysis of lymphocyte proliferative responses to food allergens in dogs with food allergy. J Vet Med Sci. 2011; 73: 1309-1317.

51. Abbott DL, McGrath JP. Evaluation of flow cytometric counting procedure for canine reticulocytes by use of thiazole orange. Am J Vet Res. 1991; 52: 723-727.

52. Wilkerson MJ, Davis E, Shuman W, Harkin K, Cox J, Rush B. Isotypespecific antibodies in horses and dogs with immune-mediated hemolytic anemia. J Vet Intern Med. 2000; 14: 190-196.

53. Quigley KA, Chelack BJ, Haines DM, Jackson ML. Application of a direct flow cytometric erythrocyte immunofluorescence assay in dogs with immune-mediated hemolytic anemia and comparison to the direct antiglobulin test. J Vet Diagn Invest. 2001; 13: 297-300.

54. Harkin KR, Hicks JA, Wilkerson MJ. Erythrocyte-bound immunoglobulin isotypes in dogs with immune-mediated hemolytic anemia: 54 cases (2001-2010). J Am Vet Med Assoc. 2012; 241: 227-232.

55. Fukata T, Ohnishi T, Okuda S, Sasai K, Baba E, Arakawa A,. Detection of canine erythrocytes infected with Babesia gibsoni by flow cytometry. J Parasitol. 1996; 82: 641-642.

56. Bicalho KA, Ribeiro MF, Martins-Filho OA. Molecular fluorescent approach to assessing intraerythrocytic hemoprotozoan Babesia canis infection in dogs. Vet Parasitol. 2004; 125: 221-235.

57. Uemura T, Suzuki S, Ohnishi T. Flow cytometric enumeration of reticulocyte in the peripheral blood from canine infected with Babesia gibsoni. Zentralbl Veterinarmed B. 1990; 37: 468-472.

58. Sánchez-Pérez A, Brown G, Malik R, Assinder SJ, Cantlon K, Gotsis C, et al. Rapid detection of haemotropic mycoplasma infection of feline erythrocytes using a novel flow cytometric approach. Parasit Vectors. 2013; 6: 158.

59. Wang C, Smith BR, Ault KA, Rinder HM. Reticulated platelets predict platelet count recovery following chemotherapy. Transfusion. 2002; 42: 368-374.

60. Robinson MS, Mackie IJ, Khair K, Liesner R, Goodall AH, Savidge GF, et al. Flow cytometric analysis of reticulated platelets: evidence for a large proportion of non-specific labelling of dense granules by fluorescent dyes. Br J Haematol. 1998; 100: 351-357.

61. Tsuchiya R, Komatsu T, Ishikawa T, Neo S, McConnell MF, Hisasue M, et al. Preparation of positive control platelets for detection of canine platelet surface-associated IgG, IgM and complement (C3) by flow cytometry. J Vet Med Sci. 2010; 72: 1063-1066.

62. Kristensen AT, Weiss DJ, Klausner JS, Laber J, Christie DJ. Comparison of microscopic and flow cytometric detection of platelet antibody in dogs suspected of having immune-mediated thrombocytopenia. Am J Vet Res. 1994; 55: 1111-1114.

63. Dircks BH, Schuberth HJ, Mischke R. Underlying diseases and clinicopathologic variables of thrombocytopenic dogs with and without platelet-bound antibodies detected by use of a flow cytometric assay: 83 cases (2004-2006). J Am Vet Med Assoc. 2009; 235: 960-966.

64. Weiss DJ, Brazzell JL. Detection of activated platelets in dogs with primary immune-mediated hemolytic anemia. J Vet Intern Med. 2006; 20: 682-686.

65. Weiss DJ. Application of flow cytometric techniques to veterinary clinical hematology. Vet Clin Pathol. 2002; 31: 72-82.

66. Moritz A, Walcheck BK, Weiss DJ. Evaluation of flow cytometric and automated methods for detection of activated platelets in dogs with inflammatory disease. Am J Vet Res. 2005; 66: 325-329.

67. Wills TB, Wardrop KJ, Meyers KM. Detection of activated platelets in canine blood by use of flow cytometry. Am J Vet Res. 2006; 67: 56-63.

68. Rivera J, Lozano ML, Navarro-Núñez L, Vicente V. Platelet receptors and signaling in the dynamics of thrombus formation. Haematologica. 2009; 94: 700-711.

69. Shattil SJ, Hoxie JA, Cunningham M, Brass LF. Changes in the platelet membrane glycoprotein IIb.IIIa complex during platelet activation. J Biol Chem. 1985; 260: 11107-11114.

70. Tomasiak M, Rusak T, Gacko M, Stelmach H. Cyclosporine enhances platelet procoagulant activity. Nephrol Dial Transplant. 2007; 22: 1750-1756.

71. Brooks MB, Catalfamo JL, Brown HA, Ivanova P, Lovaglio J. A hereditary bleeding disorder of dogs caused by a lack of platelet procoagulant activity. Blood. 2002; 99: 2434-2441.

72. Boudreaux MK, Catalfamo JL. Molecular and genetic basis for thrombasthenic thrombopathia in otterhounds. Am J Vet Res. 2001; 62: 1797-1804.

73. Boudreaux MK, Kvam K, Dillon AR, Bourne C, Scott M, Schwartz KA, Toivio-Kinnucan M. Type I Glanzmann’s thrombasthenia in a Great Pyrenees dog. Vet Pathol. 1996; 33: 503-511.

74. Weber AA, Przytulski B, Schumacher M, Zimmermann N, Gams E, Hohlfeld T, et al. Flow cytometry analysis of platelet cyclooxygenase-2 expression: induction of platelet cyclooxygenase-2 in patients undergoing coronary artery bypass grafting. Br J Haematol. 2002; 117: 424-426.

75. Thomason J, Lunsford K, Mullins K, Stokes J, Pinchuk L, Wills R, McLaughlin R. Platelet cyclooxygenase expression in normal dogs. J Vet Intern Med. 2011; 25: 1106-1112.

76. Reggeti F, Bienzle D. Flow cytometry in veterinary oncology. Vet Pathol. 2011; 48: 223-235.

77. Papakonstantinou S, Berzina I, Lawlor A, J O’Neill E, J O’Brien P. Rapid, effective and user-friendly immunophenotyping of canine lymphoma using a personal flow cytometer. Ir Vet J. 2013; 66: 6.

78. Marconato L, Martini V, Aresu L, Sampaolo M, Valentini F, Rinaldi V, Comazzi S. Assessment of bone marrow infiltration diagnosed by flow cytometry in canine large B cell lymphoma: prognostic significance and proposal of a cut-off value. Vet J. 2013; 197: 776-781.

79. Culmsee K, Nolte I. Flow cytometry and its application in small animal oncology. Methods Cell Sci. 2002; 24: 49-54.

80. Rütgen BC, Hammer SE, Gerner W, Christian M, de Arespacochaga AG, Willmann M, Kleiter M. Establishment and characterization of a novel canine B-cell line derived from a spontaneously occurring diffuse large cell lymphoma. Leuk Res. 2010; 34: 932-938.

81. Joetzke AE, Eberle N, Nolte I, Mischke R, Simon D. Flow cytometric evaluation of peripheral blood and bone marrow and fine-needle aspirate samples from multiple sites in dogs with multicentric lymphoma. Am J Vet Res. 2012; 73: 884-893.

82. Villiers E, Baines S, Law AM, Mallows V. Identification of acute myeloid leukemia in dogs using flow cytometry with myeloperoxidase, MAC387, and a canine neutrophil-specific antibody. Vet Clin Pathol. 2006; 35: 55-71.

83. Weiss DJ. Flow cytometric evaluation of canine bone marrow based on intracytoplasmic complexity and CD45 expression. Vet Clin Pathol. 2004; 33: 96-101.

84. .Takemitsu H, Zhao D, Yamamoto I, Harada Y, Michishita M, Arai T,. Comparison of bone marrow and adipose tissue-derived canine mesenchymal stem cells. BMC Vet Res. 2012; 8: 150.

85. Weiss DJ. Use of monoclonal antibodies to refine flow cytometric differential cell counting of canine bone marrow cells. Am J Vet Res. 2001; 62: 1273-1278.

86. Weiss DJ. Evaluation of proliferative disorders in canine bone marrow by use of flow cytometric scatter plots and monoclonal antibodies. Vet Pathol. 2001; 38: 512-518.

87. Niemeyer GP, Hudson J, Bridgman R, Spano J, Nash RA, Lothrop CD,. Isolation and characterization of canine hematopoietic progenitor cells. Exp Hematol. 2001; 29: 686-693.

88. Archer TM, Boothe DM, Langston VC, Fellman CL, Lunsford KV, Mackin AJ,. Oral cyclosporine treatment in dogs: a review of the literature. J Vet Intern Med. 2014; 28: 1-20.

89. Barten MJ, Rahmel A, Bocsi J, Boldt A, Garbade J, Dhein S, Mohr FW. Cytokine analysis to predict immunosuppression. Cytometry A. 2006; 69: 155-157.

90. Barten MJ, Tarnok A, Garbade J, Bittner HB, Dhein S, Mohr FW, Gummert JF. Pharmacodynamics of T-cell function for monitoring immunosuppression. Cell Prolif. 2007; 40: 50-63.

91. Oellerich M, Barten MJ, Armstrong VW. Biomarkers: the link between therapeutic drug monitoring and pharmacodynamics. Ther Drug Monit. 2006; 28: 35-38.

92. Böhler T, Nolting J, Kamar N, Gurragchaa P, Reisener K, Glander P, Neumayer HH. Validation of immunological biomarkers for the pharmacodynamic monitoring of immunosuppressive drugs in humans. Ther Drug Monit. 2007; 29: 77-86

Received : 07 Apr 2014
Accepted : 07 Jun 2014
Published : 09 Jun 2014
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X