Next Article in Journal
Comparative Metabolome and Transcriptome Analyses Reveal the Regulatory Mechanism of Purple Leafstalk Production in Taro (Colocasia esculenta L. Schott)
Previous Article in Journal
Lessons Learned from Translating Genome Sequencing to Clinical Routine: Understanding the Accuracy of a Diagnostic Pipeline
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Changes in Mastocytes and Their Significance in Mast Cell Tumor Prognosis and Treatment

by
Szymon Zmorzynski
1,*,
Aleksandra Kimicka-Szajwaj
2,
Angelika Szajwaj
3,
Joanna Czerwik-Marcinkowska
4 and
Jacek Wojcierowski
5
1
Laboratory of Genetics, Academy of Zamosc, 22-400 Zamosc, Poland
2
DVM Veterinary Hospital AMVET, 31-328 Krakow, Poland
3
St. Luke’s Hospital in Tarnow, 33-100 Tarnow, Poland
4
Institute of Biology, Jan Kochanowski University, 25-369 Kielce, Poland
5
Laboratory for Genetic Testing, 20-143 Lublin, Poland
*
Author to whom correspondence should be addressed.
Genes 2024, 15(1), 137; https://doi.org/10.3390/genes15010137
Submission received: 14 December 2023 / Revised: 12 January 2024 / Accepted: 20 January 2024 / Published: 22 January 2024
(This article belongs to the Section Molecular Genetics and Genomics)

Abstract

:
Mast cell tumors are a large group of diseases occurring in dogs, cats, mice, as well as in humans. Systemic mastocytosis (SM) is a disease involving the accumulation of mast cells in organs. KIT gene mutations are very often seen in abnormal mast cells. In SM, high KIT/CD117 expression is observed; however, there are usually no KIT gene mutations present. Mastocytoma (MCT)—a form of cutaneous neoplasm—is common in animals but quite rare in humans. KIT/CD117 receptor mutations were studied as the typical changes for human mastocytosis. In 80% of human cases, the KIT gene substitution p.D816H was present. In about 25% of MCTs, metastasis was observed. Changes in the gene expression of certain genes, such as overexpression of the DNAJ3A3 gene, promote metastasis. In contrast, the SNORD93 gene blocks the expression of metastasis genes. The panel of miR-21-5p, miR-379, and miR-885 has a good efficiency in discriminating healthy and MCT-affected dogs, as well as MCT-affected dogs with and without nodal metastasis. Further studies on the pathobiology of mast cells can lead to clinical improvements, such as better MCT diagnosis and treatment. Our paper reviews studies on the topic of mast cells, which have been carried out over the past few years.

1. Introduction

Mast cell tumors are a large group of diseases occurring in dogs, cats, mice, as well as in humans. They can be relatively benign or malignant tumors. The prognosis in benign cases can be somewhat approximated, while in malignant cases, the prognosis is rather poor. The form of cutaneous mast cell tumor—mastocytoma—is particularly common in dogs. It accounts for 16–21% of all canine skin tumors and can cause considerable frustration and sadness for afflicted dogs [1,2].

2. Characteristics of Mastocytes

Mast cells (MCs) were first identified by Paul Ehrlich in 1878 [3]. MCs are spherical cells in the cytoplasm, which contain a significant number of secretory granules. Cells similar to mast cells are basophils, which are slightly smaller in size than MCs (8 μm); their cytoplasm has fewer granules; their granules do not contain tryptase; their cell membrane has no KIT receptor but contains the FcεR1 receptor. Basophil precursors mature in the bone marrow under the influence of Il-3 [4]. The granules stain metachromatically with toluidine blue, safranin, or berberine sulfate. The function of MCs, according to Ehrlich, was initially linked to the tissue nutrition system (hence the name “mast cells”). After detecting the release of heparin and histamine from MCs in anaphylactic shock [5], mast cells have been found to play a role in type I hypersensitivity reactions (allergy) [6]. MCs play a role in regulating allergic and inflammatory processes, secrete mediators for both innate and acquired immunity [7], play a role in the maintenance of immune homeostasis [8], and play a role in wound healing processes [9,10]. MCs, together with dendritic cells (DCs) and monocytes, are the first cells of the immune system, which interact with foreign antigens. After appropriate stimulation, MCs can re-enter the cell cycle and proliferate. The increased recruitment and local maturation of mast cell progenitors can also contribute to increasing MC populations in tissues [11]. During individual biological responses, MCs can function as effector cells, immunoregulatory cells, or both [12]. MC effector functions are associated with (I) removal of pathogens via phagocytosis and/or secretion of antimicrobial proteins; (II) degradation of toxic endogenous peptides and venom components; (III) increased vascular permeability (e.g., via histamine secretion); (IV) stimulation of bronchial smooth muscle cell contraction (e.g., via leukotriene C4); (V) stimulation of collagen synthesis by fibroblasts (e.g., via tryptase) [12].
Mast cell granules contain factors and substances such as polyamines, amines (serotonin, dopamine, and histamine), enzymes (cathepsins, β-hexosaminidase, arylsulfatase, heparanase, caspase, angiogenin, and kallikreins), proteases (metalloproteinases, chymase, granzyme B, carboxypeptidase A, tryptase), proteoglycans (heparin, serglycine, and chondroitin sulfate), cytokines (TNF-α, TNF-γ, β-FGF, Il-4, SCF, and most interleukins), chemokines (CCL2, CCL5, CCL7, CCL11, and MCP4), peptides (endorphin, endothelin, cathelicidin, and VIP), metabolites (prostaglandins D2, E2, leukotrienes, and PAF), and growth factors (SCF, GM-CSF, β-FGF, NGF, PDGF, TGF-β, and VEGF). A detailed description of these factors and the literature on their discovery can also be found in excellent review papers authored by de Silva et al., 2014 and Moon et al., 2014 [13,14]. It should also be added that the content of granules depends on the tissue from which MC was isolated. Mast cells contain a variety of mediators (as mentioned above), such as heparin, histamine, tryptase, chymase, VEGF, and TNF-α, which, when released during the initial stages of wound healing, affect bleeding, followed by coagulation and acute inflammation. Various additional vasoactive and chemotactic rapidly produced mediators (C3a, C5a, PAF) contribute to these processes, while mast-cell-derived pro-inflammatory and growth-promoting mediators (VEGF, PDGF, TGF-β, NGF, IL-4, IL-8) contribute to neoangiogenesis, fibrinogenesis, or re-epithelialization during the repair process [15].
The secretion of substances accumulated inside the mast cell granules occurs via exocytosis. Several pathways of transmembrane transfers were described [14,16], e.g.,
  • anaphylactic exocytosis (degranulation without de novo synthesis), observed after treatment with C3a and C5a complement peptides (on complement receptors), snake venom, UV, acrolein, titanium nanoparticles;
  • selective exocytosis (degranulation) of cellular granules can occur after the action of specific stimulators, including antigen or IgE on FcεR1 (histamine is released), neuropeptides on NKR (cytokines, chemokines, and 5HT are released), 48/80 on Mrgprx2 (cytokines and chemokines are released), cathelicidin on G-protein-coupled receptor (GPCR, releasing histamine), defensins on GPCR (releasing histamine), pleurocidin on GPCR (releasing cytokines and chemokines), A23187 (releasing cytokines and chemokines), morphine and codeine on opioid receptor (releasing cytokines, chemokines, and hexosaminidase), and NGF on Trk receptor (releasing histamine and PGE2);
  • constitutive exocytosis is associated with granule release without degranulation after treatment with zymosan on TLR2 (releasing GM-CSF, Il-1β), poly (I-C) on TLR (releasing cytokines), LPS on TLR4 and CD14 (releasing cytokines and chemokines), SCF on MAPK (releasing cytokines), and lectin on TIM3 (releasing cytokines);
  • without degranulation and without de novo synthesis (exocytosis of exosomes). Exosomes are vesicles measuring 30 nm–100 nm in diameter formed in secretory granules. These vesicles penetrate the cell membrane, pass outside the cell, and after transferring in body fluids, they are engulfed by other cells. Exosomes are important for intercellular communication. They contain many proteins, up to 1000 different mRNAs, and more than 100 microRNA molecules [17,18,19].
The biogenesis and release of cytoplasmic granule contents require the presence of many proteins, including Munc 13-4 [20], complexin (synaphin) [21], RAB GTPase [22], lysosomal trafficking regulator protein (LYST), synaptotagmin [23], granins, RAC1/2 [24], DOCK3 [25], proteoglycans, and sensitive factor attachment protein receptor (SNARE) [26].
The origin of MCs—the effector cells of innate immunity—was initially attributed exclusively to myeloid hematopoiesis. It is now accepted that a certain amount of MC is derived from erythroid–myeloid progenitors (EMP cells) from the yolk sac [27]. The next stages of maturation in the bone marrow are (1) hematopoietic adult stem cells (HemASC); (2) multipotent progenitor containing β7-integrin, Il-33R, and active glycolysis (MPP); (3) common myeloid progenitor containing Il-7R, CD27, and active glycolysis (CMP); (4) granulocyte-macrophage progenitor containing CD34, FcγRII, and active glycolysis (GMP); (5) basophil and MC progenitor containing β7-integrin, CD16/32, and active glycolysis (B/MCP); (6) immature MC progenitor (possessing CD34, CD45, FcγRII, FcεRI, and β7-integrin, with active glycolysis); (7) MC progenitors (possessing CD34, CD45, FcγRII, FcεRI, β7-integrin, granules and glycolysis) (Figure 1). MC progenitors are present in peripheral blood and can mature in target tissues under the influence of cytokines, chemokines, and growth factors [28]. The stages of maturation are presented according to Mendoza et al., 2021 [29]. Images of mature mast cells using electron microscopy are shown in the paper by MacDonald et al. [30]. Five types of morphologically distinct granules were identified in the cytoplasm of MCs: (type I) electron-dense core surrounded by sparse particulates; (type II) less electron-dense and more electron-lucent core; (type III) uniform lumen/particulates; (type IV) a mixture of electron-dense vesicles; and (type V) particulates and scroll-like or multi-lamellar vesicles [30].
Two types of MCs have been described in rodents: mucosal mast cells (MMCs) and connective tissue mast cells (CTMCs). MMCs are found in the mucosal epithelia of the lungs and gastrointestinal tract. These cells contain the mMCP1 and mMCP2 proteases (chymases) bound to chondroitin sulfate. CTMCs are present in the submucosa of the intestines, peritoneum, and skin. These cells contain mMCP4 chymase, mMCP5 and mMCP6 tryptases, as well as mCPA carboxypeptidase bound to serglycine proteoglycans [31,32]. Three types of MCs have been described in humans: MCCT containing tryptase, chymase, and CPA in the granules; MCT containing tryptase alone [33,34]; and MCCPA3 containing CPA3 carboxypeptidase but not chymase. MCTs are present in the mucous membranes of the intestines and lungs. MCCCTs are present in the skin, lymph nodes, and submucosa of the intestines and lungs. MCCPA3s are observed in the epithelia of the lungs (in asthma) and esophagus (in eosinophilic inflammation) [35,36]. Mature tissue MCs are long-lived cells [37]. Few studies have been conducted on the differentiation and phenotypes of mature MCs in dogs.
Mast-cell-specific antigens are present on the surface of MCs—for example, CD16, CD 32, and other proteins (Table 1) [13,38,39,40,41,42,43,44].

Mast Cell Ligands and Receptors

MCs respond to multiple ligands, which stimulate or inhibit the secretion of factors stored in their granules or synthesized de novo. Two types of degranulation are distinguished: total (anaphylactic) degranulation and fragmentary degranulation [14]. In addition, growth factors, prostaglandins, eicosanoids, chemokines, and cytokines are synthesized in MCs (especially after IgE stimulation) [45]. MCs can be alternatively stimulated by pathogen-associated molecular patterns (PAMPs) acting on Toll-like receptors, growth factors, complement peptides, cytokines, and other factors. They then selectively produce factors such as TNF-α [46], histamine [47], proteases [48], VEGF, PDGF-β, Il-6 [49], and Il-1 [50]. These factors act on blood vessels, on the intercellular matrix, on T cells, NK cells, macrophages, myeloid-derived suppressor cells (MDSCs), and dendritic cells [16].
MC ligands act by binding to cell membrane receptors. Stimulation of FcεRI by IgE is the main cause of allergic reaction [51]. IgE bound to the receptor recognizes various antigens. FcεRI stimulation involves phosphorylation by Fyn kinase of the receptor’s immunoreceptor tyrosine-based activation motif (ITAM) domain. This results in the binding and autophosphorylation of Syk kinase [52]. This leads to numerous protein phosphorylations, lipid metabolism, calcium ion mobilization, and activation of transcription factors [53].
Several types of mast cell receptors have been described—for example, SCF-binding receptor, PD1/CD279 receptor, Siglec-8, mas-related G-protein-coupled receptor member X2, Toll-like receptors, thymic stromal lymphopoietin receptor, and ATP receptors.
KIT is a proto-oncogene [54] and encodes a a receptor tyrosine kinase. Stem cell factor (SCF) activates the receptor and consequently many intracellular proteins are phosphorylated. KIT enables the proliferation of HemASC but also the survival and proliferation of differentiated mast cells [55], dendritic cells, and NK cells [56]. Eosinophils [57], and especially brain cells, also show high expression of KIT [58]. KIT expression depends on the presence of microphthalmia-associated transcription factor (MITF) and Gata2 transcription factor [59]. Upon SCF stimulation, KIT dimerizes and undergoes autophosphorylation in the juxtamembrane (JM) domain [60], kinase domain, and C-terminal domain. Further signaling proceeds through mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K), phospholipase C gamma (PLC-γ), and JAK kinase [61]. KIT’s ligand is a stem cell factor (SCF), which has two isoforms: SCF220 and SCF248 [62]. SCF is synthesized by bone marrow stromal cells, eosinophils, fibroblasts, and smooth muscle cells [55].
The PD1/CD279 (programmed cell death protein) receptor is activated by ligands PDL1/CD274 and PDL2/CD273 [63].
The inhibitory receptor Siglec-8 is a sialoadhesin related to CD33. It was detected in 2000 and is expressed in MCs (but not MC precursors), eosinophils, and basophils. Its ligands are sialylated keratan sulfate, sialic acid, and glycans [64].
The mas-related G-protein-coupled receptor member X2 (MRGPRX, MrgprB2 in mice) is expressed in MCs (MCCT in mice) present in the skin and subcutaneous tissue [65], as well as in eosinophils and basophils [66]. In MCCT cells, degranulation is not dependent on FcεRI but on stimulation of MRGPRX by HDP (peptides for protection against bacterial infection), the cationic polymer C48/80, by opiates, or vancomycin [67]. Bacteria attacking the skin release quorum sensing molecules (QSMs) and competence sensing peptides (CSPs), which, through MRGPRX and MCCT degranulation, cause bacterial growth inhibition [68].
MC-type cells have been detected in invertebrates, which lived over 500 million years ago [69]. Their granules contained heparin and histamine and degranulated upon stimulation. Invertebrate MC cells may represent the oldest “part” of the innate immune system.
Toll-like receptors (TLRs) bind pathogen-associated molecular patterns (PAMPs) and house dust mites (HDM), which cause airway inflammation [70]. The main ligands of these receptors are acylated lipopeptides (binding with TLR1); peptidoglycans, mucins, hemagglutinins, and mannans (binding with TLR2); double-stranded DNA (dsDNA) (binding with TLR3); lipopolysaccharide (LPS), mouse mammary tumor virus (MMTV) (binding with TLR4); flagellin (associated with TLR5); viral single-stranded RNA (ssRNA) (binding with TLR7 and TLR8); DNA CpG islands of bacteria and viruses (binding with TLR8) [71]. Upon TLR stimulation, mast cells produce Il-37, which inhibits inflammatory processes, dimerizing under tryptase and heparin. Il-37 is a ligand of Il-18R [72]. Mouse MCs of baboon bone marrow cells (BBMC), peritoneal-cell-derived mast cells (PCDMC), fetal-skin-derived mast cells (FSDMC) stimulated with LPS and peptidoglycan (PGN) synthesize a great number of interleukins, cytokines, chemokines, TNF-α, GM-CSF, INF-α, and INF-γ (partly depending on the stimulated TLR) [73,74,75].
Thymic stromal lymphopoietin receptor (TSLPR) binds thymic stromal lymphopoietin (TSLP), which is an alarmin released from respiratory epithelial cells. TSLP plays a role in chronic skin inflammation [76]. The other alarmins include Il-25 and Il-33 [77]. Il-33 binds the ST2/lI-lR4 receptor [78] and increases the survival of “cutaneous” MCs by stimulating the anti-apoptotic protein BCL-XL [79].
ATP receptors P2X4, P2X7, and P2Y1 degranulate mouse MCs upon ATP binding, and adenosine receptors A1, A2a, A2b, and A3 (bound to G proteins) are located on the surface of lung MCs [80].
Mast cell pathology is characterized by the occurrence of degranulation. The best known is type I hypersensitivity reaction with IgE antibodies, which bind harmless antigens and Fc receptors on MCs, causing degranulation of MCs. The release of mediators can lead to a variety of effects, including edema, vasodilation, and bronchoconstriction [81].

3. Mast Cell Activation Syndrome

Mast cell activation syndrome (MCAS) depends on excessive secretion of mast cell mediators after, among others, IgE stimulation [82]. The congenital causes of MCAS are systemic mastocytosis (SM) and hereditary alpha-tryptasemia (HalfaT). Hereditary alpha-tryptasemia is caused by the presence of additional copies—in the form of duplication or amplification—of the tryptase-alpha gene (TPSAB1 gene) [83,84,85].
MCAS can manifest as symptoms of local disease, including redness, pruritus, urticaria, and conjunctivitis; mild systemic disease symptoms, including pruritus, erythema, mild hypotension, mild dyspnea, and nausea; severe systemic disease symptoms, including epidermal blisters, angioedema, fever, sweats, severe shortness of breath, vomiting, diarrhea, and collapse; and chronic systemic disease symptoms, including atopic tissue inflammation, with symptoms lasting for an extended period of time.
MCAS is not a malignancy; there is no accumulation of large numbers of mast cells or specific gene mutations, although congenital mutations of the IL13 gene have been observed in asthmatic patients [86].
Mast cells can also be independently stimulated by IgE via the receptor FcεR1; IgG through the receptor FcγR1 [87]; stem cell factor (SCF) via the receptor KIT/CD117 [88]; pathogen-associated molecular patterns (PAMPs) via TLR receptors [89]; lectins, dsRNA via TLR4 receptor, which releases TNF-α and INF-β without MC degranulation [90]; complement peptides via CR3, CR4, CR5 receptors [91]; and numerous other stimulators.

3.1. Systemic Mastocytosis

Systemic mastocytosis (SM) is a disease characterized by the accumulation of mast cells in organs and tissues [92]. In morphologically and immunotypically abnormal mast cells [93], KIT gene mutations are very often present [94].
Depending on the location, the disease is divided into
  • cutaneous mastocytosis (CM)—most common in children—manifesting as urticaria pigmentosa as a diffuse or limited form of mastocytosis. Urticaria pigmentosa has good prognosis and usually resolves itself spontaneously;
  • indolent SM (ISM), aggressive SM (ASM), bone marrow SM, mast cell leukemia (MCL) [95], as well as mast cell sarcoma [94];
  • localized mastocytoma [95].
The classification of benign human systemic mastocytosis (CM and ISM) was described by Hartmann et al., 2016 [96]. The classification of malignant ASM was proposed by Pardanani et al., 2010 [97]. Neoplastic MCs show specific expressions of tryptase [98], CD2, and CD25 [99] and less specific expressions of CD117, CD33, CD43, and CD68 antigens [98]. The expression of CD2 and CD25 is diagnostically important because it is not present in normal mast cells [100,101]. The CD30/Ki-1 antigen (encoded by the TNFRSF8 gene) is also present on the surface of malignant cells [102]. The soluble CD30 (sCD30) form in a concentration up to 130 ng/mL is present in the blood of patients with ASM and MCL [103].
Many chromosomal aberrations have also been described in systemic mastocytosis cells, including chromosome X monosomy (45,X), chromosome 7 monosomy (45,XX,-7 or 45,XY,-7), chromosome Y disomy (47,XYY), chromosome 8 trisomy (47,XX,+8 or 47,XY,+8), 46,del(12)(p13) [104]. Complex karyotypes are rarely observed [104].
The KIT receptor is constitutively expressed in mast cells. In human systemic mastocytosis (hSM), the KIT gene undergoes a frequent (in up to 80% of cases) p.D816V mutation in exon 17 [105]. However, the mutations are not present in the very early stages of the disease [106]. Other KIT-activating mutations, such as p.V560G, p.D815K, p.D816Y, p.D816H, and p.D820G, have been described, occurring in hSM in a total of 5% of all cases [107]. The blocking of KIT activity by PKC412 (midostaurin) inhibits cell proliferation in SM for only 3 years [108].
Three types of tropomyosin receptor kinases (TRKA, TRKB, and TRKC) and nerve growth factor receptor (NGFR) bind neurotrophins. The nerve growth factor (NGF) binds TRKA, a brain-derived neurotrophic factor (BNDF). Nuclear factor 4 (NF4) binds TRKB. Nuclear factor 3 (NF3) mainly binds TRKC [109]. Neurotrophins increase mast cell survival, function, and chemotaxis [110,111]. The activation of mouse TRK by tumor necrosis factor (TNF) and TRKB by BDNF quite often causes SM or even MCL [112]. TRK mutations (especially TRKB) activate tumor transformation and metastasis formation in mastocytosis [111].
Abnormal activation of the mammalian target of rapamycin (mTOR) complexes may play a role in mastocytosis [113]. Rapamycin (an mTOR kinase inhibitor) has been found to reduce the growth and viability of SM cells with the KIT p.F816V mutation [114].
Other gene mutations described in SM include IDH1 and IDH2 (encoding isocitrate decarboxylase) [115]; ERK1/ERK2 in mice [116]; SRSF2 and SF3B1 (intron excision factor) [117]; RUNX1 (intron excision factor) [118]; KRAS and NRAS [119]; STAT5, AKT [120]; TET2 [121]; SETD2 [122]; ASX1 [123]. In most cases, genetic mutations are present in somatic cells. In SM some genetic changes may occur in the germline. These include substitutions in the KIT gene (p.K509I [124], p.A533D [125], p.N822J [126]) and substitution in the IL4R gene (p.Q576R [125]).
In SM, the pathogenic variants of the CEBPA gene (rs4616402) encoding a transcription factor, TEX41 gene (rs4662380) encoding TEX41 lncRNA, and TBL1XR1 gene (rs13077541) encoding a transducin-like protein 1 and associated with the X receptor were described [127].

Targeted Therapies

Understanding the role of MCs in cancer development and progression is critical for developing new targeted therapies for human cancers [128]. The relationship between the presence of MCs in tumors, the progression of angiogenesis, and tumor development may increase the possible role of MCs in cancer biology. Therefore, blocking the release of mediators with KIT receptor tyrosine kinase inhibitors (TKIs) (for example, imatinib, mastinib) may affect MC function [128,129,130], while blocking the release of mediators with tryptase inhibitors (gabexate mesylate and nafamostat mesylate, both of which are inhibitors of trypsin-like serine proteases) [128,131] can be an important therapeutic treatment for reducing tumor growth [132].
Imatinib mesylate (STI571) is a multi-kinase inhibitor approved for clinical use in the treatment of chronic myeloid leukemia, acute lymphoblastic leukemia (Philadelphia-positive), gastrointestinal stromal tumors (CD117-positive), and myeloid/lymphoid neoplasms with PDGFR gene rearrangements [133]. The KIT wild-type receptor is taken into account as imatinib target. The data on in vitro and in vivo efficacy of imatinib in KIT-mutated SM have shown contrasting results [134,135,136,137,138]. Some rare types of KIT mutants, as well as their wild-type alleles (encoding extracellular and juxtamembrane domain), have been proven to be imatinib sensitive under in vitro conditions. In contrast, cells with the most common KIT gene mutation p.D816V (in the kinase domain) are not sensitive to the effects of imatinib [134,135,136,137,138]. The response to imatinib relies on the presence of imatinib-sensitive mutations involving KIT (e.g., juxtamembrane or transmembrane KIT mutations) or PDGFR (e.g., FIP1L1/PDGFRA rearrangement) rather than on the absence of p.D816V KIT gene mutation [134,135,136,137,138].
In addition to imatinib, both preclinical and clinical trials have analyzed the role of other TKIs. Specifically, masitinib has been shown to have in vitro activity against PDGFR, Lyn tyrosine kinase, Fyn tyrosine kinase, and wild-type KIT [139]. Clinical studies of masitinib in patients with mastocytosis have focused mainly on exploring its potential utility for treating MC mediator-associated symptoms [140,141]. Midostaurin (PCK412) is a multi-kinase inhibitor, which competitively binds to the ATP binding site in the catalytic domain of tyrosine kinases, resulting in their inhibition. In addition to its activity against FLT3, it inhibits both wild-type KIT and KIT with p.D816V mutation, as well as other protein kinases, such as kinase insert domain-containing receptor (KDR), fibroblast growth factor receptor (FGFR), vascular endothelial growth factor receptor 2 (VEGFR2), FIP1L1/PDGFR fusion protein, and members of the serine/threonine protein kinase C (PKC) family [142]. Another selective KIT inhibitor with high affinity for p.D816V mutant KIT is avapritinib (BLU-285) [143]. Ripretinib (DCC-2618) is a novel type II tyrosine switch control inhibitor for the treatment of KIT-mutated cancers, including gastrointestinal stromal tumors (GISTs). BLU-263 is an inhibitor of KIT p. D816V with minimal central nervous system penetration compared to avapritinib [144]. Another example of a highly selective TKI is bezuclastinib (CGT9486). It has potent activity against KIT p.D816V, and it does not affect the functions of other closely related kinases. Bezuclastinib has shown preliminary clinical activity and a tolerable safety profile in patients with advanced solid tumors, including GIST [145].

3.2. Canine Systemic Mastocytosis

Systemic mastocytosis in dogs is rarely described, and it is more often diagnosed as a less aggressive form of mastocytoma [146]. It usually presents as cutaneous papules distributed on the trunk, head, perineum, and extremities as cutaneous mastocytosis (CM) or pigmented urticaria with Darier’s sign [147]. The infiltrates of well-differentiated mast cells are present in the dermis. The cells show KIT/CD117 expression, but usually, there are no mutations in the KIT gene and other genes typical for human mast cells [148].
The cytoplasmic granules of mast cells contain histamine, cathepsin G, chymase, tryptase, carboxypeptidase, proteoglycans (heparin, chondroitin sulfate), TNF, interleukins (such as Il-1, Il-4, Il-5, Il-6, and Il-13), and chemokines (CCL3 and CCL4) [149]. Degranulation results in pruritus, redness, and swelling of the skin, less often in bleeding and also vomiting, diarrhea, and coughing [147].

4. Mastocytoma

Mastocytoma (MCT) is common in animals such as dogs, cats, and mice, and it accounts for 7–21% of skin cancers in these animals, but it is quite rare in humans. Mastocytoma is a neoplasm, which occurs most often in older dogs (8–10 years old) as a single skin nodule or subcutaneous tissue. The lesions are located mainly in the skin and subcutaneous tissue, and they are less common in the gastrointestinal tract, spleen, liver, bone marrow, and the nervous system. Tumors in areas other than the skin and subcutaneous tissue are usually metastatic lesions. However, the literature has reported a case of mast cell tumor diagnosis in the nasal cavity [150]. Mastocytomas present in the skin are usually present in the form of tumors, which may be devoid of hair. Swelling and redness of the skin may appear around the tumor. Importantly, mast cell tumors can also show rapid infiltrative growth [151].
Mastocytomas can be most broadly divided into minimally malignant and highly malignant. The clinical classification of MCTs was first presented by Bostock DE [152] and followed by Patnaik AK [153], as well as Kiupel AM [154].
Histopathological tumor examination is crucial for making a complete diagnosis, developing a treatment plan, and assessing prognosis. There are two commonly used grading scales for cutaneous mast cell tumors: one created by Patnaik and another by Kiupel. The grading according to Patnaik is a three-stage scale, where grade I defines well-differentiated tumors confined to the dermis; grade II denotes tumors of intermediate differentiation, extending into the subcutaneous tissue; grade III denotes tumors of low differentiation, infiltrating the subcutaneous tissue [155]. One definite disadvantage of this method is its subjectivity. Ten histopathologists evaluated the same slides from 60 mast cell tumors [156]. Agreement among pathologists was at the level of 62.1% [156]. Kiupel developed a two-stage classification, which defines skin tumors as benign (low-grade) or malignant (high-grade). High-grade lesions are characterized by karyomegaly in at least 10% of the examined cells or at least seven mitotic figures, three multi-nucleated cells, or three atypical nuclei in ten view fields [157]. Kiupel’s classification is more authoritative in assessing the malignancy and the potential for metastatic lesions [155]. According to the Kiupel scale, the average survival time for patients with benign tumors is more than 2 years, while for patients with malignant tumors, the average survival time is less than 16 weeks [154]. Many laboratories continue to report both classifications in their test results to aid the cancer therapy process. The classification of mast cell tumors according to both Patnaik and Kiupel is not applicable to histopathological evaluation of subcutaneous mastocytoma [158]. Another prognostic factor is the number of mitoses in ten fields of view. It is believed that with the number of mitoses below seven, the prognosis is good, while the number of mitoses above seven corresponds to an average survival time of 12 weeks [159].
MCT tumors contain a large number of well-differentiated, highly granular tumor cells, a small number of eosinophils and cancer-associated fibroblasts (CAFs)—especially abundant in Patnaik’s grade III—lymphocytes, and other cells. Neoplastic MC cells have enlarged cell nuclei with scattered chromatin [160]. Nuclei and sparse mitotic figures are visible in these neoplastic cells [161].
Interestingly, malignant MCTs (grade I in Patnaik’s scale) in young dogs can remain inactive for several years and even involute [161]. Patnaik’s grade I at diagnosis applied to 20% of MCTs, grade II—43.3%, and grade III—36.7%. According to the Kiupel scale, there were 43.33% benign and 56.67% malignant MCTs [162]. Surgical removal within safe limits often yields durable results. Grade II tumors have a survival time of up to 4 years, while grade III tumors have a survival time of 1–2 years [162].

4.1. Diagnosis and Prognosis

Disease diagnosis should begin with a physical examination of the patient and clinical history taking.
Fine-needle aspiration biopsy (FNAB) is an effective tool in the process of initial diagnosis of mast cell tumors. By analyzing the patient’s data, the tumor location, and the result of FNAB examination, the tumor histological type of the mast cell can be predicted. Cutaneous mast cell tumors of the tail, perineum, or the site of skin transition to mucous membranes show high metastatic potential and are histologically qualified as malignant tumors [163].
Cytoplasmic granules contain negatively charged proteoglycans [164], proteases, chymase and tryptase, and carboxypeptidase A3 (CPA3) [165]. Hyaluronate breakdown products affect MC cell activation and migration. The procedure of choice is to remove the nodule with a margin of healthy tissue. The prognosis largely depends on local lymph node status with the use of the Ki67 expression assay [166], bromo d-uridine incorporation assay [166], proliferating cell nuclear antigen (PCNA) expression assay [167], AgNoR assay [168], MCT cell ploidy assay, MCT vascularization density assay [169], p53 expression assay [170], KIT gene mutation and expression assay [171], examination of the cell nuclei morphology [172], examination of cellular infiltration depth [173], and examination of tumor localization [174].
Evaluation of abnormal expression of tyrosine kinase receptor protein (KIT) by immunohistochemistry is an important prognostic factor, since the tyrosine kinase receptor protein plays a key role in mast cell proliferation, survival, differentiation, and migration. Three patterns of KIT expression have been distinguished. The first pattern (the membranous pattern, peritumoral) involves non-tumorigenic cells, as well as well-differentiated mast cell tumors. It indicates a non-aggressive biological type of tumor [175]. The second pattern (focal cytoplasmic pattern) shows focal or striated cytoplasmic labeling. In contrast, diffuse cytoplasmic labeling is observed in the third pattern. Both the second and third patterns are associated with shorter survival times and a higher risk of local relapse [175]. Immunohistochemical detection of phosphorylated KIT in patients with MCTs may predict the prognosis and biological behavior [176].
At the diagnosis of mast cell tumors, an analysis of the nuclear protein Ki67, which is a marker of cell growth fraction, is also conducted. At the same time, AgNOR nuclear proteins, visualized by silver staining, are the markers of cell division rates. A high number of AgNOR nuclear proteins indicates an increased cell cycle rate. Therefore, Ki67 assessment, along with AgNOR, is an important prognostic factor in the evaluation of cutaneous mastocytoma. An AgNOR × Ki67 equation score greater than 54 correlates with an increased risk of metastasis or death [177]. Smith et al. described AgNOR × Ki67 values as determinants of low cell proliferation in grade II MCTs [178].
Staging assessment should always be performed to determine the stage of disease development, which has a direct impact on therapeutic decisions and prognosis [179]. To assess the cancer grade, it is helpful to examine lymph nodes for the presence of cancer cells, even if there are no palpable changes in these lymph nodes. However, selecting the appropriate lymph node can present many difficulties. Therefore, it is helpful to perform lymph node mapping. As a result of manipulations associated with surgical tumor removal, mast cell granulomas may degranulate and recruit non-cancerous mast cells into the surrounding lymph nodes. This is the reason why lymph node biopsy is recommended before surgical removal of the lesion [180]. In order to evaluate the lymph nodes, a fine-needle aspiration biopsy can be applied, or the entire node can be harvested for histopathological evaluation. This approach allows the introduction of an appropriate therapeutic protocol [20]. Patients with possible metastases should additionally undergo liver and spleen biopsy. Infiltration with cancerous mast cells is possible even if the liver and spleen show normal morphology on abdominal ultrasound. Significant difference in mean survival time has been found between dogs with metastatic lesions in the liver or spleen and those whose organs are free of tumor cells [181].

4.2. Mastocytoma Cell Proteins

Several proteins of mastocytoma cells, which play important roles in the processes of their proliferation, migration, and resistance to apoptosis, have been described. MCT cell surface contains KIT/CD117, IgE, CD11b, CD18, CD44, and CD45, and sometimes CD2, CD25, and CD34 [182]. Other mastocytoma cell proteins are described in Table 2.

4.3. Mutations in Mastocytoma Cells

KIT/CD117 receptor mutations were studied as the typical changes for human mastocytosis. In 80% of human cases, the KIT gene substitution p.D816H is present. Loss of the KIT protein function may be the cause of human piebaldism syndrome [214]. The human KIT receptor has four isoforms [215]. The activating mutations described in the tumors result in dimerization of KIT, its multiple phosphorylation, and the generation of proliferative signal transmitted via the MAPK–PI3K–Akt–PLCg–JAK–Src pathway [61]. This creates an opportunity to interrupt cell proliferation signals by using multiple inhibitors of these enzymes [216].
KIT gene mutations in canine mastocytoma occur in 20–30% of cases [217]. Internal tandem duplication (ITD) in exon 11 includes nucleotides 555–559 and 571–590. Other common changes include deletions of nucleotides 550–560 [218] and point mutations of exon 11 (p.K557insF, p.K557insP). In exon 8, ITDs of nucleotides 417–421 or point mutation p.Q430R were observed. In exon 9, two mutations in the form of substitutions were found: p.S470I and p.N508I [219]. An increase in the KIT gene copy number is often found in canine mast cell tumors [220].
Some KIT mutations occur in healthy dogs, which have never been affected by MCT. However, there are dog breeds, which are highly susceptible to MCT, such as boxers, golden retrievers, Labradors, and bull terriers. This may suggest the existence of germline mutations in these specific breeds (Table 3).

4.4. Differences in Benign and Malignant Forms of MCT (According to Kiupel)

To date, few comparisons have been made between MCT cells in the benign and malignant forms of the disease. Studies have mainly focused on mutation specificity and differences in gene expression in the two forms of the disease.
ITD in exon 11 of the KIT gene is observed more frequently in malignant cutaneous MCTs with short survival and poor prognosis [227,228], while ITD in exon 8 of the KIT gene predicts longer survival and a milder course of the disease [229]. ITDs are also observed in exons 9, 12, and 19 of the KIT gene [219].
Certain cytogenetic changes in MCT cells are observed in aggressive mast cells. These include deletions in chromosomes 5, 20, and 21 and insertions in chromosome 21 [220]. Copy number variants (CNV) in the form of PTEN and FAS gene deletions, as well as amplifications of MAPK3, WNT5B, FGF, FOXM1, and RAD51 genes, are also prognostically unfavorable [230].
It seems most important to compare the total gene expression in cells of malignant MCT forms with that of benign MCT forms. A study carried out on canine Agilent DNA arrays showed overexpression of 450 genes and lower expression of 140 genes in MCTs with a low differentiation degree [231]. The mRNA level of the GSN gene was particularly reduced in MCTs with a low differentiation degree. Overexpression of FOXM1, GSN, FEN1, and KPNA2 genes was prognostically unfavorable. The authors selected 13 genes, whose expression levels most differentiated between the malignant and benign forms of MCT (Table 4).
Subsequent comparisons of gene expressions in high-risk and low-risk MCT cells were performed utilizing the Illumina platform [242]. Differential expression was found in 71 genes, of which 68 genes had increased expression in the high-risk group, and only 3 genes had decreased expression in this group. The results supported the conclusions of studies carried out previously. In addition, an important role was found to be played by cancer-associated fibroblasts (CAFs), which produce tumor intercellular matrix proteins [243]. CAF cells in the matrix activate tumor growth and progression. Tumor-associated fibroblasts (TAFs) are similar to myofibroblasts [244], and anti-smooth muscle antibodies (ASMAs) are the markers of these cells [245].

4.5. Metastasis Formation in Mastocytoma

Metastasis is observed in about 25% of MCTs, and its formation is a multi-step process caused by changes in gene expression [246]. Genes whose expression is associated with metastasis include those encoding proteins involved in intercellular adhesion and cell–ECM binding. Downregulation of their expression facilitates metastasis [246]. The expression patterns of such genes (in humans) have been described by Daves et al. [247]. Genes with a significantly reduced expression in metastatic cancers are listed in Table 5.
Overexpression of certain genes promotes metastasis, including the DNAJ3A3 gene from the heat shock protein (HSP) family, which promotes tumor invasion, and the small nucleolar RNA, C/D Box 93 (SNORD93) gene, which blocks the expression of metastasis genes [259].
The release of a cancer cell from a primary tissue is part of the metastasis formation process. Tumor cells are transferred via lymphatic vessels to regional lymph nodes and via blood vessels to distant tissues. Tumors release cells into the bloodstream, but only cells reaching “premetastatic niches (PMNs)” can survive and proliferate. These PMNs are formed by cancer cells, inflammatory cells, cancer-associated adipocytes (CAA), tumor-associated macrophages (TAM), and cancer-associated fibroblasts (CAFs) derived in part from adipose-derived stem cells [260]. There are three types of macrophages: M1 (inhibiting and killing cancer cells), M2 (increasing metastasis formation) [261], and TAM [262].

4.6. The Role of microRNAs

Epigenetic changes refer to mechanisms connecting the genome with environmental signals to provide adaptations to various conditions, factors, and intrinsic pathologic processes [263]. There are various different types of epigenetic regulation, including DNA methylation, post-transcriptional modifications by microRNAs (miRNAs), and histone modifications [264].
MicroRNAs are small RNAs involved in the regulation of mRNA transcription [265]. MicroRNAs can circulate freely in the plasma, or they can be delivered by extracellular vesicles (EVs) and the small extracellular vesicles (sEVs) [266,267]. Extracellular vesicles are released by all cell types, including normal cells and tumor cells. Moreover, they are present in body fluids, including plasma, urine, milk, sweat, tears, saliva, and cerebrospinal fluid [268]. EVs play a role in cell-to-cell communication. They are able to carry different types of RNA, proteins, lipids, and even DNA fragments [269]. The compounds carried by EVs can affect the function of recipient cells [269]. Cancer cells communicate not only with each other but also with the surrounding cells, including immune cells, fibroblasts, and endothelial cells. This communication is possible through EVs, which play an important role in tumor progression [270]. Tumor EVs can affect the cell phenotype, and they can also affect cells in the microenvironment, which support tumor cell growth, their survival and local invasion, as well as metastasis [271].
MicroRNAs with higher expression in the exosomes (membrane-bound extracellular vesicles of MCT cells) in comparison to mast cells included hsa-miR-451, hsa-miR-503, miR-Plus_27560, miRPlus_2843, miRPlus_27564, hsa-miR-583, miRPlus_1795, miRPlus_17890, hsa-miR-663, and hsa-miR-30b [272]. The expression level of circulating sEV-miR-21-5p changes in the plasma of dogs in different pathological stages (MCT with or without nodal metastasis) [273]. The level of sEV-miR-21-5p was significantly higher in plasma collected from nodal metastatic MCT-affected dogs compared to healthy and early metastatic MCT-affected patients [273].
Deregulation in microRNAs expression is typical for various types of cancers. MicroRNAs can act as tumor suppressors or oncogenes. Their significance as prognostic or predictive factors in human and veterinary medicine has been reported by He et al., Agarwal et al., and Jain et al. [274,275,276]. The activation of murine mast cells and upregulation of the miR-221-222 family influence cell cycle checkpoints [277]. For example, miR-221 regulates mast cell degranulation, cytokine production, and cell adherence [278]. Fenger et al. found that miR-9 was significantly overexpressed in aggressive MCTs compared to benign MCTs [253].
The miRNAs profile from paraffin-fixed tissues of canine MCT has been characterized by Zamarian et al. [279]. The panel of three miRNAs, including miR-21-5p, miR-379, and miR-885, has a good efficiency in discriminating healthy and MCT-affected dogs, as well as MCT-affected dogs with and without nodal metastasis [279]. Moreover, in the saliva of dogs affected by MCT, miR-21-5p has been identified as a potential negative prognostic factor [280]. MiR-21-5p is one of the miRNAs described as upregulated in canine MCT [279]. It was one of the first miRNAs detected in humans as an oncomiR [281]. Its overexpression is associated with oncogenesis in different tumors [281].
Lee et al. found that the expression of miR-539 and miR-381 is repressed by a mutated KIT protein with p.D816V substitution. Normally, these miRNAs are involved in MITF gene expression suppression. As a result, melanocyte-inducing transcription factor is not present within the cell. Dysregulation of the miR-539 and miR-381 pathways may contribute to abnormal MC proliferation and to the development of aggressive MC diseases [282].

4.7. Treatment

Mast cell tumors are the most common skin cancer in dogs. The disease forces veterinarians to consider various treatment options, including surgical treatment, pharmacological treatment, and radiotherapy. The obtained results should be analyzed carefully, based on the patient’s condition. The most important issue is to assess the risk of local relapse and metastasis, and only on this basis select an adequate management protocol.

4.7.1. Surgical Treatment

One of the treatment options for mast cell tumors is surgical removal of the lesion [283]. Surgery is usually a fully therapeutic procedure, as long as an adequate surgical margin is maintained. A study population of 55 dogs, after complete removal of grade II mast cell tumor, showed recurrence or metastasis in 5% of cases [284]. However, de novo tumor development was observed in 11% of the studied dogs [284]. Achieving clean margins during surgical resection can often be the most difficult aspect. Moreover, the histopathological margins’ evaluation also depends on the technique used to deliver the tissue sample to the laboratory. It is important to ensure that markings of the section are made accurately. This should be achieved by using special ink or surgical threads. Attention should also be paid to maintaining accuracy when filling out the laboratory referral, as it can facilitate an accurate diagnosis for the doctor examining the lesion. Sometimes, mast cell tumors are accompanied by swelling and redness, and there may be mast cells in the tissue surrounding the tumor, which have not undergone neoplastic transformation. The doctor examining the section must distinguish between healthy mast cells and tumor-transformed mast cells. Radial sections are routinely used to make a histopathological diagnosis and to assess the margin of healthy tissue in mast cell tumors. Although this method has been shown to be effective in making a diagnosis, it can lead to difficulties when evaluating the lesion margins. In contrast, tangential sections are a much more sensitive method when assessing whether the margin of healthy tissue has been preserved. Radial sections, based on palpation of the slice, provide good-quality specimens, which indicate general information about the surgical margins. Tangential sections allow a more accurate assessment of the margins. By combining the two methods, the sensitivity in detecting “dirty” margins is 20% [285].
Assessing the risk of local relapse is important for the subsequent therapeutic process. Therefore, diagnostics can be further expanded with molecular testing [178]. The local relapse risk of low-grade tumors—according to Kiupel’s scale, with a low index of Ki67 or AgNOR × Ki67—is less than 10%. At the same time, about 40% of “high-grade” tumors with a mutation in exon 11 of the KIT gene are at risk of relapse, despite maintaining clean margins [178]. The higher number of AgNORs in silver-stained nuclei regions is associated with higher proliferation of tumor-transformed cells and the degree of tumor transformation. The number of AgNORs may be a predictor of tumor recurrence [286]. The molecular changes in cancer cells undergoing metastasis are still poorly understood. In the KIT mutational status (in exons 8, 9, and 11), 100% concordance was observed between primary and metastatic MCTs in twenty-one prospectively enrolled canine patients [287]. In contrast, ITD mutations were present in the primary tumors and were not found in relative metastasis [288].

4.7.2. Pharmacological Treatment

Pharmacological treatment is used primarily in patients in whom complete surgical resection is impossible or multiple metastases are found. In addition, therapeutic substances can be used in neoadjuvant therapy. The use of glucocorticoids in skin mastocytoma therapy makes it possible to nullify symptoms associated with degranulation of granulomas, such as swelling or redness of the skin. Glucocorticosteroids administered orally have a cytotoxic effect against tumor-transformed mast cells, and their use has few side effects [289]. Glucocorticosteroids inhibit the rate of tumor growth and slow tumor cell divisions. However, in the absence of appropriate receptors, resistance to glucocorticosteroids can develop [290].
Chemotherapy, despite its side effects, effectively controls the disease and extends the average survival time in dogs with mastocytoma [291]. Therapy with Lomustine (CCNU) has shown limited efficacy. In a group of twenty-three dogs with MCT at various stages, according to Kiupel’s classification, one dog showed complete response; seven dogs had a partial response; and in six dogs, the disease remained stable [292]. Vinblastine belongs to the group of cytostatics. It stops cell division in the metaphase. Moreover, it can lead to apoptosis of non-dividing cells, damage of tumor vascularization, and disruption of translation. Treatment regimens using vinblastine are used in patients in advanced disease stages, when the tumor is inoperable, and in dogs with cutaneous mast cell tumors with a high degree of malignancy [293].
Targeted therapy involves the administration of chemical compounds, which block tumor growth through their effects on specific molecules, such as enzymes and proteins. Targeted therapy is less harmful to normal cells and may have fewer side effects in comparison to conventional chemotherapy. Letard et al. showed that about 20–30% of mast cell tumor cells have a mutation in the KIT receptor, which is responsible for mast cell growth and differentiation [219]. In veterinary medicine, toceranib was the first drug approved for targeted treatment in animals, and it is still indicated for canine mast cell tumors [294,295,296]. It is a drug, which simultaneously targets multiple receptor tyrosine kinases (for example, vascular endothelial growth factor receptor (VEGFR), PDGFR, KIT). Another TKI approved for the treatment of canine mast cell tumors is masitinib mesylate, which, when administered orally, allows MCTs with a mutation in the KIT receptor to inhibit growth. In a study by Letard et al., dogs which received masitinib showed significantly longer survival times relative to the placebo group but only among patients with a mutated KIT receptor [219,291]. Masitinib has been shown to inhibit KIT and other tyrosine kinase receptors, such as PDGFRs and fibroblast growth factor receptor 3 (FGFR3) [139,294]. In their meta-analysis review paper, Coehlo and colleagues pointed out that, among dogs treated with TKIs, 257 dogs received the treatment under the label; 261 received the drug off-label; and 87 dogs received masitinib due to the presence of non-resectable mast cell tumors (grade II or III) with confirmed mutated KIT [133]. In the case of imatinib, this drug is not recommended in dogs [133]. Webster et al. found that treatment with vinblastine in combination with prednisone after surgery was beneficial for dogs with grade III MCT compared to those treated only with surgery. Moreover, dogs with KIT mutations, which were treated with this protocol, had a longer disease-free interval and survival duration [171]. In most cases of canine mastocytoma, molecular analyses are not carried out routinely; therefore, it is difficult to assess the effects of targeted treatment in these animals.
Electrochemotherapy is used in MCT treatment, which combines the intratumoral/intravenous application of specific drugs and treatment with electrical impulses [297]. Electrochemotherapy leads to temporary electropermeabilization of the cell membrane and entry into the cell of the chemotherapeutic agent to enhance its cytotoxic effect [297]. To perform this technique, two main drugs are used: bleomycin (intratumoral application) and cisplatin (intravenous application) [298]. To prevent the effects of mast cell degranulation, it is important to apply electrical pulses initially at the periphery of the tumor, then move toward the center [179].

4.7.3. Radiotherapy

Mast cell tumors, which have been removed with an adequate margin of healthy tissue, usually require no further treatment [284]. Unfortunately, the location of the tumor in the extremities, head, or tail area often prevents the preservation of so-called “clean” margins, and adjuvant therapy must therefore be implemented. Grade II or III mast cell tumors are treated with radiotherapy, which significantly reduces the risk of local relapse [299]. Mastocytomas respond very well to radiotherapy. In a radical protocol, fractionated irradiation is usually used. The average survival time for patients undergoing radiation therapy was between 2 and 5 years [300]. Radiotherapy treatment of mast cell tumors has additional applications as neoadjuvant treatment to facilitate complete tumor resection [300].
When using treatment with ionizing radiation, it is important to consider the possible side effects, which are divided into early and late reactions. Early side effects include swelling, redness, ulceration, and burning of the skin. Late reactions include leukotrichia, skin discoloration, and fibrosis within the irradiated area [301]. In extreme cases, it can lead to osteoradionecrosis, damage of lymphatic and blood vessels, lymphedema, as well as the formation of another tumor [302].

5. Conclusions

This review summarized the advancement in research on the genetic changes in mast cells, including point mutations (mainly), gene expression, chromosomal aberrations, and epigenetic changes in the form of microRNA expression. The prognosis depends on the histologic type of the tumor and the level of progression in the body. Increasingly advanced diagnostic techniques, combined with modern therapeutic approaches, allow long survival times. Genomic knowledge can be applied to the practice; for example, KIT gene mutations are very often present in mastocytoma cells, which affects disease prognosis. KIT/CD117 receptor mutations are typical changes for human mastocytosis. In contrast, in systemic mastocytosis, high KIT/CD117 expression is observed, but usually, there are no KIT gene mutations. This suggests that epigenetic mechanisms may play a significant role in mastocytosis development and metastasis. Moreover, the expressions of some microRNA molecules (classified as epigenetic mechanisms) have been shown to distinguish healthy and MCT-affected dogs, as well as dogs with and without nodal metastasis. Our paper reviews studies on mast cells, mastocytoma, and mastocytosis carried out over several decades and summarizes the research results conducted in this area. Further studies on the pathobiology of mast cells can lead to clinical improvements, such as better diagnosis and treatment of individuals affected by MCT.

Author Contributions

Conceptualization, J.W. and S.Z.; Writing—Original Draft Preparation, J.W., A.K.-S., A.S., J.C.-M. and S.Z.; Writing—Review and Editing, S.Z. and J.C.-M.; Funding Acquisition, J.C.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This publication was funded by grant number SUPB.RN.21.234 (J.C.M.).

Data Availability Statement

Not applicable.

Acknowledgments

The work is dedicated to Dyzia—the dog of Jacek and Alina Wojcierowscy—which sadly died from mastocytoma. Substantive review and editing provided by Paul Dudek.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

5HT5-hydroxytryptamine
ALKanaplastic lymphoma kinase
ASMaggressive systemic mastocytosis
ASMAsanti-smooth muscle antibodies
B/MCPbasophil and MC progenitor
BBMCbaboon bone marrow cells
BNDFbrain-derived neurotrophic factor
CAAcancer-associated adipocytes
CAFcancer-associated fibroblast
CAFscancer-associated fibroblasts
CCLC-C motif ligand
CMcutaneous mastocytosis
CMPcommon myeloid progenitor
CNVcopy number variants
CPA3carboxypeptidase A3
CSF-1Rcolony-stimulating factor receptor
CSPscompetence sensing peptides
CTMCsconnective tissue mast cells
DOCK3dedicator of cytokinesis 3
DSCAMDown syndrome cell adhesion molecule
DSPdesmoplakin
dsRNAdouble-stranded ribonucleic acid
ECMextracellular matrix
EMPerythroid–myeloid progenitors
EVextracellular vesicles
FGFfibroblast growth factor
FNABfine-needle aspiration biopsy
FSDMCfetal-skin-derived mast cells
GM-CSFgranulocyte-macrophage colony stimulating factor
GMPgranulocyte-macrophage progenitor
GPCRG-protein-coupled receptor
HalfaThereditary alpha-tryptasemia
HDMhouse dust mites
HDPheme detoxification protein
HemASChematopoietic adult stem cells
HR1histamine H1 receptor
hSMhuman systemic mastocytosis
HSPheat shock protein
Ilinterleukin
ISMindolent systemic mastocytosis
ITAMimmunoreceptor tyrosine-based activation motif
ITDinternal tandem duplication
JM domainjuxtamembrane domain
LPSlipopolysaccharide
LYSTlysosomal trafficking regulator protein
MAPKmitogen-activated protein kinase
MCASmast cell activation syndrome
MCLmast cell leukemia
MCP4monocyte chemoattractant protein 4
MCsmast cells
MCTmastocytoma
MDR1multi-drug resistance protein 1
MDSCsmyeloid-derived suppressor cells
miRNAmicro-ribonucleic acid
MITFmicrophthalmia-associated transcription factor
MMCsmucosal mast cells
MMTVmouse mammary tumor virus
MPPmultipotent progenitor
NF3nuclear factor 3
NF4nuclear factor 4
NGFnerve growth factor
NGFRnerve growth factor receptor
NKnatural killer
NKRnatural killer receptor
PAFplatelet-activating factor
PAMPspathogen-associated molecular patterns
PCDMCperitoneal-cell-derived mast cells
PCNAproliferating cell nuclear antigen
PD1/CD279programmed cell death protein
PDGFplatelet-derived growth factor
PDGFRplatelet-derived growth factor receptor
PD-L1programmed cell death ligand 1
PEG-ZnPPpegylated zinc protoporphyrin
PGE2prostaglandin E2
PGNpeptidoglycan
PI3Kphosphoinositide 3-kinase
PKP1plakophilin 1
PLC-γphospholipase C gamma
PMNspremetastatic niches
QSMsquorum sensing molecules
SCFstem cell factor
SDPRserum deprivation response
sEVsmall extracellular vesicle
SMsystemic mastocytosis
SNAREsensitive factor attachment protein receptor
ssRNAsingle-stranded ribonucleic acid
TAFstumor-associated fibroblasts
TAMtumor-associated macrophages
TGFtransforming growth factor
TIM3T-cell immunoglobulin and mucin domain 3
TLRsToll-like receptors
TNFtumor necrosis factor
Trktropomyosin receptor kinase
TSLPthymic stromal lymphopoietin
TSLPRthymic stromal lymphopoietin receptor
VEGFvascular endothelial growth factor
VEGFRvascular endothelial growth factor receptor
VIPvasoactive intestinal peptide

References

  1. Cohen, D.; Reif, J.S.; Brodey, R.S.; Keiser, H. Epidemiological analysis of the most prevalent sites and types of canine neoplasia observed in a veterinary hospital. Cancer Res. 1974, 34, 2859–2868. Available online: http://www.ncbi.nlm.nih.gov/pubmed/4529096 (accessed on 15 October 2023). [PubMed]
  2. Priester, W.A. Skin Tumors in Domestic Animals. Data from 12 United States and Canadian Colleges of Veterinary Medicine 2. JNCI J. Natl. Cancer Inst. 1973, 50, 457–466. [Google Scholar] [CrossRef] [PubMed]
  3. Crivellato, E.; Beltrami, C.A.; Mallardi, F.; Ribatti, D. Paul Ehrlich’s doctoral thesis: A milestone in the study of mast cells. Br. J. Haematol. 2003, 123, 19–21. [Google Scholar] [CrossRef] [PubMed]
  4. Yanase, Y.; Matsubara, D.; Takahagi, S.; Tanaka, A.; Ozawa, K.; Hide, M. Basophil Characteristics as a Marker of the Pathogenesis of Chronic Spontaneous Urticaria in Relation to the Coagulation and Complement Systems. Int. J. Mol. Sci. 2023, 24, 10320. [Google Scholar] [CrossRef] [PubMed]
  5. Rocha, M.; Silva, E.; Scroggie, A.E.; Fidlar, E.; Jaques, L.B. Liberation of Histamine and Heparin by Peptone from the Isolated Dog’s Liver. Exp. Biol. Med. 1947, 64, 141–146. [Google Scholar] [CrossRef] [PubMed]
  6. Riley, J.F.; West, G.B. Histamine in tissue mast cells. J. Physiol. 1952, 117, 72P–73P. Available online: http://www.ncbi.nlm.nih.gov/pubmed/12991263 (accessed on 20 October 2023). [CrossRef] [PubMed]
  7. von KöCkritz-Blickwede, M.; Goldmann, O.; Thulin, P.; Heinemann, K.; Norrby-Teglund, A.; Rohde, M.; Medina, E. Phagocytosis-independent antimicrobial activity of mast cells by means of extracellular trap formation. Blood 2008, 111, 3070–3080. [Google Scholar] [CrossRef] [PubMed]
  8. Sobiepanek, A.; Kuryk, Ł.; Garofalo, M.; Kumar, S.; Baran, J.; Musolf, P.; Siebenhaar, F.; Fluhr, J.W.; Kobiela, T.; Plasenzotti, R.; et al. The Multifaceted Roles of Mast Cells in Immune Homeostasis, Infections and Cancers. Int. J. Mol. Sci. 2022, 23, 2249. [Google Scholar] [CrossRef]
  9. Dong, J.; Chen, L.; Zhang, Y.; Jayaswal, N.; Mezghani, I.; Zhang, W.; Veves, A. Mast Cells in Diabetes and Diabetic Wound Healing. Adv. Ther. 2020, 37, 4519–4537. [Google Scholar] [CrossRef]
  10. Bacci, S. Fine Regulation during Wound Healing by Mast Cells, a Physiological Role Not Yet Clarified. Int. J. Mol. Sci. 2022, 23, 1820. [Google Scholar] [CrossRef]
  11. Ryan, J.J.; Kashyap, M.; Bailey, D.; Kennedy, S.; Speiran, K.; Brenzovich, J.; Barnstein, B.; Oskeritzian, C.; Gomez, G. Mast Cell Homeostasis: A Fundamental Aspect of Allergic Disease. Crit. Rev. Immunol. 2007, 27, 15–32. [Google Scholar] [CrossRef] [PubMed]
  12. Galli, S.J.; Grimbaldeston, M.; Tsai, M. Immunomodulatory mast cells: Negative, as well as positive, regulators of immunity. Nat. Rev. Immunol. 2008, 8, 478–486. [Google Scholar] [CrossRef] [PubMed]
  13. da Silva, E.Z.M.; Jamur, M.C.; Oliver, C. Mast Cell Function. J. Histochem. Cytochem. 2014, 62, 698–738. [Google Scholar] [CrossRef] [PubMed]
  14. Moon, T.C.; Befus, A.D.; Kulka, M. Mast Cell Mediators: Their Differential Release and the Secretory Pathways Involved. Front. Immunol. 2014, 5, 569. [Google Scholar] [CrossRef] [PubMed]
  15. Artuc, M.; Hermes, B.; Stckelings, U.M.; Grützkau, A.; Henz, B.M. Mast cells and their mediators in cutaneous wound healing—Active participants or innocent bystanders? Exp. Dermatol. 1999, 8, 1–16. [Google Scholar] [CrossRef] [PubMed]
  16. Lichterman, J.N.; Reddy, S.M. Mast Cells: A New Frontier for Cancer Immunotherapy. Cells 2021, 10, 1270. [Google Scholar] [CrossRef]
  17. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef]
  18. Simpson, R.J.; Kalra, H.; Mathivanan, S. ExoCarta as a resource for exosomal research. J. Extracell. Vesicles 2012, 1, 18374. [Google Scholar] [CrossRef] [PubMed]
  19. Skokos, D.; Botros, H.G.; Demeure, C.; Morin, J.; Peronet, R.; Birkenmeier, G.; Boudaly, S.; Mécheri, S. Mast Cell-Derived Exosomes Induce Phenotypic and Functional Maturation of Dendritic Cells and Elicit Specific Immune Responses In Vivo. J. Immunol. 2003, 170, 3037–3045. [Google Scholar] [CrossRef]
  20. Elstak, E.D.; Neeft, M.; Nehme, N.T.; Voortman, J.; Cheung, M.; Goodarzifard, M.; Gerritsen, H.C.; Henegouwen, P.M.P.v.B.E.; Callebaut, I.; Basile, G.d.S.; et al. The munc13-4–rab27 complex is specifically required for tethering secretory lysosomes at the plasma membrane. Blood 2011, 118, 1570–1578. [Google Scholar] [CrossRef]
  21. Tadokoro, S.; Nakanishi, M.; Hirashima, N. Complexin II facilitates exocytotic release in mast cells by enhancing Ca2+ sensitivity of the fusion process. J. Cell Sci. 2005, 118, 2239–2246. [Google Scholar] [CrossRef] [PubMed]
  22. Riedel, D.; Antonin, W.; Fernandez-Chacon, R.; de Toledo, G.A.; Jo, T.; Geppert, M.; Valentijn, J.A.; Valentijn, K.; Jamieson, J.D.; Südhof, T.C.; et al. Rab3D Is Not Required for Exocrine Exocytosis but for Maintenance of Normally Sized Secretory Granules. Mol. Cell. Biol. 2002, 22, 6487–6497. [Google Scholar] [CrossRef]
  23. Melicoff, E.; Sansores-Garcia, L.; Gomez, A.; Moreira, D.C.; Datta, P.; Thakur, P.; Petrova, Y.; Siddiqi, T.; Murthy, J.N.; Dickey, B.F.; et al. Synaptotagmin-2 Controls Regulated Exocytosis but Not Other Secretory Responses of Mast Cells. J. Biol. Chem. 2009, 284, 19445–19451. [Google Scholar] [CrossRef]
  24. Baier, A.; E Ndoh, V.N.; Lacy, P.; Eitzen, G. Rac1 and Rac2 control distinct events during antigen-stimulated mast cell exocytosis. J. Leukoc. Biol. 2014, 95, 763–774. [Google Scholar] [CrossRef] [PubMed]
  25. Ogawa, K.; Tanaka, Y.; Uruno, T.; Duan, X.; Harada, Y.; Sanematsu, F.; Yamamura, K.; Terasawa, M.; Nishikimi, A.; Côté, J.-F.; et al. DOCK5 functions as a key signaling adaptor that links FcεRI signals to microtubule dynamics during mast cell degranulation. J. Exp. Med. 2014, 211, 1407–1419. [Google Scholar] [CrossRef] [PubMed]
  26. Woska, J.R.; Gillespie, M.E. SNARE complex-mediated degranulation in mast cells. J. Cell. Mol. Med. 2012, 16, 649–656. [Google Scholar] [CrossRef]
  27. Li, Z.; Liu, S.; Xu, J.; Zhang, X.; Han, D.; Liu, J.; Xia, M.; Yi, L.; Shen, Q.; Xu, S.; et al. Adult Connective Tissue-Resident Mast Cells Originate from Late Erythro-Myeloid Progenitors. Immunity 2018, 49, 640–653.e5. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, E.; Nocka, K.; Beier, D.R.; Chu, T.-Y.; Buck, J.; Lahm, H.-W.; Wellner, D.; Leder, P.; Besmer, P. The hematopoietic growth factor KL is encoded by the SI locus and is the ligand of the c-kit receptor, the gene product of the W locus. Cell 1990, 63, 225–233. [Google Scholar] [CrossRef]
  29. Mendoza, R.P.; Fudge, D.H.; Brown, J.M. Cellular Energetics of Mast Cell Development and Activation. Cells 2021, 10, 524. [Google Scholar] [CrossRef]
  30. MacDonald, C.A.; Qian, H.; Pundir, P.; Kulka, M. Sodium butyrate supresses malignant human mast cell proliferation, downregulates expression of KIT and promotes differentiation. Front. Allergy 2023, 4, 1109717. [Google Scholar] [CrossRef]
  31. Welle, M. Development, significance, and heterogeneity of mast cells with particular regard to the mast cell-specific proteases chymase and tryptase. J. Leukoc. Biol. 1997, 61, 233–245. [Google Scholar] [CrossRef] [PubMed]
  32. Pejler, G.; Rönnberg, E.; Waern, I.; Wernersson, S. Mast cell proteases: Multifaceted regulators of inflammatory disease. Blood 2010, 115, 4981–4990. [Google Scholar] [CrossRef] [PubMed]
  33. A Irani, A.; Schechter, N.M.; Craig, S.S.; DeBlois, G.; Schwartz, L.B. Two types of human mast cells that have distinct neutral protease compositions. Proc. Natl. Acad. Sci. USA 1986, 83, 4464–4468. [Google Scholar] [CrossRef]
  34. Schwartz, L.B. Analysis of MC(T) and MC(TC) mast cells in tissue. Methods Mol. Biol. 2006, 315, 53–62. Available online: http://www.ncbi.nlm.nih.gov/pubmed/16110148 (accessed on 20 October 2023).
  35. Abonia, J.P.; Blanchard, C.; Butz, B.B.; Rainey, H.F.; Collins, M.H.; Stringer, K.; Putnam, P.E.; Rothenberg, M.E. Involvement of mast cells in eosinophilic esophagitis. J. Allergy Clin. Immunol. 2010, 126, 140–149. [Google Scholar] [CrossRef] [PubMed]
  36. Irani, A.M.; Craig, S.S.; DeBlois, G.; Elson, C.O.; Schechter, N.M.; Schwartz, L.B. Deficiency of the tryptase-positive, chymase-negative mast cell type in gastrointestinal mucosa of patients with defective T lymphocyte function. J. Immunol. 1987, 138, 4381–4386. Available online: http://www.ncbi.nlm.nih.gov/pubmed/3295046 (accessed on 20 October 2023). [CrossRef] [PubMed]
  37. Gilfillan, A.M.; Austin, S.J.; Metcalfe, D.D. Mast Cell Biology: Introduction and Overview. Adv. Exp. Med. Biol. 2011, 716, 2–12. [Google Scholar] [PubMed]
  38. Dahlin, J.S.; Hallgren, J. Mast cell progenitors: Origin, development and migration to tissues. Mol. Immunol. 2015, 63, 9–17. [Google Scholar] [CrossRef]
  39. Plum, T.; Wang, X.; Rettel, M.; Krijgsveld, J.; Feyerabend, T.B.; Rodewald, H.-R. Human Mast Cell Proteome Reveals Unique Lineage, Putative Functions, and Structural Basis for Cell Ablation. Immunity 2020, 52, 404–416.e5. [Google Scholar] [CrossRef]
  40. Kanagaratham, C.; El Ansari, Y.S.; Lewis, O.L.; Oettgen, H.C. IgE and IgG Antibodies as Regulators of Mast Cell and Basophil Functions in Food Allergy. Front. Immunol. 2020, 11, 603050. [Google Scholar] [CrossRef]
  41. Méndez-Enríquez, E.; Hallgren, J. Mast Cells and Their Progenitors in Allergic Asthma. Front. Immunol. 2019, 10, 821. [Google Scholar] [CrossRef] [PubMed]
  42. Oldford, S.A.; Marshall, J.S. Mast cells as targets for immunotherapy of solid tumors. Mol. Immunol. 2015, 63, 113–124. [Google Scholar] [CrossRef] [PubMed]
  43. Yanase, Y.; Takahagi, S.; Ozawa, K.; Hide, M. The Role of Coagulation and Complement Factors for Mast Cell Activation in the Pathogenesis of Chronic Spontaneous Urticaria. Cells 2021, 10, 1759. [Google Scholar] [CrossRef] [PubMed]
  44. Ikuno, T.; Ito, S.; Inoue, T. Human induced pluripotent stem cell-derived mast cells useful for in vitro mast cell activation assay exhibiting phenotypes and morphological characteristics of human mast cells. J. Toxicol. Sci. 2019, 44, 789–797. [Google Scholar] [CrossRef] [PubMed]
  45. Valent, P.; Akin, C.; Hartmann, K.; Nilsson, G.; Reiter, A.; Hermine, O.; Sotlar, K.; Sperr, W.R.; Escribano, L.; George, T.I.; et al. Mast cells as a unique hematopoietic lineage and cell system: From Paul Ehrlich’s visions to precision medicine concepts. Theranostics 2020, 10, 10743–10768. [Google Scholar] [CrossRef] [PubMed]
  46. Hubálek, Z.; Savage, H.; Halouzka, J.; Juăicová, Z.; Sanogo, Y.; Lusk, S. West Nile Virus Investigations in South Moravia, Czechland. Viral Immunol. 2000, 13, 427–433. [Google Scholar] [CrossRef] [PubMed]
  47. Lamas, D.J.M.; Nicoud, M.B.; A Sterle, H.; A Cremaschi, G.; A Medina, V. Histamine: A potential cytoprotective agent to improve cancer therapy? Cell Death Dis. 2015, 6, e2029. [Google Scholar] [CrossRef]
  48. Blair, R.J.; Meng, H.; Marchese, M.J.; Ren, S.; Schwartz, L.B.; Tonnesen, M.G.; Gruber, B.L. Human mast cells stimulate vascular tube formation. Tryptase is a novel, potent angiogenic factor. J. Clin. Investig. 1997, 99, 2691–2700. [Google Scholar] [CrossRef]
  49. Scott, D.W.; Gascoyne, R.D. The tumour microenvironment in B cell lymphomas. Nat. Rev. Cancer 2014, 14, 517–534. [Google Scholar] [CrossRef]
  50. Conti, P.; Caraffa, A.; Tetè, G.; Gallenga, C.E.; Ross, R.; Kritas, S.K.; Frydas, I.; Younes, A.; Di Emidio, P.; Ronconi, G. Mast cells activated by SARS-CoV-2 release histamine which increases IL-1 levels causing cytokine storm and inflammatory reaction in COVID-19. J. Biol. Regul. Homeost. Agents 2020, 34, 1629–1632. [Google Scholar] [CrossRef]
  51. Yamaguchi, M.; Lantz, C.S.; Oettgen, H.C.; Katona, I.M.; Fleming, T.; Miyajima, I.; Kinet, J.-P.; Galli, S.J. IgE Enhances Mouse Mast Cell FcεRI Expression In Vitro and In Vivo: Evidence for a Novel Amplification Mechanism in IgE-dependent Reactions. J. Exp. Med. 1997, 185, 663–672. [Google Scholar] [CrossRef] [PubMed]
  52. de Castro, R.O.; Zhang, J.; Jamur, M.C.; Oliver, C.; Siraganian, R.P. Tyrosines in the Carboxyl Terminus Regulate Syk Kinase Activity and Function. J. Biol. Chem. 2010, 285, 26674–26684. [Google Scholar] [CrossRef] [PubMed]
  53. Tkaczyk, C.; Jensen, B.M.; Iwaki, S.; Gilfillan, A.M. Adaptive and Innate Immune Reactions Regulating Mast Cell Activation: From Receptor-mediated Signaling to Responses. Immunol. Allergy Clin. N. Am. 2006, 26, 427–450. [Google Scholar] [CrossRef] [PubMed]
  54. Besmer, P.; Murphy, J.E.; George, P.C.; Qiu, F.; Bergold, P.J.; Lederman, L.; Snyder, H.W.; Brodeur, D.; Zuckerman, E.E.; Hardy, W.D. A new acute transforming feline retrovirus and relationship of its oncogene v-kit with the protein kinase gene family. Nature 1986, 320, 415–421. [Google Scholar] [CrossRef]
  55. Tsai, M.; Valent, P.; Galli, S.J. KIT as a master regulator of the mast cell lineage. J. Allergy Clin. Immunol. 2022, 149, 1845–1854. [Google Scholar] [CrossRef] [PubMed]
  56. Oriss, T.B.; Krishnamoorthy, N.; Ray, P.; Ray, A. Dendritic cell c-kit signaling and adaptive immunity. Curr. Opin. Allergy Clin. Immunol. 2014, 14, 7–12. [Google Scholar] [CrossRef] [PubMed]
  57. Yuan, Q.; Austen, K.F.; Friend, D.S.; Heidtman, M.; Boyce, J.A. Human Peripheral Blood Eosinophils Express a Functional c-kit Receptor for Stem Cell Factor that Stimulates Very Late Antigen 4 (VLA-4)–mediated Cell Adhesion to Fibronectin and Vascular Cell Adhesion Molecule 1 (VCAM-1). J. Exp. Med. 1997, 186, 313–323. [Google Scholar] [CrossRef] [PubMed]
  58. Manova, K.; Bachvarova, R.; Huang, E.; Sanchez, S.; Pronovost, S.; Velazquez, E.; McGuire, B.; Besmer, P. c-kit receptor and ligand expression in postnatal development of the mouse cerebellum suggests a function for c-kit in inhibitory interneurons. J. Neurosci. 1992, 12, 4663–4676. [Google Scholar] [CrossRef]
  59. Desai, A.; Sowerwine, K.; Liu, Y.; Lawrence, M.G.; Chovanec, J.; Hsu, A.P.; O’Connell, M.P.; Kim, J.; Boris, L.; Jones, N.; et al. GATA-2–deficient mast cells limit IgE-mediated immediate hypersensitivity reactions in human subjects. J. Allergy Clin. Immunol. 2019, 144, 613–617.e14. [Google Scholar] [CrossRef]
  60. Lemmon, M.A.; Schlessinger, J. Cell Signaling by Receptor Tyrosine Kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef]
  61. Liang, J.; Wu, Y.-L.; Chen, B.-J.; Zhang, W.; Tanaka, Y.; Sugiyama, H. The C-Kit Receptor-Mediated Signal Transduction and Tumor-Related Diseases. Int. J. Biol. Sci. 2013, 9, 435–443. [Google Scholar] [CrossRef]
  62. Anderson, D.M.; Williams, D.E.; Tushinski, R.; Gimpel, S.; Eisenman, J.; Cannizzaro, L.A.; Aronson, M.; Croce, C.M.; Huebner, K.; Cosman, D.; et al. Alternate splicing of mRNAs encoding human mast cell growth factor and localization of the gene to chromosome 12q22-q24. Cell Growth Differ. 1991, 2, 373–378. Available online: http://www.ncbi.nlm.nih.gov/pubmed/1724381 (accessed on 23 October 2023). [PubMed]
  63. Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef] [PubMed]
  64. Youngblood, B.A.; Leung, J.; Falahati, R.; Williams, J.; Schanin, J.; Brock, E.C.; Singh, B.; Chang, A.T.; O’sullivan, J.A.; Schleimer, R.P.; et al. Discovery, Function, and Therapeutic Targeting of Siglec-8. Cells 2020, 10, 19. [Google Scholar] [CrossRef]
  65. McNeil, B.D.; Pundir, P.; Meeker, S.; Han, L.; Undem, B.J.; Kulka, M.; Dong, X. Identification of a mast-cell-specific receptor crucial for pseudo-allergic drug reactions. Nature 2015, 519, 237–241. [Google Scholar] [CrossRef] [PubMed]
  66. Wedi, B.; Gehring, M.; Kapp, A. The pseudoallergen receptor MRGPRX2 on peripheral blood basophils and eosinophils: Expression and function. Allergy Eur. J. Allergy Clin. Immunol. 2020, 75, 2229–2242. [Google Scholar] [CrossRef] [PubMed]
  67. Shtessel, M.; Limjunyawong, N.; Oliver, E.T.; Chichester, K.; Gao, L.; Dong, X.; Saini, S.S. MRGPRX2 Activation Causes Increased Skin Reactivity in Patients with Chronic Spontaneous Urticaria. J. Investig. Dermatol. 2021, 141, 678–681.e2. [Google Scholar] [CrossRef]
  68. Pundir, P.; Liu, R.; Vasavda, C.; Serhan, N.; Limjunyawong, N.; Yee, R.; Zhan, Y.; Dong, X.; Wu, X.; Zhang, Y.; et al. A Connective Tissue Mast-Cell-Specific Receptor Detects Bacterial Quorum-Sensing Molecules and Mediates Antibacterial Immunity. Cell Host Microbe 2019, 26, 114–122.e8. [Google Scholar] [CrossRef] [PubMed]
  69. Lorentz, A.; Sellge, G.; Bischoff, S.C. Isolation and Characterization of Human Intestinal Mast Cells. Methods Mol. Biol. 2015, 1220, 163–177. [Google Scholar]
  70. Trompette, A.; Divanovic, S.; Visintin, A.; Blanchard, C.; Hegde, R.S.; Madan, R.; Thorne, P.S.; Wills-Karp, M.; Gioannini, T.L.; Weiss, J.P.; et al. Allergenicity resulting from functional mimicry of a Toll-like receptor complex protein. Nature 2009, 457, 585–588. [Google Scholar] [CrossRef]
  71. Sandig, H.; Bulfone-Paus, S. TLR signaling in mast cells: Common and unique features. Front. Immunol. 2012, 3, 185. [Google Scholar] [CrossRef] [PubMed]
  72. Theoharides, T.C.; Tsilioni, I.; Conti, P. Mast Cells May Regulate The Anti-Inflammatory Activity of IL-37. Int. J. Mol. Sci. 2019, 20, 3701. [Google Scholar] [CrossRef]
  73. Supajatura, V.; Ushio, H.; Nakao, A.; Okumura, K.; Ra, C.; Ogawa, H. Protective Roles of Mast Cells Against Enterobacterial Infection Are Mediated by Toll-Like Receptor 4. J. Immunol. 2001, 167, 2250–2256. [Google Scholar] [CrossRef]
  74. Supajatura, V.; Ushio, H.; Nakao, A.; Akira, S.; Okumura, K.; Ra, C.; Ogawa, H. Differential responses of mast cell Toll-like receptors 2 and 4 in allergy and innate immunity. J. Clin. Investig. 2002, 109, 1351–1359. [Google Scholar] [CrossRef] [PubMed]
  75. Gimenez, A.P.; Wu, Y.-Z.; Paya, M.; Delclaux, C.; Touqui, L.; Goossens, P.L. High Bactericidal Efficiency of Type IIA Phospholipase A2 against Bacillus anthracis and Inhibition of Its Secretion by the Lethal Toxin. J. Immunol. 2004, 173, 521–530. [Google Scholar] [CrossRef] [PubMed]
  76. Soumelis, V.; Reche, P.A.; Kanzler, H.; Yuan, W.; Edward, G.; Homey, B.; Gilliet, M.; Ho, S.; Antonenko, S.; Lauerma, A.; et al. Human epithelial cells trigger dendritic cell–mediated allergic inflammation by producing TSLP. Nat. Immunol. 2002, 3, 673–680. [Google Scholar] [CrossRef] [PubMed]
  77. Al-Sajee, D.; Oliveria, J.-P.; Sehmi, R.; Gauvreau, G.M. Antialarmins for treatment of asthma. Curr. Opin. Pulm. Med. 2018, 24, 32–41. [Google Scholar] [CrossRef] [PubMed]
  78. Schmitz, J.; Owyang, A.; Oldham, E.; Song, Y.; Murphy, E.; McClanahan, T.K.; Zurawski, G.; Moshrefi, M.; Qin, J.; Li, X.; et al. IL-33, an Interleukin-1-like Cytokine that Signals via the IL-1 Receptor-Related Protein ST2 and Induces T Helper Type 2-Associated Cytokines. Immunity 2005, 23, 479–490. [Google Scholar] [CrossRef] [PubMed]
  79. Iikura, M.; Suto, H.; Kajiwara, N.; Oboki, K.; Ohno, T.; Okayama, Y.; Saito, H.; Galli, S.J.; Nakae, S. IL-33 can promote survival, adhesion and cytokine production in human mast cells. Lab. Investig. 2007, 87, 971–978. [Google Scholar] [CrossRef]
  80. Spicuzza, L.; Di Maria, G.; Polosa, R. Adenosine in the airways: Implications and applications. Eur. J. Pharmacol. 2006, 533, 77–88. [Google Scholar] [CrossRef]
  81. Mohajeri, M.; Kovanen, P.T.; Bianconi, V.; Pirro, M.; Cicero, A.F.; Sahebkar, A. Mast cell tryptase—Marker and maker of cardiovascular diseases. Pharmacol. Ther. 2019, 199, 91–110. [Google Scholar] [CrossRef] [PubMed]
  82. Galli, S.J.; Tsai, M. IgE and mast cells in allergic disease. Nat. Med. 2012, 18, 693–704. [Google Scholar] [CrossRef] [PubMed]
  83. Sprinzl, B.; Greiner, G.; Uyanik, G.; Arock, M.; Haferlach, T.; Sperr, W.R.; Valent, P.; Hoermann, G. Genetic Regulation of Tryptase Production and Clinical Impact: Hereditary α Tryptasemia, Mastocytosis and Beyond. Int. J. Mol. Sci. 2021, 22, 2458. [Google Scholar] [CrossRef] [PubMed]
  84. Greiner, G.; Sprinzl, B.; Górska, A.; Ratzinger, F.; Gurbisz, M.; Witzeneder, N.; Schmetterer, K.G.; Gisslinger, B.; Uyanik, G.; Hadzijusufovic, E.; et al. Hereditary α tryptasemia is a valid genetic biomarker for severe mediator-related symptoms in mastocytosis. Blood 2021, 137, 238–247. [Google Scholar] [CrossRef] [PubMed]
  85. Shanmugam, G.; Schwartz, L.B.; Khan, D.A. Prolonged elevation of serum tryptase in idiopathic anaphylaxis. J. Allergy Clin. Immunol. 2006, 117, 950–951. [Google Scholar] [CrossRef] [PubMed]
  86. Omraninava, M.; Eslami, M.; Aslani, S.; Razi, B.; Imani, D.; Feyzinia, S. Interleukin 13 gene polymorphism and susceptibility to asthma: A meta-regression and meta-analysis. Eur. Ann. Allergy Clin. Immunol. 2022, 54, 150–167. [Google Scholar] [CrossRef] [PubMed]
  87. Okayama, Y.; Hagaman, D.D.; Metcalfe, D.D. A Comparison of Mediators Released or Generated by IFN-γ-Treated Human Mast Cells Following Aggregation of FcγRI or FcεRI. J. Immunol. 2001, 166, 4705–4712. [Google Scholar] [CrossRef] [PubMed]
  88. Galli, S.J.; Tsai, M.; Wershil, B.K. The c-kit receptor, stem cell factor, and mast cells. What each is teaching us about the others. Am. J. Pathol. 1993, 142, 965–974. Available online: http://www.ncbi.nlm.nih.gov/pubmed/7682764 (accessed on 23 October 2023).
  89. Marshall, J.S. Mast-cell responses to pathogens. Nat. Rev. Immunol. 2004, 4, 787–799. [Google Scholar] [CrossRef]
  90. Orinska, Z.; Bulanova, E.; Budagian, V.; Metz, M.; Maurer, M.; Bulfone-Paus, S.; Orinska, Z.; Bulanova, E.; Budagian, V.; Metz, M.; et al. TLR3-induced activation of mast cells modulates CD8+ T-cell recruitment. Blood 2005, 106, 978–987. [Google Scholar] [CrossRef]
  91. Nilsson, G.; Johnell, M.; Hammer, C.H.; Tiffany, H.L.; Nilsson, K.; Metcalfe, D.D.; Siegbahn, A.; Murphy, P.M. C3a and C5a are chemotaxins for human mast cells and act through distinct receptors via a pertussis toxin-sensitive signal transduction pathway. J. Immunol. 1996, 157, 1693–1698. Available online: http://www.ncbi.nlm.nih.gov/pubmed/8759757 (accessed on 23 October 2023). [CrossRef] [PubMed]
  92. Horny, H.-P.; Sotlar, K.; Valent, P.; Hartmann, K. Mastocytosis—A Disease of the Hematopoietic Stem Cell. Dtsch. Arztebl. Int. 2008, 105, 686–692. [Google Scholar] [CrossRef]
  93. Theoharides, T.C.; Valent, P.; Akin, C. Mast Cells, Mastocytosis, and Related Disorders. N. Engl. J. Med. 2015, 373, 1884–1886. [Google Scholar] [CrossRef]
  94. Metcalfe, D.D. Mast cells and mastocytosis. Blood 2008, 112, 946–956. [Google Scholar] [CrossRef] [PubMed]
  95. Valent, P.; Akin, C.; Metcalfe, D.D. Mastocytosis: 2016 updated WHO classification and novel emerging treatment concepts. Blood 2017, 129, 1420–1427. [Google Scholar] [CrossRef] [PubMed]
  96. Hartmann, K.; Escribano, L.; Grattan, C.; Brockow, K.; Carter, M.C.; Alvarez-Twose, I.; Matito, A.; Broesby-Olsen, S.; Siebenhaar, F.; Lange, M.; et al. Cutaneous manifestations in patients with mastocytosis: Consensus report of the European Competence Network on Mastocytosis; the American Academy of Allergy, Asthma & Immunology; and the European Academy of Allergology and Clinical Immunology. J. Allergy Clin. Immunol. 2016, 137, 35–45. [Google Scholar] [CrossRef] [PubMed]
  97. Pardanani, A.; Lim, K.-H.; Lasho, T.L.; Finke, C.M.; McClure, R.F.; Li, C.-Y.; Tefferi, A. WHO subvariants of indolent mastocytosis: Clinical details and prognostic evaluation in 159 consecutive adults. Blood 2010, 115, 150–151. [Google Scholar] [CrossRef] [PubMed]
  98. Chiu, A.; Orazi, A. Mastocytosis and related disorders. Semin. Diagn. Pathol. 2012, 29, 19–30. [Google Scholar] [CrossRef]
  99. Worobec, A.S.; Metcalfe, D.D. Mastocytosis: Current Treatment Concepts. Int. Arch. Allergy Immunol. 2002, 127, 153–155. [Google Scholar] [CrossRef]
  100. Jordan, J.-H.; Walchshofer, S.; Jurecka, W.; Mosberger, I.; Sperr, W.R.; Wolff, K.; Chott, A.; Bühring, H.-J.; Lechner, K.; Horny, H.-P.; et al. Immunohistochemical properties of bone marrow mast cells in systemic mastocytosis: Evidence for expression of CD2, CD117/Kit, and bcl-xL. Hum. Pathol. 2001, 32, 545–552. [Google Scholar] [CrossRef]
  101. Krokowski, M.; Sotlar, K.; Krauth, M.-T.; Födinger, M.; Valent, P.; Horny, H.-P. Delineation of Patterns of Bone Marrow Mast Cell Infiltration in Systemic Mastocytosis. Am. J. Clin. Pathol. 2005, 124, 560–568. [Google Scholar] [CrossRef]
  102. van Anrooij, B.; Kluin, P.M.; Elberink, J.N.O.; Kluin-Nelemans, J.C. CD30 in Systemic Mastocytosis. Immunol. Allergy Clin. N. Am. 2014, 34, 341–355. [Google Scholar] [CrossRef] [PubMed]
  103. Minato, N.; Hattori, M.; Hamazaki, Y. Physiology and pathology of T-cell aging. Int. Immunol. 2020, 32, 223–231. [Google Scholar] [CrossRef] [PubMed]
  104. Kluin-Nelemans, H.C.; Jawhar, M.; Reiter, A.; van Anrooij, B.; Gotlib, J.; Hartmann, K.; Illerhaus, A.; Elberink, H.N.O.; Gorska, A.; Niedoszytko, M.; et al. Cytogenetic and molecular aberrations and worse outcome for male patients in systemic mastocytosis. Theranostics 2021, 11, 292–303. [Google Scholar] [CrossRef] [PubMed]
  105. Arock, M.; Akin, C.; Hermine, O.; Valent, P. Current treatment options in patients with mastocytosis: Status in 2015 and future perspectives. Eur. J. Haematol. 2015, 94, 474–490. [Google Scholar] [CrossRef] [PubMed]
  106. Palacios, F.; Abreu, C.; Prieto, D.; Morande, P.; Ruiz, S.; Fernández-Calero, T.; Naya, H.; Libisch, G.; Robello, C.; I Landoni, A.; et al. Activation of the PI3K/AKT pathway by microRNA-22 results in CLL B-cell proliferation. Leukemia 2015, 29, 115–125. [Google Scholar] [CrossRef]
  107. Garcia-Montero, A.C.; Jara-Acevedo, M.; Teodosio, C.; Sanchez, M.L.; Nunez, R.; Prados, A.; Aldanondo, I.; Sanchez, L.; Dominguez, M.; Botana, L.M.; et al. KIT mutation in mast cells and other bone marrow hematopoietic cell lineages in systemic mast cell disorders: A prospective study of the Spanish Network on Mastocytosis (REMA) in a series of 113 patients. Blood 2006, 108, 2366–2372. [Google Scholar] [CrossRef]
  108. Gotlib, J.; Kluin-Nelemans, H.C.; Akin, C.; Hartmann, K.; Valent, P.; Reiter, A. Practical management of adverse events in patients with advanced systemic mastocytosis receiving midostaurin. Expert Opin. Biol. Ther. 2021, 21, 487–498. [Google Scholar] [CrossRef]
  109. Li, Z.; Beutel, G.; Rhein, M.; Meyer, J.; Koenecke, C.; Neumann, T.; Yang, M.; Krauter, J.; von Neuhoff, N.; Heuser, M.; et al. High-affinity neurotrophin receptors and ligands promote leukemogenesis. Blood 2009, 113, 2028–2037. [Google Scholar] [CrossRef]
  110. Sawada, J.; Itakura, A.; Tanaka, A.; Furusaka, T.; Matsuda, H. Nerve growth factor functions as a chemoattractant for mast cells through both mitogen-activated protein kinase and phosphatidylinositol 3-kinase signaling pathways. Blood 2000, 95, 2052–2058. Available online: http://www.ncbi.nlm.nih.gov/pubmed/10706874 (accessed on 27 October 2023). [CrossRef]
  111. Peng, W.-M.; Maintz, L.; Allam, J.-P.; Raap, U.; Gütgemann, I.; Kirfel, J.; Wardelmann, E.; Perner, S.; Zhao, W.; Fimmers, R.; et al. Increased circulating levels of neurotrophins and elevated expression of their high-affinity receptors on skin and gut mast cells in mastocytosis. Blood 2013, 122, 1779–1788. [Google Scholar] [CrossRef] [PubMed]
  112. Yang, M.; Pan, Z.; Huang, K.; Büsche, G.; Feuerhake, F.; Chaturvedi, A.; Nie, D.; Heuser, M.; Thol, F.; von Neuhoff, N.; et al. Activation of TRKA receptor elicits mastocytosis in mice and is involved in the development of resistance to KIT-targeted therapy. Oncotarget 2017, 8, 73871–73883. [Google Scholar] [CrossRef] [PubMed]
  113. Smrž, D.; Kim, M.-S.; Zhang, S.; Mock, B.A.; Smržová, Š.; DuBois, W.; Simakova, O.; Maric, I.; Wilson, T.M.; Metcalfe, D.D.; et al. mTORC1 and mTORC2 differentially regulate homeostasis of neoplastic and non-neoplastic human mast cells. Blood 2011, 118, 6803–6813. [Google Scholar] [CrossRef] [PubMed]
  114. Gabillot-Carré, M.; Lepelletier, Y.; Humbert, M.; de Sepuvelda, P.; Ben Hamouda, N.; Zappulla, J.P.; Liblau, R.; Ribadeau-Dumas, A.; Machavoine, F.; Letard, S.; et al. Rapamycin inhibits growth and survival of D816V-mutated c-kit mast cells. Blood 2006, 108, 1065–1072. [Google Scholar] [CrossRef] [PubMed]
  115. Itzykson, R.; Kosmider, O.; Fenaux, P. Somatic mutations and epigenetic abnormalities in myelodysplastic syndromes. Best Pract. Res. Clin. Haematol. 2013, 26, 355–364. [Google Scholar] [CrossRef] [PubMed]
  116. Schevzov, G.; Kee, A.J.; Wang, B.; Sequeira, V.B.; Hook, J.; Coombes, J.D.; Lucas, C.A.; Stehn, J.R.; Musgrove, E.A.; Cretu, A.; et al. Regulation of cell proliferation by ERK and signal-dependent nuclear translocation of ERK is dependent on Tm5NM1-containing actin filaments. Mol. Biol. Cell 2015, 26, 2475–2490. [Google Scholar] [CrossRef]
  117. Hanssens, K.; Brenet, F.; Agopian, J.; Georgin-Lavialle, S.; Damaj, G.; Cabaret, L.; Chandesris, M.O.; de Sepulveda, P.; Hermine, O.; Dubreuil, P.; et al. SRSF2-p95 hotspot mutation is highly associated with advanced forms of mastocytosis and mutations in epigenetic regulator genes. Haematologica 2014, 99, 830–835. [Google Scholar] [CrossRef] [PubMed]
  118. Jawhar, M.; Schwaab, J.; Schnittger, S.; Meggendorfer, M.; Pfirrmann, M.; Sotlar, K.; Horny, H.-P.; Metzgeroth, G.; Kluger, S.; Naumann, N.; et al. Additional mutations in SRSF2, ASXL1 and/or RUNX1 identify a high-risk group of patients with KIT D816V+ advanced systemic mastocytosis. Leukemia 2016, 30, 136–143. [Google Scholar] [CrossRef]
  119. Wilson, T.M.; Maric, I.; Simakova, O.; Bai, Y.; Chan, E.C.; Olivares, N.; Carter, M.; Maric, D.; Robyn, J.; Metcalfe, D.D. Clonal analysis of NRAS activating mutations in KIT-D816V systemic mastocytosis. Haematologica 2011, 96, 459–463. [Google Scholar] [CrossRef]
  120. Li, Z. New Insights into the Pathogenesis of Systemic Mastocytosis. Int. J. Mol. Sci. 2021, 22, 4900. [Google Scholar] [CrossRef]
  121. De Vita, S.; Schneider, R.K.; Garcia, M.; Wood, J.; Gavillet, M.; Ebert, B.L.; Gerbaulet, A.; Roers, A.; Levine, R.L.; Mullally, A.; et al. Loss of Function of TET2 Cooperates with Constitutively Active KIT in Murine and Human Models of Mastocytosis. PLoS ONE 2014, 9, e96209. [Google Scholar] [CrossRef] [PubMed]
  122. Martinelli, G.; Mancini, M.; De Benedittis, C.; Rondoni, M.; Papayannidis, C.; Manfrini, M.; Meggendorfer, M.; Calogero, R.; Guadagnuolo, V.; Fontana, M.C.; et al. SETD2 and histone H3 lysine 36 methylation deficiency in advanced systemic mastocytosis. Leukemia 2018, 32, 139–148. [Google Scholar] [CrossRef] [PubMed]
  123. Nagase, R.; Inoue, D.; Pastore, A.; Fujino, T.; Hou, H.-A.; Yamasaki, N.; Goyama, S.; Saika, M.; Kanai, A.; Sera, Y.; et al. Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation. J. Exp. Med. 2018, 215, 1729–1747. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, L.Y.; Smith, M.L.; Schultheis, B.; Fitzgibbon, J.; Lister, T.A.; Melo, J.V.; Cross, N.C.; Cavenagh, J.D. A novel K509I mutation of KIT identified in familial mastocytosis—In vitro and in vivo responsiveness to imatinib therapy. Leuk. Res. 2006, 30, 373–378. [Google Scholar] [CrossRef] [PubMed]
  125. Tang, X.; Boxer, M.; Drummond, A.; Ogston, P.; Hodgins, M.; Burden, A.D. A germline mutation in KIT in familial diffuse cutaneous mastocytosis. J. Med. Genet. 2004, 41, e88. [Google Scholar] [CrossRef] [PubMed]
  126. Wasag, B.; Niedoszytko, M.; Piskorz, A.; Lange, M.; Renke, J.; Jassem, E.; Biernat, W.; Debiec-Rychter, M.; Limon, J. Novel, activating KIT-N822I mutation in familial cutaneous mastocytosis. Exp. Hematol. 2011, 39, 859–865.e2. [Google Scholar] [CrossRef] [PubMed]
  127. Galatà, G.; García-Montero, A.C.; Kristensen, T.; Dawoud, A.A.; Muñoz-González, J.I.; Meggendorfer, M.; Guglielmelli, P.; Hoade, Y.; Alvarez-Twose, I.; Gieger, C.; et al. Genome-wide association study identifies novel susceptibility loci for KIT D816V positive mastocytosis. Am. J. Hum. Genet. 2021, 108, 284–294. [Google Scholar] [CrossRef]
  128. Ammendola, M.; Sacco, R.; Sammarco, G.; Luposella, M.; Patruno, R.; Gadaleta, C.D.; De Sarro, G.; Ranieri, G. Mast Cell-Targeted Strategies in Cancer Therapy. Transfus. Med. Hemotherapy 2016, 43, 109–113. [Google Scholar] [CrossRef]
  129. Faustino-Rocha, A.I.; Gama, A.; Oliveira, P.A.; Vanderperren, K.; Saunders, J.H.; Pires, M.J.; Ferreira, R.; Ginja, M. Modulation of mammary tumor vascularization by mast cells: Ultrasonographic and histopathological approaches. Life Sci. 2017, 176, 35–41. [Google Scholar] [CrossRef]
  130. Iqbal, N.; Iqbal, N. Imatinib: A Breakthrough of Targeted Therapy in Cancer. Chemother. Res. Pract. 2014, 2014, 357027. [Google Scholar] [CrossRef]
  131. Cimpean, A.M.; Raica, M. The Hidden Side of Disodium Cromolyn: From Mast Cell Stabilizer to an Angiogenic Factor and Antitumor Agent. Arch. Immunol. Ther. Exp. 2016, 64, 515–522. [Google Scholar] [CrossRef] [PubMed]
  132. Marech, I.; Ammendola, M.; Gadaleta, C.; Zizzo, N.; Oakley, C.; Gadaleta, C.D.; Ranieri, G. Possible biological and translational significance of mast cells density in colorectal cancer. World J. Gastroenterol. 2014, 20, 8910–8920. [Google Scholar] [CrossRef] [PubMed]
  133. Coelho, Y.N.B.; Soldi, L.R.; da Silva, P.H.R.; Mesquita, C.M.; Paranhos, L.R.; dos Santos, T.R.; Silva, M.J.B. Tyrosine kinase inhibitors as an alternative treatment in canine mast cell tumor. Front. Vet. Sci. 2023, 10, 1188795. [Google Scholar] [CrossRef] [PubMed]
  134. Ma, Y.; Zeng, S.; Metcalfe, D.D.; Akin, C.; Dimitrijevic, S.; Butterfield, J.H.; McMahon, G.; Longley, B.J. The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors; kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatory-type mutations. Blood 2002, 99, 1741–1744. [Google Scholar] [CrossRef] [PubMed]
  135. Droogendijk, H.J.; Kluin-Nelemans, H.J.C.; van Doormaal, J.J.; Oranje, A.P.; van de Loosdrecht, A.A.; van Daele, P.L.A. Imatinib mesylate in the treatment of systemic mastocytosis. Cancer 2006, 107, 345–351. [Google Scholar] [CrossRef]
  136. Pagano, L.; Valentini, C.G.; Caira, M.; Rondoni, M.; Van Lint, M.T.; Candoni, A.; Allione, B.; Cattaneo, C.; Marbello, L.; Caramatti, C.; et al. Advanced mast cell disease: An Italian Hematological Multicenter experience. Int. J. Hematol. 2008, 88, 483–488. [Google Scholar] [CrossRef] [PubMed]
  137. Vega-Ruiz, A.; Cortes, J.E.; Sever, M.; Manshouri, T.; Quintás-Cardama, A.; Luthra, R.; Kantarjian, H.M.; Verstovsek, S. Phase II study of imatinib mesylate as therapy for patients with systemic mastocytosis. Leuk. Res. 2009, 33, 1481–1484. [Google Scholar] [CrossRef]
  138. Pardanani, A.; Elliott, M.; Reeder, T.; Li, C.-Y.; Baxter, E.; Cross, N.; Tefferi, A. Imatinib for systemic mast-cell disease. Lancet 2003, 362, 535–537. [Google Scholar] [CrossRef]
  139. Dubreuil, P.; Letard, S.; Ciufolini, M.; Gros, L.; Humbert, M.; Castéran, N.; Borge, L.; Hajem, B.; Lermet, A.; Sippl, W.; et al. Masitinib (AB1010), a Potent and Selective Tyrosine Kinase Inhibitor Targeting KIT. PLoS ONE 2009, 4, e7258. [Google Scholar] [CrossRef]
  140. Lortholary, O.; Chandesris, M.O.; Livideanu, C.B.; Paul, C.; Guillet, G.; Jassem, E.; Niedoszytko, M.; Barete, S.; Verstovsek, S.; Grattan, C.; et al. Masitinib for treatment of severely symptomatic indolent systemic mastocytosis: A randomised, placebo-controlled, phase 3 study. Lancet 2017, 389, 612–620. [Google Scholar] [CrossRef]
  141. Paul, C.; Sans, B.; Suarez, F.; Casassus, P.; Barete, S.; Lanternier, F.; Grandpeix-Guyodo, C.; Dubreuil, P.; Palmérini, F.; Mansfield, C.D.; et al. Masitinib for the treatment of systemic and cutaneous mastocytosis with handicap: A phase 2a study. Am. J. Hematol. 2010, 85, 921–925. [Google Scholar] [CrossRef] [PubMed]
  142. Fabbro, D.; Ruetz, S.; Bodis, S.; Pruschy, M.; Csermak, K.; Man, A.; Campochiaro, P.; Wood, J.; O’Reilly, T.; Meyer, T. PKC412--a protein kinase inhibitor with a broad therapeutic potential. Anticancer. Drug Des. 2000, 15, 17–28. Available online: http://www.ncbi.nlm.nih.gov/pubmed/10888033 (accessed on 27 October 2023).
  143. Evans, E.K.; Gardino, A.K.; Kim, J.L.; Hodous, B.L.; Shutes, A.; Davis, A.; Zhu, X.J.; Schmidt-Kittler, O.; Wilson, D.; Wilson, K.; et al. A precision therapy against cancers driven by KIT/PDGFRA mutations. Sci. Transl. Med. 2017, 9, eaao1690. [Google Scholar] [CrossRef]
  144. Dave, N.; Devlin, M.; Rodstrom, J.; Yu, B.; Foley, M.; He, K.; Rasmussen, S.; Boral, A.; Si, T.D. Abstract CT122: Safety and pharmacokinetics of BLU-263, a next-generation KIT inhibitor, in normal healthy volunteers. Cancer Res 2021, 81, CT122. [Google Scholar] [CrossRef]
  145. Guarnieri, A.; Chicarelli, M.; Cable, L.; Bouhana, K.; Sullivan, F.; Ball, H.; Sachs, J.; Winski, S.; Robinson, J. Preclinical Data with KIT D816V Inhibitor Bezuclastinib (CGT9486) Demonstrates High Selectivity and Minimal Brain Penetrance. Blood 2021, 138 (Suppl. S1), 4595. [Google Scholar] [CrossRef]
  146. Sledge, D.G.; Webster, J.; Kiupel, M. Canine cutaneous mast cell tumors: A combined clinical and pathologic approach to diagnosis, prognosis, and treatment selection. Vet. J. 2016, 215, 43–54. [Google Scholar] [CrossRef] [PubMed]
  147. Aceino, A.; Jeffery, U.; Piccione, J.; Hodo, C.L. Systemic mastocytosis with subcutaneous hemorrhage and edema in a Greyhound dog: Case report and review of diagnostic criteria. J. Vet. Diagn. Investig. 2020, 33, 95–100. [Google Scholar] [CrossRef]
  148. Zorzan, E.; Hanssens, K.; Giantin, M.; Dacasto, M.; Dubreuil, P. Mutational Hotspot of TET2, IDH1, IDH2, SRSF2, SF3B1, KRAS, and NRAS from Human Systemic Mastocytosis Are Not Conserved in Canine Mast Cell Tumors. PLoS ONE 2015, 10, e0142450. [Google Scholar] [CrossRef]
  149. Pariser, M.S.; Gram, D.W. Urticaria pigmentosa-like disease in a dog. Can. Vet. J. 2015, 56, 245–248. Available online: http://www.ncbi.nlm.nih.gov/pubmed/25750443 (accessed on 27 October 2023).
  150. da Silva, M.S.; Klaser, W.; Scherer, F.; Santos, D.D.; Garcia, C.; Bortolini, E.; Costa, M. Metastatic Intranasal Mastocytoma in a Dog. Acta Sci. Vet. 2023, 51, 892. [Google Scholar] [CrossRef]
  151. Thompson, J.J.; Pearl, D.L.; Yager, J.A.; Best, S.J.; Coomber, B.L.; Foster, R.A. Canine Subcutaneous Mast Cell Tumor. Vet. Pathol. 2011, 48, 156–168. [Google Scholar] [CrossRef] [PubMed]
  152. Bostock, D.E. The prognosis following surgical removal of mastocytomas in dogs. J. Small Anim. Pract. 1973, 14, 27–40. [Google Scholar] [CrossRef] [PubMed]
  153. Patnaik, A.K.; Ehler, W.J.; MacEwen, E.G. Canine Cutaneous Mast Cell Tumor: Morphologic Grading and Survival Time in 83 Dogs. Vet. Pathol. 1984, 21, 469–474. [Google Scholar] [CrossRef] [PubMed]
  154. Kiupel, M.; Webster, J.D.; Bailey, K.L.; Best, S.; DeLay, J.; Detrisac, C.J.; Fitzgerald, S.D.; Gamble, D.; Ginn, P.E.; Goldschmidt, M.H.; et al. Proposal of a 2-Tier Histologic Grading System for Canine Cutaneous Mast Cell Tumors to More Accurately Predict Biological Behavior. Vet. Pathol. 2011, 48, 147–155. [Google Scholar] [CrossRef] [PubMed]
  155. Stefanello, D.; Buracco, P.; Sabattini, S.; Finotello, R.; Giudice, C.; Grieco, V.; Iussich, S.; Tursi, M.; Scase, T.; Di Palma, S.; et al. Comparison of 2- and 3-category histologic grading systems for predicting the presence of metastasis at the time of initial evaluation in dogs with cutaneous mast cell tumors: 386 cases (2009–2014). J. Am. Vet. Med. Assoc. 2015, 246, 765–769. [Google Scholar] [CrossRef] [PubMed]
  156. Northrup, N.C.; Howerth, E.W.; Harmon, B.G.; Brown, C.A.; Carmicheal, K.P.; Garcia, A.P.; Latimer, K.S.; Munday, J.S.; Rakich, P.M.; Richey, L.J.; et al. Variation among Pathologists in the Histologic Grading of Canine Cutaneous Mast Cell Tumors with Uniform Use of a Single Grading Reference. J. Vet. Diagn. Investig. 2005, 17, 561–564. [Google Scholar] [CrossRef]
  157. Ozaki, K.; Yamagami, T.; Nomura, K.; Narama, I. Mast cell tumors of the gastrointestinal tract in 39 dogs. Vet. Pathol. 2002, 39, 557–564. [Google Scholar] [CrossRef]
  158. Ejima, E.; Fujiyama, K.; Kiriyama, T.; Eguchi, K. [Bone changes in thyrotoxicosis]. Nihon Rinsho. 1998, 56, 1587–1590. Available online: http://www.ncbi.nlm.nih.gov/pubmed/9648486 (accessed on 28 October 2023).
  159. Elston, L.B.; Sueiro, F.A.; Cavalcanti, J.N.; Metze, K. Letter to the Editor: The Importance of the Mitotic Index as a Prognostic Factor for Survival of Canine Cutaneous Mast Cell Tumors: A Validation Study. Vet. Pathol. 2009, 46, 362–364. [Google Scholar] [CrossRef]
  160. Marcos, R.; Almeida, J.; Marques, J.; Moreira, R.; Dias-Pereira, P.; Correia-Gomes, C.; Santos, M. Canine mast cell tumors: Utility of stereologic tools in cytology. J. Vet. Diagn. Investig. 2022, 34, 263–267. [Google Scholar] [CrossRef]
  161. Hosseini, E.; Pedram, B.; Bahrami, A.M.; Moghaddam, M.H.J.; Javanbakht, J.; Ghomi, F.E.; Moghaddam, N.J.; Koohestani, M.; Shafiee, R. RETRACTED ARTICLE: Cutaneous mast cell tumor (Mastocytoma): Cyto- histopathological and haematological investigations. Diagn. Pathol. 2014, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  162. Cruz, V.S.; Borges, J.C.A.; Nepomuceno, L.L.; Gonçalves, P.A.M.; Prado, Y.C.L.; Bianchi, C.; Fioravanti, M.C.S.; Araújo, E.G. Histological classification and expression of markers of canine mast cell tumors. Vet. World 2020, 13, 1627–1637. [Google Scholar] [CrossRef] [PubMed]
  163. Hillman, L.A.; Garrett, L.D.; de Lorimier, L.-P.; Charney, S.C.; Borst, L.B.; Fan, T.M. Biological behavior of oral and perioral mast cell tumors in dogs: 44 cases (1996–2006). J. Am. Vet. Med. Assoc. 2010, 237, 936–942. [Google Scholar] [CrossRef]
  164. Graf, R.; Pospischil, A.; Guscetti, F.; Meier, D.; Welle, M.; Dettwiler, M. Cutaneous Tumors in Swiss Dogs: Retrospective Data From the Swiss Canine Cancer Registry, 2008–2013. Vet. Pathol. 2018, 55, 809–820. [Google Scholar] [CrossRef] [PubMed]
  165. Pejler, G.; Åbrink, M.; Ringvall, M.; Wernersson, S. Mast Cell Proteases. Adv. Immunol. 2007, 95, 167–255. [Google Scholar]
  166. Sakai, H.; Noda, A.; Shirai, N.; Iidaka, T.; Yanai, T.; Masegi, T. Proliferative Activity of Canine Mast Cell Tumours Evaluated by Bromodeoxyuridine Incorporation and Ki-67 Expression. J. Comp. Pathol. 2002, 127, 233–238. [Google Scholar] [CrossRef]
  167. Simoes, J.P.C.; Schoning, P.; Butine, M. Prognosis of Canine Mast Cell Tumors: A Comparison of Three Methods. Vet. Pathol. 1994, 31, 637–647. [Google Scholar] [CrossRef]
  168. Kravis, L.D.; Vail, D.M.; Kisseberth, W.C.; Ogilvie, G.K.; Volk, L.M. Frequency of argyrophilic nucleolar organizer regions in fine-needle aspirates and biopsy specimens from mast cell tumors in dogs. J. Am. Vet. Med. Assoc. 1996, 209, 1418–1420. Available online: http://www.ncbi.nlm.nih.gov/pubmed/8870737 (accessed on 28 October 2023). [CrossRef] [PubMed]
  169. Ayl, R.D.; Couto, C.G.; Hammer, A.S.; Weisbrode, S.; Ericson, J.G.; Mathes, L. Correlation of DNA Ploidy to Tumor Histologic Grade, Clinical Variables, and Survival in Dogs with Mast Cell Tumors. Vet. Pathol. 1992, 29, 386–390. [Google Scholar] [CrossRef]
  170. Jaffe, M.H.; Hosgood, G.; Taylor, H.W.; Kerwin, S.C.; Hedlund, C.S.; Lopez, M.K.; Davidson, J.R.; Miller, D.M.; Paranjpe, M. Immunohistochemical and Clinical Evaluation of p53 in Canine Cutaneous Mast Cell Tumors. Vet. Pathol. 2000, 37, 40–46. [Google Scholar] [CrossRef]
  171. Webster, J.D.; Kiupel, M.; Yuzbasiyan-Gurkan, V. Evaluation of the kinase domain of c-KIT in canine cutaneous mast cell tumors. BMC Cancer 2006, 6, 85. [Google Scholar] [CrossRef]
  172. Strefezzi, R.D.F.; Xavier, J.G.; Catão-Dias, J.L. Morphometry of Canine Cutaneous Mast Cell Tumors. Vet. Pathol. 2003, 40, 268–275. [Google Scholar] [CrossRef]
  173. Kiupel, M.; Webster, J.D.; Miller, R.A.; Kaneene, J.B. Impact of Tumour Depth, Tumour Location and Multiple Synchronous Masses on the Prognosis of Canine Cutaneous Mast Cell Tumours. J. Vet. Med. Ser. A 2005, 52, 280–286. [Google Scholar] [CrossRef]
  174. Cahalane, A.K.; Payne, S.; Barber, L.G.; Duda, L.E.; Henry, C.J.; Mauldin, G.E.; Frimberger, A.E.; Cotter, S.M.; Moore, A.S. Prognostic factors for survival of dogs with inguinal and perineal mast cell tumors treated surgically with or without adjunctive treatment: 68 cases (1994–2002). J. Am. Vet. Med. Assoc. 2004, 225, 401–408. [Google Scholar] [CrossRef]
  175. Kiupel, M.; Webster, J.D.; Kaneene, J.B.; Miller, R.; Yuzbasiyan-Gurkan, V. The Use of KIT and Tryptase Expression Patterns as Prognostic Tools for Canine Cutaneous Mast Cell Tumors. Vet. Pathol. 2004, 41, 371–377. [Google Scholar] [CrossRef] [PubMed]
  176. Thompson, J.J.; Morrison, J.A.; Pearl, D.L.; Boston, S.E.; Wood, G.A.; Foster, R.A.; Coomber, B.L. Receptor Tyrosine Kinase Expression Profiles in Canine Cutaneous and Subcutaneous Mast Cell Tumors. Vet. Pathol. 2016, 53, 545–558. [Google Scholar] [CrossRef]
  177. Webster, J.D.; Yuzbasiyan-Gurkan, V.; Miller, R.A.; Kaneene, J.B.; Kiupel, M. Cellular Proliferation in Canine Cutaneous Mast Cell Tumors: Associations with c-KIT and Its Role in Prognostication. Vet. Pathol. 2007, 44, 298–308. [Google Scholar] [CrossRef] [PubMed]
  178. Smith, J.; Kiupel, M.; Farrelly, J.; Cohen, R.; Olmsted, G.; Kirpensteijn, J.; Brocks, B.; Post, G. Recurrence rates and clinical outcome for dogs with grade II mast cell tumours with a low AgNOR count and Ki67 index treated with surgery alone. Vet. Comp. Oncol. 2017, 15, 36–45. [Google Scholar] [CrossRef] [PubMed]
  179. de Nardi, A.B.; Horta, R.d.S.; Fonseca-Alves, C.E.; de Paiva, F.N.; Linhares, L.C.M.; Firmo, B.F.; Sueiro, F.A.R.; de Oliveira, K.D.; Lourenço, S.V.; Strefezzi, R.D.F.; et al. Diagnosis, Prognosis and Treatment of Canine Cutaneous and Subcutaneous Mast Cell Tumors. Cells 2022, 11, 618. [Google Scholar] [CrossRef]
  180. Meuten, D.J. (Ed.) Tumors in Domestic Animals; Wiley: Hoboken, NJ, USA, 2016. [Google Scholar]
  181. Book, A.P.; Fidel, J.; Wills, T.; Bryan, J.; Sellon, R.; Mattoon, J. Correlation of Ultrasound Findings, Liver and Spleen Cytology, and Prognosis in the Clinical Staging of High Metastatic Risk Canine Mast Cell Tumors. Vet. Radiol. Ultrasound 2011, 52, 548–554. [Google Scholar] [CrossRef]
  182. Sulce, M.; Marconato, L.; Martano, M.; Iussich, S.; Dentini, A.; Melega, M.; Miniscalco, B.; Riondato, F. Utility of flow cytometry in canine primary cutaneous and matched nodal mast cell tumor. Vet. J. 2018, 242, 15–23. [Google Scholar] [CrossRef] [PubMed]
  183. Chiarle, R.; Podda, A.; Prolla, G.; Gong, J.; Thorbecke, G.; Inghirami, G. CD30 in Normal and Neoplastic Cells. Clin. Immunol. 1999, 90, 157–164. [Google Scholar] [CrossRef]
  184. Bauer, K.; Hadzijusufovic, E.; Cerny-Reiterer, S.; Hoermann, G.; Reifinger, M.; Pirker, A.; Valent, P.; Willmann, M. IL-4 downregulates expression of the target receptor CD30 in neoplastic canine mast cells. Vet. Comp. Oncol. 2017, 15, 1240–1256. [Google Scholar] [CrossRef] [PubMed]
  185. Uxa, S.; Castillo-Binder, P.; Kohler, R.; Stangner, K.; Müller, G.A.; Engeland, K. Ki-67 gene expression. Cell Death Differ. 2021, 28, 3357–3370. [Google Scholar] [CrossRef]
  186. Booth, D.G.; Takagi, M.; Sanchez-Pulido, L.; Petfalski, E.; Vargiu, G.; Samejima, K.; Imamoto, N.; Ponting, C.P.; Tollervey, D.; Earnshaw, W.C.; et al. Ki-67 is a PP1-interacting protein that organises the mitotic chromosome periphery. eLife 2014, 3, e01641. [Google Scholar] [CrossRef]
  187. Mrouj, K.; Andrés-Sánchez, N.; Dubra, G.; Singh, P.; Sobecki, M.; Chahar, D.; Al Ghoul, E.; Aznar, A.B.; Prieto, S.; Pirot, N.; et al. Ki-67 regulates global gene expression and promotes sequential stages of carcinogenesis. Proc. Natl. Acad. Sci. USA 2021, 118, e2026507118. [Google Scholar] [CrossRef] [PubMed]
  188. Makii, R.; Cook, H.; Louke, D.; Breitbach, J.; Jennings, R.; Premanandan, C.; Green, E.M.; Fenger, J.M. Characterization of WWOX expression and function in canine mast cell tumors and malignant mast cell lines. BMC Vet. Res. 2020, 16, 415. [Google Scholar] [CrossRef]
  189. Aqeilan, R.I.; Trapasso, F.; Hussain, S.; Costinean, S.; Marshall, D.; Pekarsky, Y.; Hagan, J.P.; Zanesi, N.; Kaou, M.; Stein, G.S.; et al. Targeted deletion of Wwox reveals a tumor suppressor function. Proc. Natl. Acad. Sci. USA 2007, 104, 3949–3954. [Google Scholar] [CrossRef]
  190. Schrock, M.S.; Batar, B.; Lee, J.; Druck, T.; Ferguson, B.; Cho, J.H.; Akakpo, K.; Hagrass, H.; A Heerema, N.; Xia, F.; et al. Wwox–Brca1 interaction: Role in DNA repair pathway choice. Oncogene 2017, 36, 2215–2227. [Google Scholar] [CrossRef]
  191. Gamperl, S.; Stefanzl, G.; Willmann, M.; Valent, P.; Hadzijusufovic, E. In vitro effects of histamine receptor 1 antagonists on proliferation and histamine release in canine neoplastic mast cells. Vet. Med. Sci. 2021, 7, 57–68. [Google Scholar] [CrossRef]
  192. Gonzalez-Maganña, A.; Blanco, F.J. Human PCNA Structure, Function and Interactions. Biomolecules 2020, 10, 570. [Google Scholar] [CrossRef] [PubMed]
  193. Gill, V.; Leibman, N.; Monette, S.; Craft, D.M.; Bergman, P.J. Prognostic Indicators and Clinical Outcome in Dogs with Subcutaneous Mast Cell Tumors Treated with Surgery Alone: 43 Cases. J. Am. Anim. Hosp. Assoc. 2020, 56, 215–225. [Google Scholar] [CrossRef] [PubMed]
  194. Takahashi, T.; Ohashi, E.; Nakagawa, T.; Mochizuki, M.; Nishimura, R.; Sasaki, N. Role of β1 Integrins in Adhesion of Canine Mastocytoma Cells to Extracellular Matrix Proteins. J. Vet. Med. Sci. 2007, 69, 495–499. [Google Scholar] [CrossRef]
  195. Houtman, R.; Koster, A.S.; Nijkamp, F.P. Integrin VLA-5: Modulator and activator of mast cells. Clin. Exp. Allergy 2001, 31, 817–822. [Google Scholar] [CrossRef] [PubMed]
  196. Foster, D.S.; Jones, R.E.; Ransom, R.C.; Longaker, M.T.; Norton, J.A. The evolving relationship of wound healing and tumor stroma. JCI Insight 2018, 3, e99911. [Google Scholar] [CrossRef] [PubMed]
  197. Hadzijusufovic, E.; Rebuzzi, L.; Gleixner, K.V.; Ferenc, V.; Peter, B.; Kondo, R.; Gruze, A.; Kneidinger, M.; Krauth, M.-T.; Mayerhofer, M.; et al. Targeting of heat-shock protein 32/heme oxygenase-1 in canine mastocytoma cells is associated with reduced growth and induction of apoptosis. Exp. Hematol. 2008, 36, 1461–1470. [Google Scholar] [CrossRef]
  198. Kondo, R.; Gleixner, K.V.; Mayerhofer, M.; Vales, A.; Gruze, A.; Samorapoompichit, P.; Greish, K.; Krauth, M.-T.; Aichberger, K.J.; Pickl, W.F.; et al. Identification of heat shock protein 32 (Hsp32) as a novel survival factor and therapeutic target in neoplastic mast cells. Blood 2007, 110, 661–669. [Google Scholar] [CrossRef]
  199. Kamal, A.; Thao, L.; Sensintaffar, J.; Zhang, L.; Boehm, M.F.; Fritz, L.C.; Burrows, F.J. A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature 2003, 425, 407–410. [Google Scholar] [CrossRef]
  200. Whitesell, L.; Lindquist, S.L. HSP90 and the chaperoning of cancer. Nat. Rev. Cancer 2005, 5, 761–772. [Google Scholar] [CrossRef]
  201. Xu, W.; Neckers, L. Targeting the Molecular Chaperone Heat Shock Protein 90 Provides a Multifaceted Effect on Diverse Cell Signaling Pathways of Cancer Cells. Clin. Cancer Res. 2007, 13, 1625–1629. [Google Scholar] [CrossRef]
  202. Lin, T.-Y.; Bear, M.; Du, Z.; Foley, K.P.; Ying, W.; Barsoum, J.; London, C. The novel HSP90 inhibitor STA-9090 exhibits activity against Kit-dependent and -independent malignant mast cell tumors. Exp. Hematol. 2008, 36, 1266–1277. [Google Scholar] [CrossRef]
  203. Akgul, C. Mcl-1 is a potential therapeutic target in multiple types of cancer. Cell. Mol. Life Sci. 2009, 66, 1326–1336. [Google Scholar] [CrossRef]
  204. Moulding, D.A.; Giles, R.V.; Spiller, D.G.; White, M.R.; Tidd, D.M.; Edwards, S.W. Apoptosis is rapidly triggered by antisense depletion of MCL-1 in differentiating U937 cells. Blood 2000, 96, 1756–1763. Available online: http://www.ncbi.nlm.nih.gov/pubmed/10961874 (accessed on 29 October 2023). [CrossRef] [PubMed]
  205. Maekawa, N.; Konnai, S.; Ikebuchi, R.; Okagawa, T.; Adachi, M.; Takagi, S.; Kagawa, Y.; Nakajima, C.; Suzuki, Y.; Murata, S.; et al. Expression of PD-L1 on Canine Tumor Cells and Enhancement of IFN-γ Production from Tumor-Infiltrating Cells by PD-L1 Blockade. PLoS ONE 2014, 9, e98415. [Google Scholar] [CrossRef] [PubMed]
  206. Geng, L.; Huang, D.; Liu, J.; Qian, Y.; Deng, J.; Li, D.; Hu, Z.; Zhang, J.; Jiang, G.; Zheng, S. B7-H1 up-regulated expression in human pancreatic carcinoma tissue associates with tumor progression. J. Cancer Res. Clin. Oncol. 2008, 134, 1021–1027. [Google Scholar] [CrossRef]
  207. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.M.; Hwu, W.-J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and Activity of Anti-PD-L1 Antibody in Patients with Advanced Cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
  208. Sharma, S.; Gao, X.; Londono, D.; Devroy, S.E.; Mauldin, K.N.; Frankel, J.T.; Brandon, J.M.; Zhang, D.; Li, Q.-Z.; Dobbs, M.B.; et al. Genome-wide association studies of adolescent idiopathic scoliosis suggest candidate susceptibility genes. Hum. Mol. Genet. 2011, 20, 1456–1466. [Google Scholar] [CrossRef] [PubMed]
  209. Tate, J.G.; Bamford, S.; Jubb, H.C.; Sondka, Z.; Beare, D.M.; Bindal, N.; Boutselakis, H.; Cole, C.G.; Creatore, C.; Dawson, E.; et al. COSMIC: The Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2019, 47, D941–D947. [Google Scholar] [CrossRef]
  210. Arendt, M.L.; Melin, M.; Tonomura, N.; Koltookian, M.; Courtay-Cahen, C.; Flindall, N.; Bass, J.; Boerkamp, K.; Megquir, K.; Youell, L.; et al. Genome-Wide Association Study of Golden Retrievers Identifies Germ-Line Risk Factors Predisposing to Mast Cell Tumours. PLoS Genet. 2015, 11, e1005647. [Google Scholar] [CrossRef]
  211. Olsson, M.; Meadows, J.R.S.; Truvé, K.; Pielberg, G.R.; Puppo, F.; Mauceli, E.; Quilez, J.; Tonomura, N.; Zanna, G.; Docampo, M.J.; et al. A Novel Unstable Duplication Upstream of HAS2 Predisposes to a Breed-Defining Skin Phenotype and a Periodic Fever Syndrome in Chinese Shar-Pei Dogs. PLoS Genet. 2011, 7, e1001332. [Google Scholar] [CrossRef]
  212. Lv, Y.S.; Yao, Y.S.; Rong, L.; Lin, M.E.; Deng, B.H.; Xie, Y.; Huang, H.; Lin, T.X.; Xu, K.W.; Huang, J. Intravesical hyaluronidase causes chronic cystitis in a rat model: A potential model of bladder pain syndrome/interstitial cystitis. Int. J. Urol. 2014, 21, 601–607. [Google Scholar] [CrossRef]
  213. Nakaichi, M.; Takeshita, Y.; Okuda, M.; Nakamoto, Y.; Itamoto, K.; Une, S.; Sasaki, N.; Kadosawa, T.; Takahashi, T.; Taura, Y. Expression of the MDR1 Gene and P-Glycoprotein in Canine Mast Cell Tumor Cell Lines. J. Vet. Med. Sci. 2007, 69, 111–115. [Google Scholar] [CrossRef] [PubMed]
  214. Spritz, R.A. Molecular Basis of Human Piebaldism. J. Investig. Dermatol. 1994, 103, 137S–140S. [Google Scholar] [CrossRef] [PubMed]
  215. Phung, B.; Steingrímsson, E.; Rönnstrand, L. Differential activity of c-KIT splice forms is controlled by extracellular peptide insert length. Cell Signal. 2013, 25, 2231–2238. [Google Scholar] [CrossRef]
  216. Morgan, E.; O’Connell, K.; Thomson, M.; Griffin, A. Canine T cell lymphoma treated with lomustine, vincristine, procarbazine, and prednisolone chemotherapy in 35 dogs. Vet. Comp. Oncol. 2018, 16, 622–629. [Google Scholar] [CrossRef] [PubMed]
  217. Chatterjee, A.; Ghosh, J.; Kapur, R. Mastocytosis: A mutated KIT receptor induced myeloproliferative disorder. Oncotarget 2015, 6, 18250–18264. [Google Scholar] [CrossRef] [PubMed]
  218. Lennartsson, J.; Rönnstrand, L. Stem Cell Factor Receptor/c-Kit: From Basic Science to Clinical Implications. Physiol. Rev. 2012, 92, 1619–1649. [Google Scholar] [CrossRef]
  219. Letard, S.; Yang, Y.; Hanssens, K.; Palmérini, F.; Leventhal, P.S.; Guéry, S.; Moussy, A.; Kinet, J.-P.; Hermine, O.; Dubreuil, P. Gain-of-Function Mutations in the Extracellular Domain of KIT Are Common in Canine Mast Cell Tumors. Mol. Cancer Res. 2008, 6, 1137–1145. [Google Scholar] [CrossRef]
  220. Mochizuki, H.; Thomas, R.; Moroff, S.; Breen, M. Genomic profiling of canine mast cell tumors identifies DNA copy number aberrations associated with KIT mutations and high histological grade. Chromosom. Res. 2017, 25, 129–143. [Google Scholar] [CrossRef]
  221. Chen, P.; Marconato, L.; Sabattini, S.; Kiupel, M. Mutations in Exons 8 and 11 of c-kit Gene in Canine Subcutaneous Mast Cell Tumors and Their Association with Cell Proliferation. Vet. Sci. 2022, 9, 493. [Google Scholar] [CrossRef]
  222. Zhou, X.; Santos, G.S.; Zhan, Y.; Oliveira, M.M.S.; Rezaei, S.; Singh, M.; Peuget, S.; Westerberg, L.S.; Johnsen, J.I.; Selivanova, G. Mutant p53 gain of function mediates cancer immune escape that is counteracted by APR-246. Br. J. Cancer 2022, 127, 2060–2071. [Google Scholar] [CrossRef]
  223. Vozdova, M.; Kubickova, S.; Fictum, P.; Fröhlich, J.; Jelinek, F.; Rubes, J. Prevalence and prognostic value of c-kit and TP53 mutations in canine mast cell tumours. Vet. J. 2019, 247, 71–74. [Google Scholar] [CrossRef] [PubMed]
  224. Biasoli, D.; Compston-Garnett, L.; Ricketts, S.L.; Birand, Z.; Courtay-Cahen, C.; Fineberg, E.; Arendt, M.; Boerkamp, K.; Melin, M.; Koltookian, M.; et al. A synonymous germline variant in a gene encoding a cell adhesion molecule is associated with cutaneous mast cell tumour development in Labrador and Golden Retrievers. PLoS Genet. 2019, 15, e1007967. [Google Scholar] [CrossRef]
  225. Amagai, Y.; Tanaka, A.; Matsuda, A.; Oida, K.; Jung, K.; Nishikawa, S.; Jang, H.; Ishizaka, S.; Matsuda, H. Increased Expression of the Antiapoptotic Protein MCL1 in Canine Mast Cell Tumors. J. Vet. Med. Sci. 2013, 75, 971–974. [Google Scholar] [CrossRef] [PubMed]
  226. Śmiech, A.; Ślaska, B.; Surdyka, M.; Grzybowska-Szatkowska, L.; Łopuszyński, W.; Różańska, D. Identification of additional mitochondrial DNA mutations in canine mast cell tumours. Acta Vet. Scand. 2015, 58, 28. [Google Scholar] [CrossRef] [PubMed]
  227. Destexhe, E.; Lespagnard, L.; Degeyter, M.; Heymann, R.; Coignoul, F. Immunohistochemical Identification of Myoepithelial, Epithelial, and Connective Tissue Cells in Canine Mammary Tumors. Vet. Pathol. 1993, 30, 146–154. [Google Scholar] [CrossRef] [PubMed]
  228. Conrad, D.; Kehl, A.; Müller, T.; Klopfleisch, R.; Aupperle-Lellbach, H. Immunohistochemical and Molecular Genetic Analysis of Canine Digital Mast Cell Tumours. Animals 2023, 13, 1694. [Google Scholar] [CrossRef] [PubMed]
  229. Brocks, B.A.W.; Bertram, C.A.; Bartel, A.; Kirpensteijn, J.; Collins-Webb, A.; Catlin, C.; Thaiwong, T.; Kiupel, M. Internal Tandem Duplication of Exon 8 of c-kit Is Associated With Longer Total Survival in Canine Cutaneous Mast Cell Tumors. Vet. Pathol. 2021, 58, 315–324. [Google Scholar] [CrossRef]
  230. Jark, P.C.; Mundin, D.B.; de Carvalho, M.; Ferioli, R.B.; A Anai, L.; Marchi, F.A.; Rogatto, S.R.; Laufer-Amorim, R.; Tinucci-Costa, M. Genomic copy number variation associated with clinical outcome in canine cutaneous mast cell tumors. Res. Vet. Sci. 2017, 111, 26–30. [Google Scholar] [CrossRef]
  231. Giantin, M.; Granato, A.; Baratto, C.; Marconato, L.; Vascellari, M.; Morello, E.M.; Vercelli, A.; Mutinelli, F.; Dacasto, M. Global Gene Expression Analysis of Canine Cutaneous Mast Cell Tumor: Could Molecular Profiling Be Useful for Subtype Classification and Prognostication? PLoS ONE 2014, 9, e95481. [Google Scholar] [CrossRef]
  232. Shubbar, E.; Kovács, A.; Hajizadeh, S.; Parris, T.Z.; Nemes, S.; Gunnarsdóttir, K.; Einbeigi, Z.; Karlsson, P.; Helou, K. Elevated cyclin B2 expression in invasive breast carcinoma is associated with unfavorable clinical outcome. BMC Cancer 2013, 13, 1. [Google Scholar] [CrossRef] [PubMed]
  233. Halasi, M.; Gartel, A.L. FOX(M1) News—It Is Cancer. Mol. Cancer Ther. 2013, 12, 245–254. [Google Scholar] [CrossRef] [PubMed]
  234. Wang, Z.; Wan, L.; Zhong, J.; Inuzuka, H.; Liu, P.; Sarkar, F.H.; Wei, W. Cdc20: A Potential Novel Therapeutic Target for Cancer Treatment. Curr. Pharm. Des. 2013, 19, 3210–3214. [Google Scholar] [CrossRef] [PubMed]
  235. Sethi, G.; Pathak, H.B.; Zhang, H.; Zhou, Y.; Einarson, M.B.; Vathipadiekal, V.; Gunewardena, S.; Birrer, M.J.; Godwin, A.K. An RNA Interference Lethality Screen of the Human Druggable Genome to Identify Molecular Vulnerabilities in Epithelial Ovarian Cancer. PLoS ONE 2012, 7, e47086. [Google Scholar] [CrossRef] [PubMed]
  236. Espinosa, A.M.; Alfaro, A.; Roman-Basaure, E.; Guardado-Estrada, M.; Palma, Í.; Serralde, C.; Medina, I.; Juárez, E.; Bermúdez, M.; Márquez, E.; et al. Mitosis Is a Source of Potential Markers for Screening and Survival and Therapeutic Targets in Cervical Cancer. PLoS ONE 2013, 8, e55975. [Google Scholar] [CrossRef]
  237. Yun, H.J.; Cho, Y.-H.; Moon, Y.; Park, Y.W.; Yoon, H.-K.; Kim, Y.-J.; Cho, S.-H.; Lee, Y.-I.; Kang, B.-S.; Kim, W.-J.; et al. Transcriptional targeting of gene expression in breast cancer by the promoters of protein regulator of cytokinesis 1 and ribonuclease reductase 2. Exp. Mol. Med. 2008, 40, 345–353. [Google Scholar] [CrossRef] [PubMed]
  238. Qiu, J.-J.; Guo, J.-J.; Lv, T.-J.; Jin, H.-Y.; Ding, J.-X.; Feng, W.-W.; Zhang, Y.; Hua, K.-Q. Prognostic value of centromere protein-A expression in patients with epithelial ovarian cancer. Tumor Biol. 2013, 34, 2971–2975. [Google Scholar] [CrossRef] [PubMed]
  239. Garnett, M.J.; Mansfeld, J.; Godwin, C.; Matsusaka, T.; Wu, J.; Russell, P.; Pines, J.; Venkitaraman, A.R. UBE2S elongates ubiquitin chains on APC/C substrates to promote mitotic exit. Nat. Cell Biol. 2009, 11, 1363–1369. [Google Scholar] [CrossRef] [PubMed]
  240. Baig, R.M.; Mahjabeen, I.; Sabir, M.; Masood, N.; Ali, K.; Malik, F.A.; Kayani, M.A. Mutational spectrum of Gelsolin and its down regulation is associated with breast cancer. Dis. Markers 2013, 34, 71–80. [Google Scholar] [CrossRef]
  241. Zhang, Y.; Liu, X.; Liu, L.; Chen, J.; Hu, Q.; Shen, S.; Zhou, Y.; Chen, S.; Xue, C.; Cui, G.; et al. Upregulation of FEN1 Is Associated with the Tumor Progression and Prognosis of Hepatocellular Carcinoma. Dis. Markers 2020, 2020, 2514090. [Google Scholar] [CrossRef]
  242. Pulz, L.H.; Barra, C.N.; Alexandre, P.A.; Huete, G.C.; Cadrobbi, K.G.; Nishiya, A.T.; de Freitas, S.H.; Fukumasu, H.; Strefezzi, R.F. Identification of two molecular subtypes in canine mast cell tumours through gene expression profiling. PLoS ONE 2019, 14, e0217343. [Google Scholar] [CrossRef] [PubMed]
  243. Kagan, H.M.; Li, W. Lysyl oxidase: Properties, specificity, and biological roles inside and outside of the cell. J. Cell. Biochem. 2003, 88, 660–672. [Google Scholar] [CrossRef] [PubMed]
  244. Kalluri, R.; Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 2006, 6, 392–401. [Google Scholar] [CrossRef]
  245. Gabbiani, G. The myofibroblast in wound healing and fibrocontractive diseases. J. Pathol. 2003, 200, 500–503. [Google Scholar] [CrossRef]
  246. Valastyan, S.; Weinberg, R.A. Tumor Metastasis: Molecular Insights and Evolving Paradigms. Cell 2011, 147, 275–292. [Google Scholar] [CrossRef] [PubMed]
  247. Daves, M.H.; Hilsenbeck, S.G.; Lau, C.C.; Man, T.-K. Meta-analysis of multiple microarray datasets reveals a common gene signature of metastasis in solid tumors. BMC Med. Genom. 2011, 4, 56. [Google Scholar] [CrossRef]
  248. Blacklock, K.B.; Birand, Z.; Biasoli, D.; Fineberg, E.; Murphy, S.; Flack, D.; Bass, J.; Di Palma, S.; Blackwood, L.; McKay, J.; et al. Identification of molecular genetic contributants to canine cutaneous mast cell tumour metastasis by global gene expression analysis. PLoS ONE 2018, 13, e0208026. [Google Scholar] [CrossRef]
  249. Hansen, C.G.; Bright, N.A.; Howard, G.; Nichols, B.J. SDPR induces membrane curvature and functions in the formation of caveolae. Nature 2009, 11, 807–814. [Google Scholar] [CrossRef]
  250. Leone, A.; Flatow, U.; VanHoutte, K.; Steeg, P.S. Transfection of human nm23-H1 into the human MDA-MB-435 breast carcinoma cell line: Effects on tumor metastatic potential, colonization and enzymatic activity. Oncogene 1993, 8, 2325–2333. Available online: http://www.ncbi.nlm.nih.gov/pubmed/8395676 (accessed on 30 October 2023).
  251. Montagner, M.; Enzo, E.; Forcato, M.; Zanconato, F.; Parenti, A.; Rampazzo, E.; Basso, G.; Leo, G.; Rosato, A.; Bicciato, S.; et al. SHARP1 suppresses breast cancer metastasis by promoting degradation of hypoxia-inducible factors. Nature 2012, 487, 380–384. [Google Scholar] [CrossRef]
  252. Chen, D.; Sun, Y.; Wei, Y.; Zhang, P.; Rezaeian, A.H.; Teruya-Feldstein, J.; Gupta, S.; Liang, H.; Lin, H.-K.; Hung, M.-C.; et al. LIFR is a breast cancer metastasis suppressor upstream of the Hippo-YAP pathway and a prognostic marker. Nat. Med. 2012, 18, 1511–1517. [Google Scholar] [CrossRef] [PubMed]
  253. Fenger, J.M.; Bear, M.D.; Volinia, S.; Lin, T.-Y.; Harrington, B.K.; A London, C.; Kisseberth, W.C. Overexpression of miR-9 in mast cells is associated with invasive behavior and spontaneous metastasis. BMC Cancer 2014, 14, 84. [Google Scholar] [CrossRef] [PubMed]
  254. Miyake, M.; Nakano, K.; Ieki, Y.; Adachi, M.; Huang, C.L.; Itoi, S.; Koh, T.; Taki, T. Motility related protein 1 (MRP-1/CD9) expression: Inverse correlation with metastases in breast cancer. Cancer Res. 1995, 55, 4127–4131. Available online: http://www.ncbi.nlm.nih.gov/pubmed/7664290 (accessed on 30 October 2023). [PubMed]
  255. Mouneimne, G.; Hansen, S.D.; Selfors, L.M.; Petrak, L.; Hickey, M.M.; Gallegos, L.L.; Simpson, K.J.; Lim, J.; Gertler, F.B.; Hartwig, J.H.; et al. Differential Remodeling of Actin Cytoskeleton Architecture by Profilin Isoforms Leads to Distinct Effects on Cell Migration and Invasion. Cancer Cell 2012, 22, 615–630. [Google Scholar] [CrossRef] [PubMed]
  256. Yang, C.; Ströbel, P.; Marx, A.; Hofmann, I. Plakophilin-associated RNA-binding proteins in prostate cancer and their implications in tumor progression and metastasis. Virchows Arch. 2013, 463, 379–390. [Google Scholar] [CrossRef] [PubMed]
  257. Akl, M.R.; Nagpal, P.; Ayoub, N.M.; Prabhu, S.A.; Gliksman, M.; Tai, B.; Hatipoglu, A.; Goy, A.; Suh, K.S. Molecular and clinical profiles of syndecan-1 in solid and hematological cancer for prognosis and precision medicine. Oncotarget 2015, 6, 28693–28715. [Google Scholar] [CrossRef]
  258. Roux, K.J.; Amici, S.A.; Fletcher, B.S.; Notterpek, L. Modulation of Epithelial Morphology, Monolayer Permeability, and Cell Migration by Growth Arrest Specific 3/Peripheral Myelin Protein 22. Mol. Biol. Cell 2005, 16, 1142–1151. [Google Scholar] [CrossRef]
  259. Brameier, M.; Herwig, A.; Reinhardt, R.; Walter, L.; Gruber, J. Human box C/D snoRNAs with miRNA like functions: Expanding the range of regulatory RNAs. Nucleic Acids Res. 2011, 39, 675–686. [Google Scholar] [CrossRef]
  260. Mileshkin, L.R.; Moore, K.N.; Barnes, E.H.; Gebski, V.; Narayan, K.; King, M.T.; Bradshaw, N.; Lee, Y.C.; Diamante, K.; Fyles, A.W.; et al. Adjuvant chemotherapy following chemoradiotherapy as primary treatment for locally advanced cervical cancer versus chemoradiotherapy alone (OUTBACK): An international, open-label, randomised, phase 3 trial. Lancet Oncol. 2023, 24, 468–482. [Google Scholar] [CrossRef]
  261. Wu, Y.-L.; Cheng, Y.; Zhou, X.; Lee, K.H.; Nakagawa, K.; Niho, S.; Tsuji, F.; Linke, R.; Rosell, R.; Corral, J.; et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): A randomised, open-label, phase 3 trial. Lancet Oncol. 2017, 18, 1454–1466. [Google Scholar] [CrossRef]
  262. Habanjar, O.; Diab-Assaf, M.; Caldefie-Chezet, F.; Delort, L. The Impact of Obesity, Adipose Tissue, and Tumor Microenvironment on Macrophage Polarization and Metastasis. Biology 2022, 11, 339. [Google Scholar] [CrossRef] [PubMed]
  263. Alegría-Torres, J.A.; Baccarelli, A.; Bollati, V.; Huang, Q.; A Vucic, E.; Brown, C.J.; Lam, W.L.; A Lewis, K.; O Tollefsbol, T.; Bartosch, C.; et al. Epigenetics lifestyle. Epigenomics 2011, 3, 267–277. [Google Scholar] [CrossRef] [PubMed]
  264. Simó-Riudalbas, L.; Esteller, M. Cancer genomics identifies disrupted epigenetic genes. Hum. Genet. 2014, 133, 713–725. [Google Scholar] [CrossRef]
  265. Kabekkodu, S.P.; Shukla, V.; Varghese, V.K.; Souza, J.D.; Chakrabarty, S.; Satyamoorthy, K. Clustered miRNAs and their role in biological functions and diseases. Biol. Rev. 2018, 93, 1955–1986. [Google Scholar] [CrossRef] [PubMed]
  266. Villa, A.; Garofalo, M.; Crescenti, D.; Rizzi, N.; Brunialti, E.; Vingiani, A.; Belotti, P.; Sposito, C.; Franzè, S.; Cilurzo, F.; et al. Transplantation of autologous extracellular vesicles for cancer-specific targeting. Theranostics 2021, 11, 2034–2047. [Google Scholar] [CrossRef] [PubMed]
  267. Jahan, S.; Mukherjee, S.; Ali, S.; Bhardwaj, U.; Choudhary, R.K.; Balakrishnan, S.; Naseem, A.; Mir, S.A.; Banawas, S.; Alaidarous, M.; et al. Pioneer Role of Extracellular Vesicles as Modulators of Cancer Initiation in Progression, Drug Therapy, and Vaccine Prospects. Cells 2022, 11, 490. [Google Scholar] [CrossRef]
  268. LeBleu, V.S.; Kalluri, R. Exosomes as a Multicomponent Biomarker Platform in Cancer. Trends Cancer 2020, 6, 767–774. [Google Scholar] [CrossRef] [PubMed]
  269. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  270. Lobb, R.J.; Lima, L.G.; Möller, A. Exosomes: Key mediators of metastasis and pre-metastatic niche formation. Semin. Cell Dev. Biol. 2017, 67, 3–10. [Google Scholar] [CrossRef]
  271. Lucchetti, D.; Tenore, C.R.; Colella, F.; Sgambato, A. Extracellular Vesicles and Cancer: A Focus on Metabolism, Cytokines, and Immunity. Cancers 2020, 12, 171. [Google Scholar] [CrossRef]
  272. Ekström, K.; Valadi, H.; Sjöstrand, M.; Malmhäll, C.; Bossios, A.; Eldh, M.; Lötvall, J. Characterization of mRNA and microRNA in human mast cell-derived exosomes and their transfer to other mast cells and blood CD34 progenitor cells. J. Extracell. Vesicles 2012, 1, 18389. [Google Scholar] [CrossRef] [PubMed]
  273. Zamboni, C.; Zamarian, V.; Stefanello, D.; Ferrari, R.; Auletta, L.; Milanesi, S.; Mauri, S.; Grieco, V.; Ceciliani, F.; Lecchi, C. Plasma small extracellular vesicles from dogs affected by cutaneous mast cell tumors deliver high levels of miR-21-5p. Front. Vet. Sci. 2023, 9, 1083174. [Google Scholar] [CrossRef]
  274. He, B.; Zhao, Z.; Cai, Q.; Zhang, Y.; Zhang, P.; Shi, S.; Xie, H.; Peng, X.; Yin, W.; Tao, Y.; et al. miRNA-based biomarkers, therapies, and resistance in Cancer. Int. J. Biol. Sci. 2020, 16, 2628–2647. [Google Scholar] [CrossRef] [PubMed]
  275. Agarwal, P.; Crepps, M.P.; Stahr, N.A.; Kretzschmar, W.P.; Harris, H.C.; Prasad, N.; Levy, S.E.; Smith, B.F. Identification of canine circulating miRNAs as tumor biospecific markers using Next-Generation Sequencing and Q-RT-PCR. Biochem. Biophys. Rep. 2021, 28, 101106. [Google Scholar] [CrossRef]
  276. Jain, M.; Ingole, S.D.; Deshmukh, R.S.; Bharucha, S.V.; Nagvekar, A.S.; Gaikwad, R.V.; Kharde, S.D. CEA, CA 15-3, and miRNA expression as potential biomarkers in canine mammary tumors. Chromosom. Res. 2021, 29, 175–188. [Google Scholar] [CrossRef]
  277. Mayoral, R.J.; Pipkin, M.E.; Pachkov, M.; van Nimwegen, E.; Rao, A.; Monticelli, S. MicroRNA-221–222 Regulate the Cell Cycle in Mast Cells. J. Immunol. 2009, 182, 433–445. [Google Scholar] [CrossRef] [PubMed]
  278. Mayoral, R.J.; Deho, L.; Rusca, N.; Bartonicek, N.; Saini, H.K.; Enright, A.J.; Monticelli, S. MiR-221 Influences Effector Functions and Actin Cytoskeleton in Mast Cells. PLoS ONE 2011, 6, e26133. [Google Scholar] [CrossRef] [PubMed]
  279. Zamarian, V.; Ferrari, R.; Stefanello, D.; Ceciliani, F.; Grieco, V.; Minozzi, G.; Chiti, L.E.; Arigoni, M.; Calogero, R.; Lecchi, C. miRNA profiles of canine cutaneous mast cell tumours with early nodal metastasis and evaluation as potential biomarkers. Sci. Rep. 2020, 10, 18918. [Google Scholar] [CrossRef]
  280. Zamarian, V.; Stefanello, D.; Ferrari, R.; Chiti, L.E.; Grieco, V.; DallaCosta, E.; Ceciliani, F.; Lecchi, C. Salivary miR-21 is a potential biomarker for canine mast cell tumors. Vet. Pathol. 2023, 60, 47–51. [Google Scholar] [CrossRef]
  281. Capodanno, A.; Boldrini, L.; Proietti, A.; Alì, G.; Pelliccioni, S.; Niccoli, C.; D’Incecco, A.; Cappuzzo, F.; Chella, A.; Lucchi, M.; et al. Let-7g and miR-21 expression in non-small cell lung cancer: Correlation with clinicopathological and molecular features. Int. J. Oncol. 2013, 43, 765–774. [Google Scholar] [CrossRef]
  282. Lee, Y.-N.; Brandal, S.; Noel, P.; Wentzel, E.; Mendell, J.T.; McDevitt, M.A.; Kapur, R.; Carter, M.; Metcalfe, D.D.; Takemoto, C.M. KIT signaling regulates MITF expression through miRNAs in normal and malignant mast cell proliferation. Blood 2011, 117, 3629–3640. [Google Scholar] [CrossRef] [PubMed]
  283. Garrett, L. Canine mast cell tumors: Diagnosis, treatment, and prognosis. Vet. Med. Res. Rep. 2014, 5, 49–58. [Google Scholar] [CrossRef]
  284. Séguin, B.; Leibman, N.F.; Bregazzi, V.S.; Ogilvie, G.K.; Powers, B.E.; Dernell, W.S.; Fettman, M.J.; Withrow, S.J. Clinical outcome of dogs with grade-II mast cell tumors treated with surgery alone: 55 cases (1996–1999). J. Am. Vet. Med. Assoc. 2001, 218, 1120–1123. [Google Scholar] [CrossRef]
  285. Dores, C.; Milovancev, M.; Russell, D. Comparison of histologic margin status in low-grade cutaneous and subcutaneous canine mast cell tumours examined by radial and tangential sections. Vet. Comp. Oncol. 2018, 16, 125–130. [Google Scholar] [CrossRef] [PubMed]
  286. Gajewska, M.; Kwiecień, I.; Rutkowska, E.; Rzepecki, P.; Sułek, K. AgNOR patterns and configurations in adult acute leukemia patients. Central Eur. J. Immunol. 2022, 47, 323–331. [Google Scholar] [CrossRef] [PubMed]
  287. Marconato, L.; Zorzan, E.; Giantin, M.; Di Palma, S.; Cancedda, S.; Dacasto, M. Concordance of c-kit Mutational Status in Matched Primary and Metastatic Cutaneous Canine Mast Cell Tumors at Baseline. J. Vet. Intern. Med. 2014, 28, 547–553. [Google Scholar] [CrossRef] [PubMed]
  288. Amagai, Y.; Tanaka, A.; Matsuda, A.; Jung, K.; Oida, K.; Nishikawa, S.; Jang, H.; Matsuda, H. Heterogeneity of internal tandem duplications in the c-kit of dogs with multiple mast cell tumours. J. Small Anim. Pract. 2013, 54, 377–380. [Google Scholar] [CrossRef] [PubMed]
  289. Bloom, F. Effect of Cortisone on Mast Cell Tumors (Mastocytoma) of the Dog. Exp. Biol. Med. 1952, 79, 651–654. [Google Scholar] [CrossRef]
  290. McCaw, D.L.; Miller, M.A.; Ogilvie, G.K.; Withrow, S.J.; Brewer, W.G.; Klein, M.K.; Bell, F.W.; Anderson, S.K. Response of Canine Mast Cell Tumors to Treatment With Oral Prednisone. J. Vet. Intern. Med. 1994, 8, 406–408. [Google Scholar] [CrossRef]
  291. Hahn, K.A.; Oglivie, G.; Rusk, T.; Devauchelle, P.; Leblanc, A.; Legendre, A.; Powers, B.; Leventhal, P.S.; Kinet, J.-P.; Palmerini, F.; et al. Masitinib is Safe and Effective for the Treatment of Canine Mast Cell Tumors. J. Veter. Intern. Med. 2008, 22, 1301–1309. [Google Scholar] [CrossRef]
  292. Rassnick, K.M.; Moore, A.S.; Williams, L.E.; London, C.A.; Kintzer, P.P.; Engler, S.J.; Cotter, S.M. Treatment of Canine Mast Cell Tumors with CCNU (Lomustine). J. Vet. Intern. Med. 1999, 13, 601. [Google Scholar] [CrossRef] [PubMed]
  293. Chun, R. Cancer Chemotherapy. In Withrow & MacEwen’s Small Animal Clinical Oncology; Elsevier: Amsterdam, The Netherlands, 2007; pp. 163–192. [Google Scholar]
  294. Hernández, I.B.; Kromhout, J.Z.; Teske, E.; Hennink, W.E.; van Nimwegen, S.A.; Oliveira, S. Molecular targets for anticancer therapies in companion animals and humans: What can we learn from each other? Theranostics 2021, 11, 3882–3897. [Google Scholar] [CrossRef]
  295. London, C.; Mathie, T.; Stingle, N.; Clifford, C.; Haney, S.; Klein, M.K.; Beaver, L.; Vickery, K.; Vail, D.M.; Hershey, B.; et al. Preliminary evidence for biologic activity of toceranib phosphate (Palladia®) in solid tumours. Vet. Comp. Oncol. 2012, 10, 194–205. [Google Scholar] [CrossRef] [PubMed]
  296. Papaetis, G.S.; Syrigos, K.N. Sunitinib. BioDrugs 2009, 23, 377–389. [Google Scholar] [CrossRef]
  297. Tozon, N.; Tratar, U.L.; Znidar, K.; Sersa, G.; Teissie, J.; Cemazar, M. Operating Procedures of the Electrochemotherapy for Treatment of Tumor in Dogs and Cats. J. Vis. Exp. 2016, 116, e54760. [Google Scholar] [CrossRef]
  298. Mir, L.M.; Gehl, J.; Sersa, G.; Collins, C.G.; Garbay, J.-R.; Billard, V.; Geertsen, P.F.; Rudolf, Z.; O’sullivan, G.C.; Marty, M. Standard operating procedures of the electrochemotherapy: Instructions for the use of bleomycin or cisplatin administered either systemically or locally and electric pulses delivered by the CliniporatorTM by means of invasive or non-invasive electrodes. Eur. J. Cancer Suppl. 2006, 4, 14–25. [Google Scholar] [CrossRef]
  299. Frimberger, A.; Moore, A.; LaRue, S.; Gliatto, J.; Bengtson, A. Radiotherapy of incompletely resected, moderately differentiated mast cell tumors in the dog: 37 cases (1989–1993). J. Am. Anim. Hosp. Assoc. 1997, 33, 320–324. [Google Scholar] [CrossRef] [PubMed]
  300. Haythornthwaite, B. What prognostic information does flow cytometry provide in canine B-cell lymphoma? Vet. Evid. 2022, 7. [Google Scholar] [CrossRef]
  301. Mason, S.L.; Pittaway, C.; Gil, B.P.; Russak, O.; Westlake, K.; Berlato, D.; Benoit, J.; Morris, J.; Dobson, J.M. Outcomes of adjunctive radiation therapy for the treatment of mast cell tumors in dogs and assessment of toxicity: A multicenter observational study of 300 dogs. J. Vet. Intern. Med. 2021, 35, 2853–2864. [Google Scholar] [CrossRef]
  302. Mellanby, R.J.; Herrtage, M.E.; Chantry, J.; Dobson, J.M. Sarcoma development after radiotherapy in two dogs. Vet. Comp. Oncol. 2003, 1, 113–119. [Google Scholar] [CrossRef]
Figure 1. Stages of MC maturation. Erythroid–myeloid progenitors and hematopoietic adult stem cells were not included in the figure. MPP—multipotent progenitor; CMP—common myeloid progenitor; GMP—granulocyte-macrophage progenitor; B/MCP—basophil and MC progenitor; MCCT—containing tryptase, chymase, and CPA in the granules; MCT—containing only tryptase in the granules; and MCCPA3—containing CPA3 carboxypeptidase in the granules.
Figure 1. Stages of MC maturation. Erythroid–myeloid progenitors and hematopoietic adult stem cells were not included in the figure. MPP—multipotent progenitor; CMP—common myeloid progenitor; GMP—granulocyte-macrophage progenitor; B/MCP—basophil and MC progenitor; MCCT—containing tryptase, chymase, and CPA in the granules; MCT—containing only tryptase in the granules; and MCCPA3—containing CPA3 carboxypeptidase in the granules.
Genes 15 00137 g001
Table 1. MC main cell membrane markers.
Table 1. MC main cell membrane markers.
MarkerTypeProtein Function
CD16
(FcγRIII, FCGR3A)
MigrationReceptor
CD32
(FcγRII, FCGR2)
MigrationReceptor
CD34Migration, Tissue specificAdhesion molecule
CD63ActivationReceptor
ENPP3 (CD203c)ActivationReceptor
FCER1 (IgE receptor, FCεRI)MaturationReceptor
ITGA4 (integrin α4, CD49d)MigrationReceptor
ITGB7 (integrin β7)Maturation, Tissue specificReceptor
KIT (CD117, c-Kit)GeneralReceptor
VCAM1 (CD106)Migration, Tissue specificReceptor
Table 2. Proteins of mastocytoma cells.
Table 2. Proteins of mastocytoma cells.
ProteinFunction
CD30/Ki-1 antigenA marker of Hodgkin’s disease and anaplastic lymphoma kinase (ALK) cells [183]. It has been shown that CD30 is expressed in human mastocytosis cells. CD30 expression in canine mastocytosis is inhibited by Il-4, which inhibits MCT cell proliferation [184].
Ki67 protein
(encoded by MKI67 gene)
The Ki67 protein is a general marker of cancer cells. Its synthesis is limited to G2 and M cell cycle phases [185]. The Ki67 protein has two isoforms (320 and 359 kDa), both of which bind and stabilize mitotic chromosomes [186]. Ki67 is an important factor for cancer development [187].
WWOX protein (oxidoreductase with a WW domain)WWOX is a tumor suppressor protein (46 kDa) in dogs [188] and mice [189]. The WWOX protein is involved in DNA repair after ionizing radiation [190]. In mastocytoma cells, the level of WWOX is always strongly reduced [188].
Histamine H1 receptor (HR1) antagonistsHR1 antagonists (loratidine, terfenadine) inhibit the proliferation and reduce the viability of mastocytoma cells. Desloraphidine, rupatadine, and cyproheptadine are particularly effective (in higher concentrations) [191].
Proliferating cell nuclear antigen (PCNA)PCNA is involved in DNA replication and DNA repair. PCNA has a PCNA-interacting protein box (PIP box) motif and forms a trimer, which slides along the DNA chain [192]. In MCT, an increased expression of PCNA is a poor prognostic factor [193].
IntegrinsMCT cells bind collagen, fibronectin, and intercellular matrix laminin via extracellular matrix (ECM) β1 and α1-6 integrins [194]. In particular, VLA5 integrins are the activators of MCT [195]. Changes in ECM are generated by cancer-associated fibroblast (CAF) cells, and the structure of ECM in mastocytoma is similar to the structure of ECM in the stroma of a healing wound [196].
Hsp32 (heme oxygenase 1)Hsp32 is synthesized by human cancerous mastocytes and dog MTCs [197]. Hsp32 protein is a cell survival factor. Its inhibitors include pegylated zinc protoporphyrin (PEG-ZnPP) and midostaurin, which inhibit tumor growth and induce apoptosis in human SM cells [198].
Hsp90The protein is a chaperone, and its levels are elevated in tumors [199]. Hsp90 is also required for cancer cell survival [200]. Geldanamycin and its derivatives are the inhibitors of Hsp90 [201]. At low concentrations, Ganetespib (STA9090) induces the apoptosis of mastocytoma cells [202].
p53The protein is expressed in mastocytoma cells, with the highest expression seen in Patnaik’s grade I and the lowest in grade II.
MCL1The anti-apoptotic protein MCL1 of the BCL2 family is present in MCT cells (independent of KIT mutations) [203]. Downregulation of MCL1 protein expression in tumors increases their susceptibility to chemotherapy. In hematopoietic U937 cells, downregulation of MCL1 via antisense strategy causes apoptosis [204].
Programmed cell death ligand 1 (PD-L1)The PD-L1 protein ligand of the programmed cell death 1 (PD-1) immunoinhibitory receptor is expressed in 66% of cases of mastocytoma and other canine cancers [205]. This is a poor prognostic factor. The PD-1 receptor causes T-cell infiltration of tumors, while PD-L1 inhibits T-cell function. Anti-PD-L1 antibodies reactivate T cells and increase IFN-γ production (also in human pancreatic cancer) [206]. The blocking of PD-L1 by antibodies can cause tumor regression [207].
Down syndrome cell adhesion molecule (DSCAM)Cell surface protein DSCAM is important for nervous system development. Mutations in humans are also associated with Down syndrome, Hirschsprung’s disease, and idiopathic scoliosis [208]. The DSCAM gene of Labrador dogs is located on chromosome 31; its mutations increase the risk of MCT up to 1.66 times. Alterations in the DSCAM gene have been described in 40 types of different cancers [209].
Proteins involved in hyaluronic acid metabolismHyaluronic acid metabolism genes (in golden retrievers) contain nucleotide polymorphisms on chromosome 14 (for HYAL4, HYALP1, and SPAM1 genes) and on chromosome 20 (for IPK1, HYAL1-3, and GNAI2 genes) [210]. Defects in hyaluronate metabolism in the skin are responsible for the risk of MCT in Shar-Pei dogs [211]. Hyaluronate breakdown products cause mast cell activation and migration [212].
Multi-drug resistance protein 1 (MDR1)The MDR1 gene encodes a glycoprotein, which is part of the pump responsible for removing foreign substances from the cell. It also removes exogenously administered drugs previously taken up by the cell. Inhibition of MDR1 gene expression may be useful in chemotherapy [213].
Table 3. Germline genetic variants in mastocytoma.
Table 3. Germline genetic variants in mastocytoma.
Gene/DNA SegmentGenetic Variant/Type of Mutation
KITGermline mutations in the KIT gene are observed infrequently, rather involving cancers other than MCT. Only KIT gene ITD mutations in exon 11 can be germline in MCT [221].
TP53TP53 mutations are present in 14.6% of MCTs [222].
GNB1In dogs, mutations in GNB1 have been found in cutaneous and subcutaneous MCTs, with a trend toward positive prognosis [223].
DSCAMGenetic variant rs850678541 inhibiting DSCAM protein synthesis [224].
Single-nucleotide polymorphisms (SNPs)SNPs in the HYAL1-4, SPAM1, and GNAI2 genes play a role in mast cell tumor development [210].
MCL1Overexpression of the myeloid target leukemia (MCL1) gene is observed in many cancers, including in mastocytoma. It encodes a labile BCL2 family protein located in the mitochondria [225].
Mitochondrial D-loop sequenceD-loop mutations in mitochondrial DNA are present in 47% of dogs with MCT and are usually homoplasmic. Six haplotypes of mitochondrial DNA sequences have been described in MCT cells [226].
Table 4. Expression of genes differing between malignant and benign forms of MCT.
Table 4. Expression of genes differing between malignant and benign forms of MCT.
GeneProduct
CCNBCyclin B is important for the transition from the G2 phase to mitosis. CCNB is an oncogene, which is important in the process of metastasis [232].
FOXM1Encodes a transcription factor with a forkhead domain. FOXM1 has high expression in proliferating tumor cells [233].
CDC20Encodes a karyokinetic spindle protein. CDC20 is an oncogene, which can initiate apoptosis [234].
CDCA8Encodes a regulator of mitosis in the centromeric CPC complex.
NUF2Encodes a protein of the NDC80 complex in the kinetochore. Silencing its expression results in apoptosis [235].
NUSAP1Encodes a karyokinetic spindle protein, which determines the survival of cancer cells [236].
PRC1Encodes a protein regulator of cytokinesis, which is present in the G2 and M phases. Overexpressed in neurons [237].
CENPPEncodes a centromere protein, plays a role in kinetochore function and during mitosis [238].
UBE2SEncodes ubiquitin-conjugating enzyme and plays a role in mitosis [239].
GSNEncodes an anti-oncogene, which plays a role in apoptosis. The only gene with lower expression in MCT and in many other cancers [240].
FEN1Encodes an endonuclease, which plays a role in DNA synthesis and is an anti-oncogene [241].
Table 5. Downregulated genes in metastatic cancers.
Table 5. Downregulated genes in metastatic cancers.
GenesProduct
Keratin genesGenes encoding keratin, mainly KRT1, KRT5, and KRT15 genes [248].
SDPRSerum deprivation response protein. SDPR is a metastasis suppressor gene [249].
NME1Co-transcription of this gene and the neighboring downstream gene (NME2) generates naturally occurring transcripts (NME1-NME2), which encode a fusion protein comprising sequences sharing identity with each individual gene product [250].
SHARP1bHLH transcription factor [251].
LIFRLeukemia inhibitory factor receptor [252].
PERPTP53 apoptosis effector. It presents low expression in mouse MCTs overexpressing miR-9 [253].
SBSN and SFNSBSN (suprabasin) and SFN (stratipin).
PSORS1C2Epithelial cell protein.
CLEC3BC-type lectin domain family 3 member B. It is an ECM biomarker protein for metastasis.
EGR1Early growth response 1 gene. Its product blocks heparanase, which increases metastasis.
CD9Encodes a four-transmembrane protein, which blocks cell motility [254].
BRAF and ADFNThe BRAF gene encodes serine/threonine kinase. The ADFN gene encodes multi-domain protein involved in signaling and the organization of cell junctions during embryogenesis. BRAF and ADFN deficiency increases cell motility.
EVL, ARHGEF10, NF2Their products activate stress fiber formation [255].
SCRIBEncodes scribble planar cell polarity protein.
PKP1 and DSPDesmosome proteins plakophilin 1 (PKP1) and desmoplakin (DSP). Their absence promotes metastasis [256].
SDC1Encodes a protein, which connects the cytoskeleton to the ECM [257].
PMP22Encodes peripheral myelin protein 22. Its overexpression decreases cell motility [258].
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

Zmorzynski, S.; Kimicka-Szajwaj, A.; Szajwaj, A.; Czerwik-Marcinkowska, J.; Wojcierowski, J. Genetic Changes in Mastocytes and Their Significance in Mast Cell Tumor Prognosis and Treatment. Genes 2024, 15, 137. https://doi.org/10.3390/genes15010137

AMA Style

Zmorzynski S, Kimicka-Szajwaj A, Szajwaj A, Czerwik-Marcinkowska J, Wojcierowski J. Genetic Changes in Mastocytes and Their Significance in Mast Cell Tumor Prognosis and Treatment. Genes. 2024; 15(1):137. https://doi.org/10.3390/genes15010137

Chicago/Turabian Style

Zmorzynski, Szymon, Aleksandra Kimicka-Szajwaj, Angelika Szajwaj, Joanna Czerwik-Marcinkowska, and Jacek Wojcierowski. 2024. "Genetic Changes in Mastocytes and Their Significance in Mast Cell Tumor Prognosis and Treatment" Genes 15, no. 1: 137. https://doi.org/10.3390/genes15010137

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