1 s2.0 S0008874924001126 Main
1 s2.0 S0008874924001126 Main
Cellular Immunology
journal homepage: www.elsevier.com/locate/ycimm
A R T I C L E I N F O A B S T R A C T
Keywords: Programmed cell death plays a relevant role in the pathogenesis of visceral Leishmaniasis. Apoptosis selects
Leishmania suitable parasites, regulating parasite density, whereas autophagy eliminates pathogens. This study aimed to
Apoptosis assess the inflammation and apoptosis in inflammatory cells and presents a unique description of the presence of
Autophagy
autophagic and apoptotic Leishmania amastigotes in naturally Leishmania-infected dogs. Fragments from seem
Pyroptosis
NETosis
ingly undamaged ear skin of sixteen Leishmania-infected dogs and seven uninfected dogs were evaluated through
histomorphometry, ultrastructural, immunohistochemical and transmission electron microscopy (TEM) analyses.
Leishmania amastigotes were present on seemingly undamaged ear skin only in clinically affected dogs. Parasite
load, morphometrical parameters of inflammation and apoptotic index of inflammatory cells were higher in
clinically affected animals and were related to clinical manifestations. Apoptotic index and morphometric pa
rameters of the inflammatory infiltrate in undamaged ear skin were positively correlated with parasite load.
Apoptotic and non-apoptotic Leishmania amastigotes were observed within neutrophils and macrophages.
Leishmania amastigotes were positive for Bax, a marker for apoptosis, by immunohistochemistry. Morphological
characteristics of apoptosis and autophagy in Leishmania amastigotes were observed only in phagocytes of
clinically affected dogs. Positive correlations were found between histomorphometry and clinical manifestations.
Our results showed that apoptosis and autophagy in Leishmania amastigotes may be related to both the increase
in parasite load and apoptotic index in inflammatory cells, and with the intensity of the inflammatory response in
clinically affected dogs. Thus, our study suggests that apoptotic and autophagy Leishmania within phagocytes
may have facilitate the survival of the parasite and it appears to play an important role in the process of
Leishmania infection.
1. Introduction its clinical characteristics, its degree of transmissibility, and its zoonotic
potential. The number of new regions endemic to CanL has grown
Leishmaniosis is still a global health problem, mainly affecting low- significantly in recent years [2]. The Canidae are considered the most
income populations worldwide, causing approximately one million in important reservoirs for zoonotic VL due to the presence of parasites in
fections and 20,000–30,000 deaths per year. Visceral leishmaniosis (VL) the skin, especially in the ears, which can infect the vector [3].
is a potentially fatal zoonosis, with an estimated incidence of Cell death is morphologically classified in six types, depending on
50,000–90,000 new cases per year in humans worldwide [1]. The World their morphophysiological and immunological characteristics: type 1)
Organization for Animal Health (OIE) recognizes the canine form of apoptosis (Programmed cell death- PCD); type 2) autophagy (PCD); type
visceral leishmaniosis (CanL) as an extremely important disease due to 3) necrosis (Accidental cell death - ACD); type 4) pyroptosis (PCD); type
* Corresponding author at: Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Av. Antônio Carlos, 6627,
Campus Pampulha, CEP: 31270-010, Belo Horizonte, Minas Gerais, Brazil.
E-mail addresses: [email protected], [email protected] (B.L.A. Verçosa).
https://doi.org/10.1016/j.cellimm.2024.104909
Received 16 September 2024; Received in revised form 30 November 2024; Accepted 6 December 2024
Available online 15 December 2024
0008-8749/© 2024 Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
5) necroptosis (PCD); and type 6) NETosis (PCD) [4,5]. Additionally, response to limited resources in the sandfly gut [31–33]. The term
these types of cell death processes can interact each other through “selfish altruism” was introduced to qualify regulated cellular death in
interconnected or even overlapping signaling pathways, and the final protozoan parasites: an altruistic phenomenon implemented for the
product is the result of the interaction of different types of cell death, entire population that is clonal [34]. Therefore, PCD appears to be an
which is closely related to the development of diseases [6], inclusive in “altruistic” mechanism for selecting suitable parasites for disease
Leishmaniosis [7]. transmission and it appears also to regulate parasite density within the
Apoptosis is morphologically characterized by generating specific host, to avoid hyperparasitism that would kill the host and thus prevent
morphological changes, such as, alteration in mitochondrial function, parasite transmission [35]. Additionally, PCD appears to maintain the
rounding of the cell, reduction in cell volume (pyknosis), condensation clonality of the parasite population, removing unsuitable cells and thus
of chromatin, and nuclear fragmentation. Membrane blebbing culmi ensuring the propagation of the fittest and most virulent cells [36].
nates in the formation of apoptotic bodies that are phagocytosed by Autophagy is a fundamental biological process that eliminates un
nearby cells [8]. In contrast, autophagy is restricted, parts of the cyto wanted cellular components and pathogens to maintain cellular ho
plasm are sequestered within double-membrane vacuoles and digested meostasis, having a relationship functionally complex with apoptosis
by lysosomal hydrolases [9,10]. However, some authors point out the and inflammatory response [37–39]. There are three forms of auto
morphological characters as insufficient to classify autophagy as death phagy: (1) autophagy mediated by chaperone, which requires interac
pathway, since its greatest characteristic is to promote cell survival tion between thermal shock protein (HSP70, heat shock protein) and
through autophagic flux [11]. altered macromolecules in the cytoplasm, to trigger the autophagic
Apoptosis or programmed cell death (PCD) is a regulated mechanism process; (2) microautophagy, in which parts of the cytoplasm are
of silent cell elimination. This phenomenon is naturally controlled, directly encompassed by lysosomes and (3) macroautophagy or auto
initiated, or inhibited by various physiological or pathological stimuli phagy, described above as a catabolic process in which a cell degrades
[12]. The apoptotic process is an intricate web of intracellular signaling and recycles its own contents and which consists of the most common
pathway that encompasses three phases: activation, execution and and best studied type [40,41].
cellular demolition that may be triggered by extrinsic or intrinsic The autophagy is a conserved process by which substrates in the
pathways [12]. This process represents a type of cellular self-destruction cytosol are transported to the lysosome via a double membrane-bound
dependent on energy, and synthesis, and degradation of proteins. As an intermediate vesicle called the autophagosome [10]. The selection of
active process, apoptosis requires ATP reserves, at least in the early autophagosomal content can occur in a relatively nonselective manner
stages, whereas necrosis occurs when ATP is depleted [13]. (known as “bulk autophagy”) or involve the tightly regulated elimina
PCD may be triggered by the extrinsic or an intrinsic apoptotic tion of individual cellular components (known as “selective auto
pathway. In the extrinsic pathway, which is triggered by death receptors phagy”), depending on the inducing factor [42].
stimulation, there are activation of caspase 8 that triggers the caspase Autophagy participates in the immune response to pathogen inva
cascade and molecules of the Bcl-2 (B-cell lymphoma-2) family, result sion. Studies show that strategies that hinder the autophagic reaction
ing in cell apoptosis [14]; Extrinsic pathway is activated through Fas predispose cells to infectious processes caused by bacteria, protozoa,
receptor (CD95/Apo-1), and by inflammatory cytokines or immune cy viruses and fungi [43–45]. It is caused probably by the multiplicity of
tokines, such as TNF and IFN-γ [15]. In the mitochondrial intrinsic actions exerted by the autophagic machinery within specialized (i.e.,
pathway, the activation of caspases occurs through the release of cyto adaptive and innate immune cells) and parenchymal cells [43,44]. This
chrome c and other pro-apoptotic proteins, primarily components of the fact can be explained by three reasons: (1) autophagy mediates quin
Bcl-2 family, and permeabilization of mitochondrial outer membrane tessential (and cell type-defining) functions in virtually all subtypes of
occurs, resulting in the release of molecules from mitochondria, such as immune cells, both at sites of hematopoiesis and in peripheral tissues
cytochrome c and activation of caspases [16]. The intrinsic pathway is [46,47]. Consequently, autophagy deficiency affects the generation,
activated by intracellular stress, as oxidative stress, and high calcium survival, maturation and effector properties of core cellular components
(Ca+) concentration in the cytoplasm. Caspase (Casp) 8 and 9 are among of innate and adaptive immunity [46–48]; (2) impaired autophagy re
initiator caspases and play a role in initial signaling events of apoptosis, sponses impair the ability of infected cells to eliminate invading path
whereas Casp 3 is an effector or executioner caspase and causes pro ogens (or components thereof) within the lysosome [43–45,48]. Finally,
teolytic cleavages [17,18]. The process is controlled by anti-apoptotic (3) cases of derailed autophagy exacerbate the organism’s response to
molecules (Bcl-2), and apoptotic ones (Bax) [19,20]. infection by altering the extinction of the inflammatory cascade, thus
Cellular apoptosis functions as a defense mechanism against viruses exacerbating the local and systemic harmful effects linked to infection
and other infectious agents, such as intracellular bacteria and some by invading pathogens [48].
protozoa [21]. Parasites interact with multiple regulatory mechanisms Autophagy is implicated in multiple aspects of processes that are
to make their host cells refractory to apoptosis during critical phases of relevant to the pathogenesis of infectious and inflammatory diseases,
infection, including cell invasion, stabilization, and pathogen growth such as, lymphocyte development, innate immune signaling and antigen
[22]. Studies revealed that Leishmania uses different survival strategies presentation [48]. Furthermore, autophagy processes are involved in
to inhibit macrophage apoptosis [23,24]. Leishmania, by preventing strategies to eliminate invasive pathogens through the production of
homooligomerization or upregulation of the Bcl-2 antiapoptotic mole reactive oxygen species, adjustment of vital nutrients and cations,
cule can impair host cell apoptosis proteins, such as Bax, which is destruction by proteolytic enzymes [49] and playing a crucial role in
required for mitochondrial permeabilization during apoptosis [25]. host-pathogen interaction, especially in intracellular pathogenesis
Evidence has demonstrated the occurrence of apoptosis in parasites [43,50].
within cells, which may represent a potential use as a tool for population The autophagy pathway is well described in Leishmania major [51],
control, which can also be generated by nutritional constraints [26,27]. Toxoplasma gondii [52], Dictyostelium discoideum [53,54], Plasmodium
Apoptosis of amastigotes was observed in hamsters during experimental falciparum [55–57], and Trypanosoma brucei [58]. Pathogen recognition-
infection with L. chagasi. Host cell apoptosis was only observed in the associated surface receptors (PRRs) such as toll-like receptors (TLRs) are
final stages of the infection, while the apoptosis process in parasites capable to associate, for example, with the beclin-1 protein and to
occurred in the early stages [28]. Some factors promote the apoptosis of induce the autophagic process. Different pro-inflammatory cytokines,
intracellular amastigotes, such as the high production of NO [29] and such as IFN-γ and TNF-α, more expressed during infection, also
hydrogen peroxide [30], both resulting from the activation of the host contribute to the activation of the autophagic pathway [59]. In macro
cell. phages infected with L. amazonensis, autophagy induces parasite pro
In Leishmania, PCD appears to regulate the parasite population in liferation. In vivo, the infection mechanism appears to be dependent on
2
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
the autophagic process, seeing an increase in LC3 expression in the autophagic pathway has also been described in metacyclogenesis, being
lesion area [60,61]. Apoptotic-like Leishmania may also uses host cell associated with the virulence of the pathogen [91,92]. However, the
autophagy machinery inhibiting T cell proliferation and favoring the apoptotic or autophagic parasite still was not shown in natural infection
overall parasite population survival [62]. in vivo. This study aimed to evaluate the apoptosis of the inflammatory
The crucial role of the autophagic pathway has been demonstrated in cells and of the Leishmania amastigotes and the inflammatory response,
apicomplexa parasites (e.g. Plasmodium sp., Toxoplasma gondii) and in and to show images of live and apoptotic and autophagic Leishmania
trypanosomatids, such as, protozoa of the genus Leishmania, T. brucei amastigotes in macrophages and neutrophils by transmission electron
and T. cruzi [63]. In pathogenic protozoa, the importance of autophagy microscopy, and to correlate these results with the parasite load and
in different processes, such as differentiation, pathogenicity and infec clinical features of stray dogs naturally infected by Leishmania.
tion, has been demonstrated. Trypanosoma cruzi, the protozoan that
causes Chagas disease, requires autophagy-mediated recruitment of ly 2. Material and methods
sosomes to the pathogen-containing vacuole to activate the necessary
virulence factors to establish infection [64]. The autophagic pathway is 2.1. Ethical issues, samples, and experimental design
a key component in the lysosomal dependent entry of Trypanosoma cruzi
into the host cell. Adult male and female stray dogs, of unknown ages, were randomly
In pyroptosis there are classical and non-classical pathways [65]. The examined for Leishmaniosis in an epidemiological study conducted by
most proteins are induced to be expressed by activation of the NF-κB the Zoonotic Disease Control Center of the city of Timon in the state of
pathway and they will form part of the inflammatory bodies in the Maranhão, Brazil. Efforts were made to avoid all unnecessary distress to
classical pathway [66]. Inflammatory bodies have a cytoplasmic pattern the dogs. All procedures concurred with the guidelines established by
recognition receptor (PRR), an adapter protein and pro-caspase-1 [67]. the local Institutional Animal Care and Use Committee, which reviewed
In the second moment, caspase-1 will cleave the N-terminal fragments of and approved this work (CETEA, Universidade Federal de Minas Gerais,
Gasdermin D (GSDMD) located in the plasma membrane, forming pores, logged under protocol number 198/2007, approved on March 27, 2008)
and consequently, leading to pyroptosis. Temporarily, the hydrolysis (Bill number 3964/97).
and activation of pro-IL-1β and pro-IL-18 will generate their pro- Sixteen positive (16/23, 69.56 %) and seven negative-tested dogs
inflammatory forms - IL-1β and IL-18, thus triggering an inflammatory were evaluated (7/23, 30.43 %). After clinical analysis, dogs were
process and immune responses [68–70]. In addition, pyroptosis can also divided into three groups: a) Eight clinically affected CanL-positive dogs
be carried out by the non-classical LPS pathway, in which Caspases- (8/23, 34.78 %), which presented clinical signs of CanL such as lym
4,5,11 are activated to cleave GSDMD to induce pyroptosis [71–73]. phadenomegaly, conjunctivitis, onychogryphosis, hepatosplenomegaly,
The caspase family of proteins has long believed to be closely related fever, skin lesions in various body regions (nose, pinna, extremities, and
only to apoptosis. However, surprisingly, caspase-8 protein can directly nail), alopecia, ulcerations, dry flaky skin, seborrhea, and discolored
cleave GSDMD to induce pyroptosis [74]. Activation of caspase-9 nose; b) Eight subclinically infected CanL positive dogs (8/23, 34.78 %),
through cleavage and activation of caspase-3 is also involved in which showed no clinical manifestation of the disease but the laboratory
pyroptosis [75]. Furthermore, the caspase-6 mediates the cleavage of tests were positive for CanL; and c) Seven CanL-negative dogs (7/23,
GSDMC [76]. 30.43 %) that showed negative clinical and laboratory test results. All of
The DNA damage and chromatin condensation are some of the the clinically affected dogs in this study presented three or more clinical
similarities between pyroptosis and apoptosis [77]. Furthermore, in manifestations of the CanL. The dogs were grouped according to the
pyroptotic cells many bleb-like protrusions appear on the surface of the Verçosa et al. [3] classification. Clinical evaluation and sample collec
cell membrane before its rupture [78]. Membrane blebbing also occurs tion for anatomopathological and sorological/parasitological analyses
during apoptosis, and caspase-3 is required for this process [78–81]. were performed concomitantly. Clinical, histopathological, and anato
However, apoptosis is considered a physiological form of death, but mopathological analyses were performed in this study.
pyroptosis can cause inflammation, activated by extracellular or intra
cellular stimulation [82], such as during infection by Leishmania [7]. 2.2. Serological and parasitological diagnosis for CanL
Unlike the explosive rupture associated with cell motility due to ne
crosis, in pyroptosis, flattening of the cytoplasm is observed due to To confirm L. infantum infection, blood samples were taken to detect
leakage of the plasma membrane [83]. Caspase inhibition does not anti-Leishmania antibodies by means of an indirect fluorescent antibody
prevent cell death caused by death receptors, and this cell death has test (IFI to Leishmaniose Visceral Canina, Bio-Manguinhos, Fiocruz, Rio
certain necrotic characteristics [84]. de Janeiro, Brasil) and enzyme-linked immunosorbent assay (ELISA,
Necroptosis has similar morphological characteristics with necrosis Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brasil). Needle aspiration and
but are regulated independently of the caspases [85]. Necrosome for culture for promastigotes in Novy-MacNeal-Nicolle (NNN) medium and
mation in necroptosis will occur under stimulation of TNF-α, which is Schneider’s liquid medium were also performed [93]. Polimerase Chain
transferred from the cytoplasm to the cell membrane or organelle Reaction (PCR) (NucleoSpin®Tissue, Macherey-Nagel, Durën, Ger
membrane to form permeable pores, destroying the integrity of the many) was performed to detect parasites in the undamaged skin, using a
membrane and thus resulting in cell death. [86–88]. target sequence of the L. donovani complex.
NETosis is characterized by the release of neutrophil extracellular
traps (NETs), which consist of decondensed chromatin associated with 2.3. Processing post-mortem samples
modified histone with bactericidal proteins from granules. During
NETosis, as in apoptosis, coordinated changes occur in the nucleus and The dogs were tranquilized with 1 % acepromazine (0.25 mg/kg IM,
cytoplasm, although the nature of the changes is different [89] and does Acepran, Univet) and anesthetized with 2.5 % sodium thiopental (25
not require caspase activation [89]. Furthermore, in NETosis there is no mg/kg IV, Thiopentax). Skin fragments from the ear region were
DNA fragmentation, as observed by the TUNEL method, and there is no collected for histological analysis. After this procedure, the animals were
exposure of phosphatidylserine before the death of the neutrophil. The euthanized with an overdose of 7.5 % sodium thiopental (75 mg/kg) for
nuclear membrane fragments into vesicles during NETosis, allowing the further clinical examination. Macroscopic abnormalities were assessed,
nuclear content to mix with the content of the granules present in the and these changes were included as parameters used in the classification
cell’s cytoplasm. But the nuclear membrane remains intact, unlike ne of animals and the subsequent distribution into study groups.
crosis [90]. After sedation with sodium thiopental, bone marrow aspirates were
In several species of the genus Leishmania, the participation of the collected for myelograms, stained with Giemsa, cultured in an NNN
3
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
culture medium enriched with Schneider’s medium, and examined different antibodies diluted in PBS in a humid atmosphere for at least 12
under an optical microscope (Olympus CX 23). Growth of the promas h. Immune expression of Caspase (Casp) 3 (Lyophilized mouse mono
tigote forms of Leishmania was observed on days 5, 8, 10, and 15. All clonal antibody CPP32, Novocastra) and Bax (Polyclonal Rabbit anti-
infected dogs (clinically affected and subclinical infected) showed pos human Bax, DAKO) antigens were assessed. Monoclonal antibodies
itive results in PCR tests and in at least one of the two parasitological were used in 1:100 dilution and a LSAB® System - HRP (Biotinylated
tests (smear stained with Giemsa and culture) in at least one of the Link Streptavidin – HRP, DAKO) was revealed with a 3.3′- dia
different organs (bone marrow, spleen, liver, skin, and lymph nodes). minobenzidine (DAB) solution (0.024 %) in PBS. The slides for negative
The animals were then euthanized with an overdose of sodium thio controls were incubated only with PBS. Counterstaining was performed
pental. After, to confirm the existence or not of infection, all Leishmania- using Harry’s hematoxylin (Sigma Chemical, USA).
infected dogs were euthanized by the Zoonotic Disease Control Center in
Teresina, PI, Brazil, according to Brazilian law (Decree n◦ 51.838 of the 2.6. Polymerase Chain Reaction (PCR)
Ministry of Health) and submitted to necropsy for sample collection. The
uninfected control animals were stray dogs from the same area, which Fragments from undamaged ear skin samples, which tested negative
were captured and euthanized for rabies control. for bacterial and fungal infection, were submitted to DNA extraction.
Prior to fixation with methyl alcohol, samples of seemingly undam The “Genomic DNA from tissue” kit (NucleoSpin®Tissue, Macherey-
aged ear skin were collected, and impression smears of the cut surface Nagel, Durën, Germany) was used, following the manufacturer’s in
were taken on clean slides to directly view the parasite [64]. After this structions. The PCR was performed with a GoTaq® Green Master Mix kit
procedure, undamaged ear skin impression smears were stained with (Promega Corporation, cat. M7122. Madison, WI. USA). It was used 1 μL
Giemsa, for Leishmania amastigotes. Skin samples were collected and of the DNA sample and 1 μL of each prime from the specific Donovani
fixed in 10 % formaldehyde buffered with 0.01 M phosphate, pH 7.4, for complex DNA sequence described by Piarroux et al. [96]. The DNA
analysis under a conventional light microscope and immunohisto samples from dog skin naturally infected with Leishmania with an
chemistry. Skin fragments were processed for histological and molecular accentuated cutaneous parasitism, as well as a L. chagasi DNA, MHOM/
analyses. Tissue (seemingly undamaged ear skin) sections (5-μm thick) BR/BH46 strain, which had been previously tested and contained 1 ng/
were stained with Hematoxylin and eosin (HE), Trichomes of Shorr (for mL, were used as PCR-generated positive controls. The DNA from un
conjunctive tissue), Giemsa Lemnert (for Leishmania amastigotes and infected dog skin was used as a negative control, along with a control
mast cells), and immunoperoxidase (for apoptotic protein - Bax and with no DNA, in which all the reaction components were present, except
Caspase 3 - and Leishmania amastigotes), and were analyzed under light the DNA. The DNA amplification was performed according to protocols
microscopy (Olympus CX23). Immunohistochemical staining was per described by Laskay et al. [97], with some alterations. The PCR-
formed by the Goodpasture method (Gram staining) and by the Grocott amplified products were analyzed through electrophoresis in non-
method (fungi staining) to discard the possibility of contamination with denaturing 5 % polyacrylamide gel in TBE buffer (89 mM Tris-Borato,
bacterial or fungal infection. 2 mM EDTA pH 8.0). Gels were transferred to a fixed solution and
Another fragment was fixed with 5 % glutaraldehyde in phosphate digitally photographed for further analysis.
buffer 0.05 M, pH 7.3, for 24 h. The fixative was then renewed after 12 h
of fixation and kept at 4 ◦ C. At the time of inclusion, 1–2 mm thick skin 2.7. Description of morphological patterns of histopathological lesions in
fragments were cut from the surface, dehydrated in an increasing series seemingly undamaged ear skin
of alcohols, and included in a plastic resin composed primarily of glycol
methacrylate (Historesine, Leica) [94]. Histological sections of 3 and 5 The composition, intensity, and distribution of the inflammatory
μm of thickness were stained with HE. response in seemingly undamaged ear skin were evaluated from sections
stained with HE. The inflammatory response was the presence of three
2.4. Immunohistochemistry to detect Leishmania amastigotes in or more inflammatory cells localized in the interstitial space. The
undamaged ear skin tissues seemingly undamaged ear skin slides were analyzed under an optical
microscope and the histopathological lesions were classified according
The Immunohistochemical technique was first used to define amas to the morphological pattern into: (a) discrete and focal: with a small,
tigote forms of Leishmania in material embedded in paraffin [95]. The isolated foci of inflammatory cells, (b) moderate and multifocal: with
slices were incubated with an anti-Leishmania chagasi polyclonal dog coalescent foci and (c) severe and diffuse: with large diffuse areas, as
antibody in a 1:100 dilution. The reaction was optimized with a LSAB® described by Solano-Gallego et al. [98].
System - HRP (Biotinylated Link Streptavidin – HRP, cat. K0675, DAKO
corporation, Carpinteria, USA), revealed with a 3.3′- diaminobenzidine 2.8. Morphometric analysis
(DAB) solution in 0.024 % PBS (Sigma Chemical, USA), and stained with
Harris hematoxylin (Sigma Chemical, USA). Fragments of dog skin The morphometric analysis was done in eight clinically affected,
infected with L. chagasi were used as positive and negative controls, and eight subclinical infected, and seven uninfected dogs in a blind assay.
were incubated with primary antibodies and only PBS, respectively. Images were digitalized in an image analyzer Kontron KS300 2.0
(extreme values of area, perimeter, and diameter of the inflammatory
2.5. Detection of Caspase 3 and Bax in undamaged ear skin fragments by lesion) and Media Cybernetics Image-Pro Plus 4.5 (cellularity, apoptotic
immunohistochemistry index in the inflammatory infiltrate, and parasite load). Cellularity of
the inflammatory infiltrate referring to the number of inflammatory
Histological slides of skin samples embedded in paraffin were cells per field. Twenty fields per animal were analyzed by morphometry,
deparaffinized in xylene and rehydrated in alcohol. After this procedure, showing a representative value according to Moro et al. [99], with
the slides were incubated with a solution containing 0.03 % hydrogen modifications. The criterion used in the selection of the samples was the
peroxide in methanol for 30 min in the dark in order to block the activity presence of inflammatory infiltrate, which, through morphometric
of endogenous peroxidase. Antigenic recovery was performed using 1.2 analysis, presented a group of three or more inflammatory cells. The cell
mg/mL Tris-HCl (pH 1.0) in a microwave oven (Sanyo, Brazil), in count of the inflammatory region was done in an area of 356,207 μm2
consecutive cycles of 10 and 5 min. After washing with 0.01 M phos per field, analyzed with an objective lens 10×, averaging out a total skin
phate buffered saline, pH 7.2 (PBS), the sections were treated with Lock area of 7,124,140 μm2.
Kit (Vector Laboratories, Inc., Burlingame, USA) and a protein block The quantification of the parasite load was performed on slides
(Dako Corporation). The slides were then incubated at 4 ◦ C with stained by immunoperoxidase under a light microscope. The 20 fields
4
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
were randomly selected, adding a total skin area of 468,752 μm2, from 3. Results
which 23,437.6 μm2 was by field.
The apoptotic index was assessed by Trichomes of Shorr, which 3.1. Leishmaniosis diagnostic and clinical analysis
shows the best characteristics of apoptotic cells. A single observer
(BLAV) performed the apoptotic cell quantification. The simultaneous In this study, the most frequent clinical manifestations of Leishmania-
presence of at least three peculiar morphological features of the process infected dogs were weight loss (7/8, 87.5 %), skin ulcerations in body
was considered: shrunken anoikic cells (cell retraction and loss of regions, except ear (7/8, 87.5 %), conjunctivitis (3/8, 37.5 %), ony
adherence between cells and basement membrane); cytoplasmatic and chogryphosis (4/8, 50.0 %), and apathy (4/8, 50.0 %). Summary of the
nuclear condensation (compression of nuclear chromatin in homoge diagnostic test by clinical groups is shown in Supplementary Table 1.
nous dense masses, aligned on the internal side of the nuclear mem
brane, sometimes forming decreasing images); nuclear fragmentation 3.2. Morphological patterns and histomorphometrical analysis of
(convolution and fragmentation of the nuclear membrane, without inflammatory response
karyorrhexis or rupture); cell fragmentation (with the formation of
apoptotic bodies). The apoptotic index (AI) was determined by the Histopathological sections of undamaged ear skin of all groups
following formula: showed inflammatory infiltrates. Inflammation was observed in all of
the evaluated dogs and was directly related to clinical signs in Leish
AI = (Ʃ#of apoptotic cells/Ʃ total#of cells) × 100
mania-infected dogs (p < 0.05, Fisher test, Supplementary Table 2). In
the clinically affected group, multifocal inflammatory infiltrate was
2.9. Detection of the apoptosis by terminal deoxynucleotidyl transferase found around the hair follicles and blood vessels, while it was focal in
(TdT)-mediated dUTP nick end labeling (TUNEL method) the subepidermal region, it was slightly diffuse in the dermis, consisting
of macrophages, lymphocytes, plasmocytes, and polymorphonuclear
Apoptotic cells were detected using a specific in situ cell death neutrophils. In subclinically infected dogs, the inflammatory infiltrate
detection kit (“Apoptag® Peroxidase in situ, Apoptosis Detection Kit”, was multifocal, perifollicular, periglandular, perivascular, and, in rare
Chemicon, Temecula, CA, USA), and the assay was performed following case, subepidermal, consisting of macrophages, lymphocytes, and plas
the protocols provided by the manufacturer. The development was then mocytes. The uninfected dogs presented focal, perifollicular, peri
performed with DAB+ (Substrate Chromogen System, DAKO Corpora vascular, and subepidermal inflammatory infiltrate, consisting of
tion, Carpinteria, USA), and stained with Methyl Green, for 3 min, at macrophages, lymphocytes, and rare plasmocytes (Fig. 1A–F).
room temperature. A negative control was performed, omitting TdT in The number of inflammatory foci, mean area, perimeter, extreme
the reaction [100]. Positive control contained in the kit was used. diameters, and cellularity of the inflammatory infiltrate in undamaged
ear skin of Leishmania-infected and uninfected dogs are shown in
2.10. Transmission Electronic Microscopy Fig. 2A–F.
Ear skin fragments were collected and processed for Transmission 3.3. Morphometric analysis
Electronic Microscopy (TEM). Initially, thick sections (1 μm) were taken
to determine the area to be sectioned for TEM analysis and examined Leishmania parasites were observed in all CanL-positive dogs by
under optical microscope. Semi-thin sections have better resolution by parasitological tests (smear stained with Giemsa and culture) in different
optical microscopy, because it favours better visualization of the organs (bone marrow, spleen, liver, skin, and lymph nodes) and the
morphology of Leishmania and its host cell [94]. The sections were made parasite load in dermis of undamaged ear skin was related to clinical
in an ultramicrotome using glass knives, placed on slides under water, manifestations (p < 0.05, Fisher test). Associations among the parasite
dried on a hot plate, and stained in HE. load, inflammation, and positive PCR results in undamaged ear skin
TEM analyses of seeming undamaged ear skin fragments of dogs fragments in all groups are shown in Supplementary Table 2. None of the
clinically affected with a high parasite load were fixed in 5 % glutaral animals in the subclinical and uninfected groups presented amastigote
dehyde in 0.1 M cacodylate buffer, pH 7.2, and processed following forms of Leishmania in the undamaged skin. Nonetheless, only in clini
standard protocols, including post-fixation in osmium tetroxide, fol cally affected CanL-positive group, Leishmania was observed in dermis of
lowed by block counterstaining with uranyl acetate and embedding in undamaged ear skin. In the clinically affected dogs, the parasite load was
Epon resin [101]. Ultrathin sections were counterstaining with lead higher than that observed in the subclinical infected and uninfected
citrate and analyzed in the FEI Tecnai G2–12 Spirit BioTwin (120 kV) animals (*p < 0,05, ANOVA and Tukey). In clinically affected dogs,
Transmission Electron Microscope. TEM was used to evaluate pro amastigote forms inside the macrophages (Fig. 3 A) and neutrophils
grammed cells death in inflammatory infiltrate, and better characterize (Fig. 3 B) were detected by an anti-Leishmania antibody, in dermis of
apoptosis and autophagy of Leishmania amastigotes. undamaged ear skin by immunohistochemistry. Neutrophil presents
segmented nuclei, a morphological characteristic that goes before
2.11. Statistical analysis NETosis. In Fig. 4, it is shown a preparation of Leishmania amastigotes in
thick sections (1 μm) stained with HE. Inflammatory cells infected with
The sample size was calculated according to the method used by Dell Leishmania amastigotes presents condensed chromatin and ruptured
et al. [102]. The power was 90 % (0.9) and α = 0.05; the standard de cytoplasmic membranes, both characteristics suggestive of pyroptosis.
viation of the variables was estimated as 20 % (0.2), and the magnitude The apoptotic index was performed in cells of the inflammatory
of the difference (d) was estimated as 40 % (0.4). The minimum number infiltrate in all of the analyzed groups. The apoptotic index of inflam
of animals per group was found to be seven. matory cells was higher in the clinically affected animals than that in
Data were submitted to the Lilliefors test for a Gaussian distribution subclinical infected and uninfected dogs (Fig. 5 A). Inflammatory cells in
and to a Cochran-Bartlett test to evaluate the homoscedasticity. Data dermis of undamaged ear skin showed morphological features of
with Gaussian distribution were analyzed by ANOVA and Tukey multi apoptosis and suggestive of pyroptosis, such as cytoplasmic and nuclear
ple comparison. Non-parametric data were analyzed by Kruskal- Wallis condensation, cell retraction, and anoikis in Leishmania-infected dogs
and Dunn tests. The Fisher test was used to test the association of vari (Fig. 5 B).
ables. To verify correlation between variables, the Pearson test was Apoptosis was confirmed by in situ fragmentation of genomic DNA
used. All statistical analyses were performed using the GraphPad Prism 5 by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick
program and were considered significant up to p < 0.05. end labeling, TUNEL reaction, within inflammatory foci in dermis of
5
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
Fig. 1. Morphological patterns of inflammatory response in the undamaged ear skin of a Leishmania-infected dog. In A, diffuse inflammatory infiltrate in the
subepidermal region and around the hair follicles and blood vessels (*) in a clinically affected dog. In B, multifocal medium perifollicular, periglandular and per
ivascular inflammatory infiltrate (*) in a subclinical infected dog. In C, focal and moderate inflammatory infiltrate consisting of macrophages, lymphocytes, plas
mocytes, and polymorphonuclear neutrophils (*) in a clinically affected dog. Shorr Trichome. In D, discrete inflammatory focus around the hair follicles and blood
vessels, composed by mononuclear cells (macrophages and lymphocytes) (*) in a subclinical infected dog. Shorr Trichome. In E, multifocal and moderate inflam
matory infiltrate consisting of macrophages, lymphocytes, plasmocytes, and polymorphonuclear neutrophils (*) in a clinically affected dog. HE. In F, Focal, peri
follicular and moderate inflammatory infiltrate mainly consisting of polymorphonuclear neutrophils (yellow arrow) and macrophages (blue arrow) in a clinically
affected dog with high parasite load. Bar = 64 μm (A and B), 32 μm (C, E and F) and 16 μm (D). f = hair follicles; b = blood vessels; ep = epidermis; g = glands. (For
interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
undamaged ear skin. Positive cells showed brownish intranuclear uninfected dogs, as shown in Supplementary Table 3.
staining. Apoptosis was confirmed by the observation of inflammatory
cells with similar morphological features, presenting brownish intra
nuclear clusters, assessed by the TUNEL method in inflammatory infil 3.4. Transmission Electronic Microscopy (TEM)
trate foci (Fig. 5 C).
Inflammatory cells in dermis of undamaged ear skin showed positive Ultrastructurally, in clinically affected dogs, apoptotic inflammatory
immunostaining by Casp 3 and Bax in Leishmania-infected dogs. In cells in dermis of undamaged ear skin were shrunk, with condensed
flammatory cells (macrophage and neutrophils) showed intense cyto nuclear chromatin and cytoplasm. Condensed nuclei were frequently
plasmic expression of Caspase 3 and Bax (Fig. 5 D and E, respectively). fragmented. Leishmania amastigotes were observed within inflammatory
Peculiar morphological features of apoptosis (anoikis, chromatin cells (neutrophils and macrophages). Representative images of cells in
condensation, nuclear fragmentation, and nuclear retraction) were dermis presenting shrunken anoikic cells, cytoplasmic and nuclear
associated with a strong expression of Bax in mononuclear cells and condensation, and nuclear convolution and fragmentation in the un
neutrophils (Fig. 5 F). In addition to the inflammatory cells, it was damaged ear skin slides of Leishmania-infected dogs by TEM are shown
observed that, in most cases, the apoptotic Leishmania were also labeled in Fig. 7. Fig. 7 shows ultrastructural morphological pattern suggestive
for Bax (Fig. 6). of pyroptosis (chromatin condensation, segmented nuclei, ruptured
Apoptotic index of inflammatory cells and morphometric parameters cytoplasmic membrane, and nuclear retraction) in inflammatory cells
of the inflammatory infiltrate in dermis of undamaged ear skin were (neutrophil). In addition, Leishmania amastigotes also show ultrastruc
correlated with parasite load in the ear skin of Leishmania-infected and tural morphological pattern of apoptosis. In clinically affected dogs with
a high parasite load, both apoptotic and non-apoptotic Leishmania
6
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
Fig. 2. The morphometric parameters of inflammatory infiltrate in undamaged ear skin fragments of CanL-positive dogs. Samples were obtained from eight clinically
affected, eight subclinical infected, and seven healthy uninfected dogs (controls). In A, the number of inflammatory foci in clinically affected dogs was higher than in
the other groups and subclinical infected dogs was higher than in uninfected groups. The average area (B), perimeter (C) and major diameter (D) of the inflammatory
infiltrate in clinically affected dogs was higher than that of the uninfected group. The minor diameter (E) and cellularity (F) of the inflammatory infiltrate in clinically
affected dogs were higher than in the other groups. *ANOVA and Tukey test p < 0.05.
amastigotes were observed within the same non-apoptotic macrophage peripheral chromatin in apoptotic Leishmania amastigote. Small blebs
and neutrophil. Apoptosis and autophagy in Leishmania amastigotes were detected attached to the cell membrane. In detail, see the cyto
were observed only in clinically affected dogs with a high parasite load. plasmic membrane intact. All these alterations are morphological
Representative images of ultrastructural morphological pattern of characteristic of apoptosis.
autophagy and apoptosis of Leishmania amastigotes in undamaged ear
skin of a clinically affected dog with a high parasite load are shown in 4. Discussion
Fig. 8 A and B, respectively. Fig. 8 A shows ultrastructural morpholog
ical pattern of autophagic Leishmania amastigote presenting appearance To the best of our knowledge, this is the first report of the occurrence
of electron-lucent areas in the cytoplasm. Intense cytoplasm vacuoliza of autophagic and apoptotic amastigotes in natural infection in vivo in
tion and the presence of structures resembling autophagosomes con Leishmania-infected dogs. This study assessed cellular death (PCD),
taining membrane profiles in Leishmania amastigotes, which are main autophagy and inflammation in undamaged ear skin of clinically
morphological characteristic of autophagy processes. The cytoplasm affected and subclinical naturally Leishmania-infected dogs and showed
seeming to be degraded, presenting areas of cytoplasm extraction is apoptotic amastigote forms of Leishmania infantum inside macrophages
shown in Fig. 8 A. In detail, see the presence of nuclear vacuolization. In and neutrophils with and without morphological characteristics of
Fig. 8 B, observe the cytoplasmatic and nuclear condensation and apoptosis. Our results demonstrate that: (a) Inflammation was observed
7
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
Fig. 3. Parasite in undamaged ear skin fragments in CanL. In A and B, focal and intense inflammatory infiltrate consisting of macrophages and polymorphonuclear
neutrophils. Leishmania amastigotes are shown, stained in brown, inside the macrophage (A) and inside the neutrophil (B) by imunoperoxidase in a clinically affected
dog with high parasite load. In detail, please, note that most of Leishmania amastigotes are in cytoplasm of macrophages and neutrophil (black arrows). In addition,
observe that in this neutrophil the nuclei appears segmented, a phenomenon that mainly occur in PMN before NETosis. (red arrow). Imunoperoxidase. Bars = 125
μm. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
in all the evaluated dogs and was directly related to clinical signs in
Leishmania-infected dogs. (b) The parasite load and apoptotic index of
inflammatory cells was higher in clinically affected dogs than the other
groups and was related to clinical manifestations. (c) Macrophage and
neutrophils showed intense cytoplasmic expression of caspase 3 and
Bax. (d) Apoptotic Leishmania were labeled for Bax. (e) Apoptotic index
of inflammatory cells and morphometric parameters of the inflamma
tory infiltrate in dermis of undamaged ear skin were positively corre
lated with parasite load. (f) Inflammatory cells (neutrophil) showed
ultrastructural morphological pattern of apoptosis (chromatin conden
sation, segmented nuclei, ruptured cytoplasmic membrane, chromatin
at the periphery of the nucleus, and nuclear retraction) by TEM; in
addition, morphological characteristics of pyroptosis and NETosis were
also observed in inflammatory cells, and (g) Leishmania amastigotes
showed ultrastructural morphological pattern of apoptosis and auto
phagy and were observed only in clinically affected dogs.
Our data showed ultrastructural morphological patterns of apoptosis
and autophagy in Leishmania amastigotes observed in infected dogs with
a high parasite load and enhanced apoptotic index in inflammatory cells
Fig. 4. Thick sections (1 μm) of undamaged ear skin fragments of clinically in the undamaged ear skin of clinically affected dogs. Only one in vitro
affected dog with CanL. Perifollicular and perivascular inflammatory response study conducted using TEM showed the presence of promastigote forms
(*) consisting of mononuclear cells (M) associated with intense parasite load. of Leishmania major with intact structures, which was viable inside PMN
Leishmania amastigotes are shown inside the macrophage in apoptosis (white cells and, the phagocytosis by macrophages of apoptotic neutrophils
arrow). However, only by the morphological aspect of the cell death, it is not [103]. Furthermore, it has been demonstrated that phagocytosis of
possible to exclude that the mechanism of the death was by pyroptosis, so that it apoptotic Leishmania induces the secretion, by macrophages, of anti-
is better to consider the possibility of apoptosis/pyroptosis. Semi-thin sections
inflammatory molecules, such as IL-10 and TGF-β, or lipids, such as
have better resolution when it is assessed by optical microscopy. HE. Bar =
lipoxin A4. In addition, it induces inhibition of the secretion of pro-
30 μm.
8
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
Fig. 5. The graph in A shows that the apoptotic index in undamaged ear skin fragments was higher in clinically affected dogs than in subclinical infected and
uninfected groups. *ANOVA and Tukey test p < 0,05. Samples were obtained from eight clinically affected, eight subclinical infected, and seven healthy uninfected
dogs (controls). Apoptotic inflammatory cells were quantified in HE stained slides, considering the simultaneous presence of at least three cells with peculiar
morphological features of apoptosis. It should be noted that for cells evaluated only by its morphological aspect, it cannot be excluded that some of these cells might
have presented the mechanisms of pyroptosis for cell death. In B, inflammatory cells, macrophage, show morphological features of apoptosis, such as cytoplasmic and
nuclear condensation, cell retraction, and anoikis (In detail, green arrow) in a Leishmania-infected dog. Shorr trichome. Please note in C that cells in apoptosis in
inflammatory infiltrate foci characterized by morphological features (cytoplasmic and nuclear condensation, cell retraction, and anoikis) present also brownish
intranuclear clusters (In detail, black arrow), the apoptosis marker by staining with the TUNEL method for in situ fragmentation of genomic DNA. In D, inflammatory
cells show positive immunostaining for Caspase 3 in a Leishmania-infected dog. Macrophage (red arrow) and neutrophil (black arrow) show intense cytoplasmic
expression of caspase 3. Imunoperoxidase. In E, the apoptotic macrophage shows a positive immunostaining for Bax (In detail, black arrow) in a Leishmania-infected
dog. Imunoperoxidase. In F, apoptotic inflammatory cells (macrophages and neutrophils) show intense cytoplasmic expression of Bax. Morphological features of
apoptosis (anoikis, chromatin condensation, nucleolar fragmentation, and nuclear retraction) are associated with a strong expression of Bax in mononuclear cells (In
detail, black arrow) and neutrophils (In detail, blue arrow). Imunoperoxidase. Bars = 10 μm (D); 12 μm (C); 16 μm (B, F) 32 μm; and (C and E). f = hair follicles; b =
blood vessel. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
inflammatory cytokines, such as TNF-α, and lipids, such as leukotriene parasite, resulting in the progression of infection and disease evolution.
B4 [104,105]. In case of to eliminate apoptotic parasites from a virulent PCD was detected in inflammatory foci in all studied groups. How
population, Leishmania do not survive within phagocytic cells in vitro ever, the apoptotic index associated with the expression of the pro-
and lose their ability to induce disease in vivo [104,105]. Phagocytosis apoptotic molecules in inflammatory cells, Bax and Casp 3, was
of apoptotic neutrophils with Leishmania also appears to decrease the enhanced only in clinically affected dogs, suggesting that the increase of
presentation of parasitic antigens by macrophages [106]. In addition, molecules that induce apoptosis in epithelial tissue may play a role in the
apoptotic cells activate the autophagic machinery of host cells, in a severity of the disease through the increase in the parasite load. HSP65
process known as LC3-associated phagocytosis, promoting an anti- expression in L. major-infected macrophages causes a dysfunction in the
inflammatory response with the production of IL-10 and TGF-β, hin control of the apoptosis mechanism [107]. Furthermore, the survival of
dering the production of pro-inflammatory cytokines, such as TNF-α, IL- Leishmania inside the cell’s host depends on the decrease of caspase-3
1β and IL-6, reducing T cell proliferation and thus favoring parasite expression, supposedly because the parasite interferes with the trans
survival [62]. Thus, apoptotic Leishmania altruistically allow the intra formation of the pro-caspase 3 enzyme into its active form [104]. Aga
cellular survival of the fittest parasites. So that, it is possible that et al. [108] showed that the infection of neutrophils with L. major pro
apoptotic Leishmania within phagocytes facilitates the survival of the mastigotes decreased caspase-3 activity and thus inhibited the
9
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
Fig. 6. Apoptotic Leishmania show intense cytoplasmic expression of Bax by immunohistochemistry. Please note in detail that a strong cytoplasmatic expression of
Bax was observed in a Leishmania amastigote (black arrow) that is not inside any cell. Imunoperoxidase. Bar = 125 μm.
10
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
Fig. 8. Ultrastructural morphological pattern of autophagy and apoptosis in Leishmania amastigotes. Ultrastructural morphological pattern of autophagy (A) and
apoptosis (B) in Leishmania amastigotes in undamaged ear skin of a clinically affected dog with a high parasite load. Please, note in A the characteristic aspect of
autophagy, which is a conserved process by which substrates in the cytosol are transported to the lysosome via a double membrane-bound intermediate vesicle called
the autophagosome. In A, observe the cytoplasm vacuolization (*) and the presence of structures resembling autophagosomes (p) and autolissosomes. The cytoplasm
seems to be degraded, presenting areas of cytoplasm extraction (+). Leishmania amastigotes present appearance of electron-lucent areas in the cytoplasm (*). In
detail, see the cytoplasmic and nuclear condensation and the presence of nuclear vacuolization (n) in the Leishmania. In B, observe the Leishmania with characteristics
of apoptosis: the cytoplasmic and nuclear condensation and peripheral chromatin in Leishmania nuclei (n). Small bleb is observed attached to the cell membrane of
Leishmania (in detail, blue arrow). Please, note that the cytoplasmic membrane of the Leishmania is intact (in detail, black arrow). Bars = 1 μm (A) and 2 μm (B). L =
Leishmania; N = Cell nuclei; n = Leishmania nuclei; K = kinetoplast and f = flagella. Transmission Electronic Microscopy. (For interpretation of the references to
colour in this figure legend, the reader is referred to the web version of this article.)
9, thereby causing cell apoptosis [117]. Evaluation by PCR and Western depends on the level of GSDME expression. When GSDME expression is
blot confirmed proper integration and expression of mLLO-Bax-Smac high, it results in pyroptosis, when expression is low, it converts to
sequence into the 18srRNA locus of L. infantum. By flow cytometry it apoptosis [125]. Thus, GSDME could be a possible biomarker and
was shown accelerated apoptosis of transgenic Leishmania-infected therapeutic target to regulate pro- and anti-inflammatory responses
macrophages compared to the wild-type parasite. In addition, transgenic related to Leishmaniasis, since its expression regulates the change be
parasites were less virulent, and a fewer parasitic burden was found in tween pyroptosis (pro-inflammatory) and apoptosis (anti-
the spleen and liver of transgenic-infected mice compared to the control inflammatory).
group [118]. Additionally, the Bec-1 protein, classified as a BH3-only Our ultrastructural results showed cytoplasm vacuolization and the
protein, interacts with Bcl-2. This interaction inhibits Bec1-dependent presence of structures resembling autophagosomes containing mem
autophagy but does not inactivate the antiapoptotic effect of Bcl-2. brane profiles in Leishmania amastigotes only in clinically affected dogs
Under normal conditions, Bec-1 is inhibited and bound by Bcl-2, how (Fig. 8A). In Leishmania, autophagy is critical for maintaining cellular
ever, for autophagy to occur, dissociation between Bec-1 and Bcl-2 must homeostasis, recycling damaged organelles, glycosome renewal through
occur [119]. Finally, in Bec1-deficient cells, the expression of Bec-1 the Atg5 protein [126] and protein turnover [127]. Although the exact
mutants that do not bind to Bcl-2, in addition to inducing more auto autophagic mechanism has not been deciphered yet, numerous studies
phagy, also promote cell death, unlike the expression of wild-type Bec-1. point to a crucial role of autophagy and the protein it produces in the
Thus, Bcl-2, in addition to acting as an antiapoptotic protein, also differentiation and infectivity of parasites [91,128]. Due to its signifi
functions as an antiautophagic protein, where it has an active role in cant influence on cell fate, autophagy still dominates many cellular
maintaining autophagy compatible with cell survival [120]. processes and signaling networks [128]. Due to the complexity of the
It could be observed in Fig. 7 that the nucleus of the neutrophil ap autophagic and immunological signaling mechanism in parasitic dis
pears segmented and the cytoplasmic membrane was ruptured, that are eases associated with the growing number of studies related to the topic,
morphological characteristics of pyroptosis. Different types of cell death it is suggested that the Leishmania autophagy process can be a successful
can interact each other, since signaling pathways are interconnected or therapeutic approach for the treatment and, subsequently, cure for
even overlapping, and the result of this relationship is closely related to Leishmaniasis [91,127,129].
the development of diseases, including Leishmaniasis. [5,6,121]. Gas In this work, the clinically affected dogs presented higher morpho
dermin E (GSDME) is an important and critical point of intersection metric parameters of the inflammatory infiltrate associated with PCD
between the processes of apoptosis and pyroptosis, acting in the con rates and parasite loads. This could result in high levels of MIP-1β
version between these two types of cell death [122]. Jiang et al. [123] secreted by infected apoptotic PMNs, the cytokine responsible for
showed that there is a type of pyroptosis caused by the connection or recruiting macrophages to the area of infection, lead to contact and
overlapping expression of GSDME and caspase-3. Caspase-3 specifically further multiplication of the parasite in the host cell [103]. Furthermore,
cleaves GSDME at the linker domain to generate N-terminal fragments of high levels of TGF-β and PGE2 may influence apoptosis, the inflamma
GSDME, forming pores and thus perforating plasma membranes to tory response, and the parasite load, as observed in L. amazonensis cul
induce pyroptosis, rather than apoptosis [124]. However, this fact tures. Just after apoptotic PMNs have been added in cultures, an
11
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
increase in the levels of TGE-β and PGE2 occurred, which further ex Declaration of competing interest
acerbates the parasite replication process [130].
Leishmania amastigotes were observed, in this study, in neutrophils The authors declare the following financial interests/personal re
and macrophages, with and without apoptosis morphology, and only in lationships which may be considered as potential competing interests:
clinically affected dogs, which showed high parasite loads, associated Anilton Cesar Vasconcelos reports financial support and equipment,
with a high apoptotic index. Afonso et al. [130] observed an increase in drugs, or supplies were provided by Minas Gerais State Foundation of
infected cells and parasite load after apoptotic PMNs were added to a Support to the Research (FAPEMIG). All authors read and approved the
human culture of macrophages infected with L. amazonensis. In addition, manuscript.
parasite load increased after the clearance of apoptotic PMN by mac
rophages, as Ribeiro-Gomes et al. [131] observed in mice infected with Data availability
L. major. Furthermore, many studies have confirmed the possibility of
PMNs actively to participate in the process of infection by Leishmania No data was used for the research described in the article.
[132].
Leishmania spp. induces an inflammatory response in the skin, of Acknowledgments
variable distribution and intensity [3]. The skin of clinically affected
dogs presented amastigote forms and inflammatory infiltrate containing The study was supported by the Fundação de Amparo a Pesquisa do
macrophages, lymphocytes, plasmacytes, and neutrophils, while the Estado de Minas Gerais - FAPEMIG, Coordenação de Aperfeiçoamento
subclinically infected animals presented a profile similar to the unin de Pessoal de Nível Superior-CAPES (Scholarship to BLAV), and Con
fected controls, as observed in the present study. Accordingly, as related selho Nacional de Desenvolvimento Científico e Tecnológico - CNPq
by Xavier et al. [133] and Giuguetti [134], no granulomatous lesions (Fellowships to MNM, AVC and MIM-J). We would like to acknowledge
were found in any animal, as it was also described by Solano-Galleno the Laboratory of Histopathology at the Department of General Pa
et al. [98] and Dos-Santos et al. [135]. thology and Laboratory of Apoptosis of Universidade Federal de Minas
For the parasites to survive in the early stages of infection, the host Gerais for their histopathologic preparations. We would also like to
cells should not be properly activated. Through phagocytosis of thank the employees of the Centro de Controle de Zoonoses (CCZ),
apoptotic inflammatory cells containing parasites, the amastigotes do Timon, Maranhao, Brazil, for supporting us with the collection of
not contact the phagocyte receptors; thus, phagocytic cells are not samples.
effectively activated, facilitating the infection and suppressing the
effective immune response to Leishmania. Our results indicate that Appendix A. Supplementary data
apoptosis is related to the increase in parasite load and to the intensity of
the inflammatory response. Association between these histo Supplementary data to this article can be found online at https://doi.
morphometry evaluations (inflammation, parasite load, and apoptosis) org/10.1016/j.cellimm.2024.104909.
and clinical manifestations was also observed. Our results show that the
apoptotic inflammatory cells and Leishmania amastigotes were labeled References
by Bax, which is a pro-apoptotic protein. Finally, our data showed ul
trastructural morphological patterns of apoptosis and autophagy in [1] World Health Organization (WHO), Leishmaniases, Disponible in, https://www.
who.int/news-room/fact-sheets/detail/leishmaniasis, 2023 (accessed 12
Leishmania amastigotes with a high parasite load and enhanced September 2024).
apoptotic index in inflammatory cells in the undamaged ear skin of [2] World Organization for Animal Health – OIE, Leishmaniosis [online]. htt
clinically affected dogs. So that, it is possible that apoptotic and auto ps://www.oie.int/fileadmin/Home/eng/Health_standards/tahm/3.01.11_LEIS
HMANIOSIS, 2018 (accessed 12 September 2024).
phagic Leishmania within phagocytes facilitate the survival of the [3] B.L. Verçosa, C.M. Lemos, I.L. Mendonça, S.M. Silva, S.M. de Carvalho, H. Goto,
parasite and plays an important role in the process of infection and F.A. Costa, Transmission potential, skin inflammatory response, and parasitism of
progression of the parasite load and disease evolution in the undamaged symptomatic and asymptomatic dogs with visceral leishmaniasis, BMC Vet. Res.
45 (2008), https://doi.org/10.1186/1746-6148-4-45, 6–4.
ear skin of dogs naturally infected with Leishmania. [4] W. Park, S. Wei, B.S. Kim, B. Kim, S.J. Bae, Y.C. Chae, D. Ryu, K.T. Ha, Diversity
The activation and overlap of different types of cell death is a com and complexity of cell death: a historical review, Exp. Mol. Med. 55 (2023)
mon means for hosts to combat disease [136]. Other studies are neces 1573–1594, https://doi.org/10.1038/s12276-023-01078-x.
[5] S.L. Kerr, C. Mathew, R. Ghildyal, Rhinovirus and cell death, Viruses 13 (2021)
sary to determine the interaction between signaling pathways in the
629, https://doi.org/10.3390/v13040629.
different mechanisms of cell death. It is essential to explore the molec [6] N. Ketelut-Carneiro, K.A. Fitzgerald, Apoptosis, pyroptosis, and necroptosis—Oh
ular mechanism that regulates the process of apoptosis, pyroptosis and my! The many ways a cell can die, J. Mol. Biol. 434 (2022) 167378, https://doi.
NETosis in Leishmaniasis and the interrelationship each other. This org/10.1016/j.jmb.2021.167378.
[7] J.C.R. Fernandes, D.S. Zamboni, Mechanisms regulating host cell death during
would allow a major advance in the understanding of the host’s defense Leishmania infection, mBio 15 (2024) e01980-23, https://doi.org/10.1128/
immunological mechanisms, the pathogenesis, prevention and clinical mbio.01980-23).
evolution of Leishmaniasis. This understanding will allow the identifi [8] D. Tang, R. Kang, T.V. Berghe, P. Vandenabeele, G. Kroemer, The molecular
machinery of regulated cell death, Cell Res. 29 (2019) 347–364, https://doi.org/
cation of new therapeutic targets, providing new strategies for the 10.1038/s41422-019-0164-5.
control and treatment of Leishmaniasis. [9] G. Kroemer, M. Jäättelä, Lysosomes and autophagy in cell death control, Nat.
Rev. Cancer 5 (2005) 886–897, https://doi.org/10.1038/nrc1738.
[10] L. Galluzzi, E.H. Baehrecke, A. Ballabio, P. Boya, J.M. Bravo-San Pedro,
CRediT authorship contribution statement F. Cecconi, A.M. Choi, C.T. Chu, P. Codogno, M.I. Colombo, A.M. Cuervo,
J. Debnath, V. Deretic, I. Dikic, E.L. Eskelinen, G.M. Fimia, S. Fulda, D.A. Gewirtz,
Barbara Laurice Araújo Verçosa: Writing – original draft, Investi D.R. Green, M. Hansen, J.W. Harper, M. Jäättelä, T. Johansen, G. Juhasz, A.
C. Kimmelman, C. Kraft, N.T. Ktistakis, S. Kumar, B. Levine, C. Lopez-Otin,
gation, Formal analysis, Data curation, Conceptualization. Maria Ima F. Madeo, S. Martens, J. Martinez, A. Melendez, N. Mizushima, C. Münz, L.
culada Muniz-Junqueira: Writing – review & editing. Ana Lys Bezerra O. Murphy, J.M. Penninger, M. Piacentini, F. Reggiori, D.C. Rubinsztein, K.
Barradas Mineiro: Data curation. Maria Norma Melo: Methodology, M. Ryan, L. Santambrogio, L. Scorrano, A.K. Simon, H.U. Simon, A. Simonsen,
N. Tavernarakis, S.A. Tooze, T. Yoshimori, J. Yuan, Z. Yue, Q. Zhong, G. Kroemer,
Investigation, Formal analysis. Anilton Cesar Vasconcelos: Validation,
Molecular definitions of autophagy and related processes, EMBO J. 36 (2017)
Resources, Project administration, Investigation, Funding acquisition, 1811–1836, https://doi.org/10.15252/embj.201796697.
Formal analysis, Conceptualization. [11] D.J. Klionsky, K. Abdelmohsen, A. Abe, M.J. Abedin, H. Abeliovich, A. Acevedo
Arozena, et al., Guidelines for the use and interpretation of assays for monitoring
autophagy (3rd edition), Autophagy 12 (2016) 1–222, https://doi.org/10.1080/
15548627.2015.1100356.
12
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
[12] S. Elmore, Apoptosis: a review of programmed cell death, Toxicol. Pathol. 35 [41] B. Loos, A.M. Engelbrecht, R.A. Lockshin, D.J. Klionsky, Z. Zakeri, The variability
(2007) 495–516, https://doi.org/10.1080/01926230701320337. of autophagy and cell death susceptibility: unanswered questions, Autophagy 9
[13] Y. Kushnareva, D.D. Newmeyer, Bioenergetics and cell death, Ann. N. Y. Acad. (2013) 1270–1285, https://doi.org/10.4161/auto.25560.
Sci. 1201 (2010) 50–57, https://doi.org/10.1111/j.1749-6632.2010.05633.x. [42] C. He, D.J. Klionsky, Regulation mechanisms and signaling pathways of
[14] C. Garrido, L. Galluzzi, M. Brunet, P.E. Puig, C. Didelot, G. Kroemer, Mechanisms autophagy, Annu. Rev. Genet. 43 (2009) 67–93, https://doi.org/10.1146/
of cytochrome c release from mitochondria, Cell Death Differ. 13 (2006) annurev-genet-102808-114910.
1423–1433, https://doi.org/10.1038/sj.cdd.4401950. [43] B. Levine, N. Mizushima, H.W. Virgin, Autophagy in immunity and inflammation,
[15] E. Obeng, Apoptosis (programmed cell death) and its signals - a review, Braz. J. Nature 469 (2011) 323–335, https://doi.org/10.1038/nature09782.
Biol. 81 (2021) 1133–1143, https://doi.org/10.1590/1519-6984.228437. [44] L.C. Gomes, I. Dikic, Autophagy in antimicrobial immunity, Mol. Cell 54 (2014)
[16] J.E. Belizário, J. Alves, J.M. Occhiucci, M. Garay-Malpartida, A. Sesso, 224–233, https://doi.org/10.1016/j.molcel.2014.03.009.
A mechanistic view of mitochondrial death decision pores, Braz. J. Med. Biol. Res. [45] M.D. Keller, V.J. Torres, K. Cadwell, Autophagy and microbial pathogenesis, Cell
40 (2007) 1011–1024, https://doi.org/10.1590/s0100-879x2006005000109. Death Differ. 27 (2020) 872–886, https://doi.org/10.1038/s41418-019-0481-8.
[17] A.B. Parrish, C.D. Freel, S. Kornbluth, Cellular mechanisms controlling caspase [46] Y. Ma, L. Galluzzi, L. Zitvogel, G. Kroemer, Autophagy and cellular immune
activation and function, Cold Spring Harb. Perspect. Biol. 5 (2013) a008672, responses, Immunity 39 (2013) 211–227, https://doi.org/10.1016/j.
https://doi.org/10.1101/cshperspect.a008672. immuni.2013.07.017.
[18] R. Jan, G.E. Chaudhry, Understanding apoptosis and apoptotic pathways targeted [47] A.J. Clarke, A.K. Simon, Autophagy in the renewal, differentiation and
cancer therapeutics, Adv. Pharm. Bull. 9 (2019) 205–218, https://doi.org/ homeostasis of immune cells, Nat. Rev. Immunol. 19 (2019) 170–183, https://
10.15171/apb.2019.024. doi.org/10.1038/s41577-018-0095-2.
[19] J.K. Brunelle, A. Letai, Control of mitochondrial apoptosis by the Bcl-2 family, [48] V. Deretic, Autophagy in inflammation, infection, and immunometabolism,
J. Cell Sci. 122 (2009) 437–441, https://doi.org/10.15171/apb.2019.024. Immunity 54 (2021) 437–453, https://doi.org/10.1016/j.immuni.2021.01.018.
[20] A. Shamas-Din, J. Kale, B. Leber, D.W. Andrews, Mechanisms of action of Bcl-2 [49] J.E. Galán, P. Cossart, Host-pathogen interactions: Adiversity of themes, a variety
family proteins, Cold Spring Harb. Perspect. Biol. 5 (2013) a008714, https://doi. of molecular machines, Curr. Opin. Microbiol. 8 (2005) 1–3, https://doi.org/
org/10.1101/cshperspect.a008714. 10.1016/j.mib.2004.12.015.
[21] D.L. Vaux, G. Haecker, A. Strasser, An evolutionary perspective on apoptosis, Cell [50] H. Zuo, C. Chen, Y. Sa, Therapeutic potential of autophagy in immunity and
76 (1994) 777–779, https://doi.org/10.1016/0092-8674(94)90350-6. inflammation: current and future perspectives, Pharmacol. Rep. 75 (2023)
[22] J.C. Carmen, A.P. Sinai, Suicide prevention: disruption of apoptotic pathways by 499–510, https://doi.org/10.1007/s43440-023-00486-0.
protozoan parasites, Mol. Microbiol. 64 (2007) 904–916, https://doi.org/ [51] V. Guhe, P. Ingale, A. Tambekar, S. Singh, Systems biology of autophagy in
10.1111/j.1365-2958.2007.05714.x. leishmanial infection and its diverse role in precision medicine, Front. Mol.
[23] P. Gupta, S. Srivastav, S. Saha, P.K. Das, A. Ukil, Leishmania donovani inhibits Biosci. 10 (2023) 1113249, https://doi.org/10.3389/fmolb.2023.1113249.
macrophage apoptosis and pro-inflammatory response through AKT-mediated [52] S. Besteiro, Which roles for autophagy in Toxoplasma gondii and related
regulation of β-catenin and FOXO-1, Cell Death Differ. 23 (2016) 1815–1826, apicomplexan parasites? Mol. Biochem. Parasitol. 184 (2012) 1–8, https://doi.
https://doi.org/10.1038/cdd.2016.101. org/10.1016/j.molbiopara.2012.04.001.
[24] S.G. Solano-Gálvez, D.A. Álvarez-Hernández, L. Gutiérrez-Kobeh, R. Vázquez- [53] J. Calvo-Garrido, R. Escalante, Autophagy dysfunction and ubiquitin-positive
López, Leishmania: manipulation of signaling pathways to inhibit host cell protein aggregates in Dictyostelium cells lacking Vmp1, Autophagy 6 (2010)
apoptosis, Ther. Adv. Infect. Dis. 8 (2021) 20499361211014977, https://doi.org/ 100–109, https://doi.org/10.4161/auto.6.1.10697.
10.1177/20499361211014977. [54] S. Bozzaro, L. Eichinger, The professional phagocyte Dictyostelium discoideum as a
[25] J. Giri, S. Srivastav, M. Basu, S. Palit, P. Gupta, A. Ukil, Leishmania donovani model host for bacterial pathogens, Curr. Drug Targets 12 (2011) 942–954,
exploits myeloid cell leukemia 1 (MCL-1) protein to prevent mitochondria- https://doi.org/10.2174/138945011795677782.
dependent host cell apoptosis, J. Biol. Chem. 291 (2016) 3496–3507, https://doi. [55] K. Kitamura, C. Kishi-Itakura, T. Tsuboi, S. Sato, K. Kita, N. Ohta, N. Mizushima,
org/10.1074/jbc.M115.672873. Autophagy-related Atg8 localizes to the apicoplast of the human malaria parasite
[26] S.C. Welburn, I. Maudlin, Control of Trypanosoma brucei brucei infections in tsetse, Plasmodium falciparum, PLoS One 7 (2012) e42977, https://doi.org/10.1371/
Glossina morsitans, Med. Vet. Entomol. 11 (1997) 286–289, https://doi.org/ journal.pone.0042977.
10.1111/j.1365-2915.1997.tb00408.x. [56] A.U. Hain, J. Bosch, Autophagy in Plasmodium, a multifunctional pathway?
[27] R.A. Knight, The archaeology of apoptosis, Cell Death Differ. 9 (2002) 1–2, Comput. Struct. Biotechnol. J. 8 (2013) e201308002 https://doi.org/10.5936/
https://doi.org/10.1038/sj.cdd.4400980. csbj.201308002.
[28] J.A. Lindoso, P.C. Cotrim, H. Goto, Apoptosis of Leishmania (Leishmania) chagasi [57] R. Navale, A.D. Atul, P.S. Sijwali Allanki, Characterization of the autophagy
amastigotes in hamsters infected with visceral leishmaniasis, Int. J. Parasitol. 34 marker protein Atg8 reveals atypical features of autophagy in Plasmodium
(2004) 1–4, https://doi.org/10.1016/j.ijpara.2003.09.010. falciparum, PLoS One 9 (2014) e113220, https://doi.org/10.1371/journal.
[29] P. Holzmuller, R. Bras-Gonçalves, J.L. Lemesre, Phenotypical characteristics, pone.0113220.
biochemical pathways, molecular targets and putative role of nitric oxide- [58] A. Barquilla, M. Navarro, Trypanosome TOR as a major regulator of cell growth
mediated programmed cell death in Leishmania, Parasitology 132 (2006) and autophagy, Autophagy 5 (2009) 256–258, https://doi.org/10.4161/
S19–S32, https://doi.org/10.1017/S0031182006000837. auto.5.2.7591.
[30] M. Das, S.B. Mukherjee, C. Shaha, Hydrogen peroxide induces apoptosis-like [59] V. Deretic, B. Levine, Autophagy, immunity, and microbial adaptations, Cell Host
death in Leishmania donovani promastigotes, J. Cell Sci. 114 (2001) 2461–2469, Microbe 5 (2009) 527–549, https://doi.org/10.1016/j.chom.2009.05.016.
https://doi.org/10.1242/jcs.114.13.2461. [60] R.O. Pinheiro, M.P. Nunes, C.S. Pinheiro, H. D’Avila, P.T. Bozza, C.M. Takiya,
[31] S.C. Welburn, M.A. Barcinski, G.T. Williams, Programmed cell death in S. Côrte-Real, C.G. Freire-de-Lima, G.A. DosReis, Induction of autophagy
trypanosomatids, Parasitol. Today 13 (1997) 22–26, https://doi.org/10.1016/ correlates with increased parasite load of Leishmania amazonensis in BALB/c but
s0169-4758(96)10076-4. not C57BL/6 macrophages, Microbes Infect. 11 (2009) 181–190, https://doi.org/
[32] N. Lee, S. Bertholet, A. Debrabant, J. Muller, R. Duncan, H.L. Nakhasi, 10.1016/j.micinf.2008.11.006.
Programmed cell death in the unicellular protozoan parasite Leishmania, Cell [61] L.T. Cyrino, A.P. Araujo, P.P. Joazeiro, C.P. Vicente, S. Giorgio, In vivo and in vitro
Death Differ. 9 (2002) 53–64, https://doi.org/10.1038/sj.cdd.4400952. Leishmania amazonensis infection induces autophagy in macrophages, Tissue Cell
[33] A. Debrabant, H. Nakhasi, Programmed cell death in trypanosomatids: is it an 44 (2012) 401–408, https://doi.org/10.1016/j.tice.2012.08.003.
altruistic mechanism for survival of the fittest? Kinetoplastid. Biol. Dis. 2 (2003) [62] P. Crauwels, R. Bohn, M. Thomas, S. Gottwalt, F. Jäckel, S. Krämer, E. Bank,
7, https://doi.org/10.1186/1475-9292-2-7. S. Tenzer, P. Walther, M. Bastian, G. van Zandbergen, Apoptotic-like Leishmania
[34] M. Duszenko, K. Figarella, E.T. Macleod, S.C. Welburn, Death of a trypanosome: a exploit the host’s autophagy machinery to reduce T-cell-mediated parasite
selfish altruism, Trends Parasitol. 22 (2006) 536–542, https://doi.org/10.1016/j. elimination, Autophagy 11 (2015) 285–297, https://doi.org/10.1080/
pt.2006.08.010. 15548627.2014.998904.
[35] I. Bruchhaus, T. Roeder, A. Rennenberg, V.T. Heussler, Protozoan parasites: [63] M. Duszenko, M.L. Ginger, A. Brennand, M. Gualdrón-López, M.I. Colombo, G.
programmed cell death as a mechanism of parasitism, Trends Parasitol. 23 (2007) H. Coombs, I. Coppens, B. Jayabalasingham, G. Langsley, S.L. de Castro,
376–383, https://doi.org/10.1016/j.pt.2007.06.004. R. Menna-Barreto, J.C. Mottram, M. Navarro, D.J. Rigden, P.S. Romano, V. Stoka,
[36] C. Shaha, Apoptosis in Leishmania species and its relevance to disease B. Turk, P.A. Michels, Autophagy in protists, Autophagy 7 (2011) 127–158,
pathogenesis, Indian J. Med. Res. 123 (2006) 233–244. https://doi.org/10.4161/auto.7.2.13310.
[37] S. Shimizu, T. Kanaseki, N. Mizushima, T. Mizuta, S. Arakawa-Kobayashi, C. [64] P.S. Romano, M.A. Arboit, C.L. Vazquez, M.I. Colombo, The autophagic pathway
B. Thompson, Y. Tsujimoto, Role of Bcl-2 family proteins in a non-apoptotic is a key component in the lysosomal dependent entry of Trypanosoma cruzi into
programmed cell death dependent on autophagy genes, Nat. Cell Biol. 6 (2004) the host cell, Autophagy 5 (2009) 6–18, https://doi.org/10.4161/auto.5.1.7160.
1221–1228, https://doi.org/10.1038/ncb1192. [65] Z.Y. Xie, Y.X. Xu, L. Yao, Angiotensin II can trigger HSC-LX2 pyroptosis through
[38] E.H. Baehrecke, Autophagy: dual roles in life and death? Nat. Rev. Mol. Cell Biol. both classical and non-classical pathways, Life Sci. 307 (2022) 120878, https://
6 (2005) 505–510, https://doi.org/10.1038/nrm1666. doi.org/10.1016/j.lfs.2022.120878.
[39] P. Boya, R.A. González-Polo, N. Casares, J.L. Perfettini, P. Dessen, N. Larochette, [66] J. Shi, Y. Zhao, K. Wang, X. Shi, Y. Wang, H. Huang, Y. Zhuang, T. Cai, F. Wang,
D. Métivier, D. Meley, S. Souquere, T. Yoshimori, G. Pierron, P. Codogno, F. Shao, Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell
G. Kroemer, Inhibition of macroautophagy triggers apoptosis, Mol. Cell. Biol. 25 death, Nature 526 (2015) 660–665, https://doi.org/10.1038/nature15514.
(2005) 1025–1040, https://doi.org/10.1128/MCB.25.3.1025-1040.2005. [67] D. Li, M. Wu, Pattern recognition receptors in health and diseases, Signal
[40] W. Li, J. Li, J. Bao, Microautophagy: lesser-known self-eating, Cell. Mol. Life Sci. Transduct. Target. Ther. 6 (2021) 291, https://doi.org/10.1038/s41392-021-
69 (2012) 1125–1136, https://doi.org/10.1007/s00018-011-0865-5. 00687-0.
[68] P. Lan, Y. Fan, Y. Zhao, X. Lou, H.P. Monsour, X. Zhang, Y. Choi, Y. Dou, N. Ishii,
R.M. Ghobrial, X. Xiao, X.C. Li, TNF superfamily receptor OX40 triggers invariant
13
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
NKT cell pyroptosis and liver injury, J. Clin. Invest. 127 (2017) 2222–2234, [93] B.L. Verçosa, M.N. Melo, H.L. Puerto, I.L. Mendonça, A.C. Vasconcelos, Apoptosis,
https://doi.org/10.1172/JCI91075. inflammatory response and parasite load in skin of Leishmania (Leishmania)
[69] K. Eichholz, T. Bru, T.T. Tran, P. Fernandes, H. Welles, F.J. Mennechet, N. Manel, chagasi naturally infected dogs: a histomorphometric analysis, Vet. Parasitol. 189
P. Alves, M. Perreau, E.J. Kremer, Immune-complexed adenovirus induce AIM2- (2012) 162–170, https://doi.org/10.1016/j.vetpar.2012.04.035.
mediated pyroptosis in human dendritic cells, PLoS Pathog. 12 (2016) e1005871, [94] H. Chiarini-Garcia, G.G. Parreira, F.R. Almeida, Glycol methacrylate embedding
https://doi.org/10.1371/journal.ppat.1005871. for improved morphological, morphometrical, and immunohistochemical
[70] W.T. He, H. Wan, L. Hu, P. Chen, X. Wang, Z. Huang, Z.H. Yang, C.Q. Zhong, investigations under light microscopy: testes as a model, Methods Mol. Biol. 689
J. Han, Gasdermin D is an executor of pyroptosis and required for interleukin-1β (2011) 3–18, https://doi.org/10.1007/978-1-60761-950-5_1.
secretion, Cell Res. 25 (2015) 1285–1298, https://doi.org/10.1038/cr.2015.139. [95] W.L. Tafuri, R.L. Santos, R.M. Arantes, R. Gonçalves, M.N. de Melo, M.
[71] D. Brough, N.J. Rothwell, Caspase-1-dependent processing of pro-interleukin-1β S. Michalick, W.L. Tafuri, An alternative immunohistochemical method for
is cytosolic and precedes cell death, J. Cell Sci. 120 (2007) 772–781, https://doi. detecting Leishmania amastigotes in paraffin-embedded canine tissues,
org/10.1242/jcs.03377. J. Immunol. Methods 292 (2004) 17–23, https://doi.org/10.1016/j.
[72] L. Franchi, T. Eigenbrod, R. Muñoz-Planillo, G. Nuñez, The inflammasome: a jim.2004.05.009.
caspase-1-activation platform that regulates immune responses and disease [96] P. Piarroux, R. Azaiez, A.M. Lossi, M. Fontes, S. Dunan, C. Mary, B. Toga,
pathogenesis, Nat. Immunol. 10 (2009) 241–247, https://doi.org/10.1038/ M. Quilici, Isolation and characterization of a repetitive DNA sequence from
ni.1703. Leishmania infantum: development of CanL polymerase chain reaction, Am. J.
[73] M. Wang, X. Ning, A. Chen, H. Huang, C. Ni, C. Zhou, Impaired formation of Trop. Med. Hyg. 49 (1993) 364–369, https://doi.org/10.4269/
homotypic cell-in-cell structures in human tumor cells lacking alpha-catenin ajtmh.1993.49.364.
expression, Sci. Rep. 5 (2015) 12223, https://doi.org/10.1038/srep12223. [97] T. Laskay, G. van Zandbergen, W. Solbach, Neutrophil granulocytes – Trojan
[74] P. Orning, D. Weng, K. Starheim, D. Ratner, Z. Best, B. Lee, A. Brooks, S. Xia, horses for Leishmania major and other intracellular microbes? Trends Microbiol.
H. Wu, M.A. Kelliher, S.B. Berger, P.J. Gough, J. Bertin, M.M. Proulx, J. 11 (2003) 210–214, https://doi.org/10.1016/S0966-842X(03)00075-18.
D. Goguen, N. Kayagaki, K.A. Fitzgerald, E. Lien, Pathogen blockade of TAK1 [98] L. Solano-Galleno, H. Fernandes-Bellon, P. Morell, D. Fondevila, J. Alberola,
triggers caspase-8-dependent cleavage of gasdermin D and cell death, Science 362 A. Ramis, L. Ferrer, Histological and immunohistochemical study of clinically
(2018) 1064–1069, https://doi.org/10.1126/science.aau2818. normal skin of L. infantum infected dogs, J. Comp. Pathol. 130 (2004) 7–12,
[75] B. Zhou, J.Y. Zhang, X.S. Liu, H.Z. Chen, Y.L. Ai, K. Cheng, R.Y. Sun, D. Zhou, https://doi.org/10.1016/s0021-9975(03)00063-x.
J. Han, Q. Wu, Tom20 senses iron-activated ROS signaling to promote melanoma [99] L. Moro, A.C. Vasconcelos, F.G.A. Santos, C.M. Alves, J.E.S. Nunes, I.B.
cell pyroptosis, Cell Res. 28 (2018) 1171–1185, https://doi.org/10.1038/s41422- M. Sampaio, Determination of the minimal representative number of microscopic
018-0090-y. fields to quantify apoptosis in canine lymph nodes, Arq. Bras. Med. Vet. Zootec.
[76] J. Hou, R. Zhao, W. Xia, C.W. Chang, Y. You, J.M. Hsu, L. Nie, Y. Chen, Y. 56 (2004) 408–410, https://doi.org/10.1590/S0102-09352004000300020.
C. Wang, C. Liu, W.J. Wang, Y. Wu, B. Ke, J.L. Hsu, K. Huang, Z. Ye, Y. Yang, [100] N. Ueda, S.V. Shah, Tubular cell damage in acute renal failure-apoptosis, necrosis,
X. Xia, Y. Li, C.W. Li, B. Shao, J.A. Tainer, M.C. Hung, PD-L1-mediated gasdermin or both, Nephrol. Dial. Transplant. 15 (2000) 318–323, https://doi.org/10.1093/
C expression switches apoptosis to pyroptosis in cancer cells and facilitates ndt/15.3.318.
tumour necrosis, Nat. Cell Biol. 22 (2020) 1264–1275, https://doi.org/10.1038/ [101] W. de Souza, J.C. Rodrigues, Sterol biosynthesis pathway as target for anti-
s41556-020-0575-z. trypanosomatid drugs, Interdiscip. Perspect. Infect. Dis. (2009) 642502, https://
[77] M. Kurokawa, S. Kornbluth, Caspases and kinases in a death grip, Cell 138 (2009) doi.org/10.1155/2009/642502.
838–854, https://doi.org/10.1016/j.cell.2009.08.021. [102] R.B. Dell, S. Holleran, R. Ramakrishnan, Sample size determination, ILAR J. 43
[78] G. Tomiyoshi, Y. Horita, M. Nishita, K. Ohashi, K. Mizuno, Caspase-mediated (2002) 207–213, https://doi.org/10.1093/ilar.43.4.207.
cleavage and activation of LIM-kinase 1 and its role in apoptotic membrane [103] G. Van Zandbergen, M. Klinger, A. Mueller, S. Dannenberg, A. Gebert,
blebbing, Genes Cells 9 (2004) 591–600, https://doi.org/10.1111/j.1356- W. Solbach, T. Laskay, Cutting edge: neutrophil granulocyte serves as a vector for
9597.2004.00745.x. Leishmania entry into macrophages, J. Immunol. 173 (2004) 6521–6525, https://
[79] K. Balasubramanian, B. Mirnikjoo, A.J. Schroit, Regulated externalization of doi.org/10.4049/jimmunol.173.11.6521.
phosphatidylserine at the cell surface: implications for apoptosis, J. Biol. Chem. [104] G. van Zandbergen, A. Bollinger, A. Wenzel, S. Kamhawi, R. Voll, M. Klinger,
282 (2007) 18357–18364, https://doi.org/10.1074/jbc.M700202200. A. Müller, C. Hölscher, M. Herrmann, D. Sacks, W. Solbach, T. Laskay, Leishmania
[80] O.T. Fackler, R. Grosse, Cell motility through plasma membrane blebbing, J. Cell disease development depends on the presence of apoptotic promastigotes in the
Biol. 181 (2008) 879–884, https://doi.org/10.1083/jcb.200802081. virulent inoculum, Proc. Natl. Acad. Sci. USA 103 (2006) 13837–13842, https://
[81] M. Sebbagh, C. Renvoizé, J. Hamelin, N. Riché, J. Bertoglio, J. Bréard, Caspase-3- doi.org/10.1073/pnas.0600843103.
mediated cleavage of ROCK I induces MLC phosphorylation and apoptotic [105] J.L.M. Wanderley, L.H. Pinto da Silva, P. Deolindo, L. Soong, V.M. Borges, D.
membrane blebbing, Nat. Cell Biol. 3 (2001) 346–352, https://doi.org/10.1038/ B. Prates, A.P.A. de Souza, A. Barral, J.M.D.F. Balanco, M.T.C.D. Nascimento, E.
35070019. M. Saraiva, M.A. Barcinski, Cooperation between apoptotic and viable
[82] R. Tang, J. Xu, B. Zhang, J. Liu, C. Liang, J. Hua, Q. Meng, X. Yu, S. Shi, metacyclics enhances the pathogenesis of Leishmaniasis, PLoS One 4 (2009)
Ferroptosis, necroptosis, and pyroptosis in anticancer immunity, J. Hematol. e5733, https://doi.org/10.1371/journal.pone.0005733.
Oncol. 13 (2020) 110, https://doi.org/10.1186/s13045-020-00946-7. [106] H. Zangger, J.C. Mottram, N. Fasel, Cell death in Leishmania induced by stress and
[83] P. Yu, X. Zhang, N. Liu, Pyroptosis: mechanisms and diseases, Sig. Transduct. differentiation: programmed cell death or necrosis? Cell Death Differ. 9 (2002)
Target. Ther. 6 (2021) 128, https://doi.org/10.1038/s41392-021-00507-5. 1126–1139, https://doi.org/10.1038/sj.cdd.4401071.
[84] L.E. Broker, F.A. Kruyt, G. Giaccone, Cell death independent of caspases: a [107] H. Ishikawa, H. Hisaeda, M. Taniguchi, T. Nakayama, T. Sakai, Y. Maekawa,
review, Clin. Cancer Res. 11 (2005) 3155–3162, https://doi.org/10.1158/1078- Y. Nakano, M. Zhang, T. Zhang, M. Nishitani, M. Takashima, K. Himeno, CD4+
0432.CCR-04-2223. Vα14 NKT cells play a crucial role in an early stage of protective immunity against
[85] M.E. Choi, D.R. Price, S.W. Ryter, A.M.K. Choi, Necroptosis: a crucial pathogenic infection with Leishmania major, Int. Immunol. 12 (2000) 1267–1274, https://doi.
mediator of human disease, JCI Insight 4 (2019) e128834, https://doi.org/ org/10.1093/intimm/12.9.1267.
10.1172/jci.insight.128834. [108] E. Aga, D.M. Katschinski, G. van Zandbergen, H. Laufs, B. Hansen, K. Müller,
[86] Z. Wang, D. Liu, A. Varin, V. Nicolas, D. Courilleau, P. Mateo, A cardiac W. Solbach, T. Laskay, Inhibition of the spontaneous apoptosis of neutrophil
mitochondrial cAMP signaling pathway regulates calcium accumulation, granulocytes by the intracellular parasite Leishmania major, J. Immunol. 169
permeability transition and cell death, Cell Death Dis. 7 (2016) e2198, https:// (2002) 898–905, https://doi.org/10.4049/jimmunol.169.2.898.
doi.org/10.1038/cddis.2016.106. [109] A.L. Bienvenu, E. Gonzalez-Rey, S. Picot, Apoptosis induced by parasitic diseases,
[87] D. Vercammen, P. Vandenabeele, R. Beyaert, W. Declercq, W. Fiers, Tumour Parasit. Vectors 3 (2010) 106, https://doi.org/10.1186/1756-3305-3-106.
necrosis factor-induced necrosis versus anti-Fas-induced apoptosis in L929 cells, [110] R. Vázquez-López, J. Argueta-Donohué, A. Wilkins-Rodríguez, A. Escalona-
Cytokine 9 (1997) 801–808, https://doi.org/10.1006/cyto.1997.0252. Montaño, M. Aguirre-García, L. Gutiérrez-Kobeh, Leishmania mexicana
[88] D. Vercammen, G. Brouckaert, G. Denecker, M. Van de Craen, W. Declercq, amastigotes inhibit p38 and JNK and activate PI3K/AKT: role in the inhibition of
W. Fiers, Dual signaling of the Fas receptor: initiation of both apoptotic and apoptosis of dendritic cells, Parasite Immunol. 37 (2015) 579–589, https://doi.
necrotic cell death pathways, J. Exp. Med. 188 (1998) 919–930, https://doi.org/ org/10.1111/pim.12275.
10.1084/jem.188.5.919. [111] C. Privé, A. Descoteaux, Leishmania donovani promastigotes evade the activation
[89] N.V. Vorobjeva, B.V. Chernyak, NETosis: molecular mechanisms, role in of mitogen-activated protein kinases p38, c-Jun N-terminal kinase, and
physiology and pathology, biochemistry, Biokhimiia 85 (2020) 1178–1190, extracellular signal-regulated kinase-1/2 during infection of naive macrophages,
https://doi.org/10.1134/S0006297920100065. Eur. J. Immunol. 30 (2000) 2235–2244, https://doi.org/10.1002/1521-4141
[90] T.A. Fuchs, U. Abed, C. Goosmann, R. Hurwitz, I. Schulze, V. Wahn, (2000)30:8<2235::AID-IMMU2235>3.0.CO;2-9.
Y. Weinrauch, V. Brinkmann, A. Zychlinsky, Novel cell death program leads to [112] M. Junghae, J.G. Raynes, Activation of p38 mitogen-activated protein kinase
neutrophil extracellular traps, J. Cell Biol. 176 (2007) 231–241, https://doi.org/ attenuates Leishmania donovani infection in macrophages, Infect. Immun. 70
10.1083/jcb.200606027. (2002) 5026–5035, https://doi.org/10.1128/IAI.70.9.5026-5035.2002.
[91] S. Besteiro, R.A.M. Williams, L.S. Morrison, G.H. Coombs, J.C. Mottram, [113] A. Sarkar, E. Aga, U. Bussmeyer, A. Bhattacharyya, S. Möller, L. Hellberg,
Endosomes orting and autophagy are essential for differentiation and virulence of M. Behnen, W. Solbach, T. Laskay, Infection of neutrophil granulocytes with
Leishmania major, J. Biol. Chem. 281 (2006) 11384–11396, https://doi.org/ Leishmania major activates ERK 1/2 and modulates multiple apoptotic pathways
10.1074/jbc.M512307200. to inhibit apoptosis, Med. Microbiol. Immunol. 202 (2013) 25–35, https://doi.
[92] R.A. Williams, K.L. Woods, L. Juliano, J.C. Mottram, G.H. Coombs, org/10.1007/s00430-012-0246-1.
Characterization of unusual families of ATG8-like proteins and ATG12 in the [114] Y.H. Huang, C.T. Yeh, Functional compartmentalization of HSP60-survivin
protozoan parasite Leishmania major, Autophagy 5 (2009) 159–172, https://doi. interaction between mitochondria and cytosol in cancer cells, Cells 9 (2019) 23,
org/10.4161/auto.5.2.7328. https://doi.org/10.3390/cells9010023.
14
B.L.A. Verçosa et al. Cellular Immunology 408 (2025) 104909
[115] M. Aghaei, H. KhanAhmad, S. Aghaei, M. Ali Nilforoushzadeh, M.A. Mohaghegh, [127] S. Besteiro, R.A.M. Williams, G.H. Coombs, J.C. Mottram, Protein turnover and
S.H. Hejazi, The role of Bax in the apoptosis of Leishmania-infected macrophages, differentiation in Leishmania, Int. J. Parasitol. 37 (2007) 1063–1075, https://doi.
Microb. Pathog. 139 (2020) 103892, https://doi.org/10.1016/j. org/10.1016/j.ijpara.2007.03.008.
micpath.2019.103892. [128] M.C. Vanrell, A.D. Losinno, J.A. Cueto, D. Balcazar, L.V. Fraccaroli, C. Carrillo, P.
[116] T.S. Wang, I. Coppens, A. Saorin, N.R. Brady, A. Hamacher-Brady, Endolysosomal S. Romano, The regulation of autophagy differentially affects Trypanosoma cruzi
targeting of mitochondria is integral to Bax-mediated mitochondrial metacyclogenesis, PLoS Neglected Trop. Dis 11 (2017), https://doi.org/10.1371/
permeabilization during apoptosis signaling, Dev. Cell 53 (2020) 627–645, journal.pntd.0006049, 00060499–e6123.
https://doi.org/10.1016/j.devcel.2020.05.014. [129] L. Gómez-Virgilio, M.D. Silva-Lucero, D.S. Flores-Morelos, J. Gallardo-Nieto,
[117] K. Ma, G. Chen, W. Li, O. Kepp, Y. Zhu, Q. Chen, Mitophagy, mitochondrial G. Lopez-Toledo, A.M. Abarca-Fernandez, A.E. Zacapala-Gómez, J. Luna-Muñoz,
homeostasis, and cell fate, Front. Cell Dev. Biol. 8 (2020) 467, https://doi.org/ F. Montiel-Sosa, L.O. Soto-Rojas, M. Pacheco-Herrero, M.D. Cardenas-Aguayo,
10.3389/fcell.2020.00467. Autophagy: a key regulator of homeostasis and disease: an overview of molecular
[118] M. Aghaei, H. Khanahmad, S. Aghaei, S.M. Hosseini, M. Farahmand, S.H. Hejazi, mechanisms and modulators, Cells 11 (2022) 2262, https://doi.org/10.3390/
Evaluation of transgenic Leishmania infantum expressing mLLO-BAX-SMAC in the cells11152262.
apoptosis of the infected macrophages in vitro and in vivo, Parasite Immunol. 42 [130] L. Afonso, V.M. Borges, H. Cruz, F.L. Ribeiro-Gomes, G.A. DosReis, N.A. Dutra,
(2020) e12726, https://doi.org/10.1111/pim.12726. J. Clarêncio, C.I. Oliveira, A. Barral, M. Barral-Neto, C.I. Brodskyn, Interactions
[119] I.A. Ciechomska, G.C. Goemans, J.N. Skepper, A.M. Tolkovsky, Bcl-2 complexed with apoptotic but not necrotic neutrophils increase parasite burden in human
with Beclin-1 maintains full anti-apoptotic function, Oncogene 28 (2009) macrophages infected with Leishmania amazonenses, J. Leukoc. Biol. 8 (2008)
2128–2141, https://doi.org/10.1038/onc.2009.60. 389–396, https://doi.org/10.1189/jlb.0108018.
[120] S. Pattingre, A. Tassa, X. Qu, R. Garuti, X.H. Liang, N. Mizushima, M. Packer, M. [131] F.L. Ribeiro-Gomes, A. Romano, S. Lee, E. Roffê, N.C. Peters, A. Debrabant,
D. Schneider, B. Levine, Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent D. Sacks, Apoptotic cell clearance of Leishmania major-infected neutrophils by
autophagy, Cell 122 (2005) 927–939, https://doi.org/10.1016/j. dendritic cells inhibits CD8+ T-cell priming in vitro by Mer tyrosine kinase-
cell.2005.07.002. dependent signaling, Cell Death Dis. 6 (2015) e2018, https://doi.org/10.1038/
[121] S. Shen, Y. Shao, C. Li, Different types of cell death and their shift in shaping cddis.2015.351.
disease, Cell Death Dis. 9 (2023) 284, https://doi.org/10.1038/s41420-023- [132] N.C. Peters, J.G. Egen, N. Secundino, A. Debrabant, D. Sacks, In vivo imaging
01581-0. revealed an essential role for neutrophils in Leishmaniasis transmitted by sand
[122] J. Ding, K. Wang, W. Liu, Y. She, Q. Sun, J. Shi, Pore-forming activity and flies, Science 321 (2008) 970–974, https://doi.org/10.1126/science.1159194.
structural autoinhibition of the gasdermin family, Nature 535 (2016) 111–116, [133] S.C. Xavier, H.M. Andrade, S.J. Hadad, I.M. Chiarelli, W.G. Lima, M.S.
https://doi.org/10.1038/nature18590. M. Michalick, W.L. Tafuri, W.L. Tafuri, Comparison of paraffin-embedded skin
[123] M. Jiang, L. Qi, L. Li, Y. Li, The caspase-3/GSDME signal pathway as a switch biopsies from different anatomical regions as sampling methods for detecting
between apoptosis and pyroptosis in cancer, Cell Death Dis. 6 (2020) 112, Leishmania infection in dogs using histological, immunohistochemical and PCR
https://doi.org/10.1038/s41420-020-00349-0. methods, BMC Vet. Res. 2 (2006) 17–21, https://doi.org/10.1186/1746-6148-2-
[124] C. Rogers, T. Fernandes-Alnemri, L. Mayes, D. Alnemri, G. Cingolani, E. 17.
S. Alnemri, Cleavage of DFNA5 by caspase-3 during apoptosis mediates [134] R.C. Giuguetti, W. Mayrink, O. Genaro, C.M. Carneiro, R. Corrêa-Oliveira, O.
progression to secondary necrotic/pyroptotic cell death, Nat. Commun. 8 (2017) A. Martins-Filho, M.J. Marques, W.L. Tafuri, A.B. Reis, Relationship between
14128, https://doi.org/10.1038/ncomms14128. canine visceral leishmaniosis and the Leishmania (Leishmania) chagasi burden in
[125] Y. Wang, W. Gao, X. Shi, J. Ding, W. Liu, H. He, Chemotherapy drugs induce dermal inflammatory foci, J. Comp. Pathol. 135 (2006) 100–107, https://doi.org/
pyroptosis through caspase-3 cleavage of a gasdermin, Nature 547 (2017) 10.1016/j.jcpa.2006.06.005.
99–103, https://doi.org/10.1038/nature22393. [135] W.L.C. Dos-Santos, J. David, R. Badaró, L.A.R. De-Freitas, Association between
[126] B. Cull, J.L.P. Godinho, J.C.F. Rodrigues, B. Frank, U. Schurigt, R.A.M. Williams, skin parasitism and granulomatous inflammatory pattern in canine visceral
G.H. Coombs, J.C. Mottram, Glycosome turnover in Leishmania major is mediated leishmaniasis, Parasitol. Res. 94 (2004) 84–94, https://doi.org/10.1007/s00436-
by autophagy, Autophagy 10 (2014) 2143–2157, https://doi.org/10.4161/ 003-1016-1.
auto.36438. [136] N. Pandian, T.D. Kanneganti, PANoptosis: a unique innate immune inflammatory
cell death modality, J. Immunol. 209 (2022) 1625–1633, https://doi.org/
10.4049/jimmunol.2200508.
15