Skip to main content
Antioxidants & Redox Signaling logoLink to Antioxidants & Redox Signaling
. 2022 May 6;36(13-15):969–983. doi: 10.1089/ars.2021.0058

Mitochondrial Ca2+ and Reactive Oxygen Species in Trypanosomatids

Roberto Docampo 1,2,, Aníbal Eugénio Vercesi 3
PMCID: PMC9125514  PMID: 34218689

Abstract

Significance:

Millions of people are infected with trypanosomatids and new therapeutic approaches are needed. Trypanosomatids possess one mitochondrion per cell and its study has led to discoveries of general biological interest. These mitochondria, as in their animal counterparts, generate reactive oxygen species (ROS) and have evolved enzymatic and nonenzymatic defenses against them. Mitochondrial calcium ion (Ca2+) overload leads to generation of ROS and its study could lead to relevant information on the biology of trypanosomatids and to novel drug targets.

Recent Advances:

Mitochondrial Ca2+ is normally involved in maintaining the bioenergetics of trypanosomes, but when Ca2+ overload occurs, it is associated with cell death. Trypanosomes lack key players in the mechanism of cell death described in mammalian cells, although mitochondrial Ca2+ overload results in collapse of their membrane potential, production of ROS, and cytochrome c release. They are also very resistant to mitochondrial permeability transition, and cell death after mitochondrial Ca2+ overload depends on generation of ROS.

Critical Issues:

In this review, we consider the mechanisms of mitochondrial oxidant generation and removal and the involvement of Ca2+ in trypanosome cell death.

Future Directions:

More studies are required to determine the reactions involved in generation of ROS by the mitochondria of trypanosomatids, their enzymatic and nonenzymatic defenses against ROS, and the occurrence and composition of a mitochondrial permeability transition pore. Antioxid. Redox Signal. 36, 969–983.

Keywords: calcium, Leishmania spp, mitochondria, Trypanosoma cruzi, Trypanosoma brucei

Introduction

Trypanosomatids include the genera, Trypanosoma and Leishmania, with species that cause several human diseases: Chagas disease (Trypanosoma cruzi), African trypanosomiasis (Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense), and leishmaniasis (several Leishmania species). These pathogens have intricate life cycles that include an insect and a mammalian host (Fig. 1).

FIG. 1.

FIG. 1.

Life cycle stages of Trypanosoma cruzi, Trypanosoma brucei, and Leishmania spp. in the vector and mammalian hosts. Color images are available online.

T. cruzi exists in two vector forms: epimastigotes and metacyclic trypomastigotes, while intracellular amastigotes and cell-derived trypomastigotes are present in the mammalian host. Metacyclic trypomastigotes are nonreplicative and are released by the vector and invade nucleated mammalian cells, where they differentiate into amastigotes. Amastigotes live in the cytosol of host cells and differentiate back into trypomastigotes. These cell-derived trypomastigotes are also nonreplicative and are released into the blood circulation to invade other cells or to be ingested by the vector (Fig. 1, T. cruzi). The T. brucei group of pathogens includes several stages, the procyclic trypomastigotes that reproduce in the insect vector, converting first into epimastigotes and then into metacyclic trypomastigotes, and the slender bloodstream trypomastigotes that replicate in the blood of the mammalian host and differentiate into stumpy forms that the vector can take up (Fig. 1, T. brucei). Leishmania spp. have three life cycle stages: the promastigotes that replicate in the vector; the metacyclic promastigotes that infect the animal host macrophages, and the amastigotes that replicate within a parasitophorous vacuole of phagocytic cells and can be released and infect other macrophages or be taken up by the vector (Fig. 1, Leishmania spp.).

All trypanosomatids possess only one mitochondrion per cell (Fig. 2), with peculiarities not found in the mammalian hosts, for example, the presence of their DNA in a network known as kinetoplast DNA (kDNA), comprising minicircles and maxicircles (31); a complex process of mitochondrial RNA editing (99); the need to import nuclear-encoded transfer RNAs (tRNAs) (134); and unique features of the mitochondrial ribosomes (36), to mention just a few. The capacity of these mitochondria to generate endogenous levels of reactive oxygen species (ROS) has not been studied with the same level of detail as that of the mammalian mitochondria, although most trypanosomes possess potential sites in their respiratory chains to generate superoxide anion (O2•−) and hydrogen peroxide (H2O2). The mitochondrial defenses against ROS are, however, better identified. Mitochondrial calcium ion (Ca2+) overload is an important mechanism involved in generation of ROS that could end up in cell death. We will review recent work on the role of the mitochondria of trypanosomes in generation of ROS, their defense mechanisms, and the relevance of Ca2+ in these pathways.

FIG. 2.

FIG. 2.

Single mitochondrion of trypanosomes. A procyclic trypomastigote is observed by DIC microscopy (left panel) or with the single mitochondrion stained with MT (in red, right panel). DAPI staining of the nucleus and kinetoplast (blue). Bars = 10 μm. DIC, differential interference contrast; MT, MitoTracker. Modified from the study by Huang et al. (77) with permission. Color images are available online.

Mitochondrial Generation of O2•− and H2O2

Mammalian cells have been proposed to possess 11 sites linked to substrate oxidation and oxidative phosphorylation that could transfer electrons to partially reduced molecular oxygen to produce O2•− or H2O2: oxoacid dehydrogenase complexes, which provide electrons to NAD+, respiratory complexes I and III, and dehydrogenases, including complex II, which uses ubiquinone as an acceptor (20). Most of these sites involve flavoprotein intermediates, which undergo one-electron reduction reactions, and it is therefore not unexpected that leak of electrons to oxygen could generate O2•−.

Trypanosomes possess several of these sites and lack others. Several 2-oxoacid dehydrogenase complexes, such as alpha-ketoglutarate dehydrogenase, pyruvate dehydrogenase, and branched-chain 2-oxoacid dehydrogenase, donate electrons to complex I through the formation of NADH (20). They have a common E3 subunit (dihydrolipoamide dehydrogenase) that transfers electrons from the dihydrolipoate of the E2 subunit to FAD and NAD+ . Production of O2•− and H2O2 is at the flavin in the matrix space (20), but this has not been tested in any trypanosomatid. Electrons from NADH enter the respiratory chain in the NADH-binding site, which was found in early work as a site of O2•− production (24). Attempts to detect the formation of ROS by complex I using dihydroethidium and flow cytometry detected some activity when the (NADH: ubiquinone oxidoreductase accessory subunit) of T. brucei procyclic forms was downregulated by RNA interference (RNAi) (153). However, this production was attributed to other metabolic processes because there were no alterations in cell growth, membrane potential, or NADH dehydrogenase activity, and it was concluded that complex I has no important function in energy metabolism of T. brucei procyclic forms (153). Similar results were found in T. cruzi epimastigotes (28).

T. brucei procyclic forms have a second NADH dehydrogenase that has flavin as a cofactor (60). The purified enzyme was able to generate O2•−, as detected by reduction of acetylated cytochrome c (59). Respiratory inhibitors were also used to produce ROS in isolated mitochondria (59). Mitochondrial membranes of T. brucei procyclic forms were also able to generate O2•− and H2O2 in the presence of NADH and their inhibition by fumarate suggested that fumarate reductase, which is able to produce succinate from fumarate using NADH, was the source of ROS (146). Similar results were obtained with T. cruzi epimastigotes (28).

In mammalian mitochondria, glycerol-3-phosphate dehydrogenase generates O2•− (20). This enzyme, which is FAD dependent, is a very important component of a shuttle between the glycosomes and mitochondria of the bloodstream forms of T. brucei and is important to maintain the glycosomal redox balance. However, there are no reports of its O2•− production activity in trypanosomes. Succinate dehydrogenase, or complex II, was able to generate O2•− in T. cruzi epimastigotes (135), but only in the presence of inhibitors. Finally, complex III or ubiquinol:cytochrome c oxidoreductase is the most important source of ROS in mammalian mitochondria (20), but mechanistic studies on generation of ROS by this complex in trypanosomes are lacking (143). Figure 3 shows a scheme of the potential or demonstrated O2•−-generating sites associated with the respiratory chain of trypanosomatids.

FIG. 3.

FIG. 3.

Potential sites of O2•− formation in the RC of trypanosomatids. Electrons that enter the RC at complex I, type II NADH-DH, and complex II, reduce UQ. G3PDH, which regenerates glycosomal G3P back to DHAP, also provides electrons to UQ. From UQ, electrons are transferred to complex III, cytochrome c (cyt c), and finally to complex IV, where they reduce O2 to H2O. Proton (H+) transfer through complexes III and IV generates an electrochemical gradient that is used to produce ATP from ADP and inorganic phosphate by complex V (ATP synthase). In Trypanosoma brucei bloodstream forms, the RC includes G3PDH and the TAO. Complexes II and III, UQ, and FR are potential sites for O2•− formation. DH, dehydrogenase; DHAP, dihydroxyacetone phosphate; FR, fumarate reductase; G3P, glycerol-3-phosphate; G3PDH, glycerol-3-phosphate dehydrogenase; O2•−, superoxide anion; RC, respiratory chain; TAO, trypanosome alternative oxidase; UQ, ubiquinone. Modified from the study by Tomás and Castro (143). Color images are available online.

Defenses Against ROS in Trypanosomatids

Trypanosomatids live in very different environments and their protection against the toxicity of ROS depends on those environmental conditions (91). The vector forms have stages exposed to low oxygen levels, such as the intestine of the vectors, and these forms were found lacking enzymes required for decomposition of H2O2 (19, 39, 40, 42, 45–47). Trypanosomatids lack genes for thioredoxin reductase, selenocysteine-dependent glutathione peroxidase (GSHPx), catalase, and glutathione reductase (79, 89). However, some defense mechanisms are present and we will describe them in more detail.

Superoxide Dismutases

Superoxide dismutases (SODs) catalyze the dismutation of O2•− to H2O2 and O2. T. brucei possesses four iron-dependent SODs: in the cytosol, SODB1; glycosomes, SODB2; and mitochondria, SODA and SODC (56, 157), where they are involved in dismutation of locally generated O2•−. Although some of the potential sources of O2•− in the mitochondria of trypanosomatids are known (Fig. 3), the cytosolic and glycosomal sources have not been identified. T. cruzi recombinant SODs have been characterized (81, 100, 141). These enzymes can be inactivated by peroxynitrite and one of them, TcSODA, has been crystallized (98). It has been proposed that SOD inhibitors, such as the iron chelator N1,N6-bis(2,3-dihydroxybenzoyl)-1,6-diaminohexane (bis-C6-DHB), could be trypanocidal agents (102). This proposal assumes that O2•− is the toxic species. Another possibility is that H2O2, the product of the SOD reaction, and the hydroxyl radical that can be produced by the Fenton reaction could be more damaging than O2•−. In this case, SOD inhibition would be protective if H2O2 is not degraded. In this regard, a hydroxyphthalazine SOD inhibitor was reported to decrease the parasitemia levels of T. cruzi in infected mice (109). The susceptibility of T. cruzi to benznidazole and crystal violet, but not to nifurtimox, increases after overexpression of cytosolic/glycosomal SOD (SODB1) (141). The reason for these effects is unknown.

As indicated above, the mitochondria of trypanosomatids possess two iron-dependent SODs (SODA and SODC) (56, 157), but they are not individually essential in T. brucei bloodstream forms, as examined by RNAi, although downregulation of SODA led to higher sensitivity to the ROS generator, paraquat (157). Overexpression of SODA also made Leishmania chagasi more resistant to paraquat (116). The mitochondrial SODA from Leishmania amazonensis has been suggested to initiate ROS-mediated signaling required for differentiation and protection of parasites against oxidative stress (103).

The Trypanothione System and Peroxidases

While intracellular thiol homeostasis in mammals is maintained by glutathione/glutathione reductases and thioredoxin/thioredoxin reductases, in trypanosomatids, thiol homeostasis is maintained by the trypanothione [T(SH)2]/trypanothione disulfide [T(S)2] reductase couple (79).

Glutathione and spermidine combine to synthesize T(SH)2 [N1,N8-bis(glutathionyl)spermidine] (Fig. 4). In a first step, an ATP-using reaction combines glutathione (GSH) and spermidine to form monoglutathionylspermidine (glutathionyl spermidine [GSH-SPD]). A second molecule of GSH is then added to generate T(SH)2 with the consumption of another ATP. Both reactions are catalyzed by trypanothione synthetase (TryS) (34, 112, 113) (Fig. 4). Oxidation of T(SH)2 results in T(S)2. T(S)2 is reduced back to T(SH)2 by a trypanothione disulfide reductase (TryR) (62, 86) in an NADPH-dependent reaction (Fig. 5).

FIG. 4.

FIG. 4.

T(SH)2 synthesis occurs by condensation of spermidine with glutathione to form N1-glutathionyl spermidine. Addition of a second glutathione leads to the formation of T(SH)2. Both reactions consume ATP and are catalyzed by T(SH)2 synthase. T(SH)2, trypanothione.

FIG. 5.

FIG. 5.

Redox metabolism in trypanosomatids. Reduction of T(S)2 to T(SH)2 is catalyzed by TryR with conversion of NADPH into NADP+. Oxidation of T(SH)2 to T(S)2 is coupled to the reduction of TryX(S)2, oxidized glutathione (GSSG), and dhA or oxidized Cc. These compounds are generated by the action of peroxidases catalyzing the decomposition of H2O2 (APx/CcPx, GSHPxs I and III, and TryXPx) and/or hydroperoxides (ROOH; GSHPxs I and II, and TryXPx, also called Prx). APx, ascorbate peroxidase; Cc, cytochrome c; CcPx, cytochrome c peroxidase; dhA, dehydroascorbate; GSHPx, glutathione peroxidase; GSSG, glutathione disulfide; H2O2, hydrogen peroxide; Prx, peroxiredoxin; TryR, trypanothione disulfide reductase; TryX, tryparedoxin; TryXPx, tryparedoxin peroxidase; TryX(S)2, tryparedoxin disulfide; T(S)2, trypanothione disulfide. Color images are available online.

TryR has homology to glutathione disulfide (GSSG) reductase and is specific for T(S)2. The enzyme was crystallized alone or bound to substrates or inhibitors (14, 82, 90, 92, 163). Several inhibitors have been tested, such as polyamine derivatives (15, 94, 107, 108), nitrofurans and naphthoquinones (2, 86), dibenzazepines (67), crystal violet (104), phenothiazines and related tricyclics (13, 32, 71, 87), bisbenzylisoquinoline alkaloids (63), acridines (18), diphenylsulfide derivatives (9), Mannich bases (93), natural products (35), and ajoene (64), as well as some terpyridine–platinum complexes (17). These compounds inhibit the activity of the enzyme and parasite growth in vitro or in vivo. The enzyme is cytosolic, but there is some evidence of its presence in the mitochondria and glycosomes (79).

T(SH)2 oxidation leads to reduction of intermediates (dehydroascorbate or oxidized cytochrome c, GSSG, or tryparedoxin disulfide); several peroxidases (ascorbate/cytochrome c peroxidase, cysteine-dependent GSHPx, and tryparedoxin peroxidase (TryXPx), respectively) use these intermediates to degrade H2O2 or organic hydroperoxides (Fig. 5).

The ascorbate peroxidase (APx) activity was first found in extracts of T. cruzi epimastigotes (41), preceding its discovery in plants (69). The enzyme belongs to Class I of the peroxidase–catalase superfamily, contains heme, and uses ascorbate or reduced cytochrome c (Cc) as the substrate to degrade H2O2, but is not active on organic hydroperoxides (19, 41, 139, 156) (Fig. 5). This APx belongs to the hybrid-type group A of APx-CcPx (cytochrome c peroxidase).

The T. cruzi APx colocalizes with BiP, an endoplasmic reticulum marker (156). Others have localized it to glycosomes (41), where ascorbate synthesis takes place (95). More recent work, using electron microscopy, has found the T. cruzi enzyme in the mitochondria and outer surface of amastigotes and trypomastigotes (78). The Leishmania major APx was found in the inner mitochondrial membrane (52). The recombinant enzyme from T. cruzi was studied in detail (78, 158). There is no correlation between its expression and metacyclogenesis or virulence (123), and the enzyme is not essential for viability in the mammalian host or to maintain the chronic infection (139). However, TcAPx null mutants are less infective in vitro and are more sensitive to exogenous H2O2 (139). T. cruzi strains resistant to benznidazole have higher expression of TcAPx (106), and as the L. major enzyme (1, 84, 161), TcAPx has cytochrome c peroxidase activity (78). The L. major APx was crystallized (83). Overexpression of this mitochondrially located enzyme protects L. major from oxidative stress (53). The enzyme is not present in T. brucei, Trypanosoma congolense, and Trypanosoma vivax. Other characteristics of the trypanosomatid peroxidases have also been reviewed (30, 121).

Cysteine-dependent GSHPxs, with some exceptions, are not able to decompose H2O2 (19, 23). In contrast to the mammalian homologs, these enzymes have cysteine instead of selenocysteine in their active site. GSHPx I from T. cruzi is localized in the glycosomes and cytosol and can use reduced tryparedoxin [TryX(SH)2] or GSH as a substrate (155, 159). The crystal structure of this enzyme has been obtained (117). GSHPx II from T. cruzi is present in the endoplasmic reticulum and can use only GSH (158). The T. brucei GSHPxs I and II localize predominantly in the cytosol (57). GSHPx III localizes to the mitochondria of T. brucei and shows strong preference for reduced tryparedoxin or thioredoxin (74). The peroxidases from T. cruzi can degrade organic hydroperoxides, but not H2O2 (155, 158). The GSHPxs I and III from T. brucei, in contrast, decompose H2O2 (74, 133).

TryXPxs, also known as peroxiredoxins, are in the family of 2-cysteine peroxiredoxins and use tryparedoxin to decompose H2O2 (70, 96, 154). Two isoforms exist, one mitochondrial and the other cytosolic (159). They can degrade peroxynitrite (120, 124, 142, 144). The TryXPx crystal structure has been determined (126). These peroxiredoxins play an important role in the defense mechanism against macrophage-generated nitrosative and oxidative stresses (4). The peroxidase activity of these enzymes depends on T(SH)2 (Fig. 5) (123). The mitochondria of T. brucei also have a TryXPx (or peroxiredoxin). Interestingly, the mitochondrial TryXPx of Leishmania infantum (140) and the cytosolic TryXPx of T. cruzi (125) also function as chaperones. Recent work using a tryparedoxin-coupled biosensor revealed that T. brucei harbors a mitochondrial T(SH)2/T(S)2 system (57). However, synthesis of T(SH)2 and reduction of T(S)2 occur in the cytosol (112, 137), and it was suggested that trypanosomes might possess an exchanger of T(S)2 and T(SH)2 and an oxidoreductase to transfer reducing equivalents from T(SH)2 onto the mitochondrial peroxidases (57) (Fig. 6).

FIG. 6.

FIG. 6.

H2O2 metabolism in mitochondria of trypanosomes. RC-generated O2•− is dismutated to H2O2 via SODs A and C. H2O2 is metabolized by APx/CcPx (absent in Trypanosoma brucei), GSHPx III, and TryXPx (or peroxiredoxin), with concomitant oxidation of ascorbate or cytochrome c (A/Ccr), GSH, and tryparedoxin [TryX(SH)2], respectively. These compounds are reduced by T(SH)2. There is probably a [T(S)2)/(T(SH)2] exchanger, and T(SH)2 is regenerated by cytosolic T(SH)2 reductase using NADPH. Ccr, reduced cytochrome c; GSH, glutathione; SOD, superoxide dismutase. Color images are available online.

Transformation of epimastigotes of T. cruzi into metacyclic trypomastigotes is accompanied by increased expression of several antioxidant enzymes, such as APx, TryXPx, tryparedoxin, TryS, and iron SOD (8). It was proposed that the change indicates preparation of the infective forms for potential damage generated by the respiratory burst of mammalian phagocytic cells (8).

In conclusion, past (143) and current evidence indicates that mitochondria of trypanosomatids have enzymatic defenses against ROS, such as SOD (SODA and SODC), APxCcpx (Leishmania spp.), GSHPx III (T. brucei), and TryXPx (T. cruzi, T. brucei, and Leishmania spp.) (Fig. 6).

Other Thiols

T. cruzi possesses T(SH)2 analogs derived from spermine or other polyamines such as homotrypanothione, N1,N12-bis(glutathionyl)spermine, N1-glutathionyl-N8-acetylspermidine, and N1-glutathionyl-N12-acetylspermine (7, 111). These compounds are the result of condensation of GSH with cadaverin, spermine, N-acetyl spermine, and N1- and N8-acetylspermine catalyzed by TryS (7), but their importance has not been elucidated. Ovothiol (N1-methyl-4-mercaptohistidine) (6) also occurs in different life cycle stages of T. cruzi and Leishmania spp. (138), but its relevance is not known. The ovothiol biosynthetic enzyme, 5-histidylcysteine sulfoxide synthase, was characterized in T. cruzi (21).

Mitochondrial Ca2+ Transport

The mitochondrial Ca2+ uniporter (MCU) was discovered 60 years ago as a result of studies with rat kidney mitochondria (38, 147). When MCU was found to be absent in Saccharomyces cerevisiae mitochondria (25), it was thought that this transporter was not present in nonanimal species (26, 101). However, an MCU was described in T. cruzi (48, 49). T. cruzi mitochondrial Ca2+ transport was found to have the same properties of the animal MCU: electrogenicity, low affinity for Ca2+, high capacity, and inhibition by ruthenium red (48, 49).

The MCU was later found in several Leishmania spp. (12, 149, 151) and T. brucei (150, 152, 160). Interestingly, it was found that mitochondria of the bloodstream form of T. brucei were also able to transport Ca2+, although they lack a conventional respiratory chain and oxidative phosphorylation (150). These mitochondria use the ATP synthase, working in reverse, as an ATPase to generate the electrochemical gradient needed to transport Ca2+, and this transport is inhibited by oligomycin (150).

The lack of an MCU in yeast (25) and its presence in trypanosomes (48) and mammals (90) led to the comparison of the mitochondrial proteomes of yeast, trypanosomes, and humans to identify mitochondrial proteins present in trypanosomes and humans and absent in yeast, resulting in first, the identification of the modulator of MCU in mammals, the mitochondrial calcium uptake 1 (MICU1) (118), and later, of the MCU pore subunit (11, 37, 44).

Several components, in addition to the pore-forming subunit MCU, form part of the MCU complex (11, 37, 51), including the MCU paralog, MCUb, which is a dominant negative subunit (130); the mitochondrial calcium uniporter regulator 1 (MCUR1), which is a scaffolding subunit (97); the essential mitochondrial regulator (EMRE) (132); and MICU1 (118), 2 (MICU2), and 3 (MICU3) (128) (Fig. 7A).

FIG. 7.

FIG. 7.

MCU complex organization. (A) In animals, the MCU complex is constituted by the pore-forming subunit MCU and regulator proteins MCUb, MICU1, MICU2, EMRE, and MCUR1. (B) The MCU complex in trypanosomes comprises the pore-forming subunits MCU, MCUb, MCUc, and MCUd, probably forming a hetero-hexamer, and Ca2+-sensing subunits MICU1 and MICU2. Black balls, Ca2+ ions; dark orange circular sectors: EF hand domains, S letters enclosed in the circle are cysteine residues that can form a disulfide bridge. Ca2+, calcium ion; EMRE, essential mitochondrial regulator; IMS, inter membrane space; MCU, mitochondrial calcium uniporter; MCUR1, mitochondrial calcium uniporter regulator 1; MICU, mitochondrial calcium uptake. Reprinted from the study by Docampo et al. (51) with permission from Elsevier. Color images are available online.

The trypanosome MCU complex has a different composition (Fig. 7B) and characteristics that differ from those of the mammalian complex. (i) The MCU subunit is essential for T. brucei growth and infectivity in vivo (77), while MCU knockout mice survive with only alterations in skeletal muscle function under strenuous exercise (115). (ii) There are paralogs of the trypanosome MCU, named MCUb, MCUc, and MCUd, which are necessary for mitochondrial Ca2+ transport. MCUc and MCUd are only present in trypanosomatids (75). These two subunits, together with MCU and MCUb, form hetero-hexameric complexes in membranes (75). In contrast, the recombinant MCUs described in fungi (10, 58, 105, 162) and zebra fish (10) form homotetramers, while the recombinant protein of Caenorhabditis elegans forms homopentamers (110). These differences were considered as evidence of parallel evolution of the uniporter in animals and trypanosomes (127). (iii) While in vertebrate cells, a fundamental function of mitochondrial Ca2+ transport is regulation of oxidative phosphorylation (27), T. brucei bloodstream forms are the only cells that have a functional MCU complex, but do not have a conventional respiratory chain and oxidative phosphorylation, and its function in these stages warrants further exploration (150). (iv) The trypanosome MCU interacts with the ATP synthase subunit c (76). This interaction could have potential implications for the understanding of mitochondrial physiology. The biological significance of this interaction is evidenced by the formation of a megacomplex by the ATP synthase, the ADP/ATP translocator (adenine nucleotide translocase [ANT]), the inorganic phosphate carrier (PiC), and the MCU complex. The ATP synthase uses ADP, transported by the ADP/ATP translocator, and Pi, transported by the PiC, to synthesize ATP, and this activity is stimulated by Ca2+ transported by the MCU complex. (v) The inositol 1,4,5-trisphosphate receptor (IP3R) of trypanosomes localizes to the acidocalcisome (33, 77), the most important Ca2+ store in trypanosomes (50). This IP3R is essential in the bloodstream forms of T. brucei (77). There are also membrane contact sites between acidocalcisomes and mitochondria (131). Ca2+ release from acidocalcisomes mediated by the IP3R can be taken up by the mitochondria to stimulate mitochondrial bioenergetics of T. cruzi (33).

Mitochondrial Ca2+ release in mammalian cells is by either an H+- or Na+-coupled antiporter. The molecular identity of the Na+-coupled antiporter was recently discovered (114), but trypanosomes lack homolog genes. However, a Ca2+/H+ antiporter was proposed to be present in T. cruzi mitochondria based on their response to addition of Ca2+ and EGTA (49). A protein named Letm1 was proposed to act as a Ca2+/H+ antiporter in the mitochondria of mammalian cells (85, 145). Studies in T. brucei suggested that TbLetm1 is involved in maintaining mitochondrial volume via K+/H+ exchange across the inner membrane (72). However, recent work has shown that T. cruzi Letm1 is important for Ca2+ uptake and release (54). Both mitochondrial Ca2+ influx and efflux are reduced in TcLetm1 knockdown trypanosomes. These changes lead to Ca2+ overload and increased autophagy, as well as reduced invasion of host cells and intracellular replication (54).

Mitochondrial Ca2+ Overload, ROS Generation, and Cell Death in Trypanosomes

Early work on T. cruzi (42, 43) described that treatment of different life cycle stages of the parasite with naphthoquinones that generate ROS resulted in cell death and morphological changes that are now considered typical of mammalian apoptosis, such as mitochondrial disruption, chromatin condensation, and plasma membrane blebbing. Later reports in the mid 1990s suggested that T. cruzi epimastigotes can undergo apoptosis under starvation or after complement exposure (5), and this led to subsequent studies centered on apoptosis in parasitic protozoa (136). It has been indicated that robust experimental evidence demonstrating regulated or programmed cell death in parasitic protozoa is still lacking (129). However, some characteristic features of apoptosis such as release of cytochrome c from the mitochondria, DNA fragmentation, and exposure of phosphatidylserine in the plasma membrane have been observed in T. cruzi subjected to different stresses (55, 80, 88, 119, 122). Their relevance as markers of apoptosis in protozoa has been extensively discussed (129).

Trypanosomatids lack homologs to proteins involved in apoptosis in animal cells, such as the TNF-related family of receptors, Bcl-2 family members, and caspases (136). However, metacaspases and cathepsin-like proteases (88), as well as nucleases such as endonuclease G (Endo G) (66) and TaD (65), have been shown to play roles in some of the apoptosis-like phenotypic changes that accompany cell death in different trypanosomatids. What appears to be clear is that there is a mitochondrial pathway of cell death in trypanosomatids. For example, the interplay between mitochondrial Ca2+ overload, ROS generation, and mitochondrial dysfunction upon complement-dependent cell death has been described in T. cruzi (80). A similar interplay between Ca2+ overload, ROS generation, and mitochondrial alteration upon overexpression of MCU in T. brucei has been reported (77). In contrast to mitochondrial Ca2+ overload, decreased mitochondrial Ca2+ uptake leads to increase in the AMP/ATP ratio and autophagy, suggesting that a delicate bioenergetic balance is required to keep the cells alive (77) (Fig. 8).

FIG. 8.

FIG. 8.

Consequences of decreased and increased mitochondrial Ca2+ uptake. Reduced Ca2+ uptake leads to decreased ATP synthesis, resulting in a higher AMP/ATP ratio and autophagy, while increased Ca2+ uptake leads to mitochondrial Ca2+ overload, ROS generation, and cell death. Background shows a procyclic form of Trypanosoma brucei. ROS, reactive oxygen species. Color images are available online.

Opening of the Mitochondrial Permeability Transition Pore and Cell Death in Trypanosomes

The mitochondrial permeability transition pore (mPTP) is an alternative mechanism to the mitochondrial (intrinsic) pathway of apoptosis and a mediator of cell death mechanisms in mammals (29). The mPTP is a nonselective channel of high conductance located at contact sites between the inner and outer mitochondrial membranes. Several proteins have been shown to be components of this channel, including voltage-dependent anion channels (VDAC), ANT, PiC, cyclophilin D (CypD), and other proteins such as dimers of the ATP synthase (68). Opening of the mPTP is increased by Ca2+ overload, thiol oxidation, pyridine nucleotide oxidation, oxidative stress, alkalinization, or low transmembrane potential. Cyclosporin A, which binds to CypD, inhibits its opening. Opening of this pore determines mitochondrial dysfunction and cell death by either apoptosis or necrosis (61). The intramitochondrial Ca2+ plays an important role in control of the redox balance that affects the opening of the PTP (148).

It has been suggested that an mPTP may be formed in T. cruzi based on the loss of mitochondrial membrane potential and cell viability when a CypD homolog (TcPyD22) is overexpressed and H2O2 is present. This mechanism is sensitive to cyclosporin A (22, 23).

The c-ring of the ATP synthase (3, 16) or a channel inside ATP synthase dimers has been suggested as forming the pore (16, 68) of the mPTP. However, ATP synthases devoid of c subunits maintain mPTP formation characteristics (73), suggesting that the c-ring is not the channel. The interaction between the MCU and the c subunit of the ATP synthase described above suggests a role for the MCU complex in formation of the mPTP, which needs to be investigated.

Conclusions

The mitochondria of trypanosomatids have mechanisms for ROS generation and defenses against their toxicity, and Ca2+ overload has a role in ROS generation and cell death. However, there are still several gaps in our knowledge of these mechanisms and roles. The mitochondrial sites for ROS generation have not been characterized in detail, and while defenses against ROS production depend on the T(SH)2 system, it is not known whether the T(SH)2/T(S)2 couple has access to the mitochondrial matrix or they exchange with their cytosolic pools. It is not yet clear whether opening of an mPTP occurs in these mitochondria, and which is its composition.

Acknowledgments

The authors thank the members of their laboratories for useful discussions. R.D. is a Barbara and Sanford Orkin/Georgia Research Alliance Eminent Scholar. A.E.V. is Doctor Honoris Causa of the Department of Biochemistry, Medical School, University of the Republic, Uruguay.

Abbreviations Used

ANT

adenine nucleotide translocase

APx

ascorbate peroxidase

Ca2+

calcium ion

CcPx

cytochrome c peroxidase

Ccr

reduced cytochrome c

CypD

cyclophilin D

DH

dehydrogenase

dhA

dehydroascorbate

DHAP

dihydroxyacetone phosphate

DIC

differential interference contrast

EMRE

essential mitochondrial regulator

FR

fumarate reductase

G3P

glycerol-3-phosphate

G3PDH

glycerol-3-phosphate dehydrogenase

GSH

glutathione

GSHPx

glutathione peroxidase

GSSG

glutathione disulfide

H2O2

hydrogen peroxide

IMS

intermembrane space;

IP3R

inositol 1,4,5-trisphosphate receptor

MCU

mitochondrial calcium uniporter

MCUR1

mitochondrial calcium uniporter regulator 1

MICU

mitochondrial calcium uptake

mPTP

mitochondrial permeability transition pore

MT

MitoTracker

O2•−

superoxide anion

PiC

inorganic phosphate carrier

Prx

peroxiredoxin

RC

respiratory chain

RNAi

RNA interference

ROS

reactive oxygen species

SOD

superoxide dismutase

TryR

trypanothione disulfide reductase

TryS

trypanothione synthetase

TryX

tryparedoxin

TryXPx

tryparedoxin peroxidase

TryX(S)2

tryparedoxin disulfide

T(S)2

trypanothione disulfide

T(SH)2

trypanothione

UQ

ubiquinone

Authors' Contributions

R.D. was involved in conceptualization, writing—draft, and writing—review and editing; and A.E.V. was involved in conceptualization, writing—draft, and writing—review and editing.

Author Disclosure Statement

The authors declare no conflicts of interest.

Funding Information

Work in the authors' laboratories is funded by the U.S. National Institutes of Health (grant AI108222 to R.D.) and the Brazilian State Agency Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP; grant 2017/17728-8 to A.E.V.).

References

  • 1. Adak S and Datta AK. Leishmania major encodes an unusual peroxidase that is a close homologue of plant ascorbate peroxidase: a novel role of the transmembrane domain. Biochem J 390: 465–474, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Aguirre G, Cabrera E, Cerecetto H, Di Maio R, Gonzalez M, Seoane G, Duffaut A, Denicola A, Gil MJ, and Martinez-Merino V. Design, synthesis and biological evaluation of new potent 5-nitrofuryl derivatives as anti-Trypanosoma cruzi agents. Studies of trypanothione binding site of trypanothione reductase as target for rational design. Eur J Med Chem 39: 421–431, 2004. [DOI] [PubMed] [Google Scholar]
  • 3. Alavian KN, Beutner G, Lazrove E, Sacchetti S, Park HA, Licznerski P, Li H, Nabili P, Hockensmith K, Graham M, Porter GA Jr., and Jonas EA. An uncoupling channel within the c-subunit ring of the F1F0 ATP synthase is the mitochondrial permeability transition pore. Proc Natl Acad Sci U S A 111: 10580–10585, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Alvarez MN, Peluffo G, Piacenza L, and Radi R. Intraphagosomal peroxynitrite as a macrophage-derived cytotoxin against internalized Trypanosoma cruzi: consequences for oxidative killing and role of microbial peroxiredoxins in infectivity. J Biol Chem 286: 6627–6640, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Ameisen JC, Idziorek T, Billaut-Mulot O, Loyens M, Tissier JP, Potentier A, and Ouaissi A. Apoptosis in a unicellular eukaryote (Trypanosoma cruzi): implications for the evolutionary origin and role of programmed cell death in the control of cell proliferation, differentiation and survival. Cell Death Differ 2: 285–300, 1995. [PubMed] [Google Scholar]
  • 6. Ariyanayagam MR and Fairlamb AH. Ovothiol and trypanothione as antioxidants in trypanosomatids. Mol Biochem Parasitol 115: 189–198, 2001. [DOI] [PubMed] [Google Scholar]
  • 7. Ariyanayagam MR, Oza SL, Mehlert A, and Fairlamb AH. Bis(glutathionyl)spermine and other novel trypanothione analogues in Trypanosoma cruzi. J Biol Chem 278: 27612–27619, 2003. [DOI] [PubMed] [Google Scholar]
  • 8. Atwood JA, 3rd, Weatherly DB, Minning TA, Bundy B, Cavola C, Opperdoes FR, Orlando R, and Tarleton RL. The Trypanosoma cruzi proteome. Science 309: 473–476, 2005. [DOI] [PubMed] [Google Scholar]
  • 9. Baillet S, Buisine E, Horvath D, Maes L, Bonnet B, and Sergheraert C. 2-Amino diphenylsulfides as inhibitors of trypanothione reductase: modification of the side chain. Bioorg Med Chem 4: 891–899, 1996. [DOI] [PubMed] [Google Scholar]
  • 10. Baradaran R, Wang C, Siliciano AF, and Long SB. Cryo-EM structures of fungal and metazoan mitochondrial calcium uniporters. Nature 559: 580–584, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Baughman JM, Perocchi F, Girgis HS, Plovanich M, Belcher-Timme CA, Sancak Y, Bao XR, Strittmatter L, Goldberger O, Bogorad RL, Koteliansky V, and Mootha VK. Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature 476: 341–345, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Benaim G, Bermudez R, and Urbina JA. Ca2+ transport in isolated mitochondrial vesicles from Leishmania braziliensis promastigotes. Mol Biochem Parasitol 39: 61–68, 1990. [DOI] [PubMed] [Google Scholar]
  • 13. Benson TJ, McKie JH, Garforth J, Borges A, Fairlamb AH, and Douglas KT. Rationally designed selective inhibitors of trypanothione reductase. Phenothiazines and related tricyclics as lead structures. Biochem J 286: 9–11, 1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Bond CS, Zhang Y, Berriman M, Cunningham ML, Fairlamb AH, and Hunter WN. Crystal structure of Trypanosoma cruzi trypanothione reductase in complex with trypanothione, and the structure-based discovery of new natural product inhibitors. Structure 7: 81–89, 1999. [DOI] [PubMed] [Google Scholar]
  • 15. Bonnet B, Soullez D, Davioud-Charvet E, Landry V, Horvath D, and Sergheraert C. New spermine and spermidine derivatives as potent inhibitors of Trypanosoma cruzi trypanothione reductase. Bioorg Med Chem 5: 1249–1256, 1997. [DOI] [PubMed] [Google Scholar]
  • 16. Bonora M, Bononi A, De Marchi E, Giorgi C, Lebiedzinska M, Marchi S, Patergnani S, Rimessi A, Suski JM, Wojtala A, Wieckowski MR, Kroemer G, Galluzzi L, and Pinton P. Role of the c subunit of the F0 ATP synthase in mitochondrial permeability transition. Cell Cycle 12: 674–683, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Bonse S, Richards JM, Ross SA, Lowe G, and Krauth-Siegel RL. (2,2′:6′,2″-Terpyridine)platinum(II) complexes are irreversible inhibitors of Trypanosoma cruzi trypanothione reductase but not of human glutathione reductase. J Med Chem 43: 4812–4821, 2000. [DOI] [PubMed] [Google Scholar]
  • 18. Bonse S, Santelli-Rouvier C, Barbe J, and Krauth-Siegel RL. Inhibition of Trypanosoma cruzi trypanothione reductase by acridines: kinetic studies and structure-activity relationships. J Med Chem 42: 5448–5454, 1999. [DOI] [PubMed] [Google Scholar]
  • 19. Boveris A, Sies H, Martino EE, Docampo R, Turrens JF, and Stoppani AO. Deficient metabolic utilization of hydrogen peroxide in Trypanosoma cruzi. Biochem J 188: 643–648, 1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Brand MD. Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling. Free Radic Biol Med 100: 14–31, 2016. [DOI] [PubMed] [Google Scholar]
  • 21. Braunshausen A and Seebeck FP. Identification and characterization of the first ovothiol biosynthetic enzyme. J Am Chem Soc 133: 1757–1759, 2011. [DOI] [PubMed] [Google Scholar]
  • 22. Bustos PL, Perrone AE, Milduberger NA, and Bua J. Mitochondrial permeability transition in protozoan parasites: what we learned from Trypanosoma cruzi. Cell Death Dis 8: e3057, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Bustos PL, Perrone AE, Milduberger N, Postan M, and Bua J. Oxidative stress damage in the protozoan parasite Trypanosoma cruzi is inhibited by Cyclosporin A. Parasitology 142: 1024–1032, 2015. [DOI] [PubMed] [Google Scholar]
  • 24. Cadenas E, Boveris A, Ragan CI, and Stoppani AO. Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria. Arch Biochem Biophys 180: 248–257, 1977. [DOI] [PubMed] [Google Scholar]
  • 25. Carafoli E, Balcavage WX, Lehninger AL, and Mattoon JR. Ca2+ metabolism in yeast cells and mitochondria. Biochim Biophys Acta 205: 18–26, 1970. [DOI] [PubMed] [Google Scholar]
  • 26. Carafoli E and Lehninger AL. A survey of the interaction of calcium ions with mitochondria from different tissues and species. Biochem J 122: 681–690, 1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Cardenas C, Miller RA, Smith I, Bui T, Molgo J, Muller M, Vais H, Cheung KH, Yang J, Parker I, Thompson CB, Birnbaum MJ, Hallows KR, and Foskett JK. Essential regulation of cell bioenergetics by constitutive InsP3 receptor Ca2+ transfer to mitochondria. Cell 142: 270–283, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Carranza JC, Kowaltowski AJ, Mendonca MA, de Oliveira TC, Gadelha FR, and Zingales B. Mitochondrial bioenergetics and redox state are unaltered in Trypanosoma cruzi isolates with compromised mitochondrial complex I subunit genes. J Bioenerg Biomembr 41: 299–308, 2009. [DOI] [PubMed] [Google Scholar]
  • 29. Carraro M and Bernardi P. Calcium and reactive oxygen species in regulation of the mitochondrial permeability transition and of programmed cell death in yeast. Cell Calcium 60: 102–107, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Castro H and Tomas AM. Peroxidases of trypanosomatids. Antioxid Redox Signal 10: 1593–1606, 2008. [DOI] [PubMed] [Google Scholar]
  • 31. Cavalcanti DP and de Souza W. The kinetoplast of trypanosomatids: from early studies of electron microscopy to recent advances in atomic force microscopy. Scanning 2018: 9603051, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Chan C, Yin H, Garforth J, McKie JH, Jaouhari R, Speers P, Douglas KT, Rock PJ, Yardley V, Croft SL, and Fairlamb AH. Phenothiazine inhibitors of trypanothione reductase as potential antitrypanosomal and antileishmanial drugs. J Med Chem 41: 148–156, 1998. [DOI] [PubMed] [Google Scholar]
  • 33. Chiurillo MA, Lander N, Vercesi AE, and Docampo R. IP3 receptor-mediated Ca2+ release from acidocalcisomes regulates mitochondrial bioenergetics and prevents autophagy in Trypanosoma cruzi. Cell Calcium 92: 102284, 2020. [DOI] [PubMed] [Google Scholar]
  • 34. Comini M, Menge U, and Flohe L. Biosynthesis of trypanothione in Trypanosoma brucei brucei. Biol Chem 384: 653–656, 2003. [DOI] [PubMed] [Google Scholar]
  • 35. Cota BB, Rosa LH, Fagundes EM, Martins-Filho OA, Correa-Oliveira R, Romanha AJ, Rosa CA, and Zani CL. A potent trypanocidal component from the fungus Lentinus strigosus inhibits trypanothione reductase and modulates PBMC proliferation. Mem Inst Oswaldo Cruz 103: 263–270, 2008. [DOI] [PubMed] [Google Scholar]
  • 36. Dass S, Mather MW, and Ke H. Divergent mitochondrial ribosomes in unicellular parasitic protozoans. Trends Parasitol 36: 318–321, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. De Stefani D, Raffaello A, Teardo E, Szabo I, and Rizzuto R. A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporter. Nature 476: 336–340, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Deluca HF and Engstrom GW. Calcium uptake by rat kidney mitochondria. Proc Natl Acad Sci U S A 47: 1744–1750, 1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Docampo R. Sensitivity of parasites to free radical damage by antiparasitic drugs. Chem Biol Interact 73: 1–27, 1990. [DOI] [PubMed] [Google Scholar]
  • 40. Docampo R, Cruz FS, Boveris A, Muniz RP, and Esquivel DM. Lipid peroxidation and the generation of free radicals, superoxide anion, and hydrogen peroxide in beta-lapachone-treated Trypanosoma cruzi epimastigotes. Arch Biochem Biophys 186: 292–297, 1978. [DOI] [PubMed] [Google Scholar]
  • 41. Docampo R, de Boiso JF, Boveris A, and Stoppani AO. Localization of peroxidase activity in Trypanosoma cruzi microbodies. Experientia 32: 972–975, 1976. [DOI] [PubMed] [Google Scholar]
  • 42. Docampo R, De Souza W, Cruz FS, Roitman I, Cover B, and Gutteridge WE. Ultrastructural alterations and peroxide formation induced by naphthoquinones in different stages of Trypanosoma cruzi. Z Parasitenkd 57: 189–198, 1978. [DOI] [PubMed] [Google Scholar]
  • 43. Docampo R, Lopes JN, Cruz FS, and Souza W. Trypanosoma cruzi: ultrastructural and metabolic alterations of epimastigotes by beta-lapachone. Exp Parasitol 42: 142–149, 1977. [DOI] [PubMed] [Google Scholar]
  • 44. Docampo R and Lukes J. Trypanosomes and the solution to a 50-year mitochondrial calcium mystery. Trends Parasitol 28: 31–37, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Docampo R and Moreno SN. Free radical metabolites in the mode of action of chemotherapeutic agents and phagocytic cells on Trypanosoma cruzi. Rev Infect Dis 6: 223–238, 1984. [DOI] [PubMed] [Google Scholar]
  • 46. Docampo R and Moreno SN. Free radical metabolism of antiparasitic agents. Fed Proc 45: 2471–2476, 1986. [PubMed] [Google Scholar]
  • 47. Docampo R and Moreno SN. Biochemical toxicology of antiparasitic drugs used in the chemotherapy and chemoprophylaxis of American trypanosomiasis (Chagas' disease). Rev Biochem Toxicol 7: 159–204, 1985. [Google Scholar]
  • 48. Docampo R and Vercesi AE. Ca2+ transport by coupled Trypanosoma cruzi mitochondria in situ. J Biol Chem 264: 108–111, 1989. [PubMed] [Google Scholar]
  • 49. Docampo R and Vercesi AE. Characteristics of Ca2+ transport by Trypanosoma cruzi mitochondria in situ. Arch Biochem Biophys 272: 122–129, 1989. [DOI] [PubMed] [Google Scholar]
  • 50. Docampo R, Vercesi AE, and Huang G. Mitochondrial calcium transport in trypanosomes. Mol Biochem Parasitol 196: 108–116, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Docampo R, Vercesi AE, Huang G, Lander N, Chiurillo MA, and Bertolini M. Mitochondrial Ca2+ homeostasis in trypanosomes. Inter Rev Cell Mol Biol 362: 261–289, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Dolai S, Yadav RK, Pal S, and Adak S. Leishmania major ascorbate peroxidase overexpression protects cells against reactive oxygen species-mediated cardiolipin oxidation. Free Radic Biol Med 45: 1520–1529, 2008. [DOI] [PubMed] [Google Scholar]
  • 53. Dolai S, Yadav RK, Pal S, and Adak S. Overexpression of mitochondrial Leishmania major ascorbate peroxidase enhances tolerance to oxidative stress-induced programmed cell death and protein damage. Eukaryot Cell 8: 1721–1731, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. dos Santos GRR, Rezende Leite AC, Lander N, Chiurillo MA, Vercesi AE, and Docampo R. Trypanosoma cruzi Letm1 is involved in mitochondrial Ca2+ transport, and is essential for replication, differentiation, and host cell invasion. FASEB J 35: e21685, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Dotiwala F, Mulik S, Polidoro RB, Ansara JA, Burleigh BA, Walch M, Gazzinelli RT, and Lieberman J. Killer lymphocytes use granulysin, perforin and granzymes to kill intracellular parasites. Nat Med 22: 210–216, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Dufernez F, Yernaux C, Gerbod D, Noel C, Chauvenet M, Wintjens R, Edgcomb VP, Capron M, Opperdoes FR, and Viscogliosi E. The presence of four iron-containing superoxide dismutase isozymes in trypanosomatidae: characterization, subcellular localization, and phylogenetic origin in Trypanosoma brucei. Free Radic Biol Med 40: 210–225, 2006. [DOI] [PubMed] [Google Scholar]
  • 57. Ebersoll S, Bogacz M, Gunter LM, Dick TP, and Krauth-Siegel RL. A tryparedoxin-coupled biosensor reveals a mitochondrial trypanothione metabolism in trypanosomes. Elife 9: e53227, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Fan C, Fan M, Orlando BJ, Fastman NM, Zhang J, Xu Y, Chambers MG, Xu X, Perry K, Liao M, and Feng L. X-ray and cryo-EM structures of the mitochondrial calcium uniporter. Nature 559: 575–579, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Fang J and Beattie DS. Rotenone-insensitive NADH dehydrogenase is a potential source of superoxide in procyclic Trypanosoma brucei mitochondria. Mol Biochem Parasitol 123: 135–142, 2002. [DOI] [PubMed] [Google Scholar]
  • 60. Fang J and Beattie DS. Identification of a gene encoding a 54 kDa alternative NADH dehydrogenase in Trypanosoma brucei. Mol Biochem Parasitol 127: 73–77, 2003. [DOI] [PubMed] [Google Scholar]
  • 61. Figueira TR, Barros MH, Camargo AA, Castilho RF, Ferreira JC, Kowaltowski AJ, Sluse FE, Souza-Pinto NC, and Vercesi AE. Mitochondria as a source of reactive oxygen and nitrogen species: from molecular mechanisms to human health. Antioxid Redox Signal 18: 2029–2074, 2013. [DOI] [PubMed] [Google Scholar]
  • 62. Flohe L, Hecht HJ, and Steinert P. Glutathione and trypanothione in parasitic hydroperoxide metabolism. Free Radic Biol Med 27: 966–984, 1999. [DOI] [PubMed] [Google Scholar]
  • 63. Fournet A, Inchausti A, Yaluff G, Rojas De Arias A, Guinaudeau H, Bruneton J, Breidenbach MA, Karplus PA, and Faerman CH. Trypanocidal bisbenzylisoquinoline alkaloids are inhibitors of trypanothione reductase. J Enzyme Inhib 13: 1–9, 1998. [DOI] [PubMed] [Google Scholar]
  • 64. Gallwitz H, Bonse S, Martinez-Cruz A, Schlichting I, Schumacher K, and Krauth-Siegel RL. Ajoene is an inhibitor and subversive substrate of human glutathione reductase and Trypanosoma cruzi trypanothione reductase: crystallographic, kinetic, and spectroscopic studies. J Med Chem 42: 364–372, 1999. [DOI] [PubMed] [Google Scholar]
  • 65. Gannavaram S and Debrabant A. Involvement of TatD nuclease during programmed cell death in the protozoan parasite Trypanosoma brucei. Mol Microbiol 83: 926–935, 2012. [DOI] [PubMed] [Google Scholar]
  • 66. Gannavaram S, Vedvyas C, and Debrabant A. Conservation of the pro-apoptotic nuclease activity of endonuclease G in unicellular trypanosomatid parasites. J Cell Sci 121: 99–109, 2008. [DOI] [PubMed] [Google Scholar]
  • 67. Garforth J, Yin H, McKie JH, Douglas KT, and Fairlamb AH. Rational design of selective ligands for trypanothione reductase from Trypanosoma cruzi. Structural effects on the inhibition by dibenzazepines based on imipramine. J Enzyme Inhib 12: 161–173, 1997. [DOI] [PubMed] [Google Scholar]
  • 68. Giorgio V, von Stockum S, Antoniel M, Fabbro A, Fogolari F, Forte M, Glick GD, Petronilli V, Zoratti M, Szabo I, Lippe G, and Bernardi P. Dimers of mitochondrial ATP synthase form the permeability transition pore. Proc Natl Acad Sci U S A 110: 5887–5892, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Groden D and Beck E. H2O2 destruction by ascorbate-dependent systems from chloroplasts. Biochim Biophys Acta 546: 426–435, 1979. [DOI] [PubMed] [Google Scholar]
  • 70. Guerrero SA, Lopez JA, Steinert P, Montemartini M, Kalisz HM, Colli W, Singh M, Alves MJ, and Flohe L. His-tagged tryparedoxin peroxidase of Trypanosoma cruzi as a tool for drug screening. Appl Microbiol Biotechnol 53: 410–414, 2000. [DOI] [PubMed] [Google Scholar]
  • 71. Gutierrez-Correa J, Fairlamb AH, and Stoppani AO. Trypanosoma cruzi trypanothione reductase is inactivated by peroxidase-generated phenothiazine cationic radicals. Free Radic Res 34: 363–378, 2001. [DOI] [PubMed] [Google Scholar]
  • 72. Hashimi H, McDonald L, Stribrna E, and Lukes J. Trypanosome Letm1 protein is essential for mitochondrial potassium homeostasis. J Biol Chem 288: 26914–26925, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. He J, Carroll J, Ding S, Fearnley IM, and Walker JE. Permeability transition in human mitochondria persists in the absence of peripheral stalk subunits of ATP synthase. Proc Natl Acad Sci U S A 114: 9086–9091, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Hillebrand H, Schmidt A, and Krauth-Siegel RL. A second class of peroxidases linked to the trypanothione metabolism. J Biol Chem 278: 6809–6815, 2003. [DOI] [PubMed] [Google Scholar]
  • 75. Huang G and Docampo R. The mitochondrial Ca2+ uniporter complex (MCUC) of Trypanosoma brucei is a hetero-oligomer that contains novel subunits essential for Ca2+ uptake. mBio 9: e01700-18, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Huang G and Docampo R. The mitochondrial calcium uniporter interacts with subunit c of the ATP synthase of trypanosomes and humans. mBio 11: e00268-20, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Huang G, Vercesi AE, and Docampo R. Essential regulation of cell bioenergetics in Trypanosoma brucei by the mitochondrial calcium uniporter. Nat Commun 4: 2865, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Hugo M, Martinez A, Trujillo M, Estrada D, Mastrogiovanni M, Linares E, Augusto O, Issoglio F, Zeida A, Estrin DA, Heijnen HF, Piacenza L, and Radi R. Kinetics, subcellular localization, and contribution to parasite virulence of a Trypanosoma cruzi hybrid type A heme peroxidase (TcAPx-CcP). Proc Natl Acad Sci U S A 114: E1326–E1335, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Irigoin F, Cibils L, Comini MA, Wilkinson SR, Flohe L, and Radi R. Insights into the redox biology of Trypanosoma cruzi: trypanothione metabolism and oxidant detoxification. Free Radic Biol Med 45: 733–742, 2008. [DOI] [PubMed] [Google Scholar]
  • 80. Irigoin F, Inada NM, Fernandes MP, Piacenza L, Gadelha FR, Vercesi AE, and Radi R. Mitochondrial calcium overload triggers complement-dependent superoxide-mediated programmed cell death in Trypanosoma cruzi. Biochem J 418: 595–604, 2009. [DOI] [PubMed] [Google Scholar]
  • 81. Ismail SO, Paramchuk W, Skeiky YA, Reed SG, Bhatia A, and Gedamu L. Molecular cloning and characterization of two iron superoxide dismutase cDNAs from Trypanosoma cruzi. Mol Biochem Parasitol 86: 187–197, 1997. [DOI] [PubMed] [Google Scholar]
  • 82. Jacoby EM, Schlichting I, Lantwin CB, Kabsch W, and Krauth-Siegel RL. Crystal structure of the Trypanosoma cruzi trypanothione reductase.mepacrine complex. Proteins 24: 73–80, 1996. [DOI] [PubMed] [Google Scholar]
  • 83. Jasion VS, Doukov T, Pineda SH, Li H, and Poulos TL. Crystal structure of the Leishmania major peroxidase-cytochrome c complex. Proc Natl Acad Sci U S A 109: 18390–18394, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Jasion VS and Poulos TL. Leishmania major peroxidase is a cytochrome c peroxidase. Biochemistry 51: 2453–2460, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Jiang D, Zhao L, and Clapham DE. Genome-wide RNAi screen identifies Letm1 as a mitochondrial Ca2+/H+ antiporter. Science 326: 144–147, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Jockers-Scherubl MC, Schirmer RH, and Krauth-Siegel RL. Trypanothione reductase from Trypanosoma cruzi. Catalytic properties of the enzyme and inhibition studies with trypanocidal compounds. Eur J Biochem 180: 267–272, 1989. [DOI] [PubMed] [Google Scholar]
  • 87. Khan MO, Austin SE, Chan C, Yin H, Marks D, Vaghjiani SN, Kendrick H, Yardley V, Croft SL, and Douglas KT. Use of an additional hydrophobic binding site, the Z site, in the rational drug design of a new class of stronger trypanothione reductase inhibitor, quaternary alkylammonium phenothiazines. J Med Chem 43: 3148–3156, 2000. [DOI] [PubMed] [Google Scholar]
  • 88. Kosec G, Alvarez VE, Aguero F, Sanchez D, Dolinar M, Turk B, Turk V, and Cazzulo JJ. Metacaspases of Trypanosoma cruzi: possible candidates for programmed cell death mediators. Mol Biochem Parasitol 145: 18–28, 2006. [DOI] [PubMed] [Google Scholar]
  • 89. Krauth-Siegel RL and Comini MA. Redox control in trypanosomatids, parasitic protozoa with trypanothione-based thiol metabolism. Biochim Biophys Acta 1780: 1236–1248, 2008. [DOI] [PubMed] [Google Scholar]
  • 90. Krauth-Siegel RL, Sticherling C, Jost I, Walsh CT, Pai EF, Kabsch W, and Lantwin CB. Crystallization and preliminary crystallographic analysis of trypanothione reductase from Trypanosoma cruzi, the causative agent of Chagas' disease. FEBS Lett 317: 105–108, 1993. [DOI] [PubMed] [Google Scholar]
  • 91. Lander N, Chiurillo MA, and Docampo R. Signaling pathways involved in environmental sensing in Trypanosoma cruzi. Mol Microbiol 115: 819–828, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Lantwin CB, Schlichting I, Kabsch W, Pai EF, and Krauth-Siegel RL. The structure of Trypanosoma cruzi trypanothione reductase in the oxidized and NADPH reduced state. Proteins 18: 161–173, 1994. [DOI] [PubMed] [Google Scholar]
  • 93. Lee B, Bauer H, Melchers J, Ruppert T, Rattray L, Yardley V, Davioud-Charvet E, and Krauth-Siegel RL. Irreversible inactivation of trypanothione reductase by unsaturated Mannich bases: a divinyl ketone as key intermediate. J Med Chem 48: 7400–7410, 2005. [DOI] [PubMed] [Google Scholar]
  • 94. Li Z, Fennie MW, Ganem B, Hancock MT, Kobaslija M, Rattendi D, Bacchi CJ, and O'Sullivan MC. Polyamines with N-(3-phenylpropyl) substituents are effective competitive inhibitors of trypanothione reductase and trypanocidal agents. Bioorg Med Chem Lett 11: 251–254, 2001. [DOI] [PubMed] [Google Scholar]
  • 95. Logan FJ, Taylor MC, Wilkinson SR, Kaur H, and Kelly JM. The terminal step in vitamin C biosynthesis in Trypanosoma cruzi is mediated by a FMN-dependent galactonolactone oxidase. Biochem J 407: 419–426, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Lopez JA, Carvalho TU, de Souza W, Flohe L, Guerrero SA, Montemartini M, Kalisz HM, Nogoceke E, Singh M, Alves MJ, and Colli W. Evidence for a trypanothione-dependent peroxidase system in Trypanosoma cruzi. Free Radic Biol Med 28: 767–772, 2000. [DOI] [PubMed] [Google Scholar]
  • 97. Mallilankaraman K, Cardenas C, Doonan PJ, Chandramoorthy HC, Irrinki KM, Golenar T, Csordas G, Madireddi P, Yang J, Muller M, Miller R, Kolesar JE, Molgo J, Kaufman B, Hajnoczky G, Foskett JK, and Madesh M. MCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolism. Nat Cell Biol 14: 1336–1343, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Martinez A, Peluffo G, Petruk AA, Hugo M, Pineyro D, Demicheli V, Moreno DM, Lima A, Batthyany C, Duran R, Robello C, Marti MA, Larrieux N, Buschiazzo A, Trujillo M, Radi R, and Piacenza L. Structural and molecular basis of the peroxynitrite-mediated nitration and inactivation of Trypanosoma cruzi iron-superoxide dismutases (Fe-SODs) A and B: disparate susceptibilities due to the repair of Tyr35 radical by Cys83 in Fe-SODB through intramolecular electron transfer. J Biol Chem 289: 12760–12778, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Maslov DA. Separating the wheat from the chaff: RNA editing and selection of translatable mRNA in trypanosome mitochondria. Pathogens 8: 105, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Mateo H, Marin C, Perez-Cordon G, and Sanchez-Moreno M. Purification and biochemical characterization of four iron superoxide dismutases in Trypanosoma cruzi. Mem Inst Oswaldo Cruz 103: 271–276, 2008. [DOI] [PubMed] [Google Scholar]
  • 101. McCormack JG and Denton RM. Ca2+ as a second messenger within mitochondria. Trends Biochem Sci 11: 258–262, 1986. [Google Scholar]
  • 102. Meshnick SR, Kitchener KR, and Trang NL. Trypanosomatid iron-superoxide dismutase inhibitors. Selectivity and mechanism of N1,N6-bis(2,3-dihydroxybenzoyl)-1,6-diaminohexane. Biochem Pharmacol 34: 3147–3152, 1985. [DOI] [PubMed] [Google Scholar]
  • 103. Mittra B, Laranjeira-Silva MF, Miguel DC, Perrone Bezerra de Menezes J, and Andrews NW. The iron-dependent mitochondrial superoxide dismutase SODA promotes Leishmania virulence. J Biol Chem 292: 12324–12338, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Moreno SN, Carnieri EG, and Docampo R. Inhibition of Trypanosoma cruzi trypanothione reductase by crystal violet. Mol Biochem Parasitol 67: 313–320, 1994. [DOI] [PubMed] [Google Scholar]
  • 105. Nguyen NX, Armache JP, Lee C, Yang Y, Zeng W, Mootha VK, Cheng Y, Bai XC, and Jiang Y. Cryo-EM structure of a fungal mitochondrial calcium uniporter. Nature 559: 570–574, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Nogueira FB, Rodrigues JF, Correa MM, Ruiz JC, Romanha AJ, and Murta SM. The level of ascorbate peroxidase is enhanced in benznidazole-resistant populations of Trypanosoma cruzi and its expression is modulated by stress generated by hydrogen peroxide. Mem Inst Oswaldo Cruz 107: 494–502, 2012. [DOI] [PubMed] [Google Scholar]
  • 107. O'Sullivan MC, Dalrymple DM, and Zhou Q. Inhibiting effects of spermidine derivatives on Trypanosoma cruzi trypanothione reductase. J Enzyme Inhib 11: 97–114, 1996. [DOI] [PubMed] [Google Scholar]
  • 108. O'Sullivan MC, Zhou Q, Li Z, Durham TB, Rattendi D, Lane S, and Bacchi CJ. Polyamine derivatives as inhibitors of trypanothione reductase and assessment of their trypanocidal activities. Bioorg Med Chem 5: 2145–2155, 1997. [DOI] [PubMed] [Google Scholar]
  • 109. Olmo F, Urbanova K, Rosales MJ, Martin-Escolano R, Sanchez-Moreno M, and Marin C. An in vitro iron superoxide dismutase inhibitor decreases the parasitemia levels of Trypanosoma cruzi in BALB/c mouse model during acute phase. Int J Parasitol Drugs Drug Resist 5: 110–116, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Oxenoid K, Dong Y, Cao C, Cui T, Sancak Y, Markhard AL, Grabarek Z, Kong L, Liu Z, Ouyang B, Cong Y, Mootha VK, and Chou JJ. Architecture of the mitochondrial calcium uniporter. Nature 533: 269–273, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Oza SL, Ariyanayagam MR, and Fairlamb AH. Characterization of recombinant glutathionylspermidine synthetase/amidase from Crithidia fasciculata. Biochem J 364: 679–686, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Oza SL, Shaw MP, Wyllie S, and Fairlamb AH. Trypanothione biosynthesis in Leishmania major. Mol Biochem Parasitol 139: 107–116, 2005. [DOI] [PubMed] [Google Scholar]
  • 113. Oza SL, Tetaud E, Ariyanayagam MR, Warnon SS, and Fairlamb AH. A single enzyme catalyses formation of trypanothione from glutathione and spermidine in Trypanosoma cruzi. J Biol Chem 277: 35853–35861, 2002. [DOI] [PubMed] [Google Scholar]
  • 114. Palty R, Silverman WF, Hershfinkel M, Caporale T, Sensi SL, Parnis J, Nolte C, Fishman D, Shoshan-Barmatz V, Herrmann S, Khananshvili D, and Sekler I. NCLX is an essential component of mitochondrial Na+/Ca2+ exchange. Proc Natl Acad Sci U S A 107: 436–441, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Pan X, Liu J, Nguyen T, Liu C, Sun J, Teng Y, Fergusson MM, Rovira, II, Allen M, Springer DA, Aponte AM, Gucek M, Balaban RS, Murphy E, and Finkel T. The physiological role of mitochondrial calcium revealed by mice lacking the mitochondrial calcium uniporter. Nat Cell Biol 15: 1464–1472, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Paramchuk WJ, Ismail SO, Bhatia A, and Gedamu L. Cloning, characterization and overexpression of two iron superoxide dismutase cDNAs from Leishmania chagasi: role in pathogenesis. Mol Biochem Parasitol 90: 203–221, 1997. [DOI] [PubMed] [Google Scholar]
  • 117. Patel S, Hussain S, Harris R, Sardiwal S, Kelly JM, Wilkinson SR, Driscoll PC, and Djordjevic S. Structural insights into the catalytic mechanism of Trypanosoma cruzi GPXI (glutathione peroxidase-like enzyme I). Biochem J 425: 513–522, 2010. [DOI] [PubMed] [Google Scholar]
  • 118. Perocchi F, Gohil VM, Girgis HS, Bao XR, McCombs JE, Palmer AE, and Mootha VK. MICU1 encodes a mitochondrial EF hand protein required for Ca2+ uptake. Nature 467: 291–296, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Piacenza L, Irigoin F, Alvarez MN, Peluffo G, Taylor MC, Kelly JM, Wilkinson SR, and Radi R. Mitochondrial superoxide radicals mediate programmed cell death in Trypanosoma cruzi: cytoprotective action of mitochondrial iron superoxide dismutase overexpression. Biochem J 403: 323–334, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Piacenza L, Peluffo G, Alvarez MN, Kelly JM, Wilkinson SR, and Radi R. Peroxiredoxins play a major role in protecting Trypanosoma cruzi against macrophage- and endogenously-derived peroxynitrite. Biochem J 410: 359–368, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Piacenza L, Peluffo G, Alvarez MN, Martinez A, and Radi R. Trypanosoma cruzi antioxidant enzymes as virulence factors in Chagas disease. Antioxid Redox Signal 19: 723–734, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Piacenza L, Peluffo G, and Radi R. L-arginine-dependent suppression of apoptosis in Trypanosoma cruzi: contribution of the nitric oxide and polyamine pathways. Proc Natl Acad Sci U S A 98: 7301–7306, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Piacenza L, Zago MP, Peluffo G, Alvarez MN, Basombrio MA, and Radi R. Enzymes of the antioxidant network as novel determiners of Trypanosoma cruzi virulence. Int J Parasitol 39: 1455–1464, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Pineyro MD, Arcari T, Robello C, Radi R, and Trujillo M. Tryparedoxin peroxidases from Trypanosoma cruzi: high efficiency in the catalytic elimination of hydrogen peroxide and peroxynitrite. Arch Biochem Biophys 507: 287–295, 2011. [DOI] [PubMed] [Google Scholar]
  • 125. Pineyro MD, Arias D, Ricciardi A, Robello C, and Parodi-Talice A. Oligomerization dynamics and functionality of Trypanosoma cruzi cytosolic tryparedoxin peroxidase as peroxidase and molecular chaperone. Biochim Biophys Acta Gen Subj 1863: 1583–1594, 2019. [DOI] [PubMed] [Google Scholar]
  • 126. Pineyro MD, Pizarro JC, Lema F, Pritsch O, Cayota A, Bentley GA, and Robello C. Crystal structure of the tryparedoxin peroxidase from the human parasite Trypanosoma cruzi. J Struct Biol 150: 11–22, 2005. [DOI] [PubMed] [Google Scholar]
  • 127. Pittis AA, Goh V, Cebrian-Serrano A, Wettmarshausen J, Perocchi F, and Gabaldon T. Discovery of EMRE in fungi resolves the true evolutionary history of the mitochondrial calcium uniporter. Nat Commun 11: 4031, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Plovanich M, Bogorad RL, Sancak Y, Kamer KJ, Strittmatter L, Li AA, Girgis HS, Kuchimanchi S, De Groot J, Speciner L, Taneja N, Oshea J, Koteliansky V, and Mootha VK. MICU2, a paralog of MICU1, resides within the mitochondrial uniporter complex to regulate calcium handling. PLoS One 8: e55785, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Proto WR, Coombs GH, and Mottram JC. Cell death in parasitic protozoa: regulated or incidental? Nat Rev Microbiol 11: 58–66, 2013. [DOI] [PubMed] [Google Scholar]
  • 130. Raffaello A, De Stefani D, Sabbadin D, Teardo E, Merli G, Picard A, Checchetto V, Moro S, Szabo I, and Rizzuto R. The mitochondrial calcium uniporter is a multimer that can include a dominant-negative pore-forming subunit. EMBO J 32: 2362–2376, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Ramakrishnan S, Asady B, and Docampo R. Acidocalcisome-mitochondrion membrane contact sites in Trypanosoma brucei. Pathogens 7: 33, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Sancak Y, Markhard AL, Kitami T, Kovacs-Bogdan E, Kamer KJ, Udeshi ND, Carr SA, Chaudhuri D, Clapham DE, Li AA, Calvo SE, Goldberger O, and Mootha VK. EMRE is an essential component of the mitochondrial calcium uniporter complex. Science 342: 1379–1382, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Schlecker T, Schmidt A, Dirdjaja N, Voncken F, Clayton C, and Krauth-Siegel RL. Substrate specificity, localization, and essential role of the glutathione peroxidase-type tryparedoxin peroxidases in Trypanosoma brucei. J Biol Chem 280: 14385–14394, 2005. [DOI] [PubMed] [Google Scholar]
  • 134. Shikha S, Brogli R, Schneider A, and Polacek N. tRNA biology in trypanosomes. Chimia (Aarau) 73: 395–405, 2019. [DOI] [PubMed] [Google Scholar]
  • 135. Silva TM, Peloso EF, Vitor SC, Ribeiro LH, and Gadelha FR. O2 consumption rates along the growth curve: new insights into Trypanosoma cruzi mitochondrial respiratory chain. J Bioenerg Biomembr 43: 409–417, 2011. [DOI] [PubMed] [Google Scholar]
  • 136. Smirlis D and Soteriadou K. Trypanosomatid apoptosis: ‘Apoptosis’ without the canonical regulators. Virulence 2: 253–256, 2011. [DOI] [PubMed] [Google Scholar]
  • 137. Smith K, Opperdoes FR, and Fairlamb AH. Subcellular distribution of trypanothione reductase in bloodstream and procyclic forms of Trypanosoma brucei. Mol Biochem Parasitol 48: 109–112, 1991. [DOI] [PubMed] [Google Scholar]
  • 138. Steenkamp DJ. Trypanosomal antioxidants and emerging aspects of redox regulation in the trypanosomatids. Antioxid Redox Signal 4: 105–121, 2002. [DOI] [PubMed] [Google Scholar]
  • 139. Taylor MC, Lewis MD, Fortes Francisco A, Wilkinson SR, and Kelly JM. The Trypanosoma cruzi vitamin C dependent peroxidase confers protection against oxidative stress but is not a determinant of virulence. PLoS Negl Trop Dis 9: e0003707, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Teixeira F, Castro H, Cruz T, Tse E, Koldewey P, Southworth DR, Tomas AM, and Jakob U. Mitochondrial peroxiredoxin functions as crucial chaperone reservoir in Leishmania infantum. Proc Natl Acad Sci U S A 112: E616–E624, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Temperton NJ, Wilkinson SR, Meyer DJ, and Kelly JM. Overexpression of superoxide dismutase in Trypanosoma cruzi results in increased sensitivity to the trypanocidal agents gentian violet and benznidazole. Mol Biochem Parasitol 96: 167–176, 1998. [DOI] [PubMed] [Google Scholar]
  • 142. Thomson L, Denicola A, and Radi R. The trypanothione-thiol system in Trypanosoma cruzi as a key antioxidant mechanism against peroxynitrite-mediated cytotoxicity. Arch Biochem Biophys 412: 55–64, 2003. [DOI] [PubMed] [Google Scholar]
  • 143. Tomás AM and Castro H. Redox metabolism in mitochondria of trypanosomatids. Antioxid Redox Signal 19: 696–707, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Trujillo M, Budde H, Pineyro MD, Stehr M, Robello C, Flohe L, and Radi R. Trypanosoma brucei and Trypanosoma cruzi tryparedoxin peroxidases catalytically detoxify peroxynitrite via oxidation of fast reacting thiols. J Biol Chem 279: 34175–34182, 2004. [DOI] [PubMed] [Google Scholar]
  • 145. Tsai MF, Jiang D, Zhao L, Clapham D, and Miller C. Functional reconstitution of the mitochondrial Ca2+/H+ antiporter Letm1. J Gen Physiol 143: 67–73, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Turrens JF. Possible role of the NADH-fumarate reductase in superoxide anion and hydrogen peroxide production in Trypanosoma brucei. Mol Biochem Parasitol 25: 55–60, 1987. [DOI] [PubMed] [Google Scholar]
  • 147. Vasington FD and Murphy JV. Ca ion uptake by rat kidney mitochondria and its dependence on respiration and phosphorylation. J Biol Chem 237: 2670–2677, 1962. [PubMed] [Google Scholar]
  • 148. Vercesi AE, Castilho RF, Kowaltowski AJ, de Oliveira HCF, de Souza-Pinto NC, Figueira TR, and Busanello ENB. Mitochondrial calcium transport and the redox nature of the calcium-induced membrane permeability transition. Free Radic Biol Med 129: 1–24, 2018. [DOI] [PubMed] [Google Scholar]
  • 149. Vercesi AE and Docampo R. Ca2+ transport by digitonin-permeabilized Leishmania donovani. Effects of Ca2+, pentamidine and WR-6026 on mitochondrial membrane potential in situ. Biochemical J 284: 463–467, 1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Vercesi AE, Docampo R, and Moreno SN. Energization-dependent Ca2+ accumulation in Trypanosoma brucei bloodstream and procyclic trypomastigotes mitochondria. Mol Biochem Parasitol 56: 251–257, 1992. [DOI] [PubMed] [Google Scholar]
  • 151. Vercesi AE, Macedo DV, Lima SA, Gadelha FR, and Docampo R. Ca2+ transport in digitonin-permeabilized trypanosomatids. Mol Biochem Parasitol 42: 119–124, 1990. [DOI] [PubMed] [Google Scholar]
  • 152. Vercesi AE, Moreno SN, Bernardes CF, Meinicke AR, Fernandes EC, and Docampo R. Thapsigargin causes Ca2+ release and collapse of the membrane potential of Trypanosoma brucei mitochondria in situ and of isolated rat liver mitochondria. J Biol Chem 268: 8564–8568, 1993. [PubMed] [Google Scholar]
  • 153. Verner Z, Cermakova P, Skodova I, Kriegova E, Horvath A, and Lukes J. Complex I (NADH:ubiquinone oxidoreductase) is active in but non-essential for procyclic Trypanosoma brucei. Mol Biochem Parasitol 175: 196–200, 2011. [DOI] [PubMed] [Google Scholar]
  • 154. Wilkinson SR, Meyer DJ, and Kelly JM. Biochemical characterization of a trypanosome enzyme with glutathione-dependent peroxidase activity. Biochem J 352 Pt 3: 755–761, 2000. [PMC free article] [PubMed] [Google Scholar]
  • 155. Wilkinson SR, Meyer DJ, Taylor MC, Bromley EV, Miles MA, and Kelly JM. The Trypanosoma cruzi enzyme TcGPXI is a glycosomal peroxidase and can be linked to trypanothione reduction by glutathione or tryparedoxin. J Biol Chem 277: 17062–17071, 2002. [DOI] [PubMed] [Google Scholar]
  • 156. Wilkinson SR, Obado SO, Mauricio IL, and Kelly JM. Trypanosoma cruzi expresses a plant-like ascorbate-dependent hemoperoxidase localized to the endoplasmic reticulum. Proc Natl Acad Sci U S A 99: 13453–13458, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Wilkinson SR, Prathalingam SR, Taylor MC, Ahmed A, Horn D, and Kelly JM. Functional characterisation of the iron superoxide dismutase gene repertoire in Trypanosoma brucei. Free Radic Biol Med 40: 198–209, 2006. [DOI] [PubMed] [Google Scholar]
  • 158. Wilkinson SR, Taylor MC, Touitha S, Mauricio IL, Meyer DJ, and Kelly JM. TcGPXII, a glutathione-dependent Trypanosoma cruzi peroxidase with substrate specificity restricted to fatty acid and phospholipid hydroperoxides, is localized to the endoplasmic reticulum. Biochem J 364: 787–794, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Wilkinson SR, Temperton NJ, Mondragon A, and Kelly JM. Distinct mitochondrial and cytosolic enzymes mediate trypanothione-dependent peroxide metabolism in Trypanosoma cruzi. J Biol Chem 275: 8220–8225, 2000. [DOI] [PubMed] [Google Scholar]
  • 160. Xiong ZH, Ridgley EL, Enis D, Olness F, and Ruben L. Selective transfer of calcium from an acidic compartment to the mitochondrion of Trypanosoma brucei. Measurements with targeted aequorins. J Biol Chem 272: 31022–31028, 1997. [DOI] [PubMed] [Google Scholar]
  • 161. Yadav RK, Dolai S, Pal S, and Adak S. Role of tryptophan-208 residue in cytochrome c oxidation by ascorbate peroxidase from Leishmania major-kinetic studies on Trp208Phe mutant and wild type enzyme. Biochim Biophys Acta 1784: 863–871, 2008. [DOI] [PubMed] [Google Scholar]
  • 162. Yoo J, Wu M, Yin Y, Herzik MA Jr., Lander GC, and Lee SY.. Cryo-EM structure of a mitochondrial calcium uniporter. Science 361: 506–511, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Zhang Y, Bond CS, Bailey S, Cunningham ML, Fairlamb AH, and Hunter WN. The crystal structure of trypanothione reductase from the human pathogen Trypanosoma cruzi at 2.3 A resolution. Protein Sci 5: 52–61, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Antioxidants & Redox Signaling are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES