Role of mitochondria in the leishmanicidal effects and toxicity of acyl phloroglucinol derivatives: nemorosone and guttiferone A

Parasitology ◽  
2015 ◽  
Vol 142 (9) ◽  
pp. 1239-1248 ◽  
Author(s):  
LIANET MONZOTE ◽  
ALEXANDRA LACKOVA ◽  
KATRIN STANIEK ◽  
OSMANY CUESTA-RUBIO ◽  
LARS GILLE

SUMMARYNemorosone (Nem) and guttiferone A (GutA) are acyl phloroglucinol derivatives (APD) that are present in different natural products. For both compounds anti-cancer and anti-microbial properties have been reported. In particular, an anti-leishmanial activity of both compounds was demonstrated. The aim of this study was to explore the possible role of mitochondria in the anti-leishmanial activity of Nem and GutA in comparison with their action on mammalian mitochondria. Both APD inhibited the growth of promastigotes ofLeishmania tarentolae(LtP) with half maximal inhibitory concentration (IC50) values of 0·67 ± 0·17 and 6·2 ± 2·6μm; while IC50values for cytotoxicity against peritoneal macrophages from BALB/c mice were of 29·5 ± 3·7 and 9·2 ± 0·9μm, respectively. Nemorosone strongly inhibited LtP oxygen consumption, caused species-specific inhibition (P< 0·05) of succinate:ubiquinone oxidoreductase (complex II) from LtP-mitochondria and significantly increased (P< 0·05) the mitochondrial superoxide production. In contrast, GutA caused only a moderate reduction of respiration in LtP and triggered less superoxide radical production in LtP compared with Nem. In addition, GutA inhibited mitochondrial complex III in bovine heart submitochondrial particles, which is possibly involved in its mammalian toxicity. Both compounds demonstrated at low micromolar concentrations an effect on the mitochondrial membrane potential in LtP. The present study suggests that Nem caused its anti-leishmanial action due to specific inhibition of complexes II/III of mitochondrial respiratory chain ofLeishmaniaparasites that could be responsible for increased production of reactive oxygen species that triggers parasite death.

2021 ◽  
Vol 12 (12) ◽  
Author(s):  
Xuelian Zhang ◽  
Bin Zhang ◽  
Chenyang Zhang ◽  
Guibo Sun ◽  
Xiaobo Sun

AbstractTribbles homolog 1 (TRIB1) belongs to the Tribbles family of pseudokinases, which plays a key role in tumorigenesis and inflammation. Although genome-wide analysis shows that TRIB1 expression is highly correlated with blood lipid levels, the relationship between TRIB1 and adipose tissue metabolism remains unclear. Accordingly, the aim of the present study was to explore the role of TRIB1 on mitochondrial function in the brown adipose tissue (BAT). Trib1-knockout mice were established using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology. The metabolic function of the BAT was induced by a β3-adrenoceptor agonist and the energy metabolism function of mitochondria in the BAT of mice was evaluated. Trib1-knockout mice exhibited obesity and impaired BAT thermogenesis. In particular, Trib1 knockout reduced the ability of the BAT to maintain body temperature, inhibited β3-adrenoceptor agonist-induced thermogenesis, and accelerated lipid accumulation in the liver and adipose tissues. In addition, Trib1 knockout reduced mitochondrial respiratory chain complex III activity, produced an imbalance between mitochondrial fusion and fission, caused mitochondrial structural damage and dysfunction, and affected heat production and lipid metabolism in the BAT. Conversely, overexpression of Trib1 in 3T3-L1 adipocytes increased the number of mitochondria and improved respiratory function. These findings support the role of Trib1 in regulating the mitochondrial respiratory chain and mitochondrial dynamics by affecting mitochondrial function and thermogenesis in the BAT.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3268-3268
Author(s):  
Margaret Nieborowska-Skorska ◽  
Mateusz Koptyra ◽  
Elisabeth Bolton ◽  
Regina Ray ◽  
Danielle Ngaba ◽  
...  

Abstract Abstract 3268 Poster Board III-1 BCR/ABL kinase transforms hematopoietic stem cells to induce chronic myelogenous leukemia (CML). CML in chronic phase (CML-CP) is a leukemia stem cell (LSC)-derived but leukemia progenitor cell (LPC)-driven disease, which is, in most cases, sensitive to ABL tyrosine kinase inhibitors (TKIs) monotherapy. TKIs do not eradicate the leukemia but instead usually render the disease ‘inactive', since the residual quiescent LSCs are intrinsically insensitive to BCR-ABL inhibition and, in a significant cohort of CML patients, LPCs are also refractory or acquire resistance to TKIs due to mutations in BCR/ABL kinase. In the post-imatinib era, these cells may eventually undergo transformation and initiate fatal CML blast crisis (CML-BC). The malignant progression is usually associated with enhanced expression of BCR/ABL and accumulation of additional genetic aberrations, such as TKI-resistant mutations and chromosomal aberrations. In CML-CP, LSCs and LPCs reside in the CD34+CD38- and CD34+CD38+ populations, respectively, whereas in CML-BC, LSCs are also found in the CD34+CD38+ population. In addition, LSCs and LPCs usually belong to quiescent (CFSEmax) and proliferative (CFSElow) populations, respectively. However, the origin of CML-BC clone and the role of BCR/ABL “dosage” are not known. Since genomic instability usually results from DNA damage, we investigated the mechanisms responsible for enhanced DNA damage in CML cells. Much endogenous DNA damage arises from free radicals such as reactive oxygen species (ROS). Here we show that LSCs-enriched CD34+CD38- and quiescent (CFSEmax) CML cells and LPCs-enriched CD34+CD38+ cells contain higher levels of ROS (superoxide anion, hydrogen peroxide, and hydroxyl radical) than corresponding cells from normal donors (CML-BC>CML-CP>Normal). Interestingly, CFSEmax and CFSElow CML cells displayed similar elevation of ROS indicating that the presence of BCR/ABL and not the proliferative status enhances ROS. In addition, total cellular ROS and mitochondrial ROS levels were proportional to the expression of BCR/ABL kinase implicating the role of BCR/ABL kinase “dosage”. Higher levels of ROS caused more oxidative DNA lesions, such as 8-oxoG and DNA double-strand breaks (DSBs) in CD34+ and also in CD34+CD38- CML cells than in normal counterparts (CML-BC>CML-CP>Normal). Inhibition of BCR/ABL kinase with imatinib partially reduced ROS and oxidative DNA damage in CD34+ CML-CP cells, implicating BCR/ABL-dependent and -;independent mechanisms. Our previous studies showed that elevated levels of oxidative DNA damage in BCR/ABL-transformed cells were responsible for accumulation of TKI-resistant BCR/ABL mutants and chromosomal aberrations (Blood, 2006; Leukemia, 2008), highlighting the importance of identification of the sources of ROS in CML. Mitochondrial respiratory chain (MRC) is a major site of ATP production via oxidative phosphorylation, which is associated with electron flux through MRC. Some of the electrons may escape and react with molecular oxygen to form ROS. To shut down MRC, cells were depleted of mitochondrial DNA (mtDNA) by long-term exposure to ethidium bromide in the presence of uridine and pyruvate as confirmed by RT-PCR showing the absence/reduction of mtDNA-coded Cox II gene transcript. The absence of functional MRC reduced the level of ROS by 40% and 20% in CD34+ CML-CP cells and normal counterparts, respectively, suggesting that MRC is an important source of ROS in leukemia cells. Using selective inhibitors of various MRC complexes we identified complex III as major producer of ROS in LSCs and LPCs in CML-CP. The role of complex III in CML-BC cells is somehow diminished in concordance with the observation that prolonged exposure of MRC to elevated levels of ROS results in “mitochondrial injury” and reduction of MRC activity in advanced stages of cancer. In summary, we postulate that BCR/ABL kinase generates ROS and oxidative DNA damage in a dose-dependent manner not only in LPCs-enriched CD34+CD38+ and CFSElow cells, but also in LSCs-enriched CD34+CD38- and CFSEmax cells, and that MRC complex III generates significant amount of ROS in CML-CP cells. Thus, genomic instability causing TKI resistance and progression to CML-BC may originate in LSCs as well as in LPCs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1211-1211
Author(s):  
Piotr Kopinski ◽  
Margaret Nieborowska-Skorska ◽  
Grazyna Hoser ◽  
Danielle Ngaba ◽  
Regina Ray ◽  
...  

Abstract Abstract 1211 Background: BCR-ABL1 –positive chronic myeloid leukemia in chronic phase (CML-CP) is a leukemia stem cell (LSC)-derived but leukemia progenitor cell (LPC)-driven disease, which may eventually progress to fatal CML blast phase (CML-BP). In CML-CP, LSCs and LPCs reside in the CD34+CD38- and CD34+CD38+ populations, respectively. In addition, majority of LSCs and LPCs belong to quiescent (CFSEmax) and proliferative (CFSElow) populations, respectively. Tyrosine kinase inhibitors (TKIs) such as imatinib, dasatinib and nilotinib do not eradicate the leukemia but instead render the disease ‘dormant’. Residual quiescent LSCs are intrinsically insensitive to TKIs and, in a significant cohort of CML patients, LPCs acquire resistance to TKIs. In the TKI era, these cells may eventually initiate disease relapse and progression to CML-BP, which is associated with genomic instability manifested by accumulation of TKI-resistant BCR-ABL1 kinase mutations and chromosomal aberrations. Previously we showed that BCR-ABL1 kinase stimulates reactive oxygen species (ROS)-dependent oxidative DNA damage resulting in genomic instability (Nowicki et al., Blood, 2005; Koptyra et al., Blood, 2006; Koptyra et al., Leukemia, 2008). These studies highlighted the importance of identification of cellular lineage origin and mechanisms responsible for generation of ROS-mediated oxidative DNA damage in CML. Result: Here we show that LSC-enriched CD34+CD38- and quiescent (CFSEmax) CML-CP cells and LPC-enriched CD34+CD38+ and proliferating (CFSElow) CML-CP cells contain higher levels of ROS (superoxide anion, hydrogen peroxide, and hydroxyl radical) and oxidative DNA lesions (8-oxoG and DNA double-strand breaks) than corresponding cells from healthy donors. Non-mutated and TKI-resistant BCR-ABL1 kinase mutants (Y253F, T315I, H396P) stimulated ROS-induced oxidative DNA damage in a BCR-ABL1 dosage-dependent manner. Inhibition of BCR-ABL1 kinase with imatinib only partially reduced ROS and oxidative DNA damage in CD34+ CML-CP cells, implicating kinase-dependent and –independent mechanisms. Mitochondrial respiratory chain (MRC) is a major site of ATP production via oxidative phosphorylation, which is associated with electron flux through MRC. Some of the electrons may escape and react with molecular oxygen to form ROS. We detected that CD34+ CML-CP cells displayed lower mitochondrial potential than normal counterparts, which is indicative of enhanced ROS production. To determine the role of MRC in ROS-induced oxidative DNA damage, cells were depleted of mitochondrial DNA (mtDNA) by ethidium bromide, as confirmed by RT-PCR showing the absence/reduction of mtDNA-coded Cox II gene transcript (Rho0 cells). The absence of functional MRC reduced ROS and oxidative DNA damage in CD34+ CML-CP Rho0 cells and 32Dcl3 Rho0 cells transformed by non-mutated and TKI-resistant BCR-ABL1 kinase mutants, but not in normal counterparts, implicating a specific role of MRC in genomic instability in leukemia cells. In concordance, BCR-ABL1 –positive 32Dcl3 Rho0 cells accumulated fewer TKI-resistant BCR-ABL1 kinase mutants than cells with functional MRC. Using selective inhibitors of various MRC complexes we identified complex III as major producer of ROS and oxidative DNA damage in CD34+CD38- and quiescent LSCs and in CD34+CD38+ and proliferating LPCs in CML-CP. Moreover, BCR-ABL1 –positive cells in which complex III was inactive due to a single base substitution within the cytochrome b gene displayed diminished capability to generate ROS. In contrast, ROS was not affected in cells lacking complex I due to a mutation in the ND6 gene. In addition to BCR-ABL1 –positive CML-CP complex III also appeared to play a major role in generation of ROS in FLT3(ITD)-positive acute myeloid leukemia cells and in JAK2(V617F)-positive polycythemia vera cells. Conclusion: In summary, we postulate that enhanced production of ROS by MRC complex III induces genomic instability in LSC-enriched CD34+CD38- and quiescent cells, and also in LPC-enriched CD34+CD38+ and proliferating cells. Thus, genomic instability causing TKI resistance, disease relapse and progression to CML-BP may originate in LSCs as well as in LPCs. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Michele Brischigliaro ◽  
Elena Frigo ◽  
Samantha Corrà ◽  
Cristiano De Pittà ◽  
Ildikò Szabò ◽  
...  

AbstractMutations in BCS1L are the most frequent cause of human mitochondrial disease linked to complex III deficiency. Different forms of BCS1L-related diseases and more than 20 pathogenic alleles have been reported to date. Clinical symptoms are highly heterogenous, and multisystem involvement is often present, with liver and brain being the most frequently affected organs. BCS1L encodes a mitochondrial AAA + -family member with essential roles in the latest steps in the biogenesis of mitochondrial respiratory chain complex III. Since Bcs1 has been investigated mostly in yeast and mammals, its function in invertebrates remains largely unknown. Here, we describe the phenotypical, biochemical and metabolic consequences of Bcs1 genetic manipulation in Drosophila melanogaster. Our data demonstrate the fundamental role of Bcs1 in complex III biogenesis in invertebrates and provide novel, reliable models for BCS1L-related human mitochondrial diseases. These models recapitulate several features of the human disorders, collectively pointing to a crucial role of Bcs1 and, in turn, of complex III, in development, organismal fitness and physiology of several tissues.


Cancers ◽  
2021 ◽  
Vol 13 (9) ◽  
pp. 2036
Author(s):  
Luigi Marongiu ◽  
Markus Burkard ◽  
Sascha Venturelli ◽  
Heike Allgayer

Natural compounds such as essential oils and tea have been used successfully in naturopathy and folk medicine for hundreds of years. Current research is unveiling the molecular role of their antibacterial, anti-inflammatory, and anticancer properties. Nevertheless, the effect of these compounds on bacteriophages is still poorly understood. The application of bacteriophages against bacteria has gained a particular interest in recent years due to, e.g., the constant rise of antimicrobial resistance to antibiotics, or an increasing awareness of different types of microbiota and their potential contribution to gastrointestinal diseases, including inflammatory and malignant conditions. Thus, a better knowledge of how dietary products can affect bacteriophages and, in turn, the whole gut microbiome can help maintain healthy homeostasis, reducing the risk of developing diseases such as diverse types of gastroenteritis, inflammatory bowel disease, or even cancer. The present review summarizes the effect of dietary compounds on the physiology of bacteriophages. In a majority of works, the substance class of polyphenols showed a particular activity against bacteriophages, and the primary mechanism of action involved structural damage of the capsid, inhibiting bacteriophage activity and infectivity. Some further dietary compounds such as caffeine, salt or oregano have been shown to induce or suppress prophages, whereas others, such as the natural sweeter stevia, promoted species-specific phage responses. A better understanding of how dietary compounds could selectively, and specifically, modulate the activity of individual phages opens the possibility to reorganize the microbial network as an additional strategy to support in the combat, or in prevention, of gastrointestinal diseases, including inflammation and cancer.


Antioxidants ◽  
2021 ◽  
Vol 10 (5) ◽  
pp. 759
Author(s):  
David Mantle ◽  
Robert A. Heaton ◽  
Iain P. Hargreaves

Coenzyme Q10 (CoQ10) has a number of important roles in the cell that are required for optimal functioning of the immune system. These include its essential role as an electron carrier in the mitochondrial respiratory chain, enabling the process of oxidative phosphorylation to occur with the concomitant production of ATP, together with its role as a potential lipid-soluble antioxidant, protecting the cell against free radical-induced oxidation. Furthermore, CoQ10 has also been reported to have an anti-inflammatory role via its ability to repress inflammatory gene expression. Recently, CoQ10 has also been reported to play an important function within the lysosome, an organelle central to the immune response. In view of the differing roles CoQ10 plays in the immune system, together with the reported ability of CoQ10 supplementation to improve the functioning of this system, the aim of this article is to review the current literature available on both the role of CoQ10 in human immune function and the effect of CoQ10 supplementation on this system.


Sign in / Sign up

Export Citation Format

Share Document