• No results found

University of Groningen The role of small conductance calcium-activated potassium channels in mitochondrial dysfunction Krabbendam, Inge

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen The role of small conductance calcium-activated potassium channels in mitochondrial dysfunction Krabbendam, Inge"

Copied!
21
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

The role of small conductance calcium-activated potassium channels in mitochondrial dysfunction

Krabbendam, Inge DOI:

10.33612/diss.144370526

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Krabbendam, I. (2020). The role of small conductance calcium-activated potassium channels in mitochondrial dysfunction: Targeting metabolic reprogramming and calcium homeostasis. University of Groningen. https://doi.org/10.33612/diss.144370526

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 213PDF page: 213PDF page: 213PDF page: 213

Ca

CaCa

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

(3)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 214PDF page: 214PDF page: 214PDF page: 214

214

Preface

Neurodegenerative diseases affect millions of people worldwide and seriously degrade the quality of life. Alterations of potassium channels in the brain have been linked to the pathogenesis of neurodegeneration. For instance, genetic evidence links calcium-activated potassium (KCa) channels to Alzheimer’s

Disease (AD) (1,2). KCa channels also gained attention in the field of ischemia and

cancer. In neurons, plasma membrane KCa channels regulate excitability, while

mitochondrial located KCa (mitoKCa) channels play a role in balancing levels of

mitochondrial reactive oxygen species (ROS) and mitochondrial calcium (Ca2+),

leading to protection against oxidative stress. Oxidative stress and mitochondrial dysfunction serve nowadays as pathological markers for neurodegeneration, next to neuronal hyperexcitability and Ca2+ deregulation, all

contributing to cell death (3–9). Several types of cell death can be related to neurodegeneration, including excitotoxicity, oxytosis, ferroptosis and inflammaging (10–12). As a response mechanism to protect against defects in cellular energy metabolism and cell death, compounds that are able to mediate metabolic (re)programming gained interest also in the field of neuroscience, besides long-standing studies in oncology (13,14). Interestingly, previous data of our group showed that small conductance Ca2+-activated K+ (SK) channels

modulate mitochondrial Ca2+ levels and protect neurons against glutamate

excitotoxicity and glutamate-induced oxytosis, of which the latter is a well-established in vitro model to study mitochondrial dysfunction associated with oxidative stress. Moreover, pharmacological SK channel modulators also attenuated inflammation. It is not yet understood precisely how SK channel modulators affect mitochondrial function and inflammation and how they are able to confer cellular protection. The objective of this thesis was to investigate the role of SK channels on mitochondrial dysfunction and inflammation, thereby focusing on Ca2+, ROS and mitochondrial metabolism.

Mitochondrial calcium-activated potassium channels

As reviewed in chapter 2, KCa channels are found in the central nervous system

(CNS), in the cardiovascular system, particularly in the heart, and in several other organs. KCa channels are expressed at the plasma membrane and in membranes

(4)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 215PDF page: 215PDF page: 215PDF page: 215

215

8

(BK, IK and SK) at the inner mitochondrial membrane (IMM) of several cell types has gained a lot of interest in cardiovascular, neurological and cancer research (15–19), attributed to the involvement of the channels in mitochondrial function and Ca2+ homeostasis. K

Ca channels located at the IMM facilitate K+ entry into

the mitochondrial matrix, leading to depolarization of the mitochondrial membrane and a reduced driving force for Ca2+ influx into mitochondria (19–

21). Under oxidative stress conditions, mitochondrial dysfunction and subsequent cell death is a consequence of elevated mitochondrial calcium (mito[Ca2+]) levels, higher probability of mitochondrial permeability transition

pore (mPTP) formation, enhanced ROS production, and inhibition of the respiratory chain in the mitochondria. A mechanism that obtained interest in the search for protection against diseases that involve oxidative stress, is mitochondrial preconditioning or mitohormesis, a process where cells undergo adaptation to oxidative stress. This process is attributed to a mild stressor that increases the resistance of both mitochondria and the entire cell to subsequent insults (22,23). Notably, pharmacological activation of mitoKCa channels

contribute to cell survival by lowering mito[Ca2+], attenuating formation of

mitochondrial ROS, and restoring the mitochondrial membrane potential (MMP) (24,25). Moreover, mitoKCa activation also prevents the loss of ATP

production in the mitochondrial electron transport chain. Several studies identified mitoKCa channels as central players in the molecular mechanisms

underlying preconditioning in cardiac ischemia and in different neurodegenerative conditions. Preconditioning and postconditioning mechanisms promoted by activation of mitoBK and mitoSK channels decreased the infarct size in conditions of ischemic stress via attenuation of mitochondrial ROS generation, attenuating mito[Ca2+] and inhibiting mPTP formation (26–28).

Both neuronal mitoBK- and mitoSK channels provoke protection by maintaining mitochondrial function. Importantly, however, mitoSK channels were shown to directly influence mitochondrial respiration (24). Among the different subcellular localizations of SK channels, in particular the mitochondrial-located SK2 channels are conferring protection against oxidative stress (24). With respect to SK channels, we investigated the impact of SK channel activation on mitochondrial Ca2+ regulation in oxidative stress paradigms (chapter 3, 4 and 5).

In addition, we studied whether the channels are involved in mitochondrial preconditioning and ROS production, mitochondrial complex activity and metabolic shifts, as a part of potential protective mechanisms against

(5)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 216PDF page: 216PDF page: 216PDF page: 216

216

ferroptosis in neurons and inflammatory processes in macrophages (chapter 5

and 6).

SK channel activation and mitochondrial calcium in oxidative stress

The impact of ER – mitochondria coupling on mitochondrial Ca2+ homeostasis

and oxytosis in neuronal cell survival

Neuronal cell survival is impacted by Ca2+ regulation, since Ca2+ ions are involved

in action potential transduction, neurotransmitter release, and several downstream signalling pathways such as mitochondrial metabolism (29). Intracellular Ca2+ stores including those in the ER and mitochondria play a critical

role in maintaining the intracellular Ca2+ homeostasis. Moreover, dysfunctional

Ca2+ signalling has been linked to neurodegeneration (30). The physical

connection between ER and mitochondria is established by ER-located IP3R,

GRP75 and OMM-located VDAC. These mitochondria-associated ER membranes (MAMs) were found to be critical for intracellular calcium [Ca2+]

i signalling and

regulation of metabolism. Under healthy conditions, Ca2+ is transferred from the

ER along the MAM into the mitochondrial matrix where it facilitates ATP production through promotion of the TCA cycle and mitochondrial respiration (31–33). Disrupted Ca2+ homeostasis can lead to mito[Ca2+] overload, and

subsequently to impaired mitochondrial metabolism and respiration (34,35). In addition, enhanced MAM-mediated mito[Ca2+] uptake leads to MMP loss, ROS

production and eventually cell death (36,37). Several studies have proven that enhanced or weakened ER-mitochondrial coupling (EMC) and associated mitochondrial damage is detrimental. Moreover, different diseases either have decreased or enhanced EMC (40,41), which can be explained by the fact that both diminishing basal mito[Ca2+] uptake as well as inducing mito[Ca2+] overload

can lead to cell death. Using genetically encoded linkers sensitive to rapamycin, we provided evidence that EMC strengthening increased the vulnerability of HT22 cells to glutamate-induced oxytosis and cell death, through an increase in mito[Ca2+] uptake and impaired mitochondrial respiration (chapter 3). The

linkers used in our study consist of ER-and OMM targeted anchors whose endogenous function is not related to EMC and which rapidly form a bridge upon rapamycin treatment (39). Although this type of physical connection is artificial,

(6)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 217PDF page: 217PDF page: 217PDF page: 217

217

8

similar endogenous mechanisms regulating EMC are observed in neurodegenerative diseases.

Recent research showed that chronic incubations with high glucose of either insulin-secreting cells or pancreatic islets reduced ER Ca2+ store, increased basal

mito[Ca2+], and reduced ATP-stimulated ER-mitochondria Ca2+ exchanges, in

conditions of increased organelle interactions (38). Under our experimental conditions, we enhanced EMC and proved that SK channel activation still mediated protection against glutamate challenge which induced cellular damage in the presence of ER-mitochondrial linkers. Interestingly, overexpression of either SK2 or mitoSK2 together with pharmacological positive modulator CyPPA treatment further enhanced cell survival compared to the GFP/mitoGFP overexpression, and this was independent of any influence on IP3R

– VDAC1 interaction (chapter 3). Thus, these data indicate that the overexpression of SK2 channels increased CyPPA-mediated protection against glutamate toxicity. In addition, these neuroprotective actions of SK2 channel activation were independent of the physical EMC.

Modulation of mitochondrial calcium uptake in ferroptosis

IP3R-mediated release of Ca2+ from the ER is subsequently transferred by the

mitochondrial Ca2+ uniporter (MCU) to facilitate mitochondrial Ca2+ uptake.

Located in the IMM, MCU is the true entry point of Ca2+ into the mitochondrial

matrix. Therefore, we hypothesized that besides modulating ER – mitochondria contact points, antagonism of MCU may influence the cells’ vulnerability to oxidative stress or ferroptosis in HT22 cells (chapter 4). We demonstrated that inhibition of mito[Ca2+] uptake using ruthenium red (RR) and mitoxantrone (MX)

restored cell viability in ferroptosis and attenuated mitochondrial Ca2+ levels

following ferroptosis. Both compounds attenuated lipid peroxidation levels induced by erastin, an inducer of ferroptosis, and co-treatment with RR reduced mitochondrial ROS levels similar to ferrostatin, a classical inhibitor of ferroptosis. While both RR and MX reduced mitochondrial Ca2+ overload, they

were not able to rescue the decrease in mitochondrial respiration due to ferroptosis. Interestingly, Arduino and colleagues postulated that MX-mediated inhibition of MCU Ca2+ currents did not influence oxidative phosphorylation

(OXPHOS) (42). However, their experiment was performed using acute treatment. In our study, MX treatment alone resulted in a reduction in cell

(7)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 218PDF page: 218PDF page: 218PDF page: 218

218

proliferation, a decrease in OXPHOS and an increase in early-apoptosis. Interestingly, co-treatment with SK channel activator, CyPPA, potentiated the protective effect of RR but demonstrated no significant additive effect to the protective action of MX. For further understanding of this additive effect, future research could investigate possible physical proximity or interaction between the two proteins. Notably, it has been suggested that opening of ATP-sensitive potassium (mitoKATP) channels by diazoxide in heart causes a drop in Ψm, thereby

limiting mitochondrial Ca2+ entry through MCU and consequent Ca2+ overload,

providing protection against ischemia-reperfusion injury (43,44). Furthermore, we showed that MICU1 knockdown cells were more sensitive to erastin treatment compared to control. In conclusion, we demonstrated that MCU antagonism and mito[Ca2+] reduction is protective against ferroptosis, and that

potentially MICU1 plays a regulatory role.

The differential impacts of ROS on cell survival Mitohormesis or mitochondrial preconditioning

Apart from preservation of mito[Ca2+] levels, mitochondrial K

Ca channels have

been shown to modulate mitochondrial ROS production, thereby inducing mitochondrial preconditioning or mitohormesis. By mildly increasing ROS production at the level of mitochondrial complexes, these channels have been shown to provide neuro- and cardioprotection, effects that could be abrogated by ROS scavengers (chapter 2). There is an increasing number of studies showing the ROS-dependent BK channel-mediated protection in for example ischemia reperfusion (I/R) (45,46). Moreover, mitoKATP opening evoked protective effects

against I/R and cerebral ischemia (47,48), with both ROS and nitric oxide (NO) production participating in the observed protection (49).

Interestingly, a study by Dai and colleagues found that simultaneous administration of cell-permeant superoxide dismutase (SOD) mimetic MnTBAP

([5,10,15,20-Tetrakis(4-carboxyphenyl)-21H,23H-porphine]manganese(III)chloride) with BK channel opener NS1619, reduced the NS1619-induced protective effect against ischemic preconditioning (45), which is in line with our observation described in chapter 5. Notably, mucosal

(8)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 219PDF page: 219PDF page: 219PDF page: 219

219

8

permeability-sparing and anti-inflammatory effects triggered by NS1619 to prevent I/R was found to be ROS-dependent (45).

There is extensive evidence that preconditioning effects in the CNS mediate neuroprotection, for instance shown by one study of an in vitro model of oxygen-glucose deprivation/re-oxygenation in neuron-astrocyte co-cultures subjected to ischemic preconditioning (50). To our knowledge, we are the first to show SK channel-induced preconditioning effects in neurons against oxidative stress. In chapter 5 we elucidated that SK channel activation by CyPPA slightly reduced mitochondrial complex I and II activity, which is in line with a mild increase in mitochondrial ROS production that was observed after long-term treatment with CyPPA. Similarly, CyPPA used at low concentrations promoted cell survival against H2O2 damage, and opening of SK channels induced a mild

reduction of the mitochondrial membrane potential (MMP) and a slight increase in mitochondrial ROS formation (51). In our study, CyPPA rescued neuronal cells from ferroptosis, while scavenging mitochondrial ROS with MnTBAP reduced both mitochondrial ROS production induced by CyPPA and also its protection. Thus, our findings indicate that SK channel activation provoked preconditioning effects that play a role in the neuronal protection against ferroptosis. This is in line with previous observations showing that increasing superoxide levels to a threshold level that does not affect cell survival protects SH-SY5Y neuroblastoma cells against oxidative stress through a mitohormetic process (52). Mitohormesis has been shown to be a process also occurring in vivo (53), for example SOD2 depletion in embryonic mice resulted in mitochondrial adjustments and enhanced antioxidant capacity, leading to resistance to subsequent oxidant challenges in later life (54). Furthermore, deletion of SOD2 extended lifespan in Caenorhabditis elegans (C. elegans) by altering mitochondrial function (55). We found an SK channel-mediated complex I activity reduction in HT22 cells and increased mean lifespan in C. elegans (chapter 5). In line with these findings, mild complex I inhibition with low rotenone concentrations reversed aging-related regulation of gene expression and prolonged lifespan in killifish (56). Future studies should investigate whether indeed SK channel activation leads to mild mitochondrial ROS production in vivo, for instance using longevity models in C. elegans, killifish or

(9)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 220PDF page: 220PDF page: 220PDF page: 220

220

Reverse electron transport

The current literature indicates that low ROS levels are beneficial, mediating adaptation to stress via signalling, while high levels of ROS are detrimental because they evoke oxidative stress. Recent studies suggest that the site at which ROS are produced is also important in assessing effects on cellular homeostasis. An established mechanism of site-specific ROS signalling is reverse electron transport (RET). RET occurs when electrons from coenzyme Q are transferred back to complex I, generating a significant amount of ROS (57,58). RET has been shown to stimulate lifespan in D. melanogaster (59), and is crucial for the activation of macrophages in response to bacterial infection (60), since blocking mitochondrial ROS arising from complex I abrogated the energy switch from OXPHOS to glycolysis in macrophages (61). In chapter 6, we aimed to elucidate potential effects of SK channel activation on inflammation and whether it affects ROS-RET. SK channel activation reduces ROS, potentially also ROS induced by RET, via inhibiting complex I. Future experiments would further substantiate this hypothesis. We demonstrated a reduction in cellular ROS with CyPPA upon LPS stimulation. In line, activation of mitochondrial BK channels attenuated ROS-RET in cardiac mitochondria (62). The effect of SK channels and RET in inflammation needs more investigation in the future. In addition, it would be interesting to study mitochondrial KCa channels and ROS-RET in other disease

models, such as diabetes type 2, where RET could be a potential target (63), and also in the context of neuronal- and cardiac function.

Novel strategy to improve ROS-targeted therapy in brain cancer

Oxidative stress plays an important role in cancer cell proliferation. Recently, mitohormesis was also identified in cancer cells, where it stimulated a subpopulation of cancer cells to basally upregulate mitochondrial stress responses, providing an adaptive metastatic advantage (64). Whereas mitohormesis is linked to longevity in non-disease conditions, it is thus used by cancer cells to contribute to tumour development. Although cancer cells are equipped with enhanced antioxidant defence capacities to benefit from these higher ROS levels, it has been observed that these cells are sensitive to further increases in ROS levels (65,66). Even though many recent studies focus on targeting ROS (67–70), resistance is still a big problem in the treatment of

(10)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 221PDF page: 221PDF page: 221PDF page: 221

221

8

glioblastoma.

Since in healthy brain cells, pharmacological SK channel activation with CyPPA, led to a moderate reduction in mitochondrial respiration and a mild increase in mitochondrial ROS (33 and chapter 5), we hypothesized that current cancer therapies targeting ROS could benefit from this. In chapter 7, we reported that combined treatment of auranofin, inhibiting thioredoxin reductases, and CyPPA, induced mitochondrial damage, ROS production and potentiated auranofin-induced toxicity in neuroblastoma cells, glioblastoma cells, and neurospheres generated from patient-derived glioblastoma cells. On the other hand, inhibition, and not activation, of mitochondrial K+ channel (mitoKv1.3) was also

leading to ROS-mediated apoptosis of cancer cells in vivo, via the same downstream events provoked by Bax. Inhibiting the depolarizing K+ influx causes

IMM hyperpolarization, thereby promoting increased ROS levels, loss of Ψm,

mPTP activation, and cytochrome c and further ROS release (71,72). In addition, several studies proposed IK channel inhibition as a target for cancer treatment (73,74). More studies are needed to investigate how inhibition of these ion channels differ from the treatment that we proposed in chapter 7.

Furthermore, we found that healthy (HT22) cells were less affected, which is in line with studies showing no toxicity in neurons and glia cells in the brain compared to cancer cells, with mitochondrial ROS being involved in this differential effect (75). Testing whether SK2/3 channel-specific inhibitors will protect against auranofin-induced cell death in cells overexpressing either SK2 or SK3 channels in cancer cells or using SK channel knockout cells might provide us more knowledge on understanding why cancer cells were found to be more vulnerable than HT22 cells, that only express SK2 channels (18).

SK channels mediate metabolic reprogramming Neuroprotection

Since mitochondrial dysfunction is a characteristic of neurodegeneration, we focused on exploring the possibility of metabolic reprogramming in neurons as a potential therapeutic action. As described before, we have found that SK channel activation attenuates mitochondrial respiration (chapter 5). Hence, we studied in addition whether SK channel activation could have concomitant effects on glycolysis that could contribute to the observed neuroprotection.

(11)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 222PDF page: 222PDF page: 222PDF page: 222

222

Indeed, we found that an initial increase in glycolytic activity is crucial for CyPPA-mediated protection against oxidative stress in HT22 cells (chapter 5). Also in

vivo, CyPPA facilitated increased glycolysis and protected against paraquat and

slightly extended mean lifespan. Though mildly increasing ROS production at the level of mitochondrial complexes has been shown to be an established characteristic of all mitochondrial KCa channels, to our knowledge we are the

first to report that SK channel activation at the same time also influences glycolytic activity. We also showed that lactate plays an important role in neuroprotection, and this mechanism has also been demonstrated in CNS cells. For instance in neurons, the presence of L-lactate as an alternative source to drive ATP production, protected against glutamate-induced neuronal death (76,77). Interestingly, in skin fibroblasts it has been demonstrated that intermittent exposure to L-lactate triggered mitohormesis and prevented aged-related mitochondrial dysfunction and other aging-aged-related mechanisms (78). In alignment with what we observed in chapter 5, it was recently also shown in C.

elegans and in SH-SY5Y neuroblastoma cells that L-lactate mildly promoted ROS,

that triggered antioxidant defense- and pro-survival pathways (79). Similar to our data on SK channel-mediated complex I inhibition and metabolic shifts, more papers discuss the link between inhibition of mitochondrial OXPHOS and glycolysis. Interestingly, metformin has been shown to suppress mitochondrial respiration and promote glycolysis. However, only its effect on mitochondrial respiration, and not on glycolysis, played a role in the protection against hepatotoxicity (80). In addition, loss of mitochondrial function as a result of NO production in dendritic cells commits these cells to sustain enhanced glycolytic activity to meet their energy demands (81). In astrocytes, NO-induced upregulation of glycolysis through activation of energy-sensor AMP-activated protein kinase (AMPK) was shown to be protective (82). In contrast, promotion of longevity in C. elegans due to complex I inhibition occurred independently of AMPK activation (83). Further studies could include the investigation of SK channel effects on NO and AMPK expression and signalling in neurons.

Immune cells

The switch from oxidative phosphorylation to glycolysis is a well-known mechanism involved in activation of immune cells via Toll-like receptors (TLRs), notably TLR4 (84). For example LPS that activates TLR4 strongly increased the

(12)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 223PDF page: 223PDF page: 223PDF page: 223

223

8

TCA cycle intermediate succinate (85) that facilitated glycolysis via HIF-1α stabilisation (60). Glutamine-dependent anerplerosis was the major source of succinate accumulation and metabolic alterations (85). In bone marrow-derived macrophages, succinate strongly boosted IL-1β expression in the presence of LPS (60). Although as described in chapter 6 macrophage activation in terms of shape expansion and IL-1β gene expression was not enhanced by succinate in the presence of LPS compared to LPS alone in RAW macrophage cells, we did observe increases in glycolytic activity in these conditions. Though we studied yet RAW macrophages only, future studies should enrol for instance human microglia or other human immune cells.

In agreement with data revealing that SK3 channel activation inhibited inflammatory responses of primary microglial cells after exposure to LPS (86), we have shown here, in addition, that SK2/SK3 channel activation in macrophages reduced the increase in glycolysis facilitated by LPS and succinate (chapter 6). Notably, CyPPA reduces NO release in microglia (86). Since NO is known to upregulate glycolysis (82,87), it would be interesting to study CyPPA effects on NO related to a possible downregulation of glycolysis in inflammatory macrophages. Interestingly, in contrast to SK channels, inhibition, and not activation, of BK channels has been found to suppress LPS-induced microglia activation and contributed to NO production. Here, both plasma membrane- and nuclear BK channels were involved, of which the latter location was evident after long-term LPS treatment (88).

Cancer cells

Similar to immune cells, tumour cells make a switch from OXPHOS to glycolysis, a metabolic adaptation that facilitates tumour growth and cancer progression. Although it was not the focus of the investigation and auranofin is supposed to suppress tumour activity, in chapter 7 we observed slight increases in glycolytic activity upon the treatment with auranofin in neuroblastoma cells. However, we did demonstrate detrimental effects of auranofin on OXPHOS, that was further enhanced by SK channel activation to levels that would induce cell death. In our study, SK channel activation by CyPPA without auranofin had no detrimental effects on cell survival. In contrast, opening of mitochondrial BK channel alone was shown to induce glioma cell death by increasing mitochondrial respiration (89). Thus, pharmacological activation of SK channels in combination with

(13)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 224PDF page: 224PDF page: 224PDF page: 224

224

auranofin induced brain cancer cell death, via suppressing mitochondrial function and potentiating ROS production.

Main conclusions

In this thesis, the role of small-conductance calcium-activated potassium channels in Ca2+ homeostasis and oxidative stress in mitochondrial function has

been studied. More specifically, we looked at the regulatory effects of SK channels on neurodegeneration, inflammation and brain cancer. Collectively, our studies have revealed the following findings:

Mitochondrial KCa channels mediate cellular protection against oxidative

stress through mitochondrial mechanisms of preconditioning, in several disease models including neurodegeneration and ischemia/reperfusion injury (chapter 2).

SK channel activation was neuroprotective against oxytosis in conditions of enhanced ER-mitochondrial coupling, that induced mitochondrial Ca2+ overload, impaired bioenergetics and cell death (chapter 3).

Attenuation of mitochondrial calcium uptake using ruthenium red (RR) and mitoxantrone (MX) restored cell viability in ferroptosis and decreased oxidative stress, while metabolism was not improved (chapter 4)

Co-treatment with SK channel activator CyPPA potentiated the protective effect of RR but demonstrated no significant additive effect to the protective action of MX (chapter 4).

Mild ROS induction by SK channel opening plays an important role in neuroprotection against oxidative stress in healthy cells but can promote anti-tumour capacities of thioredoxin reductase inhibitor auranofin in brain cancer cells (chapter 5 and chapter 7).

Neuroprotective effects by CyPPA against ferroptosis involve an initial, fast metabolic shift towards glycolysis (chapter 5)

Lower mitochondrial complex I activity by SK channel activation plays a role in cellular protection via reducing forward electron transfer but possibly also reverse electron transfer (chapter 5 and chapter 6)

Auranofin toxicity was potentiated by SK channel activation due to enhanced oxidative stress and mitochondrial dysfunction (chapter 7)

(14)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 225PDF page: 225PDF page: 225PDF page: 225

225

8

References

1. Beecham GW, Hamilton K, Naj AC, Martin ER, Huentelman M, Myers AJ, et al. Genome-Wide Association Meta-analysis of Neuropathologic Features of Alzheimer’s Disease and Related Dementias. PLoS Genet. 2014;10(9).

2. Heinzen EL, Yoon W, Weale ME, Sen A, Wood NW, Burke JR, et al. Alternative ion channel splicing in mesial temporal lobe epilepsy and Alzheimer’s disease. Genome Biol. 2007;8(3).

3. Arun S, Liu L, Donmez G. Mitochondrial Biology and Neurological Diseases. Curr Neuropharmacol. 2015;14(2):143–54.

4. Onyango IG, Khan SM, Jr JPB, Biotechnology G, St BF, Therapeutics N. Mitochondria in the pathophysiology of Alzheimer ’ s and Parkinson ’ s diseases. 2017;(4):854–72. 5. Pchitskaya E, Popugaeva E BI. Calcium signaling and molecular mechanisms underlying

neurodegenerative diseases. Cell Calcium. 2018;70:87–94.

6. Bondy SC. The relation of oxidative stress and hyperexcitation to neurological disease. Proc Soc Exp Biol Med. 1995 Apr;208(4):337–45.

7. Jenwitheesuk A, Nopparat C, Mukda S, Wongchitrat P, Govitrapong P. Melatonin regulates aging and neurodegeneration through energy metabolism, epigenetics, autophagy and circadian rhythm pathways. Int J Mol Sci. 2014;15:16848–84.

8. Quansah E, Peelaerts W, Langston JW, Simon DK, Colca J, Brundin P. Targeting energy metabolism via the mitochondrial pyruvate carrier as a novel approach to attenuate neurodegeneration. Mol Neurodegener. 2018;13:1–12.

9. Beal MF, Hyman BT, Koroshetz W. Do defects in mitochondrial energy metabolism underlie the pathology of neurodegenerative diseases? Trends Neurosci. 1993;16:125– 31.

10. Gaki GS, Papavassiliou AG. Oxidative stress-induced signaling pathways implicated in the pathogenesis of Parkinson’s disease. NeuroMolecular Med. 2014;16(2):217–30. 11. Rojas-Gutierrez E, Muñoz-Arenas G, Treviño S, Espinosa B, Chavez R, Rojas K, et al.

Alzheimer’s disease and metabolic syndrome: A link from oxidative stress and inflammation to neurodegeneration. Synapse. 2017;71(10).

12. Guiney SJ, Adlard PA, Bush AI, Finkelstein DI, Ayton S. Ferroptosis and cell death mechanisms in Parkinson’s disease. Neurochem Int. 2017;104:34–48.

13. Gohil VM, Sheth SA, Nilsson R, Wojtovich AP, Lee JH, Perocchi F, et al. Discovery and therapeutic potential of drugs that shift energy metabolism from mitochondrial respiration to glycolysis. Nat Biotechnol. 2010;28:249–55.

14. Gohil VM, Offner N, Walker JA, Sheth SA, Fossale E, Gusella JF, et al. Meclizine is neuroprotective in models of Huntington’s disease. Hum Mol Genet. 2011;20:294–300. 15. Siemen D, Loupatatzis C, Borecky J, Gulbins E, Lang F. Ca 2+ -Activated K Channel of the

BK-Type in the Inner Mitochondrial Membrane of a Human Glioma Cell Line. 1999;554:549–54.

16. Skalska J, Piwo M, Wyroba E, Surmacz L, Wieczorek R, Koszela-piotrowska I, et al. Biochimica et Biophysica Acta A novel potassium channel in skeletal muscle mitochondria. 2008;1777(2008):651–9.

17. Sassi N, Marchi U De, Fioretti B, Biasutto L, Gulbins E, Franciolini F, et al. Biochimica et Biophysica Acta An investigation of the occurrence and properties of the mitochondrial intermediate-conductance Ca 2 + -activated K + channel mtK Ca 3 . 1. BBA - Bioenerg. 2009;1797(6–7):1260–7.

18. Dolga AM, Netter MF, Perocchi F, Doti N, Meissner L, Tobaben S, et al. Mitochondrial small conductance SK2 channels prevent glutamate-induced oxytosis and mitochondrial dysfunction. J Biol Chem. 2013;288:10792–804.

(15)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 226PDF page: 226PDF page: 226PDF page: 226

226

19. Yang M, Camara AKS, Aldakkak M, Kwok W-M, Stowe DF. Identity and function of a cardiac mitochondrial small conductance Ca2+-activated K+ channel splice variant. Biochim Biophys Acta. 2017 Jun;1858(6):442–58.

20. Xu W, Liu Y, Wang S, Mcdonald T, Eyk JE Van, Sidor A, et al. Cytoprotective Role of Ca 2 ؉ - Activated K ؉ Channels in the Cardiac Inner Mitochondrial Membrane. 2002. 21. Bednarczyk P, Koziel A, Jarmuszkiewicz W, Szewczyk A. Large-conductance

Ca2+-activated potassium channel in mitochondria of endothelial EA.hy926 cells. Am J Physiol - Hear Circ Physiol. 2013;304(11):1415–27.

22. Yun J FT. Mitohormesis. Cell Metab. 2014;19:757–66.

23. Ristow M, Schmeisser K. Mitohormesis: Promoting health and lifespan by increased levels of reactive oxygen species (ROS). Dose-Response. 2014;12(2):288–341.

24. Honrath B, Matschke L, Meyer T, Magerhans L, Perocchi F, Ganjam GK, et al. SK2 channels regulate mitochondrial respiration and mitochondrial Ca2+ uptake. Cell Death Differ. 2017;(2017):1–13.

25. Piwonska M, Szewczyk A, Schroder UH, Reymann KG, Bednarczyk I, Piwońska M, et al. Effectors of large-conductance calcium-activated potassium channel modulate glutamate excitotoxicity in organotypic hippocampal slice cultures. Acta Neurobiol Exp (Wars). 2016;76(1689-0035 (Electronic)):20–31.

26. Fretwell L, Dickenson JM. Role of large-conductance Ca 2 + -activated potassium channels in adenosine A 1 receptor-mediated pharmacological preconditioning in H9c2 cells. Eur J Pharmacol. 2008;618(1–3):37–44.

27. Stowe DF, Aldakkak M, Camara AKS, Riess ML, Heinen A, Varadarajan SG, et al. Cardiac mitochondrial preconditioning by Big Ca2+-sensitive K+ channel opening requires superoxide radical generation. Am J Physiol Hear Circ Physiol. 2006;53226:434–40. 28. Stowe DF. Protection against cardiac injury by small Ca2+-sensitive K+ channels

identified in guinea pig cardiac inner mitochondrial membrane. Biochem Biophys Acta. 2013;1828(2):427–42.

29. Südhof TC. Calcium control of neurotransmitter release. Cold Spring Harb Perspect Biol. 2012;4(1).

30. Naia L, Ferreira IL, Ferreiro E, Rego AC. Mitochondrial Ca2+ handling in Huntington’s and Alzheimer’s diseases – Role of ER-mitochondria crosstalk. Biochem Biophys Res Commun. 2017;483(4):1069–77.

31. Cárdenas C, Miller RA, Smith I, Bui T, Molgó J, Müller M, et al. Essential Regulation of Cell Bioenergetics By Constitutive InsP3 Receptor Ca2+ Transfer to Mitochondria. Cell. 2011;142(2):270–83.

32. Denton RM, McCormack JG. The role of calcium in the regulation of mitochondrial metabolism. Biochem Soc Tran. 1980;8(3):266–8.

33. Hansford RG. Physiological role of mitochondrial Ca2+ transport. J Bioenerg Biomembr. 1994 Oct;26(5):495–508.

34. Lemasters J, Theruvath T, Zhong Z, Nieminen a. Mitochondrial Calcium and the Permeability Transition in Cell Death. Biochim Biophys Acta. 2010;1787(11):1395–401. 35. Chacon E, Acosta D. Mitochondrial regulation of superoxide by Ca2+: an alternate

mechanism for the cardiotoxicity of doxorubicin. Toxicol Appl Pharmacol. 1991 Jan;107(1):117–28.

36. Egnatchik RA, Leamy AK, Jacobson DA, Shiota M, Young JD. ER calcium release promotes mitochondrial dysfunction and hepatic cell lipotoxicity in response to palmitate overload. Mol Metab. 2014;3(5):544–53.

37. De Stefani D, Bononi A, Romagnoli a, Messina A, De Pinto V, Pinton P, et al. VDAC1 selectively transfers apoptotic Ca(2+) signals to mitochondria. Cell Death Differ. 2012;19(2):267–73.

(16)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 227PDF page: 227PDF page: 227PDF page: 227

227

8

38. Dingreville F, Panthu B, Thivolet C, Ducreux S, Gouriou Y, Pesenti S, et al. Differential

effect of glucose on ER-mitochondria Ca2+ exchange participates in insulin secretion and glucotoxicity-mediated dysfunction of β-cells. Diabetes. 2019;68(9):1778–94.

39. Csordas G, Varnai P, Golenar T, Roy S, Purkins G, Schneider TG, Balla T HG. Imaging interorganelle contacts and local calcium dynamics at the ER-mitochondrial interface. Mol Cell. 2010;39(1):121–32.

40. Area-Gomez E, Del Carmen Lara Castillo M, Tambini MD, Guardia-Laguarta C, De Groof AJC, Madra M, et al. Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. EMBO J. 2012;31(21):4106–23

41. Ottolini D, Calì T, Negro A, Brini M. The Parkinson disease-related protein DJ-1 counteracts mitochondrial impairment induced by the tumour suppressor protein p53 by enhancing endoplasmic reticulum-mitochondria tethering. Hum Mol Genet. 2013;22(11):2152–68.

42. Arduino DM, Wettmarshausen J, Vais H, Navas-Navarro P, Cheng Y, Leimpek A, Ma Z, Delrio-Lorenzo A, Giordano A, Garcia-Perez C, Medard G, Kuster B, Garcia-Sancho J, Mokranjac D, Foskett JK, Teresa Alonso M PF. Systematic Identification of MCU Modulators by Orthogonal Interspecies Chemical Screening. Mol Cell. 2017;67(4):711– 23.

43. Sarre A, Gardier S, Maurer F, Bonny C, Raddatz E. Modulation of the c-Jun N-terminal kinase activity in the embryonic heart in response to anoxia-reoxygenation: Involvement of the Ca2+ and mitoKATP channels. Mol Cell Biochem. 2008;313(1–2):133–8.

44. Zhang L, Gao X, Yuan X, Dong H, Zhang Z, Wang S. Mitochondrial calcium uniporter opener spermine attenuates the cerebral protection of diazoxide through apoptosis in rats. J Stroke Cerebrovasc Dis. 2014;23(5):829–35.

45. Dai H, Wang M, Patel PN, Kalogeris T, Liu Y, Durante W, et al. Preconditioning with the BKCa channel activator NS-1619 prevents ischemiareperfusion- induced inflammation and mucosal barrier dysfunction: Roles for ROS and heme oxygenase-1. Am J Physiol - Hear Circ Physiol. 2017;313(5):H988–99.

46. Raupach, Reinle, Stroethoff, Mathes, Heinen, Hollmann, et al. Milrinone-Induced Pharmacological Preconditioning in Cardioprotection: Hints for a Role of Mitochondrial Mechanisms. J Clin Med. 2019;8(4):507.

47. Pertiwi KR, Hillman RM, Scott CA, Chilton EL. Ischemia Reperfusion Injury Produces, and Ischemic Preconditioning Prevents, Rat Cardiac Fibroblast Differentiation: Role of KATP Channels. J Cardiovasc Dev Dis. 2019;6(2):22.

48. Arabian M, Aboutaleb N, Soleimani M, Ajami M, Habibey R, Pazoki-Toroudi H. Activation of mitochondrial KATP channels mediates neuroprotection induced by chronic morphine preconditioning in hippocampal CA-1 neurons following cerebral ischemia. Adv Med Sci. 2018;63(2):213–9.

49. Doul J, Miková D, Rašková M, Ošťádalová I, Maxová H, Ošťádal B, et al. Possible role of mitochondrial K-ATP channel and nitric oxide in protection of the neonatal rat heart. Mol Cell Biochem. 2019;450(1–2):35–42.

50. Ma D, Feng L, Cheng Y, Xin M, You J, Yin X, et al. Astrocytic gap junction inhibition by carbenoxolone enhances the protective effects of ischemic preconditioning following cerebral ischemia. J Neuroinflammation. 2018;15(1):1–12.

51. Richter M, Nickel C, Apel L, Kaas A, Dodel R, Culmsee C, et al. SK channel activation modulates mitochondrial respiration and attenuates neuronal HT-22 cell damage induced by H2O2. Neurochem Int. 2015;81:63–75.

52. Murakami Y, Ito M, Ohsawa I. Molecular hydrogen protects against oxidative stress-induced SH-SY5Y neuroblastoma cell death through the process of mitohormesis. PLoS One. 2017;12(5):1–14.

(17)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 228PDF page: 228PDF page: 228PDF page: 228

228

53. Xiong LG, Chen YJ, Tong JW, Gong YS, Huang JA, Liu ZH. Epigallocatechin-3-gallate promotes healthy lifespan through mitohormesis during early-to-mid adulthood in Caenorhabditis elegans. Redox Biol. 2018;14(September 2017):305–15.

54. Cox CS, McKay SE, Holmbeck MA, Christian BE, Scortea AC, Tsay AJ, et al. Mitohormesis in Mice via Sustained Basal Activation of Mitochondrial and Antioxidant Signaling. Cell Metab. 2018;28(5):776-786.e5.

55. Van Raamsdonk JM, Hekimi S. Deletion of the mitochondrial superoxide dismutase sod-2 extends lifespan in Caenorhabditis elegans. PLoS Genet. sod-2009;5:e1000361.

56. Baumgart M, Priebe S, Groth M, Hartmann N, Menzel U, Pandolfini L, et al. Longitudinal RNA-seq analysis of vertebrate aging identifies mitochondrial complex i as a small-molecule-sensitive modifier of lifespan. Cell Syst. 2016;2(2):122–32.

57. Robb EL, Hall AR, Prime TA, Eaton S, Szibor M, Viscomi C, et al. Control of mitochondrial superoxide production by reverse electron transport at complex I. J Biol Chem. 2018;293(25):9869–79.

58. Dubouchaud H, Walter L, Rigoulet M, Batandier C. Mitochondrial NADH redox potential impacts the reactive oxygen species production of reverse Electron transfer through complex I. J Bioenerg Biomembr. 2018;50(5):367–77.

59. Scialò F, Sriram A, Fernández-Ayala D, Gubina N, Lõhmus M, Nelson G, et al. Mitochondrial ROS Produced via Reverse Electron Transport Extend Animal Lifespan. Cell Metab. 2016;23(4):725–34.

60. Mills EL, Kelly B, Logan A, Costa ASH, Varma M, Bryant CE, et al. Succinate Dehydrogenase Supports Metabolic Repurposing of Mitochondria to Drive Inflammatory Macrophages. Cell. 2016;167(2):457-470.e13.

61. Sun W, Pang Y, Liu Z, Sun L, Liu B, Xu M, et al. Macrophage inflammasome mediates hyperhomocysteinemia-aggravated abdominal aortic aneurysm. J Mol Cell Cardiol. 2015;81:96–106.

62. Heinen AA, Aldakkak M, Stowe DF, Rhodes SS, Riess ML, Varadarajan SG, et al. Reverse electron flow-induced ROS production is attenuated by activation of mitochondrial Ca2+-sensitive K+ channels. Am J Physiol Heart Circ Physiol. 2007;293(3):H1400–7. 63. Ren W, Xia Y, Chen S, Wu G, Bazer FW, Zhou B, et al. Glutamine Metabolism in

Macrophages: A Novel Target for Obesity/Type 2 Diabetes. Adv Nutr. 2019;10(2):221– 30.

64. Kenny TC, Craig AJ, Villanueva A, Germain D. Mitohormesis Primes Tumor Invasion and Metastasis. Cell Rep. 2019;27(8):2292-2303.e6.

65. Zou Z, Chang H, Li H, Wang S. Induction of reactive oxygen species : an emerging approach for cancer therapy. Apoptosis. 2017;22(11):1321–35.

66. Hassan M, Sami R, Abida S, Usman A. ROS ‑ modulated therapeutic approaches in cancer treatment. J Cancer Res Clin Oncol. 2017;143(9):1789–809.

67. Su BC, Pan CY, Chen JY. Antimicrobial peptide TP4 induces ROS-mediated necrosis by triggering mitochondrial dysfunction in wild-type and mutant p53 glioblastoma cells. Cancers (Basel). 2019;11(2):1–18.

68. Gersey ZC, Rodriguez GA, Barbarite E, Sanchez A, Walters WM, Ohaeto KC, et al. Curcumin decreases malignant characteristics of glioblastoma stem cells via induction of reactive oxygen species. BMC Cancer. 2017;17(1):1–11.

69. Singer E, Judkins J, Salomonis N, Matlaf L, Soteropoulos P, McAllister S, et al. Reactive oxygen species-mediated therapeutic response and resistance in glioblastoma. Cell Death Dis. 2015;6(1):e1601-11.

70. Bijangi-Vishehsaraei K, Saadatzadeh MR, Wang H, Nguyen A, Kamocka MM, Cai W, et al. Sulforaphane suppresses the growth of glioblastoma cells, glioblastoma stem cell-like spheroids, and tumor xenografts through multiple cell signaling pathways. J Neurosurg. 2017;127(6):1219–30.

(18)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 229PDF page: 229PDF page: 229PDF page: 229

229

8

71. Leanza L, Romio M, Becker KA, Azzolini M, Trentin L, Managò A, et al. Direct

Pharmacological Targeting of a Mitochondrial Ion Channel Selectively Kills Tumor Cells In Vivo. Cancer Cell. 2017;31(4):516-531.e10.

72. Szabò I, Bock J, Grassmé H, Soddemann M, Wilker B, Lang F, et al. Mitochondrial potassium channel Kv1.3 mediates Bax-induced apoptosis in lymphocytes. Proc Natl Acad Sci U S A. 2008;105(39):14861–6.

73. Catacuzzeno L, Franciolini F. Role of KCa3.1 channels in modulating ca2+ oscillations during glioblastoma cell migration and invasion. Int J Mol Sci. 2018;19(10):1–15. 74. Mohr CJ, Steudel FA, Gross D, Ruth P, Lo WY, Hoppe R, et al. Cancer-associated

intermediate conductance Ca 2+ -activated K + channel K Ca 3.1. Cancers (Basel). 2019;11(1):1–22.

75. Janiszewska J, Posadas I, Jativa P, Bugaj-Zarebska M, Urbanczyk-Lipkowska Z, Ceña V. Second generation amphiphilic poly-lysine dendrons inhibit glioblastoma cell proliferation without toxicity for neurons or astrocytes. PLoS One. 2016;11(11):1–21. 76. Llorente-Folch I, Rueda CB, Pérez-Liébana I, Satrústegui J, Pardo B. L-lactate-mediated

neuroprotection against glutamate- induced excitotoxicity requires ARALAR/AGC1. J Neurosci. 2016;36(16):4443–56.

77. Jourdain P, Allaman I, Rothenfusser K, Fiumelli H, Marquet P, Magistretti PJ. L-Lactate protects neurons against excitotoxicity: Implication of an ATP-mediated signaling cascade. Sci Rep. 2016;6(September 2015):1–13.

78. Zelenka J, Dvořák A, Alán L. L-Lactate protects skin fibroblasts against aging-Associated mitochondrial dysfunction via mitohormesis. Oxid Med Cell Longev. 2015;2015. 79. Tauffenberger A, Fiumelli H, Almustafa S, Magistretti PJ. Lactate and pyruvate promote

oxidative stress resistance through hormetic ROS signaling. Cell Death Dis. 2019;10(9). 80. Ling S, Shan Q, Liu P, Feng T, Zhang X, Xiang P, et al. Metformin ameliorates arsenic

trioxide hepatotoxicity via inhibiting mitochondrial complex i. Cell Death Dis. 2017;8(11). 81. Thwe PM, Amiel E. The role of nitric oxide in metabolic regulation of Dendritic cell

immune function. Cancer Lett. 2018;412:236–42.

82. Almeida A, Cidad P, Delgado-Esteban M, Fernández E, García-Nogales P, Bolaños JP. Inhibition of mitochondrial respiration by nitric oxide: Its role in glucose metabolism and neuroprotection. J Neurosci Res. 2005;79(1–2):166–71.

83. Schmeisser S, Priebe S, Groth M, Monajembashi S, Hemmerich P, Guthke R, et al. Neuronal ROS signaling rather than AMPK/sirtuin-mediated energy sensing links dietary restriction to lifespan extension. Mol Metab. 2013;2(2):92–102.

84. Krawczyk CM, Holowka T, Sun J, Blagih J, Amiel E, DeBerardinis RJ, et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood. 2010;115(23):4742–9.

85. Tannahill GM, Curtis AM, Adamik J, Palsson-Mcdermott EM, McGettrick AF, Goel G, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496(7444):238–42.

86. Dolga AM, Letsche T, Gold M, Doti N, Bacher M, Chiamvimonvat N, et al. Activation of KCNN3/SK3/KCa2.3 channels attenuates enhanced calcium influx and inflammatory cytokine production in activated microglia. Glia. 2012;60(12):2050–64.

87. Brix B, Mesters JR, Pellerin L, Jöhren O. Endothelial cell-derived nitric oxide enhances aerobic glycolysis in astrocytes via HIF-1α-mediated target gene activation. J Neurosci. 2012;32(28):9727–35.

88. Yang X, Wang G, Cao T, Zhang L, Ma Y, Jiang S, et al. Large-conductance calcium-activated potassium channels mediate lipopolysaccharide-induced activation of murine microglia. J Biol Chem. 2019;294(35):12921–32.

(19)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 230PDF page: 230PDF page: 230PDF page: 230

230

89. Debska-Vielhaber G, Godlewski MM, Kicinska A, Skalska J, Kulawiak B, Piwonska M, et al. Large-conductance K+ channel openers induce death of human glioma cells. 2009;(8):27–36.

(20)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 231PDF page: 231PDF page: 231PDF page: 231

231

(21)

549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam 549886-L-bw-Krabbendam Processed on: 27-10-2020 Processed on: 27-10-2020 Processed on: 27-10-2020

Processed on: 27-10-2020 PDF page: 232PDF page: 232PDF page: 232PDF page: 232

Ca

Ca

Ca

2+

Ca

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

Ca

Ca

Ca

2+

Referenties

GERELATEERDE DOCUMENTEN

Once they are within the dopamine neurons, they selectively inhibit mitochondrial complex I and the inhibition of which affects respiratory chain function resulting in the

Preconditioning and postconditioning mechanisms elicited by activated mitoBK channels reduce the infarct size due to ischemic stress via attenuation of mitochondrial ROS

To study the consequences of increased EMC on neuronal cell death signaling, we induced oxidative stress and mitochondrial damage in neuronal HT22 cells by glutamate (22).. In

Since inhibition of the glycolytic activity only partially reduced the neuroprotection mediated by CyPPA and since opening of the channels slightly decreased complex I and

Annexin/PI analysis showed that CyPPA alone had no impact on cell viability in HT22 cells in a concentration range of 10–50 μM, while in SK-N-AS cells it reduced

dat behandeling met SK-kanaal modulator CyPPA morfologische veranderingen bij activatie van macrofagen verminderde. Bovendien zorgde activatie van SK- kanalen voor verlaging in

The role of small conductance calcium-activated potassium channels in mitochondrial dysfunction – Targeting metabolic reprogramming and

CI inhibitors caused a potent decrease in OCR, decreased mitochondrial membrane potential, increased ECAR and increased lactate pro- duction in both cell types..