Sten Orrenius’s research while affiliated with Karolinska Institutet and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (393)


Role of Cell Death in Toxicology: Does It Matter How Cells Die?
  • Literature Review

January 2019

·

11 Reads

·

19 Citations

Annual Review of Pharmacology

Sten Orrenius

My research activity started with studies on drug metabolism in rat liver microsomes in the early 1960s. The CO-binding pigment (cytochrome P450) had been discovered a few years earlier and was subsequently found to be involved in steroid hydroxylation in adrenal cortex microsomes. Our early studies suggested that it also participated in the oxidative demethylation of drugs catalyzed by liver microsomes, and that prior treatment of the animals with phenobarbital caused increased levels of the hemoprotein in the liver, and similarly enhanced rates of drug metabolism. Subsequent studies of cytochrome P450-mediated metabolism of toxic drugs in freshly isolated rat hepatocytes characterized critical cellular defense systems and identified mechanisms by which accumulating toxic metabolites could damage and kill the cells. These studies revealed that multiple types of cell death could result from the toxic injury, and that it is important to know which type of cell death results from the toxic injury. Expected final online publication date for the Annual Review of Pharmacology and Toxicology Volume 59 is January 6, 2019. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.


Mitochondria – a switch board between various cell death modalities

January 2016

·

43 Reads

For many years, mitochondria were regarded exclusively as cellular power plants, destined to provide the cell with energy in the form of ATP. Mitochondrial deterioration can have fatal consequences for cellular physiology and be a cause of various diseases. Subsequent studies revealed the involvement of mitochondria in the regulation and execution of multiple modalities of cell death. Analysis of the involvement of mitochondria in the control of various modes of cell death led to the conclusion that mitochondria might play a leading role as a “switch-board” between various cell death modalities.


Calcium and mitochondria in the regulation of cell death

April 2015

·

233 Reads

·

483 Citations

Biochemical and Biophysical Research Communications

The calcium ion has long been known to play an important role in cell death regulation. Hence, necrotic cell death was early associated with intracellular Ca(2+) overload, leading to mitochondrial permeability transition and functional collapse. Subsequent characterization of the signaling pathways in apoptosis revealed that Ca(2+)/calpain was critically involved in the processing of the mitochondrially localized, Apoptosis Inducing Factor. More recently, the calcium ion has been demonstrated to play important regulatory roles also in other cell death modalities, notably autophagic cell death and anoikis. In this review, we summarize current knowledge about the mechanisms involved in Ca(2+) regulation of these various modes of cell death with a focus on the importance of the mitochondria. Copyright © 2015 Elsevier Inc. All rights reserved.


Figure 2. Role of Ca 2+ in As 2 O 3-induced apoptosis. (A) Effects of the BAPTA-AM on DNA fragmentation. WT and DKO MEFs were loaded with 10 µM BAPTA-AM for 30 min and then were exposed to 10 µM As 2 O 3 for 8 h. Cells were harvested and DNA fragmentation was quantified by PI/FACS. Mean ± SD, n = 6. (B) Effects of BAPTA-AM on cytochrome c release. Cells loaded with or without 10 µM BAPTA-AM were exposed to 10 µM As 2 O 3 for 8 h. Cytosolic extracts from these cells were prepared and cytochrome c content in the extracts was measured by immunoblotting as described in Experimental Procedures. (C) Quantitative analysis of cytochrome c release. Relative cytochrome c/GAPDH band intensities were determined in the immunoblot displayed in (B) by densitometry (AIDA, BioRad, Inc.).
Figure 3. Concentration-dependent effects of As 2 O 3 on necrosis. WT and DKO MEFs were treated with As 2 O 3 for the indicated periods of time, and necrotic cell death was quantified by propidium iodide uptake and flow cytometry. (A) 125 µM As 2 O 3 ; (B) 500 µM As 2 O 3 ; (C) 1 mM As 2 O 3. (D) Effects of As 2 O 3 on cytochrome c release. WT and DKO MEFs were treated with 1 mM As 2 O 3 , and cytochrome c was detected in cytosolic extracts by immunoblotting as described in Experimental Procedures. Results shown are representative of those obtained in four independent experiments.
Figure 4. Effects of As 2 O 3 on isolated mitochondria. (A) Effects of As2O3 on mitochondrial respiration. Rotenone was used to inhibit complex I and antimycin A (AA) was used to block complex III. ADP was added to stimulate state 3 respiration. Bak -/-mouse liver mitochondria (1 mg/ml) were incubated in an oxygen electrode chamber with or without 125 mM As2O3, and oxygen consumption was measured using a Clark electrode as described in Experimental Procedures. In all of the experiments identical results were obtained with WT and Bak -/-mitochondria. Complex substrates: malate /pyruvate for complex I, succinate for complex II, TMPD/ascorbate (asc) for Complex IV. (B) Effects of As2O3 on respiratory control ratio (RCR). RCR was estimated by measuring the ratio between state 3 respiration and state 4 respiration . Uncoupled respiration was estimated in the presence of 1 µM CCCP with either pyruvate/malate or succinate as a substrate. Bak -/-mitochondria were treated with 50 µM, 500 µM, or 1000 µM As 2 O 3 with or without 1 mM DTT present. Results were identical in Wt mitochondria. (C) Cooperative effects of As2O3 and Ca 2+ on cytochrome c release. Bak -/mitochondria oxidizing succinate were incubated for 15 min in the absence or presence of 125 µM As2O3 with or without 20 nmol CaCl 2 and 1 mM DTT. Cytochrome c release was quantified by immunoblotting. The results shown are reflective of four different experiments. Indistinguishable results were obtained with WT mitochondria.  
Figure 7. As 2 O 3-induced ROS-production and caspase-3 inactivation. (A) Effects of As 2 O 3 on ROS production. MEFs were treated with 1 mM As 2 O 3 for the times indicated and then stained with dihydroethidium to measure superoxide (O 2-) production by flow cytometry. Percentages of HE-positive cells are shown. Mean ± SD, n = 5. (B) Effects of As 2 O 3 on caspase activation in whole cells. DKO cells were treated with As 2 O 3 for 3 h and caspase-3 activity was measured as described in Experimental Procedures. Where indicated, cells were incubated in caspase reaction buffer containing 5 mM DTT for 1 h at 30˚C and then analyzed for DEVD-AMC cleavage. The data are from one representative experiment out of three: , untreated; , 500 µM As 2 O 3 ; 1 mM As 2 O 3 ; and +, positive control (1 mM dATP and 4 µM cytochrome c added to untreated, permeabilized cells). (C) Direct effects of As 2 O 3 on caspase-3. Active recombinant caspase-3 (rCasp-3) was treated with 500 µM or 1 mM As 2 O 3 and 5 mM DTT, and caspase activity was measured using DEVD-AMC as described in Experimental Procedure. Results shown are from one experiment that was representative of three independent replicates.
Bax and Bak are required for cytochrome c release during arsenic trioxide-induced apoptosis
  • Article
  • Full-text available

October 2014

·

75 Reads

·

21 Citations

Leta K. Nutt

·

Vladimir Gogvadze

·

·

[...]

·

Sten Orrenius

Arsenic trioxide is a potent chemotherapeutic agent that selectively triggers apoptosis in tumor cells. Previous studies have demonstrated that arsenicals cause direct damage to mitochondria, but it is not clear that these effects initiate apoptosis. Here we used Bak-/- mouse liver mitochondria and virally immortalized Bax-/-Bak-/- mouse embryonic fibroblasts (MEFs) to investigate whether or not multidomain proapoptotic Bcl-2 family proteins were required for arsenic-induced mitochondrial damage and cell death. Near clinically achievable concentrations, arsenic stimulated cytochrome c release and apoptosis via a Bax/Bak- dependent mechanism. At higher concentrations (125 mM- 1 mM), cells died via a Bax/Bak-independent mechanism mediated by oxidative stress that resulted in necrosis. Consistent with previous reports, arsenic directly inhibited complex I of the mitochondrial electron transport chain, which resulted in mitochondrial permeability transition (MPT), the generation of reactive oxygen species (ROS), and thiol oxidation. However, these effects only occurred at concentrations of arsenic trioxide of 50 mM and higher, and the oxidative stress associated with them blocked caspase activation. Our data demonstrate for the first time that the cytochrome c release which initiates apoptosis in cells exposed to this classic mitochondrial poison occurs indirectly via the activation of Bax/Bak rather than via direct mitochondrial damage. Furthermore, the results implicate reactive oxygen species in a concentration-dependent mechanistic switch between apoptosis and necrosis.

Download

Targeting Hepatoma Using Nitric Oxide Donor Strategies

February 2013

·

140 Reads

·

28 Citations

Antioxidants and Redox Signaling

Aims: The study evaluated the role of increased intracellular nitric oxide (NO) concentration using NO donors or stably NO synthase-3 (NOS-3) overexpression during CD95-dependent cell death in hepatoma cells. The expression of cell death receptors and caspase activation, RhoA kinase activity, NOS-3 expression/activity, oxidative/nitrosative stress, and p53 expression were analyzed. The antitumoral activity of NO was also evaluated in the subcutaneous implantation of NOS-3-overexpressing hepatoma cells, as well NO donor injection into wild-type hepatoma-derived tumors implanted in xenograft mouse models. Results: NO donor increased CD95 expression and activation of caspase-8 and 3 in HepG2, Huh7, and Hep3B cells. NOS-3 overexpression increased oxidative/nitrosative stress, p53 and CD95 expression, cellular Fas-associated death domain (FADD)-like IL-1beta converting enzyme (FLICE) inhibitory protein long (cFLIP(L)) and its short isoform (cFLIP(S)) shift, and cell death in HepG2 (4TO-NOS) cells. The inhibition of RhoA kinase and p53 knockdown using RNA interference reduced cell death in 4TO-NOS cells. The supplementation with hydrogen peroxide (H(2)O(2)) increased NOS-3 activity and cell death in 4TO-NOS cells. NOS-3 overexpression or NO donor injection into hepatoma-derived tumors reduced the size and increased p53 and cell death receptor expression in nude mice. Innovation and conclusions: The increase of intracellular NO concentration promoted oxidative and nitrosative stress, Rho kinase activity, p53 and CD95 expression, and cell death in cultured hepatoma cells. NOS-3-overexpressed HepG2 cells or intratumoral NO donor administration reduced tumor cell growth and increased the expression of p53 and cell death receptors in tumors developed in a xenograft mouse model.


Reactive Oxygen Species generated in different compartments induce cell death, survival or senescence.

January 2013

·

202 Reads

·

145 Citations

Free Radical Biology and Medicine

Although reactive oxygen species (ROS) are well established mediators of oxidative damage and cell demise, the mechanisms by which they trigger specific cell death modalities and the temporal/spatial requirements underlying this phenomenon are largely unknown. Yet, it is well established that most anticancer therapies depend on ROS production for efficient tumor eradication. Using several non-small cell lung cancer cell lines, we have dissected how the site of ROS production and accumulation in various cell compartments affect cell fate. We demonstrate that high levels of exogenously generated H(2)O(2)induce extensive DNA damage, ATP depletion and severe cytotoxicity. While these effects were independent of caspase activity, they could - at least in part - be prevented by RIP1 kinase inhibition. In contrast, low levels of exogenously produced H(2)O(2) triggered a modest drop in ATP level, delayed toxicity, G2/M arrest and cell senescence. Mitochondrially-produced H(2)O(2) induced a reversible ATP drop without affecting cell viability. Instead, the cells accumulated in the G1/S phase of the cell cycle and became senescent. Concomitant inhibition of glycolysis was found to markedly sensitize cells to death in the presence of otherwise nontoxic concentrations of H(2)O(2,) presumably by the inhibition of ATP restoring mechanisms. Combined, our data provide evidence that ROS might dictate different cellular consequences depending on their overall concentration at steady state levels and on their site of generation.


Autophagy in Toxicology: Cause or Consequence?

October 2012

·

104 Reads

·

71 Citations

Annual Review of Pharmacology

Research on autophagy and its effects on cell metabolism and physiology has increased dramatically during recent years. Multiple forms of autophagy have been characterized, and many of the genes involved in the regulation of this process have been identified. The importance of autophagy for embryonic development and maintenance of tissue homeostasis in the adult organism has been demonstrated convincingly, and several human diseases have been linked to deficiencies in autophagy. Most often, autophagy serves as a protective mechanism, but persistent activation of autophagy can result in cell death. This is true for many toxic agents. In fact, there are ample examples of cross talk between autophagy and other modes of cell death after exposure to toxicants. However, the relative contribution of autophagy to the overall toxicity of these compounds is not always clear, and further research is needed to clarify the toxicological significance of this process. Expected final online publication date for the Annual Review of Pharmacology and Toxicology Volume 53 is January 06, 2013. Please see http://www.annualreviews.org/catalog/pubdates.aspx for revised estimates.


Citrate kills tumor cells through activation of apical caspases

October 2012

·

62 Reads

·

45 Citations

Cellular and Molecular Life Sciences

Most tumor cells exhibit a glycolytic phenotype. Thus, inhibition of glycolysis might be of therapeutic value in antitumor treatment. Among the agents that can suppress glycolysis is citrate, a member of the Krebs cycle and an inhibitor of phosphofructokinase. Here, we show that citrate can trigger cell death in multiple cancer cell lines. The lethal effect of citrate was found to be related to the activation of apical caspases-8 and -2, rather than to the inhibition of cellular energy metabolism. Hence, increasing concentrations of citrate induced characteristic manifestations of apoptosis, such as caspase-3 activation, and poly-ADP-ribose polymerase cleavage, as well as the release of cytochrome c. Apoptosis induction did not involve the receptor-mediated pathway, since the processing of caspase-8 was not attenuated in cells deficient in Fas-associated protein with Death Domain. We propose that the activation of apical caspases by citrate could be explained by its kosmotropic properties. Caspase-8 is activated by proximity-induced dimerization, which might be facilitated by citrate through the stabilization of intermolecular interactions between the proteins.


Mitochondrial Involvement in the Execution of Cell Death

June 2012

·

6 Reads

Investigation of different forms of cell death has become an important area of biomedical research. Recently, several cell death modalities, in addition to necrosis and apoptosis, have been described and characterized based on morphological and biochemical criteria. In 2009, the Nomenclature Committee on Cell Death proposed unified criteria for the definition of 12 cell death modalities (Kroemer et al. 2009). Among the best characterized of these modes of cell death are apoptosis, autophagy, cornification, and necrosis. Until recently, a requirement for gene expression was documented only for apoptotic and autophagic cell death. Cornification is a special form of programmed cell death in the epidermis. To some extent, it represents an example of terminal differentiation, similar to the maturation of red blood cells or lens epithelium, although there are significant differences between them at the biochemical level. The interaction between the different forms of cell death is complex and is still a matter of debate. However, recently, the mitochondria have been demonstrated to play a crucial role in the effectuation of several cell death modalities, although the precise mechanisms of their involvement are still unclear. In this review, we focus on the mitochondrial involvement in four cell death modalities, namely, necrosis, apoptosis, autophagy, and anoikis (Figure 2.1).



Citations (91)


... Extensive injury leads to organ dysfunction or failure. Various types of cell death processes including apoptosis, necrosis, ferroptosis, pyroptosis and autophagy are in a dynamic flux from the time of injury initiation to the final outcome of injury and can be modeled based on the available knowledge on "Mechanisms of Toxicity" in a number of target organs (Naughton and Terry, 2018;Orrenius, 2019;Tahrir et al., 2019;Dong et al., 2019;Rusyn et al., 2021;Ajoolabady et al., 2023;Costa and Gołembiowska, 2022;Perazella, 2019;Barnett and Cummings, 2019;Bhagat and Kleinerman, 2020;Ray et al., 2019, Ray et al., 2021, Ray et al., 2022Skála et al., 2019;Yang et al., 2015;Ketelut-Carneiro and Fitzgerald, 2022;Patel and Karch, 2020;Lu et al., 2021;Christidi et al., 2018;Li et al., 2022a, Li et al., 2022bRayego-Mateos et al., 2023;Maremonti et al., 2022;Shi et al., 2021;Wesley et al., 2021;Kasture et al., 2021). production. ...

Reference:

Mechanisms of toxicity
Role of Cell Death in Toxicology: Does It Matter How Cells Die?
  • Citing Article
  • January 2019

Annual Review of Pharmacology

... Cytochrome-c teams up with PROCASPASE-9 and APAF-1 to create the apoptosome, a crucial complex in the cell. This powerful formation triggers the auto-activation of CASPASE-9, which then turns-on the effector CASPASE-3, driving the cell towards apoptosis 119,120 . The mRNA expression of gadd34 surged by an impressive 45.8 ± 1.20 and 11.40 ± 2.51 fold, indicating a substantial upregulation in T1 and T2 compared to control group after 6 weeks. ...

Calcium and mitochondria in the regulation of cell death
  • Citing Article
  • April 2015

Biochemical and Biophysical Research Communications

... growth inhibition and several physiological disorders of plants, and can ultimately lead to death [14,15]. Arsenic toxicity induces the production and accumulation of ROS that damage biomolecules (proteins, DNA and lipids) and eventually cause cell death [16,17]. ...

Bax and Bak are required for cytochrome c release during arsenic trioxide-induced apoptosis

... Another cardioprotective mechanism of miR-335 during sepsis may lie in its ability to reduce apoptosis (59). A large number of studies has demonstrated that myocardial infarction, myocardial hypertrophy and heart failure induce excessive apoptosis and that myocardial apoptosis is involved in SIMI (37,(60)(61)(62). Sepsis-associated inflammatory factors induce cardiomyocyte apoptosis by activating the caspase pathway (63). ...

The Potential Role of Apoptosis in Human Disease
  • Citing Article
  • May 2000

Medical Principles and Practice

... 29,34 Induction of HSP27 has been shown to protect cells against stress-induced apoptosis. [34][35][36] In addition, HSP27 interacts with actin to maintain cytoskeletal integrity and promote survival in the face of stress stimuli. 2,30 sHSPs are ATP-independent chaperones which form dynamic oligomeric structures around unfolded proteins to hold these client proteins in a partially competent state 37 until transfer to ATP dependent chaperone complexes like HSP70/40 to refold the protein. ...

Hsp27 protects mitochondria of thermotolerant cells against apoptotic stimuli

Cell Stress and Chaperones

... The superoxide anion radical (O 2 À ) formed in aerobic metabolism upon reduction of dioxygen, as a kind of reactive oxygen species (ROS), is an important biological substance in living organisms. 1 In healthy systems, the appropriate concentration of ROS plays a role in cellular signal pathways of biological processes, killing the invading funguses, viruses and bacteria. 2 However, excess of generated ROS in vivo can oxidize proteins, lipids and nucleic acids, and eventually lead to a variety of diseases, for example, Parkinson's and Alzheimer's, diabetes, inflammation and age-related diseases. ...

Free Radicals in the 20th Century
  • Citing Article
  • June 1999

Science

... Panieri et al. reported that mitochondrially derived H 2 O 2 induced G1 phase cell cycle arrest, while low exogenous H 2 O 2 caused G2/M phase accumulation. These findings suggest that mitochondrial mechanisms and intracellular H 2 O 2 levels play essential roles in cell cycle regulation and DNA damage response [102]. In an in vivo study, chelidonic acid was found to have similar effects to our study on MDA levels, SOD and GSH activities [77]. ...

Reactive Oxygen Species generated in different compartments induce cell death, survival or senescence.
  • Citing Article
  • January 2013

Free Radical Biology and Medicine

... Several other non-apoptotic programmed cell death modalities have also been reported following ionophore exposure. Activation of canonical and noncanonical autophagy pathways [16,[19][20][21]53], associated with impaired endolysosomal functions [61,62], osmotic imbalances [41], ER stress [63][64][65], ROS production, and activation of protein kinase signalling has been shown.in numerous cell lines [16, 19-21, 41, 53, 61-65]. ...

Autophagy in Toxicology: Cause or Consequence?
  • Citing Article
  • October 2012

Annual Review of Pharmacology

... This relationship between cfDNA and disease has been a subject of study ever since. There are multiple possible pathways for the release of DNA fragments from cells, including apoptosis, necrosis, and exosome secretion [3][4][5][6]. Processes that increase the release of cfDNA include disease, inflammation, tissue injury, and exercise [7,8]. In healthy individuals, haematopoietic maturation is a major contributor to the normal cfDNA pool. ...

Current Protocols in Toxicology
  • Citing Article
  • May 2001

Current Protocols in Toxicology

... The specific MMP required for ECD sPD-1 production warrants further study. We also observed FL sPD-1 production along with PD-1 + T cell death; this is not surprising, because all cell components, including membrane-bound proteins, would be released when cells die 8,41 . Moreover, when lymphoma cells or activated T cells begin to die, ECD sPD-1 production begins to decline, but FL sPD-1 increases; and vice versa, mice producing high levels of ECD sPD-1 generally have low FL sPD-1 levels. ...

Current Concepts in Cell Toxicity
  • Citing Article
  • May 2001

Current Protocols in Toxicology