Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sougat Misra is active.

Publication


Featured researches published by Sougat Misra.


Nutrients | 2015

Redox-Active Selenium Compounds—From Toxicity and Cell Death to Cancer Treatment

Sougat Misra; Mallory Boylan; Arun Selvam; Julian E. Spallholz; Mikael Björnstedt

Selenium is generally known as an antioxidant due to its presence in selenoproteins as selenocysteine, but it is also toxic. The toxic effects of selenium are, however, strictly concentration and chemical species dependent. One class of selenium compounds is a potent inhibitor of cell growth with remarkable tumor specificity. These redox active compounds are pro-oxidative and highly cytotoxic to tumor cells and are promising candidates to be used in chemotherapy against cancer. Herein we elaborate upon the major forms of dietary selenium compounds, their metabolic pathways, and their antioxidant and pro-oxidant potentials with emphasis on cytotoxic mechanisms. Relative cytotoxicity of inorganic selenite and organic selenocystine compounds to different cancer cells are presented as evidence to our perspective. Furthermore, new novel classes of selenium compounds specifically designed to target tumor cells are presented and the potential of selenium in modern oncology is extensively discussed.


Journal of Cellular and Molecular Medicine | 2014

Selenium induces a multi‐targeted cell death process in addition to ROS formation

Marita Wallenberg; Sougat Misra; Agata M. Wasik; Cristina Marzano; Mikael Björnstedt; Valentina Gandin; Aristi P. Fernandes

Selenium compounds inhibit neoplastic growth. Redox active selenium compounds are evolving as promising chemotherapeutic agents through tumour selectivity and multi‐target response, which are of great benefit in preventing development of drug resistance. Generation of reactive oxygen species is implicated in selenium‐mediated cytotoxic effects on cancer cells. Recent findings indicate that activation of diverse intracellular signalling leading to cell death depends on the chemical form of selenium applied and/or cell line investigated. In the present study, we aimed at deciphering different modes of cell death in a single cell line (HeLa) upon treatment with three redox active selenium compounds (selenite, selenodiglutathione and seleno‐DL‐cystine). Both selenite and selenodiglutathione exhibited equipotent toxicity (IC50 5 μM) in these cells with striking differences in toxicity mechanisms. Morphological and molecular alterations provided evidence of necroptosis‐like cell death in selenite treatment, whereas selenodiglutathione induced apoptosis‐like cell death. We demonstrate that selenodiglutathione efficiently glutathionylated free protein thiols, which might explain the early differences in cytotoxic effects induced by selenite and selenodiglutathione. In contrast, seleno‐DL‐cystine treatment at an IC50 concentration of 100 μM induced morphologically two distinct different types of cell death, one with apoptosis‐like phenotype, while the other was reminiscent of paraptosis‐like cell death, characterized by induction of unfolded protein response, ER‐stress and occurrence of large cytoplasmic vacuoles. Collectively, the current results underline the diverse cytotoxic effects and variable potential of redox active selenium compounds on the survival of HeLa cells and thereby substantiate the potential of chemical species‐specific usage of selenium in the treatment of cancers.


Basic & Clinical Pharmacology & Toxicology | 2014

Selenium Cytotoxicity in Cancer

Marita Wallenberg; Sougat Misra; Mikael Björnstedt

Selenium is an essential trace element with growth-modulating properties. Decades of research clearly demonstrate that selenium compounds inhibit the growth of malignant cells in diverse experimental model systems. However, the growth-modulating and cytotoxic mechanisms are diverse and far from clear. Lately, a remarkable tumour selective cytotoxicity of selenium compounds has been shown, indicating the potential of selenium in the treatment of cancer. Of particular interest are the redox-active selenium compounds exhibiting cytotoxic potential to tumour cells. These selenium compounds elicit complex patterns of pharmacodynamics and pharmacokinetics, leading to cell death pathways that differ among compounds. Modern oncology often focuses on targeted ligand-based therapeutic strategies that are specific to their molecular targets. These drugs are initially efficient, but the tumour cells often rapidly develop resistance against these drugs. In contrast, certain redox-active selenium compounds induce complex cascades of pro-death signalling at pharmacological concentrations with superior tumour specificity. The target molecules are often the ones that are important for the survival of cancer cells and often implicated in drug resistance. Therefore, the chemotherapeutic applications of selenium offer great possibilities of multi-target attacks on tumour cells. This MiniReview focuses on the tumour-specific cytotoxic effects of selenium, with special emphasis on cascades of cellular events induced by the major groups of pharmacologically active selenium compounds. Furthermore, the great pharmacological potential of selenium in the treatment of resistant cancers is discussed.


Nutrients | 2015

Pharmacokinetics and Toxicity of Sodium Selenite in the Treatment of Patients with Carcinoma in a Phase I Clinical Trial: The SECAR Study

Ola Brodin; Staffan Eksborg; Marita Wallenberg; Charlotte Asker-Hagelberg; Erik Huusfeldt Larsen; Dag Mohlkert; Clara Lenneby-Helleday; Hans Jacobsson; Stig Linder; Sougat Misra; Mikael Björnstedt

Background: Sodium selenite at high dose exerts antitumor effects and increases efficacy of cytostatic drugs in multiple preclinical malignancy models. We assessed the safety and efficacy of intravenous administered sodium selenite in cancer patients’ refractory to cytostatic drugs in a phase I trial. Patients received first line of chemotherapy following selenite treatment to investigate altered sensitivity to these drugs and preliminary assessment of any clinical benefits. Materials and Methods: Thirty-four patients with different therapy resistant tumors received iv sodium selenite daily for consecutive five days either for two weeks or four weeks. Each cohort consisted of at least three patients who received the same daily dose of selenite throughout the whole treatment. If 0/3 patients had dose-limiting toxicities (DLTs), the study proceeded to the next dose-level. If 2/3 had DLT, the dose was considered too high and if 1/3 had DLT, three more patients were included. Dose-escalation continued until the maximum tolerated dose (MTD) was reached. MTD was defined as the highest dose-level on which 0/3 or 1/6 patients experienced DLT. The primary endpoint was safety, dose-limiting toxic effects and the MTD of sodium selenite. The secondary endpoint was primary response evaluation. Results and Conclusion: MTD was defined as 10.2 mg/m2, with a calculated median plasma half-life of 18.25 h. The maximum plasma concentration of selenium from a single dose of selenite increased in a nonlinear pattern. The most common adverse events were fatigue, nausea, and cramps in fingers and legs. DLTs were acute, of short duration and reversible. Biomarkers for organ functions indicated no major systemic toxicity. In conclusion, sodium selenite is safe and tolerable when administered up to 10.2 mg/m2 under current protocol. Further development of the study is underway to determine if prolonged infusions might be a more effective treatment strategy.


PLOS ONE | 2012

Methylselenol Formed by Spontaneous Methylation of Selenide Is a Superior Selenium Substrate to the Thioredoxin and Glutaredoxin Systems

Aristi P. Fernandes; Marita Wallenberg; Valentina Gandin; Sougat Misra; Francesco Tisato; Cristina Marzano; Maria Pia Rigobello; Sushil Kumar; Mikael Björnstedt

Naturally occurring selenium compounds like selenite and selenodiglutathione are metabolized to selenide in plants and animals. This highly reactive form of selenium can undergo methylation and form monomethylated and multimethylated species. These redox active selenium metabolites are of particular biological and pharmacological interest since they are potent inducers of apoptosis in cancer cells. The mammalian thioredoxin and glutaredoxin systems efficiently reduce selenite and selenodiglutathione to selenide. The reactions are non-stoichiometric aerobically due to redox cycling of selenide with oxygen and thiols. Using LDI-MS, we identified that the addition of S-adenosylmethionine (SAM) to the reactions formed methylselenol. This metabolite was a superior substrate to both the thioredoxin and glutaredoxin systems increasing the velocities of the nonstoichiometric redox cycles three-fold. In vitro cell experiments demonstrated that the presence of SAM increased the cytotoxicity of selenite and selenodiglutathione, which could neither be explained by altered selenium uptake nor impaired extra-cellular redox environment, previously shown to be highly important to selenite uptake and cytotoxicity. Our data suggest that selenide and SAM react spontaneously forming methylselenol, a highly nucleophilic and cytotoxic agent, with important physiological and pharmacological implications for the highly interesting anticancer effects of selenium.


Oncotarget | 2016

Selenite promotes all- trans retinoic acid-induced maturation of acute promyelocytic leukemia cells

Sougat Misra; Arun Kumar Selvam; Marita Wallenberg; Aditya Ambati; András Matolcsy; Isabelle Magalhaes; Gilbert Lauter; Mikael Björnstedt

Selective targeting of the PML/RARα oncoprotein demonstrates a successful molecular targeted therapy in acute promyelocytic leukemia (APL) with a typical t(15:17) chromosomal translocation. The zinc-thiolate coordination is critical for structural stability of zinc finger proteins, including the PML moiety of PML/RARα. Based on the known interaction of redox-active selenium compounds with thiolate ligands of zinc, we herein have investigated the abrogatory effects of selenite alone or in combination with all-trans retinoic acid on PML/RARα and the possible effects on differentiation in these cells. At pharmacological concentrations, selenite inhibited the proliferation and survival of APL originated NB4 cells. In combination with ATRA, it potentiated the differentiation of NB4 cells without any differentiating effects of its own as a single agent. Concordant with our hypothesis, PML/RARα oncoprotein expression was completely abrogated by selenite. Increased expression of RAR, PU.1 and FOXO3A transcription factors in the combined treatment suggested the plausible basis for increased differentiation in these cells. We show that selenite at clinically achievable dose targets PML/RARα oncoprotein for degradation and potentiates differentiation of promyelocytic leukemic cells in combination with ATRA. The present investigation reveals the hitherto unknown potential of selenite in targeted abrogation of PML/RARα in APL cells with prospective therapeutic value.


Archive | 2017

Chapter 14. Metabolism of Selenium/Selenocystine and Their Roles in the Prevention and Treatment of Human Cancer

Sougat Misra; Mikael Björnstedt

Selenium and sulfur share many common chemical properties. There are many biologically active selenium analogues of naturally occurring sulfur compounds. Based on the structural similarities, some of these selenium compounds are known to be taken up by transporters involved in uptake of cognate sulfur-containing substrates. In a similar way, they share some of the common metabolic transformations. Selenium compounds are unique in their redox chemistry in comparison to their sulfur counterparts. Such distinct chemical properties result in the unique biological activities of selenium. While selenium functions as an antioxidant at low doses, high doses elicit pro-oxidant effects. Manifestation of these dual biological functions is exploited in cancer chemoprevention as well as in prospective cancer therapy. Efficacy of these selenium compounds is attributed towards their notable differences in the metabolic pathways and their modes and mechanisms of actions.


Archive | 2018

Therapeutic Potential of Selenium Compounds in the Treatment of Cancer

Arun Selvam; Mikael Björnstedt; Sougat Misra

The potential applications of different selenium compounds as cancer chemotherapeutic agents is an active area of research within the field of cancer drug discovery. The antineoplastic efficacies of many of these small molecules have been extensively investigated, mainly in multiple preclinical models of cancer. Sodium selenite and Se-methylselenocysteine represent two of such selenium compounds, the cytotoxic and antiproliferative efficacies of which are discussed herein. These compounds differ in their mechanisms of action. Sodium selenite exerts its cytotoxic effects by directly oxidizing cellular free thiol pools. In contrast, Se-methylselenocysteine undergoes enzymatic transformation into methylselenol which is cytotoxic due to its ability to redox cycle with cellular thiols. Despite the inherent differences in their metabolic transformations, the disruption of the cellular redox balance and the activation of pro-death intracellular signaling pathways have been implicated as the most prevalent mechanisms of their cytotoxic effects. Both of these selenium compounds exert synergistic toxic effects with certain cancer chemotherapeutics. Together, the well-documented tumor-specific cytotoxic and antiproliferative effects of these compounds have paved the path for their clinical translation. In a phase I clinical trial, it has been shown that sodium selenite is well tolerated in human up to a dose of 10.2 mg/m2 when administered daily for 5 days a week for 2 weeks. Similarly, Se-methylselenocysteine exhibits a favorable pharmacokinetic and safety profile during prolonged oral administration in healthy subjects. Further studies are warranted to investigate their cancer chemotherapeutic efficacies in clinical settings.


Archive | 2018

Selenocystine and Cancer

Sougat Misra; Mikael Björnstedt

The diselenide compound selenocystine is a selenium analog of cystine. It is more reactive compared to cystine due to intrinsic differences in chemical properties between sulfur and selenium. Thioredoxin reductase or excess cysteine and glutathione reduces selenocystine to highly reactive selenolate. When selenocystine is present at high concentration, selenolate-mediated biochemical reactions perturb cellular redox homeostasis and induce oxidative stress. Limited pharmacokinetic studies indicate rather long half-life and biphasic elimination kinetics of selenocystine. It is well tolerated in mice and rats with a narrow window between no observed effects and toxicity. Several preclinical studies have interrogated its redox modulatory effects as an anticancer modality. Reported cytotoxic effects include DNA damage, S-phase arrest, activation of P53, alteration of MAPK and PI3K-AKT signaling pathways, loss of mitochondrial membrane potential, and release of cytochrome C. So far, findings from published studies suggest limited antineoplastic effects of selenocystine in various animal models of cancer.


BMC Genomics | 2018

Characterization of the human RFX transcription factor family by regulatory and target gene analysis

Debora Sugiaman-Trapman; Morana Vitezic; Eeva-Mari Jouhilahti; Anthony Mathelier; Gilbert Lauter; Sougat Misra; Carsten O. Daub; Juha Kere; Peter Swoboda

BackgroundEvolutionarily conserved RFX transcription factors (TFs) regulate their target genes through a DNA sequence motif called the X-box. Thereby they regulate cellular specialization and terminal differentiation. Here, we provide a comprehensive analysis of all the eight human RFX genes (RFX1–8), their spatial and temporal expression profiles, potential upstream regulators and target genes.ResultsWe extracted all known human RFX1–8 gene expression profiles from the FANTOM5 database derived from transcription start site (TSS) activity as captured by Cap Analysis of Gene Expression (CAGE) technology. RFX genes are broadly (RFX1–3, RFX5, RFX7) and specifically (RFX4, RFX6) expressed in different cell types, with high expression in four organ systems: immune system, gastrointestinal tract, reproductive system and nervous system. Tissue type specific expression profiles link defined RFX family members with the target gene batteries they regulate. We experimentally confirmed novel TSS locations and characterized the previously undescribed RFX8 to be lowly expressed. RFX tissue and cell type specificity arises mainly from differences in TSS architecture. RFX transcript isoforms lacking a DNA binding domain (DBD) open up new possibilities for combinatorial target gene regulation. Our results favor a new grouping of the RFX family based on protein domain composition. We uncovered and experimentally confirmed the TFs SP2 and ESR1 as upstream regulators of specific RFX genes. Using TF binding profiles from the JASPAR database, we determined relevant patterns of X-box motif positioning with respect to gene TSS locations of human RFX target genes.ConclusionsThe wealth of data we provide will serve as the basis for precisely determining the roles RFX TFs play in human development and disease.

Collaboration


Dive into the Sougat Misra's collaboration.

Top Co-Authors

Avatar

Mikael Björnstedt

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlos Fernández Moro

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Del Chiaro

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge