Network


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

Hotspot


Dive into the research topics where Felix J. Kim is active.

Publication


Featured researches published by Felix J. Kim.


Molecular Pharmacology | 2013

Sequential cytoprotective responses to Sigma1 ligand induced endoplasmic reticulum stress

Joel M. Schrock; Christina M. Spino; Charles G. Longen; Stacy M. Stabler; Jacqueline C. Marino; Gavril W. Pasternak; Felix J. Kim

The Sigma1 receptor (Sigma1) is an endoplasmic reticulum (ER) integral membrane protein that is highly expressed in a number of cancer cell lines. Small molecule compounds targeting Sigma1 (Sigma1 ligands) inhibit cancer cell proliferation and induce apoptotic cell death in vitro and inhibit tumor growth in xenograft experiments. However, the cellular pathways activated by Sigma1 protein-ligand interaction are not well defined. Here, we find that treatment with some Sigma1 ligands induces ER stress and activates the unfolded protein response (UPR) in a dose- and time-responsive manner in a range of adenocarcinoma cell lines. Autophagy is engaged after extended treatment with Sigma1 ligands, which suggests that protracted UPR results in autophagy as a secondary response. Inhibition of UPR by RNAi-mediated knockdown of inositol-requiring enzyme 1α and activating transcription factor 4 abrogates autophagosome formation, as does knockdown of essential autophagy gene products Beclin1 and autophagy protein 5. Knockdown of Sigma1 also suppresses IPAG [1-(4-iodophenyl)-3-(2-adamantyl) guanidine] induced UPR marker and autophagosome levels, indicating that this response is indeed Sigma1mediated. We find that UPR activation precedes autophagosome formation and autophagy precedes apoptosis in Sigma1 ligand-treated cells. These processes are reversible, and washout of IPAG before cell death results in a return of autophagosomes and UPR markers toward basal levels. However, inhibition of Sigma1 ligand–induced UPR or autophagy accelerates apoptotic cell death. Together, these data suggest that UPR and autophagy are engaged as primary and secondary cytoprotective responses, respectively, to Sigma1 ligand–induced disruption of cancer cell protein homeostasis.


Biochemical and Biophysical Research Communications | 2012

Inhibition of tumor cell growth by Sigma1 ligand mediated translational repression

Felix J. Kim; Joel M. Schrock; Christina M. Spino; Jacqueline C. Marino; Gavril W. Pasternak

Treatment with sigma1 receptor (Sigma1) ligands can inhibit cell proliferation in vitro and tumor growth in vivo. However, the cellular pathways engaged in response to Sigma1 ligand treatment that contribute to these outcomes remain largely undefined. Here, we show that treatment with putative antagonists of Sigma1 decreases cell mass. This effect corresponds with repressed cap-dependent translation initiation in multiple breast and prostate cancer cell lines. Sigma1 antagonist treatment suppresses phosphorylation of translational regulator proteins p70S6K, S6, and 4E-BP1. RNAi-mediated knockdown of Sigma1 also results in translational repression, consistent with the effects of antagonist treatment. Sigma1 antagonist mediated translational repression and decreased cell size are both reversible. Together, these data reveal a role for Sigma1 in tumor cell protein synthesis, and demonstrate that small molecule Sigma1 ligands can be used as modulators of protein translation.


Cancer Research | 2017

Sigma1 Targeting to Suppress Aberrant Androgen Receptor Signaling in Prostate Cancer

Jeff D. Thomas; Charles G. Longen; Halley M. Oyer; Nan Chen; Christina M. Maher; Joseph M. Salvino; Blase Kania; Kelsey N. Anderson; William F. Ostrander; Karen E. Knudsen; Felix J. Kim

Suppression of androgen receptor (AR) activity in prostate cancer by androgen depletion or direct AR antagonist treatment, although initially effective, leads to incurable castration-resistant prostate cancer (CRPC) via compensatory mechanisms including resurgence of AR and AR splice variant (ARV) signaling. Emerging evidence suggests that Sigma1 (also known as sigma-1 receptor) is a unique chaperone or scaffolding protein that contributes to cellular protein homeostasis. We reported previously that some Sigma1-selective small molecules can be used to pharmacologically modulate protein homeostasis pathways. We hypothesized that these Sigma1-mediated responses could be exploited to suppress AR protein levels and activity. Here we demonstrate that treatment with a small-molecule Sigma1 inhibitor prevented 5α- dihydrotestosterone-mediated nuclear translocation of AR and induced proteasomal degradation of AR and ARV, suppressing the transcriptional activity and protein levels of both full-length and splice-variant AR. Consistent with these data, RNAi knockdown of Sigma1 resulted in decreased AR levels and transcriptional activity. Furthermore, Sigma1 physically associated with ARV7 and ARv567es as well as full-length AR. Treatment of mice xenografted with ARV-driven CRPC tumors with a drug-like small-molecule Sigma1 inhibitor significantly inhibited tumor growth associated with elimination of AR and ARV7 in responsive tumors. Together, our data show that Sigma1 modulators can be used to suppress AR/ARV-driven prostate cancer cells via regulation of pharmacologically responsive Sigma1-AR/ARV interactions, both in vitro and in vivoCancer Res; 77(9); 2439-52. ©2017 AACR.


Molecular Cancer Research | 2017

Small-Molecule Sigma1 Modulator Induces Autophagic Degradation of PD-L1

Christina M. Maher; Jeff D. Thomas; Derick A. Haas; Charles G. Longen; Halley M. Oyer; Jane Y. Tong; Felix J. Kim

Emerging evidence suggests that Sigma1 (SIGMAR1, also known as sigma-1 receptor) is a unique ligand-regulated integral membrane scaffolding protein that contributes to cellular protein and lipid homeostasis. Previously, we demonstrated that some small-molecule modulators of Sigma1 alter endoplasmic reticulum (ER)–associated protein homeostasis pathways in cancer cells, including the unfolded protein response and autophagy. Programmed death-ligand 1 (PD-L1) is a type I integral membrane glycoprotein that is cotranslationally inserted into the ER and is processed and transported through the secretory pathway. Once at the surface of cancer cells, PD-L1 acts as a T-cell inhibitory checkpoint molecule and suppresses antitumor immunity. Here, we demonstrate that in Sigma1-expressing triple-negative breast and androgen-independent prostate cancer cells, PD-L1 protein levels were suppressed by RNAi knockdown of Sigma1 and by small-molecule inhibition of Sigma1. Sigma1-mediated action was confirmed by pharmacologic competition between Sigma1-selective inhibitor and activator ligands. When administered alone, the Sigma1 inhibitor decreased cell surface PD-L1 expression and suppressed functional interaction of PD-1 and PD-L1 in a coculture of T cells and cancer cells. Conversely, the Sigma1 activator increased PD-L1 cell surface expression, demonstrating the ability to positively and negatively modulate Sigma1 associated PD-L1 processing. We discovered that the Sigma1 inhibitor induced degradation of PD-L1 via autophagy, by a mechanism distinct from bulk macroautophagy or general ER stress–associated autophagy. Finally, the Sigma1 inhibitor suppressed IFNγ-induced PD-L1. Our data demonstrate that small-molecule Sigma1 modulators can be used to regulate PD-L1 in cancer cells and trigger its degradation by selective autophagy. Implications: Sigma1 modulators sequester and eliminate PD-L1 by autophagy, thus preventing functional PD-L1 expression at the cell surface. This posits Sigma1 modulators as novel therapeutic agents in PD-L1/PD-1 blockade strategies that regulate the tumor immune microenvironment. Visual Overview: http://mcr.aacrjournals.org/content/molcanres/16/2/243/F1.large.jpg. Mol Cancer Res; 16(2); 243–55. ©2017 AACR. Visual Overview


Archive | 2017

Sigma1 Pharmacology in the Context of Cancer

Felix J. Kim; Christina M. Maher

Sigma1 (also known as sigma-1 receptor, Sig1R, σ1 receptor) is a unique pharmacologically regulated integral membrane chaperone or scaffolding protein. The majority of publications on the subject have focused on the neuropharmacology of Sigma1. However, a number of publications have also suggested a role for Sigma1 in cancer. Although there is currently no clinically used anti-cancer drug that targets Sigma1, a growing body of evidence supports the potential of Sigma1 ligands as therapeutic agents to treat cancer. In preclinical models, compounds with affinity for Sigma1 have been reported to inhibit cancer cell proliferation and survival, cell adhesion and migration, tumor growth, to alleviate cancer-associated pain, and to have immunomodulatory properties. This review will highlight that although the literature supports a role for Sigma1 in cancer, several fundamental questions regarding drug mechanism of action and the physiological relevance of aberrant SIGMAR1 transcript and Sigma1 protein expression in certain cancers remain unanswered or only partially answered. However, emerging lines of evidence suggest that Sigma1 is a component of the cancer cell support machinery, that it facilitates protein interaction networks, that it allosterically modulates the activity of its associated proteins, and that Sigma1 is a selectively multifunctional drug target.


Journal of Biological Chemistry | 2014

Cyclopamine Modulates γ-Secretase-mediated Cleavage of Amyloid Precursor Protein by Altering Its Subcellular Trafficking and Lysosomal Degradation

Anna G. Vorobyeva; Randall Lee; Sean Miller; Charles G. Longen; Michal Sharoni; Preeti J. Kandelwal; Felix J. Kim; Daniel R. Marenda; Aleister J. Saunders

Background: Sterols can alter APP metabolism. Results: Cyclopamine, a phytosterol, alters APP-CTF degradation rate, decreases APP-CTF bioavailability for γ-secretase cleavage, and reduces Aβ and AICD generation. Conclusion: Cyclopamine decreases Aβ and AICD production by altering APP-CTF retrograde trafficking. Significance: Cyclopamine is a novel modulator of APP metabolism and trafficking, which can illuminate new avenues for Alzheimer disease treatment. Alzheimer disease (AD) is a progressive neurodegenerative disease leading to memory loss. Numerous lines of evidence suggest that amyloid-β (Aβ), a neurotoxic peptide, initiates a cascade that results in synaptic dysfunction, neuronal death, and eventually cognitive deficits. Aβ is generated by the proteolytic processing of the amyloid precursor protein (APP), and alterations to this processing can result in Alzheimer disease. Using in vitro and in vivo models, we identified cyclopamine as a novel regulator of γ-secretase-mediated cleavage of APP. We demonstrate that cyclopamine decreases Aβ generation by altering APP retrograde trafficking. Specifically, cyclopamine treatment reduced APP-C-terminal fragment (CTF) delivery to the trans-Golgi network where γ-secretase cleavage occurs. Instead, cyclopamine redirects APP-CTFs to the lysosome. These data demonstrate that cyclopamine treatment decreases γ-secretase-mediated cleavage of APP. In addition, cyclopamine treatment decreases the rate of APP-CTF degradation. Together, our data demonstrate that cyclopamine alters APP processing and Aβ generation by inducing changes in APP subcellular trafficking and APP-CTF degradation.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Cloning the sigma2 receptor: Wandering 40 years to find an identity

Felix J. Kim; Gavril W. Pasternak

Scientists have endeavored to understand sigma receptors for over 40 y. Although most agree that they are important, there is little agreement on anything else. In their behavioral classification of opioid receptors in 1976, Martin et al. (1) proposed three groups of compounds illustrating three distinct opioid receptor classes (mu, kappa, and sigma) based upon morphine, ketocyclazocine, and SKF10,047, respectively, and noted that the opioid antagonist naltrexone antagonized them all. Since then, the sigma receptor story has undergone many twists and turns. Although the SKF10,047 stereoisomer used in the initial description is not stated, subsequent investigators used (+)SKF10,047 to define sigma receptors, identifying sites that clearly were not opioid. Extensive binding studies associated (+)SKF10,047 with many putative receptors, including phencyclidine (2), but these ligands proved quite promiscuous, labeling a multitude of sites. As more ligands became available, sigma receptors were dissociated into two categories: sigma1 and sigma2 (3). Functional studies strongly suggested that both classes were important. Our understanding of sigma1 receptors took a major leap forward with the cloning of the protein in 1996 (4) and its subsequent crystallization in 2016 (5). However, these structural insights have not answered many fundamental questions regarding how these proteins work. In PNAS, Alon et al. (6) present compelling information for the cloning of the sigma2 receptor, completing the molecular characterization of this class of receptor and …


Handbook of experimental pharmacology | 2017

Introduction to Sigma Proteins: Evolution of the Concept of Sigma Receptors

Felix J. Kim

For over 40 years, scientists have endeavored to understand the so-called sigma receptors. During this time, the concept of sigma receptors has continuously and significantly evolved. With thousands of publications on the subject, these proteins have been implicated in various diseases, disorders, and physiological processes. Nevertheless, we are just beginning to understand what sigma proteins do and how they work. Two subtypes have been identified, Sigma1 and Sigma2. Whereas Sigma1 (also known as sigma-1 receptor, Sig1R, σ1 receptor, and several other names) was cloned over 20 years ago, Sigma2 (sigma-2 receptor, σ2 receptor) was cloned very recently and had remained a pharmacologically defined entity. In this volume, we will focus primarily on Sigma1. We will highlight several key subject areas in which Sigma1 has been well characterized as well as (re)emerging areas of interest. Despite the large number of publications regarding Sigma1, several fundamental questions remain unanswered or only partially answered. Most of what we know about Sigma1 comes from pharmacological studies; however, a clearly defined molecular mechanism of action remains elusive. One concept has become clear; Sigma1 is not a traditional receptor. Sigma1 is now considered a unique pharmacologically regulated integral membrane chaperone or scaffolding protein. A number of landmark discoveries over the past decade have begun to reshape the concept of sigma receptors. With the rapid emergence of new information, development of new tools, and changing conceptual frameworks, the field is poised for a period of accelerated progress.


Archive | 2017

Sigma Proteins: Evolution of the Concept of Sigma Receptors

Felix J. Kim; Gavril W. Pasternak

For over 40 years, scientists have endeavored to understand the so-called sigma receptors. During this time, the concept of sigma receptors has continuously and significantly evolved. With thousands of publications on the subject, these proteins have been implicated in various diseases, disorders, and physiological processes. Nevertheless, we are just beginning to understand what sigma proteins do and how they work. Two subtypes have been identified, Sigma1 and Sigma2. Whereas Sigma1 (also known as sigma-1 receptor, Sig1R, σ1 receptor, and several other names) was cloned over 20 years ago, Sigma2 (sigma-2 receptor, σ2 receptor) was cloned very recently and had remained a pharmacologically defined entity. In this volume, we will focus primarily on Sigma1. We will highlight F.J. Kim (*) Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA Sidney Kimmel Cancer Center, Philadelphia, PA, USA e-mail: [email protected] # Springer International Publishing AG 2017 F.J. Kim, G.W. Pasternak (eds.), Sigma Proteins: Evolution of the Concept of Sigma Receptors, Handbook of Experimental Pharmacology 244, DOI 10.1007/164_2017_41 1 several key subject areas in which Sigma1 has been well characterized as well as (re)emerging areas of interest. Despite the large number of publications regarding Sigma1, several fundamental questions remain unanswered or only partially answered. Most of what we know about Sigma1 comes from pharmacological studies; however, a clearly defined molecular mechanism of action remains elusive. One concept has become clear; Sigma1 is not a traditional receptor. Sigma1 is now considered a unique pharmacologically regulated integral membrane chaperone or scaffolding protein. A number of landmark discoveries over the past decade have begun to reshape the concept of sigma receptors. With the rapid emergence of new information, development of new tools, and changing conceptual frameworks, the field is poised for a period of accelerated progress.


Cancer Research | 2017

Abstract 3202: Pharmacologic modulation of Sigma1 induces autophagic degradation of programmed death-ligand 1 in cancer cells

Christina M. Maher; Jeff D. Thomas; Charles G. Longen; Derick A. Haas; Halley M. Oyer; Jane Y. Tong; Felix J. Kim

Emerging evidence suggests that Sigma1 (also known as sigma1 receptor) is a unique ligand-operated integral membrane chaperone or scaffolding protein that contributes to cellular protein homeostasis. Previously, we found that treatment of various cancer cell lines with some prototypic small molecule modulators of Sigma1 can engage endoplasmic reticulum (ER) associated protein homeostasis pathways including the unfolded protein response and autophagy. Programmed death-ligand 1 (PD-L1) is a type 1 integral membrane glycoprotein that is processed and transported through the ER and secretory pathway of tumor cells. PD-L1 expressed at the surface of tumor cells can act as a T-cell inhibitory checkpoint molecule that inactivates tumor infiltrating immune cells that express PD-1, its cognate receptor. Here, we show that Sigma1 physically associates with PD-L1. In triple negative breast and androgen-independent prostate cancer cells, PD-L1 protein levels are suppressed by both RNAi mediated knockdown of Sigma1 and pharmacological modulation of Sigma1. We observe decreased cell surface and intracellular levels of PD-L1 by flow cytometry and biochemical subcellular fractionation respectively, which corresponds with a dose-responsive decrease in functional PD-L1/PD-1 interaction in a co-culture of cancer cells and T-cells. Inhibitors of autophagy block this suppression of PD-L1 protein levels, suggesting PD-L1 is degraded away by autophagy after Sigma1 modulation. Through confocal microscopy, we show that Sigma1 modulation results in colocalization of PD-L1 and GFP-LC3, a marker of autophagosomes. From these conclusions, we hypothesize that autophagic degradation of nascent PD-L1 after Sigma1 modulation plays a key role in preventing the transport of functional PD-L1 to the plasma membrane. Together, these data demonstrate that Sigma1 modulators have the potential to act as novel therapeutic agents in PD-1/PD-L1 blockade strategies. Citation Format: Christina M. Maher, Jeffrey D. Thomas, Charles G. Longen, Derick A. Haas, Halley M. Oyer, Jane Y. Tong, Felix J. Kim. Pharmacologic modulation of Sigma1 induces autophagic degradation of programmed death-ligand 1 in cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3202. doi:10.1158/1538-7445.AM2017-3202

Collaboration


Dive into the Felix J. Kim's collaboration.

Top Co-Authors

Avatar

Gavril W. Pasternak

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joel M. Schrock

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge