Network


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

Hotspot


Dive into the research topics where Leonid A. Sitailo is active.

Publication


Featured researches published by Leonid A. Sitailo.


Cancer Research | 2006

Enhanced Killing of Melanoma Cells by Simultaneously Targeting Mcl-1 and NOXA

Jian-Zhong Qin; Hong Xin; Leonid A. Sitailo; Mitchell F. Denning; Brian J. Nickoloff

By deciphering the dysregulation of apoptosis in melanoma cells, new treatment approaches exploiting aberrant control mechanisms regulating cell death can be envisioned. Among the Bcl-2 family, a BH3-only member, NOXA, functions in a specific mitochondrial-based cell death pathway when melanoma cells are exposed to a proteasome inhibitor (e.g., bortezomib). Some therapeutic agents, such as bortezomib, not only induce proapoptotic Bcl-2 family members and active conformational changes in Bak and Bax but also are associated with undesirable effects, including accumulation of antiapoptotic proteins, such as Mcl-1. To enhance the bortezomib-mediated killing of melanoma cells, the apoptotic pathway involving NOXA was further investigated, leading to identification of an important target (i.e., the labile Bcl-2 homologue Mcl-1 but not other survival proteins). To reduce Mcl-1 levels, melanoma cells were pretreated with several different agents, including Mcl-1 small interfering RNA (siRNA), UV light, or the purine nucleoside analogue fludarabine. By simultaneously triggering production of NOXA (using bortezomib) as well as reducing Mcl-1 levels (using siRNA, UV light, or fludarabine), significantly enhanced killing of melanoma cells was achieved. These results show binding interactions between distinct Bcl-2 family members, such as NOXA and Mcl-1, in melanoma cells, paving the way for novel and rational therapeutic combination strategies, which target guardians of the proapoptotic Bak- and Bax-mediated pathways, against this highly aggressive and often fatal malignancy.


Journal of Biological Chemistry | 2006

The Protein Kinase Cδ Catalytic Fragment Targets Mcl-1 for Degradation to Trigger Apoptosis

Leonid A. Sitailo; Shalini S. Tibudan; Mitchell F. Denning

Proteolytic cleavage and subsequent activation of protein kinase C (PKC) δ is required for apoptosis induced by a variety of genotoxic agent, including UV radiation. In addition, overexpression of the constitutively active PKCδ catalytic fragment (PKCδ-cat) is sufficient to trigger Bax activation, cytochrome c release, and apoptosis. While PKCδ is a key apoptotic effector, the downstream target(s) responsible for the mitochondrial apoptotic cascade are not known. We found that expression of the active PKCδ-cat in HaCaT cells triggers a reduction in the anti-apoptotic protein Mcl-1, similar to UV radiation. The down-regulation of Mcl-1 induced by PKCδ-cat was not at the mRNA level but was due to decreased protein half-life. Overexpression of Mcl-1 protected HaCaT cells from both UV and PKCδ-cat-induced apoptosis and blocked the release of cytochrome c from the mitochondria, indicating that Mcl-1 down-regulation was required for apoptosis signaling. Indeed, down-regulation of Mcl-1 with siRNA slightly increased the basal apoptotic rate of HaCaT cells and dramatically sensitized them to UV or PKCδ-cat-induced apoptosis. HaCaT cells with down-regulated Mcl-1 had higher activated Bax protein, as measured by Bax cross-linking, indicating that Mcl-1 down-regulation is sufficient for Bax activation. Finally, recombinant PKCδ could phosphorylate Mcl-1 in vitro, identifying Mcl-1 as a direct target for PKCδ. Overall our results identify Mcl-1 as an important target for PKCδ-cat that can mediate its pro-apoptotic effects on mitochondria to amplify the apoptotic signaling induced by a wide range of apoptotic stimuli.


Oncogene | 2005

Knockdown of p53 levels in human keratinocytes accelerates Mcl-1 and Bcl-xL reduction thereby enhancing UV-light induced apoptosis

Vijaya Chaturvedi; Leonid A. Sitailo; Jian Zhong Qin; Barbara Bodner; Mitchell F. Denning; Jonathan L. Curry; Wengeng Zhang; Douglas E. Brash; Brian J. Nickoloff

Ultraviolet (UV) light exposure is a common cause of epithelial-derived skin cancers, and the epidermal response to UV-light has been extensively studied using both mouse models and cultured human keratinocytes (KCs). Elimination of cells with UV-induced DNA damage via apoptosis provides a powerful mechanism to minimize retention or expansion of genetically abnormal cells. This cell editing function has largely been ascribed to the biological role of the p53 tumor suppressor gene, as mutations or deletions involving p53 have been linked to skin cancer development. Rather than introducing mutations, or using cells with complete loss of wild-type p53, we used an siRNA-based approach to knockdown, but not eliminate, p53 levels in primary cultures of human KCs followed by UV-irradiation. Surprisingly, when p53 levels were reduced by 50–80% the apoptosis induced by exposure to UV-light was accelerated and markedly enhanced (two- to three- fold) compared to control siRNA treated KCs. The p53 siRNA treated KCs were characterized by elevated E2F-1 levels accompanied by accelerated elimination of the Mcl-1 and Bcl-xL antiapoptotic proteins, as well as enhanced Bax oligomerization. Forced overexpression of either Mcl-1 or Bcl-xL reduced the UV-light enhanced apoptotic response in p53 siRNA treated KCs. We conclude that p53 not only can provide proapoptotic signals but also regulates a survival pathway influencing Mcl-1 and Bcl-xL levels. This overlooked survival function of p53 may explain previous paradoxical responses noted by investigators using p53 heterozygous and knockout mouse models, and opens up the possibility that not all liaisons within the cell involving p53 necessarily represent fatal attractions.


Journal of Cellular Physiology | 2004

Low‐dose UV‐radiation sensitizes keratinocytes to TRAIL‐induced apoptosis

Jian-Zhong Qin; Patricia Bacon; Jeffrey Panella; Leonid A. Sitailo; Mitchell F. Denning; Brian J. Nickoloff

The impact of low‐dose ultraviolet light (UV‐light) on apoptotic susceptibility of keratinocytes (KCs) induced by TRAIL is unclear. Skin expresses a functional form of TRAIL, and while sun exposure influences TRAIL death receptors, a role for decoy receptors has not been evaluated. Unraveling mechanisms involving apoptotic sensitivity of KCs is important because skin is the first target of UV‐light, and a site for commonly occurring cancers. Since apoptosis is a homeostatic process eliminating UV‐light induced DNA damaged cells, elucidating molecular events regulating apoptosis enhances understanding of cutaneous photocarcinogenesis. Here we demonstrate low‐dose UV‐light enhances susceptibility of KCs to TRAIL‐induced apoptosis. Low‐dose UV‐light selectively reduces decoy receptors, without influencing death receptor levels. UV‐induced enhanced apoptotic susceptibility was reduced by over‐expression of decoy receptor TRAIL‐R4, but not TRAIL‐R3; or treatment with thiol compound pyrrolidine dithiocarbamate (PDTC), which also enhanced TRAIL‐R4 levels. Besides influencing decoy receptors, low‐dose UV‐light plus TRAIL also synergistically promoted cytochrome c and Smac release from mitochondria. Inhibitors directed against caspases 2, 3, 8, and 9 reduced the synergistic apoptotic response following low‐dose UV‐light plus TRAIL exposure; as did forced over‐expression of Bcl‐x and dominant negative (DN) constructs of FADD and caspase 9. Thus, relative levels of decoy receptors significantly influence susceptibility of KCs to TRAIL‐induced apoptosis with concomitant low‐dose UV‐light exposure; in addition to the apoptotic pathway mediated by mitochondrial permeabilization.


Journal of Investigative Dermatology | 2009

Mcl-1 Functions as Major Epidermal Survival Protein Required for Proper Keratinocyte Differentiation

Leonid A. Sitailo; Anita Jerome-Morais; Mitchell F. Denning

Rapid downregulation of the antiapoptotic Bcl-2 family protein myeloid cell leukemia 1 (Mcl-1) is required for UV-induced apoptosis, underlining an important role for Mcl-1 in epidermal pathology. To determine if Mcl-1 has a specific role in normal keratinocyte (KC) biology, Mcl-1 was downregulated in human KCs by RNAi and these KCs were induced to differentiate in organotypic raft cultures. Mcl-1 shRNA organotypic cultures showed increased levels of spontaneous premature apoptosis, implicating Mcl-1 as an essential KC survival protein. Mcl-1-downregulated cultures also had reduced granular and cornified layers, and produced lower levels of cross-linked protein and cornified envelopes. Cornification could only partially be rescued with the general caspase inhibitor z-VAD, suggesting that reduced cornification was not entirely because of premature apoptosis. Differentiation markers (K1, K10, filaggrin, loricrin, cleaved caspase-14) were normally expressed in control organotypic cultures, but were expressed at reduced levels in organotypic cultures with downregulated Mcl-1. The defect in differentiation marker expression was independent of apoptosis as it could not be rescued by z-VAD. Thus, Mcl-1 serves two important, independent functions in epidermal KCs: acting as a major survival protein by inhibiting premature apoptosis in the spinous and granular layers to promote conification, and promoting the robust induction of KC differentiation markers.


Experimental Dermatology | 2006

Defining the caspase‐containing apoptotic machinery contributing to cornification in human epidermal equivalents

Vijaya Chaturvedi; Leonid A. Sitailo; Barbara Bodner; Mitchell F. Denning; Brian J. Nickoloff

Abstract:  Whether terminal differentiation/stratum corneum formation of keratinocytes (KCs) represents a form of programmed cell death, utilizing mediators of classical apoptosis, is unclear. Apoptosis, an evolutionarily conserved death process, is comprised of extrinsic and intrinsic pathways, which converge using caspase 3. To define upstream and downstream caspases involved in terminal differentiation, we utilized human epidermal equivalents (EEs). Using submerged cultures comprised of human KCs, EEs were sequentially analyzed before and after being raised to an air/liquid (A/L) interface at 3–24 h intervals. At each time point, EEs were analyzed morphologically and for specific enzyme activity to distinguish different initiator (caspases 1, 2, 8, 9) and effector caspases (3, 6, 7). Terminal differentiation began at 6–8 h, as defined by stratum corneum with loricirin expression and completed at 18–24 h producing an epidermis resembling normal skin. Enzyme activity for caspases 1, 2, 3, 6, 7, 8, and 9 (but not 4, 5) was enhanced (>two‐fold nmol/mg/h) at 3–6 h compared with submerged cultures. Processing of caspase 14 occurred at 18 h, and cleaved caspase 14 was increased at 24 h. Activated caspase 3‐positive and terminal deoxynucleotidyl transferase‐mediated nick end labeling‐positive KCs were identified in EEs at 3–6 h corresponding to initiation sites of terminal differentiation. Addition of caspase inhibitors reduced levels of involucrin and loricrin in EEs raised to an A/L interface. We conclude caspases function as important death effectors strategically positioned at intersection of intrinsic and extrinsic pathways in KCs undergoing stratum corneum formation.


Journal of Biological Chemistry | 2010

The Protein Kinase Cδ Catalytic Fragment Is Critical for Maintenance of the G2/M DNA Damage Checkpoint

Edward L. LaGory; Leonid A. Sitailo; Mitchell F. Denning

Protein kinase Cδ (PKCδ) is an essential component of the intrinsic apoptotic program. Following DNA damage, such as exposure to UV radiation, PKCδ is cleaved in a caspase-dependent manner, generating a constitutively active catalytic fragment (PKCδ-cat), which is necessary and sufficient for keratinocyte apoptosis. We found that in addition to inducing apoptosis, expression of PKCδ-cat caused a pronounced G2/M cell cycle arrest in both primary human keratinocytes and immortalized HaCaT cells. Consistent with a G2/M arrest, PKCδ-cat induced phosphorylation of Cdk1 (Tyr15), a critical event in the G2/M checkpoint. Treatment with the ATM/ATR inhibitor caffeine was unable to prevent PKCδ-cat-induced G2/M arrest, suggesting that PKCδ-cat is functioning downstream of ATM/ATR in the G2/M checkpoint. To better understand the role of PKCδ and PKCδ-cat in the cell cycle response to DNA damage, we exposed wild-type and PKCδ null mouse embryonic fibroblasts (MEFs) to UV radiation. Wild-type MEFs underwent a pronounced G2/M arrest, Cdk1 phosphorylation, and induction of apoptosis following UV exposure, whereas PKCδ null MEFs were resistant to these effects. Expression of PKCδ-green fluorescent protein, but not caspase-resistant or kinase-inactive PKCδ, was able to restore G2/M checkpoint integrity in PKCδ null MEFs. The function of PKCδ in the DNA damage-induced G2/M cell cycle checkpoint may be a critical component of its tumor suppressor function.


Pigment Cell & Melanoma Research | 2010

Functional alterations in protein kinase C beta II expression in melanoma

John P. Voris; Leonid A. Sitailo; Heidi R. Rahn; Ann Defnet; Aaron T. Gerds; Robert Sprague; Vipin Yadav; I. Caroline Le Poole; Mitchell F. Denning

Protein kinase C (PKC) is a heterogeneous family of serine/threonine protein kinases that have different biological effects in normal and neoplastic melanocytes (MCs). To explore the mechanism behind their differential response to PKC activation, we analyzed the expression profile of all nine PKC isoforms in normal human MCs, HPV16 E6/E7 immortalized MCs, and a panel of melanoma cell lines. We found reduced PKCβ and increased PKCζ and PKCι expression at both the protein and mRNA levels in immortalized MCs and melanoma lines. We focused on PKCβ as it has been functionally linked to melanin production and oxidative stress response. Re‐expression of PKCβ in melanoma cells inhibited colony formation in soft agar, indicating that PKCβ loss in melanoma is important for melanoma growth. PKCβII, but not PKCβI, was localized to the mitochondria, and inhibition of PKCβ significantly reduced UV‐induced reactive oxygen species (ROS) in MCs with high PKCβ expression. Thus alterations in PKCβ expression in melanoma contribute to their neoplastic phenotype, possibly by reducing oxidative stress, and may constitute a selective therapeutic target.


Cancer Research | 2010

Abstract 1026: A non-apoptotic function for Mcl-1 as an activator of canonical Wnt signaling in keratinocytes

Leonid A. Sitailo; Periannan Sethupathi; Mitchell F. Denning

Mcl-1 is an anti-apoptotic Bcl-2 family member whose rapid down-regulation is required for UV apoptosis. We recently showed that not only is Mcl-1 a major human keratinocyte (KC) survival protein, but it is required for proper KC differentiation in organotypic culture, implicating a non-apoptotic function for Mcl-1 involved in regulating epidermal morphogenesis. To further explore the function of Mcl-1 in vivo, we generated and analyzed transgenic mice expressing human Mcl-1 under control of the keratin 5 (K5) promoter. K5-Mcl-1 transgenic mice had >2-fold increase in epidermal thickness (p 3 H-thymidine incorporation (3.4- and 2.9-fold respectively), indicating that the mitogenic activity of Mcl-1 resides in its N-terminal region. Since β-catenin signaling is activated in cutaneous SCC and involved in stem cell maintenance, this non-apoptotic function of Mcl-1 represents a novel and potentially oncogenic activity in addition to its well characterized anti-apoptotic function. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1026.


Journal of Biological Chemistry | 2002

Activation of Caspase-9 Is Required for UV-induced Apoptosis of Human Keratinocytes

Leonid A. Sitailo; Shalini S. Tibudan; Mitchell F. Denning

Collaboration


Dive into the Leonid A. Sitailo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shalini S. Tibudan

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Barbara Bodner

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Jian-Zhong Qin

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Vijaya Chaturvedi

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Aaron T. Gerds

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Anita Jerome-Morais

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ann Defnet

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge