Network


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

Hotspot


Dive into the research topics where Brian A. Salvatore is active.

Publication


Featured researches published by Brian A. Salvatore.


British Journal of Cancer | 2003

Vitamin E analogues as inducers of apoptosis: structure-function relation.

Marc Birringer; J H EyTina; Brian A. Salvatore; Jiri Neuzil

Recent results show that α-tocopheryl succinate (α-TOS) is a proapoptotic agent with antineoplastic activity. As modifications of the vitamin E (VE) molecule may affect its apoptogenic activity, we tested a number of newly synthesised VE analogues using malignant cell lines. Analogues of α-TOS with lower number of methyl substitutions on the aromatic ring were less active than α-TOS. Replacement of the succinyl group with a maleyl group greatly enhanced the activity, while it was lower for the glutaryl esters. Methylation of the free succinyl carboxyl group on α-TOS and δ-TOS completely prevented the apoptogenic activity of the parent compounds. Both Trolox and its succinylated derivative were inactive. α-tocotrienol (α-T3 H) failed to induce apoptosis, while γ-T3 H was apoptogenic, and more so when succinylated. Shortening the aliphatic side chain of γ-T3 by one isoprenyl unit increased its activity. Neither phytyl nor oleyl succinate caused apoptosis. These findings show that modifications of different functional moieties of the VE molecule can enhance apoptogenic activity. It is hoped that these observations will lead to the synthesis of analogues with even higher apoptogenic and, consequently, antineoplastic efficacy.


Journal of Biological Chemistry | 2011

Mitochondrial Targeting of Vitamin E Succinate Enhances Its Pro-apoptotic and Anti-cancer Activity via Mitochondrial Complex II

Lan-Feng Dong; Victoria J.A. Jameson; David Patrice Tilly; Jiri Cerny; Elahe Mahdavian; Alvaro Marín-Hernández; Luz Hernández-Esquivel; Sara Rodríguez-Enríquez; Jan Stursa; Paul K. Witting; Jakub Rohlena; Jaroslav Truksa; Katarina Kluckova; Jeffrey Clifford Dyason; Miroslav Ledvina; Brian A. Salvatore; Rafael Moreno-Sánchez; Mark J. Coster; Stephen John Ralph; Robin A. J. Smith; Jiri Neuzil

Mitochondrial complex II (CII) has been recently identified as a novel target for anti-cancer drugs. Mitochondrially targeted vitamin E succinate (MitoVES) is modified so that it is preferentially localized to mitochondria, greatly enhancing its pro-apoptotic and anti-cancer activity. Using genetically manipulated cells, MitoVES caused apoptosis and generation of reactive oxygen species (ROS) in CII-proficient malignant cells but not their CII-dysfunctional counterparts. MitoVES inhibited the succinate dehydrogenase (SDH) activity of CII with IC50 of 80 μm, whereas the electron transfer from CII to CIII was inhibited with IC50 of 1.5 μm. The agent had no effect either on the enzymatic activity of CI or on electron transfer from CI to CIII. Over 24 h, MitoVES caused stabilization of the oxygen-dependent destruction domain of HIF1α fused to GFP, indicating promotion of the state of pseudohypoxia. Molecular modeling predicted the succinyl group anchored into the proximal CII ubiquinone (UbQ)-binding site and successively reduced interaction energies for serially shorter phytyl chain homologs of MitoVES correlated with their lower effects on apoptosis induction, ROS generation, and SDH activity. Mutation of the UbQ-binding Ser68 within the proximal site of the CII SDHC subunit (S68A or S68L) suppressed both ROS generation and apoptosis induction by MitoVES. In vivo studies indicated that MitoVES also acts by causing pseudohypoxia in the context of tumor suppression. We propose that mitochondrial targeting of VES with an 11-carbon chain localizes the agent into an ideal position across the interface of the mitochondrial inner membrane and matrix, optimizing its biological effects as an anti-cancer drug.


Molecular Pharmacology | 2007

Vitamin E Analogs, a Novel Group of “Mitocans,” as Anticancer Agents: The Importance of Being Redox-Silent

Jiri Neuzil; Marco Tomasetti; Yan Zhao; Lan-Feng Dong; Marc Birringer; Xiu-Fang Wang; Pauline Low; Kun Wu; Brian A. Salvatore; Stephen John Ralph

The search for a selective and efficient anticancer agent for treating all neoplastic disease has yet to deliver a universally suitable compound(s). The majority of established anticancer drugs either are nonselective or lose their efficacy because of the constant mutational changes of malignant cells. Until recently, a largely neglected target for potential anticancer agents was the mitochondrion, showing a considerable promise for future clinical applications. Vitamin E (VE) analogs, epitomized by α-tocopheryl succinate, belong to the group of “mitocans” (mitochondrially targeted anticancer drugs). They are selective for malignant cells, cause destabilization of their mitochondria, and suppress cancer in preclinical models. This review focuses on our current understanding of VE analogs in the context of their proapoptotic/anticancer efficacy and suggests that their effect on mitochondria may be amplified by modulation of alternative pathways operating in parallel. We show here that the analogs of VE that cause apoptosis (which translates into their anticancer efficacy) generally do not possess antioxidant (redox) activity and are prototypical of the mitocan group of anticancer compounds. Therefore, by analogy to Oscar Wildes play The Importance of Being Earnest, we use the motto in the title “the importance of being redox-silent” to emphasize an essentially novel paradigm for cancer therapy, in which redox-silence is a prerequisite property for most of the anticancer activities described in this communication.


Current Cancer Drug Targets | 2004

Vitamin E analogues: a new class of inducers of apoptosis with selective anti-cancer effects.

Jiri Neuzil; Marco Tomasetti; Albert Sleiman Mellick; Renata Alleva; Brian A. Salvatore; Marc Birringer; Marc W. Fariss

In spite of unrelenting effort, the net incidence of neoplastic diseases appears not to have been curbed. While some types of cancer have been suppressed significantly, others are either stagnating or on the increase. Therefore, the need for a cure is imperative, in particularly a drug or combination of drugs that would be selective for malignant cells, i.e. with as low secondary toxicity as possible. Recent data strongly suggest that analogues of vitamin E, epitomised by the most studied alpha-tocopheryl succinate (alpha-TOS), may meet the need for the coveted drugs with a selective anti-neoplastic effect. The reasons for this optimism are reviewed in this article.


Free Radical Biology and Medicine | 2011

Mitochondrial targeting of α-tocopheryl succinate enhances its pro-apoptotic efficacy: A new paradigm for effective cancer therapy

Lan-Feng Dong; Victoria J.A. Jameson; David Patrice Tilly; Lubomir Prochazka; Jakub Rohlena; Karel Valis; Jaroslav Truksa; Renata Zobalova; Elahe Mahdavian; Katarina Kluckova; Marina Stantic; Jan Stursa; Ruth Freeman; Paul K. Witting; Erik Norberg; Jacob Goodwin; Brian A. Salvatore; Jana Novotná; Jaroslav Turánek; Miroslav Ledvina; Pavel Hozák; Boris Zhivotovsky; Mark J. Coster; Stephen John Ralph; Robin A. J. Smith; Jiri Neuzil

Mitochondria are emerging as intriguing targets for anti-cancer agents. We tested here a novel approach, whereby the mitochondrially targeted delivery of anti-cancer drugs is enhanced by the addition of a triphenylphosphonium group (TPP(+)). A mitochondrially targeted analog of vitamin E succinate (MitoVES), modified by tagging the parental compound with TPP(+), induced considerably more robust apoptosis in cancer cells with a 1-2 log gain in anti-cancer activity compared to the unmodified counterpart, while maintaining selectivity for malignant cells. This is because MitoVES associates with mitochondria and causes fast generation of reactive oxygen species that then trigger mitochondria-dependent apoptosis, involving transcriptional modulation of the Bcl-2 family proteins. MitoVES proved superior in suppression of experimental tumors compared to the untargeted analog. We propose that mitochondrially targeted delivery of anti-cancer agents offers a new paradigm for increasing the efficacy of compounds with anti-cancer activity.


Journal of Neurochemistry | 2005

α-Tocopheryl succinate selectively induces apoptosis in neuroblastoma cells: potential therapy of malignancies of the nervous system?

Emma Swettenham; Paul K. Witting; Brian A. Salvatore; Jiri Neuzil

Vitamin E (VE) analogues, epitomized by α‐tocopheryl succinate (α‐TOS), are potent inducers of apoptosis and anti‐cancer agents. Here, we tested their effect on the highly malignant N‐type neuroblastoma (Nb) cells and their differentiated, neurone‐like counterparts. Nb cells were highly susceptible to several VE analogues, while differentiated Nb cells were relatively resistant to α‐TOS. The importance of caspase‐9 rather than caspase‐8, as judged by specific siRNAs studies, together with the loss of the inner mitochondrial potential, suggests that α‐TOS triggers apoptosis in Nb cells via the mitochondrial pathway. Cultured Nb cells were sensitized to α‐TOS by pre‐treatment with Bcl‐2, Bcl‐xL or Mcl‐1 siRNAs, while the malignant cell line was more resistant to the vitamin E analogue when Bax was knocked down. In contrast, overexpression of Bcl‐2 in Nb cells rendered them more resistant to α‐TOS‐induced apoptosis. The resistance of differentiated Nb cells to α‐TOS‐mediated apoptosis occurred via two modes: first, by up‐regulation of the anti‐apoptotic Bcl‐2 family proteins and second, by accumulation of decreased levels of reactive oxygen species when challenged with α‐TOS. We conclude that α‐TOS is highly selective in killing malignant brain cancer cells while relatively inert toward differentiated neuronal cells, and that vitamin E analogues may be novel therapeutics for the treatment of tumours such as neuroblastomas.


International Journal of Cancer | 2005

Vitamin E amides, a new class of vitamin E analogues with enhanced proapoptotic activity

Adisa Tomic-Vatic; John EyTina; James M. Chapman; Elahe Mahdavian; Jiri Neuzil; Brian A. Salvatore

Vitamin E (VE) analogues, epitomized by α‐tocopheryl succinate (α‐TOS), are proapoptotic agents with selective antineoplastic activity. The molecule of α‐TOS comprises several structurally and functionally distinct moieties that can be modified in order to yield analogues with higher activity. In order to find analogues with higher apoptogenic efficacy, we prepared novel compounds where the ester bond was replaced by an amide bond. All of these analogues were significantly more proapoptotic than their ester counterparts, with α‐tocopheryl maleyl amide being the most effective. Importantly, methylation of the free carboxylic group completely obliterated apoptogenic activity of the compounds. Similarly as shown for the ester analogues, the amides induced apoptosis by mitochondrial destabilization. Superiority of amides over the ester analogues may be due to their higher partitioning into the lipid phase, as suggested by the log p‐values that were lower for the amides than the corresponding esters. In conclusion, we present evidence that modification of the ester bond of agents such as α‐TOS can be used as a basis for generating novel analogues with higher efficacy of killing malignant cells, an activity that suggests anticancer effect of the agents.


Toxicology and Applied Pharmacology | 2009

Liposomal formulation of α-tocopheryl maleamide: In vitro and in vivo toxicological profile and anticancer effect against spontaneous breast carcinomas in mice

Jaroslav Turánek; Xiu-Fang Wang; Pavlína Knötigová; Štěpán Koudelka; Lan-Feng Dong; Eva Vrublová; Elahe Mahdavian; Lubomir Prochazka; Smink Sangsura; Antonín Vacek; Brian A. Salvatore; Jiri Neuzil

The vitamin E analogue alpha-tocopheryl succinate (alpha-TOS) is an efficient anti-cancer drug. Improved efficacy was achieved through the synthesis of alpha-tocopheryl maleamide (alpha-TAM), an esterase-resistant analogue of alpha-tocopheryl maleate. In vitro tests demonstrated significantly higher cytotoxicity of alpha-TAM towards cancer cells (MCF-7, B16F10) compared to alpha-TOS and other analogues prone to esterase-catalyzed hydrolysis. However, in vitro models demonstrated that alpha-TAM was cytotoxic to non-malignant cells (e.g. lymphocytes and bone marrow progenitors). Thus we developed lyophilized liposomal formulations of both alpha-TOS and alpha-TAM to solve the problem with cytotoxicity of free alpha-TAM (neurotoxicity and anaphylaxis), as well as the low solubility of both drugs. Remarkably, neither acute toxicity nor immunotoxicity implicated by in vitro tests was detected in vivo after application of liposomal alpha-TAM, which significantly reduced the growth of cancer cells in hollow fiber implants. Moreover, liposomal formulation of alpha-TAM and alpha-TOS each prevented the growth of tumours in transgenic FVB/N c-neu mice bearing spontaneous breast carcinomas. Liposomal formulation of alpha-TAM demonstrated anti-cancer activity at levels 10-fold lower than those of alpha-TOS. Thus, the liposomal formulation of alpha-TAM preserved its strong anti-cancer efficacy while eliminating the in vivo toxicity found of the free drug applied in DMSO. Liposome-based targeted delivery systems for analogues of vitamin E are of interest for further development of efficient and safe drug formulations for clinical trials.


International Journal of Molecular Medicine | 2014

Caspase-dependent signaling underlies glioblastoma cell death in response to the fungal metabolite, fusarochromanone

Elahe Mahdavian; Monique Marshall; Patrick M. Martin; Patrice Cagle; Brian A. Salvatore; Quincy A. Quick

Fungal metabolites continue to show promise as a viable class of anticancer agents. In the present study, we investigated the efficacy of the fungal metabolite, fusarochromanone (FC101), for its antitumor activities in glioblastomas, which have a median survival of less than two years and a poor clinical response to surgical resection, radiation therapy and chemotherapy. Using clinically applicable doses, we demonstrated that FC101 induced glioblastoma apoptotic cell death via caspase dependent signaling, as indicated by the cleavage of poly(ADP-ribose) polymerase, glioblastoma (PARP). FC101 also induced differential reactive oxygen species (ROS) levels in glioblastoma cells, contrasting a defined role of oxidative stress in apoptotic cell death observed with other fungal metabolites. Furthermore, the antitumorigenic effects of FC101 on tumor cell migration were assessed. Cell migration assays revealed that FC101 significantly reduced the migratory capacity of glioblastomas, which are incredibly invasive tumors. Taken together, the present study establishes FC101 as a candidate anticancer agent for the cooperative treatment of glioblastomas.


PLOS ONE | 2014

Fusarochromanone Induces G1 Cell Cycle Arrest and Apoptosis in COS7 and HEK293 Cells

Ying Gu; Xin Chen; Chaowei Shang; Karnika Singh; Mansoureh Barzegar; Elahe Mahdavian; Brian A. Salvatore; Shanxiang Jiang; Shile Huang

Fusarochromanone (FC101), a mycotoxin produced by the fungus Fusarium equiseti, is frequently observed in the contaminated grains and feedstuffs, which is toxic to animals and humans. However, the underlying molecular mechanism remains to be defined. In this study, we found that FC101 inhibited cell proliferation and induced cell death in COS7 and HEK293 cells in a concentration-dependent manner. Flow cytometric analysis showed that FC101 induced G1 cell cycle arrest and apoptosis in the cells. Concurrently, FC101 downregulated protein expression of cyclin D1, cyclin-dependent kinases (CDK4 and CDK6), and Cdc25A, and upregulated expression of the CDK inhibitors (p21Cip1 and p27Kip1), resulting in hypophosphorylation of Rb. FC101 also inhibited protein expression of Bcl-2, Bcl-xL, Mcl-1 and survivin, and induced expression of BAD, leading to activation of caspase 3 and cleavage of PARP, indicating caspase-dependent apoptosis. However, Z-VAD-FMK, a pan-caspase inhibitor, only partially prevented FC101-induced cell death, implying that FC101 may induce cell death through both caspase-dependent and -independent mechanisms. Our results support the notion that FC101 executes its toxicity at least by inhibiting cell proliferation and inducing cell death.

Collaboration


Dive into the Brian A. Salvatore's collaboration.

Top Co-Authors

Avatar

Elahe Mahdavian

Louisiana State University in Shreveport

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

Tara Williams-Hart

Louisiana State University in Shreveport

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge