Network


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

Hotspot


Dive into the research topics where Julian J. Raffoul is active.

Publication


Featured researches published by Julian J. Raffoul.


BMC Cancer | 2006

Genistein inhibits radiation-induced activation of NF-κB in prostate cancer cells promoting apoptosis and G2/M cell cycle arrest

Julian J. Raffoul; Yu Wang; Omer Kucuk; Jeffrey D. Forman; Fazlul H. Sarkar; Gilda G. Hillman

BackgroundNew cancer therapeutic strategies must be investigated that enhance prostate cancer treatment while minimizing associated toxicities. We have previously shown that genistein, the major isoflavone found in soy, enhanced prostate cancer radiotherapy in vitro and in vivo. In this study, we investigated the cellular and molecular interaction between genistein and radiation using PC-3 human prostate cancer cells.MethodsTumor cell survival and progression was determined by clonogenic analysis, flow cytometry, EMSA analysis of NF-κB, and western blot analysis of cyclin B1, p21WAF1/Cip1, and cleaved PARP protein.ResultsGenistein combined with radiation caused greater inhibition in PC-3 colony formation compared to genistein or radiation alone. Treatment sequence of genistein followed by radiation and continuous exposure to genistein showed optimal effect. Cell cycle analysis demonstrated a significant dose- and time-dependent G2/M arrest induced by genistein and radiation that correlated with increased p21WAF1/Cip1 and decreased cyclin B1 expression. NF-κB activity was significantly decreased by genistein, yet increased by radiation. Radiation-induced activation of NF-κB activity was strongly inhibited by genistein pre-treatment. A significant and striking increase in cleaved PARP protein was measured following combined genistein and radiation treatment, indicating increased apoptosis.ConclusionA mechanism of increased cell death by genistein and radiation is proposed to occur via inhibition of NF-κB, leading to altered expression of regulatory cell cycle proteins such as cyclin B and/or p21WAF1/Cip1, thus promoting G2/M arrest and increased radiosensitivity. These findings support the important and novel strategy of combining genistein with radiation for the treatment of prostate cancer.


Cancer Research | 2007

Down-regulation of Apurinic/Apyrimidinic Endonuclease 1/Redox Factor-1 Expression by Soy Isoflavones Enhances Prostate Cancer Radiotherapy In vitro and In vivo

Julian J. Raffoul; Sanjeev Banerjee; Vinita Singh-Gupta; Zvi E. Knoll; Alemu Fite; Hao Zhang; Judith Abrams; Fazlul H. Sarkar; Gilda G. Hillman

We previously showed that genistein, the major bioactive component of soy isoflavones, acts as a radiosensitizer and potentiates prostate tumor cell killing by radiation in vitro and in animal tumor models in vivo. However, when given alone in vivo, pure genistein promoted increased lymph node metastasis, which was not observed with a soy isoflavone mixture consisting of genistein, daidzein, and glycitein. In this study, we show that soy inhibit tumor cell growth and potentiates radiation-induced cell killing in vitro like pure genistein. In an orthotopic model, combining soy isoflavones with tumor irradiation inhibited prostate tumor growth. To determine the molecular mechanisms by which soy isoflavones potentiate radiotherapy, we investigated apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) and nuclear factor kappaB (NF-kappaB), two signaling molecules involved in survival pathways. Soy isoflavones decreased APE1/Ref-1 expression in vitro, whereas radiation up-regulated it. Pretreatment with soy isoflavones followed by radiation inhibited APE1/Ref-1 expression. APE1/Ref-1 decrease correlated with decreased DNA-binding activity of NF-kappaB mediated by soy isoflavones and radiation, thus promoting cell killing. In vivo treatment of prostate tumors with soy isoflavones and radiation down-regulated APE1/Ref-1 protein expression and NF-kappaB activity, confirming the molecular alterations observed in vitro. The down-regulation of APE1/Ref-1 and NF-kappaB by isoflavones, in vitro and in vivo, supports our hypothesis that these markers represent biological targets of isoflavones. Indeed, a 2-fold increase in APE1/Ref-1 expression, obtained by cDNA transfection, resulted in a 2-fold increase in NF-kappaB DNA-binding activity, and both of which were down-regulated by soy isoflavones, confirming the cross-talk between these molecules and, in turn, causing radiosensitization.


DNA Repair | 2003

Caloric restriction promotes genomic stability by induction of base excision repair and reversal of its age-related decline

Diane C. Cabelof; Sunitha Yanamadala; Julian J. Raffoul; ZhongMao Guo; Abdulsalam Soofi; Ahmad R. Heydari

Caloric restriction is a potent experimental manipulation that extends mean and maximum life span and delays the onset and progression of tumors in laboratory rodents. While caloric restriction (CR) clearly protects the genome from deleterious damage, the mechanism by which genomic stability is achieved remains unclear. We provide evidence that CR promotes genomic stability by increasing DNA repair capacity, specifically base excision repair (BER). CR completely reverses the age-related decline in BER capacity (P<0.01) in all tissues tested (brain, liver, spleen and testes) providing aged, CR animals with the BER phenotype of young, ad libitum-fed animals. This CR-induced reversal of the aged BER phenotype is accompanied by a reversal in the age-related decline in DNA polymerase beta (beta-pol), a rate-limiting enzyme in the BER pathway. CR significantly reversed the age-related loss of beta-pol protein levels (P<0.01), mRNA levels (P<0.01) and enzyme activity (P<0.01) in all tissues tested. Additionally, in young (4-6-month-old) CR animals a significant up-regulation in BER capacity, beta-pol protein and beta-pol mRNA is observed (P<0.01), demonstrating an early effect of CR that may provide insight in distinguishing the anti-tumor from the anti-aging effects of CR. This up-regulation in BER by caloric restriction in young animals corresponds to increased protection from carcinogen exposure, as mutation frequency is significantly reduced in CR animals exposed to either DMS or 2-nitropropane (2-NP) (P<0.01). Overall the data suggest an important biological consequence of moderate BER up-regulation and provides support for the hormesis theory of caloric restriction.


Mutation Research | 2002

Attenuation of DNA polymerase β-dependent base excision repair and increased DMS-induced mutagenicity in aged mice

Diane C. Cabelof; Julian J. Raffoul; Sunitha Yanamadala; Cirlette Ganir; ZhongMao Guo; Ahmad R. Heydari

The biological mechanisms responsible for aging remain poorly understood. We propose that increases in DNA damage and mutations that occur with age result from a reduced ability to repair DNA damage. To test this hypothesis, we have measured the ability to repair DNA damage in vitro by the base excision repair (BER) pathway in tissues of young (4-month-old) and old (24-month-old) C57BL/6 mice. We find in all tissues tested (brain, liver, spleen and testes), the ability to repair damage is significantly reduced (50-75%; P<0.01) with age, and that the reduction in repair capacity seen with age correlates with decreased levels of DNA polymerase beta (beta-pol) enzymatic activity, protein and mRNA. To determine the biological relevance of this age-related decline in BER, we measured spontaneous and chemically induced lacI mutation frequency in young and old animals. In line with previous findings, we observed a three-fold increase in spontaneous mutation frequency in aged animals. Interestingly, lacI mutation frequency in response to dimethyl sulfate (DMS) does not significantly increase in young animals whereas identical exposure in aged animals results in a five-fold increase in mutation frequency. Because DMS induces DNA damage processed by the BER pathway, it is suggested that the increased mutagenicity of DMS with age is related to the decline in BER capacity that occurs with age. The inability of the BER pathway to repair damages that accumulate with age may provide a mechanistic explanation for the well-established phenotype of DNA damage accumulation with age.


International Journal of Cancer | 2009

Radiation‐induced HIF‐1α cell survival pathway is inhibited by soy isoflavones in prostate cancer cells

Vinita Singh-Gupta; Hao Zhang; Sanjeev Banerjee; Dejuan Kong; Julian J. Raffoul; Fazlul H. Sarkar; Gilda G. Hillman

We previously showed that treatment of prostate cancer cells with soy isoflavones and radiation resulted in greater cell killing in vitro, and caused downregulation of NF‐κB and APE1/Ref‐1. APE1/Ref‐1 functions as a redox activator of transcription factors, including NF‐κB and HIF‐1α. These molecules are upregulated by radiation and implicated in radioresistance of cancer cells. We extended our studies to investigate the role of HIF‐1α survival pathway and its upstream Src and STAT3 molecules in isoflavones and radiation interaction. Radiation induced phosphorylation of Src and STAT3 leading to induction of HIF‐1α. Genistein, daidzein or a mixture of soy isoflavones did not activate this pathway. These data were observed both in PC‐3 (AR‐) and C4‐2B (AR+) androgen‐independent cell lines. Pretreatment with isoflavones inhibited Src/STAT3/HIF‐1α activation by radiation and nuclear translocation of HIF‐1α. These findings correlated with decreased expression of APE1/Ref‐1 and DNA binding activity of HIF‐1α and NF‐κB. In APE1/Ref‐1 cDNA transfected cells, radiation caused a greater increase in HIF‐1α and NF‐κB activities but this effect was inhibited by pretreatment with soy prior to radiation. Transfection experiments indicate that APE1/Ref‐1 inhibition by isoflavones impairs the radiation‐induced transcription activity of NF‐κB and HIF‐1α. This mechanism could result in the inhibition of genes essential for tumor growth and angiogenesis, as demonstrated by inhibition of VEGF production and HUVECs tube formation. Our novel findings suggest that the increased responsiveness to radiation mediated by soy isoflavones could be due to pleiotropic effects of isoflavones blocking cell survival pathways induced by radiation including Src/STAT3/HIF‐1α, APE1/Ref‐1 and NF‐κB.


International Journal of Cancer | 2007

Soy isoflavones enhance radiotherapy in a metastatic prostate cancer model

Julian J. Raffoul; Sanjeev Banerjee; Mingxin Che; Zvi E. Knoll; Daniel R. Doerge; Judith Abrams; Omer Kucuk; Fazlul H. Sarkar; Gilda G. Hillman

We previously reported that genistein, the bioactive isoflavone of soybeans, acts as a radiosensitizer for prostate cancer. Pretreatment of tumor cells with genistein potentiated radiation‐induced killing in vitro and in orthotopic models in vivo. However, pure genistein promoted increased lymph node metastasis, when administered alone in vivo. We investigated in vitro and in vivo the effects of soy isoflavones (genistein, daidzein and glycitein) as soy pills of similar composition are used in human interventions but not pure genistein. Soy isoflavones inhibited cell survival and potentiated radiation cell killing in PC‐3 tumor cells, in vitro. Increased cell killing correlated with inhibition of antiapoptotic molecules Bcl‐xL and survivin, upregulation of proapoptotic Bax molecule and PARP cleavage, suggesting activation of apoptotic pathways. In vivo, using the PC‐3 orthotopic metastatic mouse model, soy isoflavones and prostate tumor irradiation led to enhanced control of primary tumor growth and metastasis, as observed with pure genistein and radiation. Interestingly, treatment with soy isoflavones did not increase metastasis to para‐aortic lymph nodes in contrast to the consistent increase caused by pure genistein. Histologically prostate tumors, treated with soy isoflavones and radiation, showed tumor destruction and in situ tissue alterations, comparable with genistein and radiation effects. However, genistein, but not soy isoflavones, caused induction of HIF1‐α in prostate tumors, suggesting that induction of hypoxia by pure genistein could contribute to increased metastasis. Our studies demonstrate the safety and potential role of soy isoflavones for enhancing the therapeutic effect of radiotherapy in prostate cancer.


BMC Cancer | 2007

Progression of renal cell carcinoma is inhibited by genistein and radiation in an orthotopic model

Gilda G. Hillman; Yu Wang; Mingxin Che; Julian J. Raffoul; Mark Yudelev; Omer Kucuk; Fazlul H. Sarkar

BackgroundWe have previously reported the potentiation of radiotherapy by the soy isoflavone genistein for prostate cancer using prostate tumor cells in vitro and orthotopic prostate tumor models in vivo. However, when genistein was used as single therapy in animal models, it promoted metastasis to regional para-aortic lymph nodes. To clarify whether these intriguing adverse effects of genistein are intrinsic to the orthotopic prostate tumor model, or these results could also be recapitulated in another model, we used the orthotopic metastatic KCI-18 renal cell carcinoma (RCC) model established in our laboratory.MethodsThe KCI-18 RCC cell line was generated from a patient with papillary renal cell carcinoma. Following orthotopic renal implantation of KCI-18 RCC cells and serial in vivo kidney passages in nude mice, we have established a reliable and predictable metastatic RCC tumor model. Mice bearing established kidney tumors were treated with genistein combined with kidney tumor irradiation. The effect of the therapy was assessed on the primary tumor and metastases to various organs.ResultsIn this experimental model, the karyotype and histological characteristics of the human primary tumor are preserved. Tumor cells metastasize from the primary renal tumor to the lungs, liver and mesentery mimicking the progression of RCC in humans. Treatment of established kidney tumors with genistein demonstrated a tendency to stimulate the growth of the primary kidney tumor and increase the incidence of metastasis to the mesentery lining the bowel. In contrast, when given in conjunction with kidney tumor irradiation, genistein significantly inhibited the growth and progression of established kidney tumors. These findings confirm the potentiation of radiotherapy by genistein in the orthotopic RCC model as previously shown in orthotopic models of prostate cancer.ConclusionOur studies in both RCC and prostate tumor models demonstrate that the combination of genistein with primary tumor irradiation is a more effective and safer therapeutic approach as the tumor growth and progression are inhibited both in the primary and metastatic sites.


Radiation Research | 2006

Prostate Cancer Treatment is Enhanced by Genistein In Vitro and In Vivo in a Syngeneic Orthotopic Tumor Model

Yu Wang; Julian J. Raffoul; Mingxin Che; Daniel R. Doerge; Michael C. Joiner; Omer Kucuk; Fazlul H. Sarkar; Gilda G. Hillman

Abstract Wang, Y., Raffoul, J. J., Che, M., Doerge, D. R., Joiner, M. C., Kucuk, O., Sarkar, F. H. and Hillman, G. G. Prostate Cancer Treatment is Enhanced by Genistein In Vitro and In Vivo in a Syngeneic Orthotopic Tumor Model. Radiat. Res. 166, 73–80 (2006). Pretreatment with genistein, a bioactive component of soy isoflavones, potentiated cell killing induced by radiation in human PC-3 prostate cancer cells in vitro. Using an orthotopic xenograft in nude mice, we demonstrated that genistein combined with prostate tumor irradiation caused greater inhibition of primary tumor growth and increased control of spontaneous metastasis to para-aortic lymph nodes, increasing mouse survival. Paradoxically, treatment with genistein alone increased metastasis to lymph nodes. This observation is of concern in relation to soy-based clinical trials for cancer patients. To address whether this observation is because nude mice have an impaired immune system, these studies were repeated in orthotopic RM-9 prostate tumors in syngeneic C57BL/6 mice. The combination of genistein with radiation in this model also caused a greater inhibition of primary tumor growth and spontaneous metastasis to regional para-aortic lymph nodes, whereas treatment with genistein alone showed a trend to increased lymph node metastasis. Data from the syngeneic and xenograft models are comparable and indicate that the combination of genistein with radiotherapy is more effective and safer for prostate cancer treatment than genistein alone, which promotes metastatic spread to regional lymph nodes.


Current Cancer Drug Targets | 2007

Radiosensitization of prostate cancer by soy isoflavones

Julian J. Raffoul; Fazlul H. Sarkar; Gilda G. Hillman

A trend in investigating the use of several nutritional compounds for cancer chemoprevention has revealed that phytochemicals demonstrated anti-cancer properties by inhibiting signal transduction pathways essential for cancer cell proliferation, tumor growth, invasion and metastasis. Emerging evidence suggests that the anti-proliferative and anti-oxidant effects of some of these dietary agents could be utilized to both potentiate the response of cancer cells to radiotherapy and reduce radiation-induced toxicity to normal surrounding tissues. Using pre-clinical orthotopic models of prostate cancer, studies on the combination of soy isoflavones with tumor irradiation demonstrate a synergistic anti-cancer effect between these two modalities and emphasize the potential and safety of dietary factors to improve conventional radiotherapy for a better control of tumor growth and metastasis. The goal of this review is to focus on the role of soy isoflavones as potent radiosensitizers for prostate cancer and other malignancies. We will discuss molecular pathways regulated by soy isoflavones that inhibit survival pathways activated by radiation and ultimately drive the cells to cell death both in vitro and in vivo in pre-clinical models.


Cancer Research | 2003

Base excision repair deficiency caused by polymerase β haploinsufficiency: Accelerated DNA damage and increased mutational response to carcinogens

Diane C. Cabelof; ZhongMao Guo; Julian J. Raffoul; Robert W. Sobol; Samuel H. Wilson; Arlan Richardson; Ahmad R. Heydari

Collaboration


Dive into the Julian J. Raffoul's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

ZhongMao Guo

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Mingxin Che

Wayne State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yu Wang

Wayne State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge