Network


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

Hotspot


Dive into the research topics where Chad N. Hancock is active.

Publication


Featured researches published by Chad N. Hancock.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC.

Wei Liu; Anne Le; Chad N. Hancock; Andrew N. Lane; Chi V. Dang; Teresa W.-M. Fan; James M. Phang

In addition to glycolysis, the oncogenic transcription factor c-MYC (MYC) stimulates glutamine catabolism to fuel growth and proliferation of cancer cells through up-regulating glutaminase (GLS). Glutamine is converted to glutamate by GLS, entering the tricarboxylic acid cycle as an important energy source. Less well-recognized, glutamate can also be converted to proline through Δ1-pyrroline-5-carboxylate (P5C) and vice versa. This study suggests that some MYC-induced cellular effects are due to MYC regulation of proline metabolism. Proline oxidase, also known as proline dehydrogenase (POX/PRODH), the first enzyme in proline catabolism, is a mitochondrial tumor suppressor that inhibits proliferation and induces apoptosis. MiR-23b* mediates POX/PRODH down-regulation in human kidney tumors. MiR-23b* is processed from the same transcript as miR-23b; the latter inhibits the translation of GLS. Using MYC-inducible human Burkitt lymphoma model P493 and PC3 human prostate cancer cells, we showed that MYC suppressed POX/PRODH expression primarily through up-regulating miR-23b*. The growth inhibition in the absence of MYC was partially reversed by POX/PRODH knockdown, indicating the importance of suppression of POX/PRODH in MYC-mediated cellular effects. Interestingly, MYC not only inhibited POX/PRODH, but also markedly increased the enzymes of proline biosynthesis from glutamine, including P5C synthase and P5C reductase 1. MYC-induced proline biosynthesis from glutamine was directly confirmed using 13C,15N-glutamine as a tracer. The metabolic link between glutamine and proline afforded by MYC emphasizes the complexity of tumor metabolism. Further studies of the relationship between glutamine and proline metabolism should provide a deeper understanding of tumor metabolism while enabling the development of novel therapeutic strategies.


Current Opinion in Clinical Nutrition and Metabolic Care | 2015

Proline metabolism and cancer: emerging links to glutamine and collagen.

James M. Phang; Wei Liu; Chad N. Hancock; Joseph W. Fischer

Purpose of reviewProline metabolism impacts a number of regulatory targets in both animals and plants and is especially important in cancer. Glutamine, a related amino acid, is considered second in importance only to glucose as a substrate for tumors. But proline and glutamine are interconvertible and linked in their metabolism. In animals, proline and glutamine have specific regulatory functions and their respective physiologic sources. A comparison of the metabolism of proline and glutamine would help us understand the importance of these two nonessential amino acids in cancer metabolism. Recent findingsThe regulatory functions of proline metabolism proposed 3 decades ago have found relevance in many areas. For cancer, these functions play a role in apoptosis, autophagy and in response to nutrient and oxygen deprivation. Importantly, proline-derived reactive oxygen species served as a driving signal for reprogramming. This model has been applied by others to metabolic regulation for the insulin-prosurvival axis, induction of adipose triglyceride lipase for lipid metabolism and regulation of embryonic stem cell development. Of special interest, modulatory proteins such as parkinson protein 7 and oral cancer overexpressed 1 interact with pyrroline-5-carboxylate reductase, a critical component of the proline regulatory axis. Although the interconvertibility of proline and glutamine has been long established, recent findings showed that the proto-oncogene, cellular myelocytomatosis oncogene, upregulates glutamine utilization (glutaminase) and routes glutamate to proline biosynthesis (pyrroline-5-carboxylate synthase, pyrroline-5-carboxylate reductases). Additionally, collagen, which contains large amounts of proline, may be metabolized to serve as a reservoir for proline. This metabolic relationship as well as the new regulatory targets of proline metabolism invites an elucidation of the differential effects of these nonessential amino acids and their production, storage and mobilization. SummaryMechanisms by which the proline regulatory axis modulates the cancer phenotype are being revealed. Proline can be synthesized from glutamine as well as derived from collagen degradation. The metabolism of proline serves as a source of energy during stress, provides signaling reactive oxygen species for epigenetic reprogramming and regulates redox homeostasis.


Frontiers in Oncology | 2012

The proline regulatory axis and cancer.

James M. Phang; Wei Liu; Chad N. Hancock; Kyle J. Christian

Studies in metabolism and cancer have characterized changes in core pathways involving glucose and glutamine, emphasizing the provision of substrates for building cell mass. But recent findings suggest that pathways previously considered peripheral may play a critical role providing mechanisms for cell regulation. Several of these mechanisms involve the metabolism of non-essential amino acids, for example, the channeling of glycolytic intermediates into the serine pathway for one-carbon transfers. Historically, we proposed that the proline biosynthetic pathway participated in a metabolic interlock with glucose metabolism. The discovery that proline degradation is activated by p53 directed our attention to the initiation of apoptosis by proline oxidase/dehydrogenase. Now, however, we find that the biosynthetic mechanisms and the metabolic interlock may depend on the pathway from glutamine to proline, and it is markedly activated by the oncogene MYC. These findings add a new dimension to the proline regulatory axis in cancer and present attractive potential targets for cancer treatment.


Epigenetics | 2013

Bridging epigenetics and metabolism

James M. Phang; Wei Liu; Chad N. Hancock

Recent research suggests that chromatin-modifying enzymes are metabolic sensors regulating gene expression. Epigenetics is linked to metabolomics in response to the cellular microenvironment. Specific metabolites involved in this sensing mechanism include S-adenosylmethionine, acetyl-CoA, alphaketoglutarate and NAD+. Although the core metabolic pathways involving glucose have been emphasized as the source of these metabolites, the reprogramming of pathways involving non-essential amino acids may also play an important role, especially in cancer. Examples include metabolic pathways for glutamine, serine and glycine. The coupling of these pathways to the intermediates affecting epigenetic regulation occurs by “parametabolic” mechanisms. The metabolism of proline may play a special role in this parametabolic linkage between metabolism and epigenetics. Both proline degradation and biosynthesis are robustly affected by oncogenes or suppressor genes, and they can modulate intermediates involved in epigenetic regulation. A number of mechanisms in a variety of animal species have been described by our laboratory and by others. The challenge we now face is to identify the specific chromatin-modifying enzymes involved in coupling of proline metabolism to altered reprogramming of gene expression.


Scientific Reports | 2015

Proline biosynthesis augments tumor cell growth and aerobic glycolysis: involvement of pyridine nucleotides.

Wei Liu; Chad N. Hancock; Joseph W. Fischer; Meredith Harman; James M. Phang

The metabolism of the nonessential amino acid proline contributes to tumor metabolic reprogramming. Previously we showed that MYC increases proline biosynthesis (PB) from glutamine. Here we show MYC increases the expression of the enzymes in PB at both protein and mRNA levels. Blockade of PB decreases tumor cell growth and energy production. Addition of Δ1-pyrroline-5-carboxylate (P5C) or proline reverses the effects of P5C synthase knockdown but not P5C reductases knockdown. Importantly, the reversal effect of proline was blocked by concomitant proline dehydrogenase/oxidase (PRODH/POX) knockdown. These findings suggest that the important regulatory contribution of PB to tumor growth derives from metabolic cycling between proline and P5C rather than product proline or intermediate P5C. We further document the critical role of PB in maintaining pyridine nucleotide levels by connecting the proline cycle to glycolysis and to the oxidative arm of the pentose phosphate pathway. These findings establish a novel function of PB in tumorigenesis, linking the reprogramming of glucose, glutamine and pyridine nucleotides, and may provide a novel target for antitumor therapy.


Amino Acids | 2016

Co-regulation of mitochondrial respiration by proline dehydrogenase/oxidase and succinate.

Chad N. Hancock; Wei Liu; W. Gregory Alvord; James M. Phang

Proline dehydrogenase/oxidase (PRODH/POX) is a mitochondrial protein critical to multiple stress pathways. Because of the roles of PRODH/POX in signaling, and its shared localization to the mitochondrial inner membrane with the electron transport chain (ETC), we investigated whether there was a direct relationship between PRODH/POX and regulation of the ETC. We found that PRODH/POX binds directly to CoQ1 and that CoQ1-dependent PRODH/POX activity required functional Complex III and Complex IV. PRODH/POX supported respiration in living cells during nutrient stress; however, expression of PRODH/POX resulted in an overall decrease in respiratory fitness. Effects on respiratory fitness were inhibited by DHP and NAC, indicating that these effects were mediated by PRODH/POX-dependent reactive oxygen species (ROS) generation. PRODH/POX expression resulted in a dose-dependent down-regulation of Complexes I–IV of the ETC, and this effect was also mitigated by the addition of DHP and NAC. We found that succinate was an uncompetitive inhibitor of PRODH/POX activity, inhibited ROS generation by PRODH/POX, and alleviated PRODH/POX effects on respiratory fitness. The findings demonstrate novel cross-talk between proline and succinate respiration in vivo and provide mechanistic insights into observations from previous animal studies. Our results suggest a potential regulatory loop between PRODH/POX and succinate in regulation of mitochondrial respiration.


Amino Acids | 2016

Erratum to: Co-regulation of mitochondrial respiration by proline dehydrogenase/oxidase and succinate

Chad N. Hancock; Wei Liu; W. Gregory Alvord; James M. Phang

Abbreviations AA5 Atpenin A5 AMA Antimycin A CIII-R Complex III Rieske subunit CM-H2DCFDA 5 ( a n d 6 ) C h l o r o m e t h y l 2 ′ , 7 ′ dichlorodihydrofluorescein diacetate, acetyl ester CoQ1 Coenzyme Q1 COX IV Cytochrome C oxidase subunit IV DCIP 2,6-Dichlorophenolindophenol DHP 3,4-Dehydro-l-proline DMSO Dimethyl sulfoxide DOX Doxycycline DSP Dithiobis(succinimidylpropionate) ETC Electron transport chain FCCP Carbonyl cyanide p-trifluoromethoxyphenylhydrazone NAC N-acetyl-l-cysteine NDUFA10 NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 10 OAB 2-Aminobenzaldehyde OCR Oxygen consumption rate OLIGO Oligomycin P5C Pyrroline-5-carboxylate PRODH/POX Proline dehydrogenase/proline oxidase ROS Reactive oxygen species ROT Rotenone SDHA Succinate dehydrogenase subunit A Abstract Proline dehydrogenase/oxidase (PRODH/ POX) is a mitochondrial protein critical to multiple stress pathways. Because of the roles of PRODH/POX in signaling, and its shared localization to the mitochondrial inner membrane with the electron transport chain (ETC), we investigated whether there was a direct relationship between PRODH/POX and regulation of the ETC. We found that PRODH/POX binds directly to CoQ1 and that CoQ1-dependent PRODH/POX activity required functional Complex III and Complex IV. PRODH/POX supported respiration in living cells during nutrient stress; however, expression of PRODH/POX resulted in an overall decrease in respiratory fitness. Effects on respiratory fitness were inhibited by DHP and NAC, indicating that these effects were mediated by PRODH/POX-dependent reactive oxygen species (ROS) generation. PRODH/POX expression resulted in a dose-dependent down-regulation of Complexes I–IV of the ETC, and this effect was also mitigated by the addition of DHP and NAC. We found that succinate was an uncompetitive inhibitor of PRODH/POX activity, inhibited ROS generation by PRODH/POX, and alleviated PRODH/POX effects on respiratory fitness. The findings demonstrate novel cross-talk between proline and succinate respiration in vivo and provide mechanistic insights into


Cancer Research | 2013

Abstract 4613: Effects of co-regulation of proline oxidase and succinate dehydrogenase on oxidative respiration.

Chad N. Hancock; James M. Phang

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Proline oxidase (POX) is an inner mitochondrial membrane protein that oxidizes proline to generate superoxide as well as ATP. Work in our laboratory has documented that POX activity is highly regulated during genotoxic and metabolic stress. Inducers of POX include p53, and proline-dependent ROS play a role in p53- and mitochondrial-mediated apoptosis. Succinate dehydrogenase, also known as Complex II of the electron transport chain (ETC), oxidizes the TCA cycle metabolite succinate to fumarate as it reduces ubiquinone to ubiquinol, generating ATP through oxidative respiration. Complex II and the p53-DNA damage repair pathway are intimately linked. Mutations in Complex II have been shown to increase risk of breast and thyroid cancer through destabilization of p53, and tocopherol-succinate has been demonstrated to reduce radiation-induced apoptosis. Thus, the ETC and activities of p53 appear to be linked through Complex II. Because POX can generate ATP from proline, we explored the relationship between POX and the ETC. We found that POX binds directly to ubiquinone, and that ubiquinone is a very effective acceptor of proline-derived electrons. Increased POX catalytic activity due to ubiquinone required Complex III and Complex IV of the ETC to be functional, indicating that ATP generated by POX is through oxidative respiration. In addition, experiments using the Seahorse XF24 analyzer confirmed that while POX can utilize proline as a short term source of electrons during nutrient stress conditions, long term expression of POX results in reactive oxygen species (ROS)-dependent suppression of respiration, consistent with its role in mitochondrial-mediated apoptosis. We found that the POX and Complex II were linked functionally, and that the presence of succinate dramatically affects POX activity. Lineweaver-Burk analysis showed that succinate inhibits POX catalytic activity using anti-competitive inhibition, a rare mechanism of enzymatic inhibition. Two highly selective inhibitors of the Complex II holoenzyme, TTFA and carboxin, inhibit the ubiquinone-mediated activities of POX; in addition, POX co-immunoprecipitates with Complex II, indicating a physical interaction between these two enzymes. Interestingly, proline appears to stimulate oxidation of succinate by Complex II in vitro, and acute co-administration of proline and succinate to POX-expressing cells results in a burst of oxidative respiration well above that seen with either substrate alone. Finally, addition of succinate to cell culture inhibits ROS generation by POX and inhibits the long-term effects of POX on respiration. Together, these data support a model in which POX is an integral contributor in oxidative respiration during nutrient stress, and in which respiration is regulated by level and duration of POX expression, as well as levels of proline relative to the TCA cycle metabolite, and Complex II substrate, succinate. Citation Format: Chad N. Hancock, James M. Phang. Effects of co-regulation of proline oxidase and succinate dehydrogenase on oxidative respiration. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4613. doi:10.1158/1538-7445.AM2013-4613


Cancer Research | 2012

Abstract 1119: The oxidation of proline by proline oxidase provides a regulated source of ROS for mitochondria derived cellular signaling

Chad N. Hancock; James M. Phang

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL A role for reactive oxygen species (ROS) in intracellular signaling has been generally accepted, but the organellar localization, the enzymatic source and the regulation of ROS generation are not well understood. Recent work by others has focused on mitochondria as a source of ROS and specifically on the reduction of oxygen by electrons from complex III to form superoxide. The accessibility of complex III to the mitochondrial intermembrane space and cytosol provides an attractive hypothesis for mitochondrial ROS as regulators of cellular redox signaling. We now report that the regulated metabolism of proline makes it a special substrate for this process. Proline oxidase (POX) is an inner mitochondrial membrane protein that oxidizes proline to generate superoxide as well as ATP. Work in our laboratory has documented that POX activity is highly regulated during genotoxic and metabolic stress. Inducers of POX include p53, and proline-dependent ROS play a role in mitochondrial apoptosis. PPAR-γ and its ligands also induce POX but depending on the metabolic context, POX initiates autophagy as well as apoptosis. Mechanistic models for ROS generation by POX have come from microorganisms. PutA, the prokaryotic homolog of POX can generate ROS by electrons reducing oxygen directly from its catalytic center. On the other hand, in S. cerevisiae, the POX homolog, Put1, has been shown to directly interact with ubiquinone, thereby contributing electrons derived from proline oxidation into the electron transport chain. In this work, we now report that human POX interacts directly with ubiquinone and that POX activity is markedly increased by the presence of ubiquinone. In addition, by using selective inhibitors of electron transport, we demonstrate that electrons derived from proline can be converted to superoxide by complex III of the electron transport chain. These radicals are directed into both the mitochondrial matrix as well as into the intermembrane space and the cytosol. Thus, POX is not only specifically regulated but also specially located and coupled to the mitochondrial electron transport chain to mediate ROS signaling. These POX- mediated mechanisms provide important insights into its novel apoptotic and metabolic tumor suppressive functions. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1119. doi:1538-7445.AM2012-1119


Archive | 2015

C URRENT OPINION Proline metabolism and cancer: emerging links to glutamine and collagen

James M. Phang; Wei Liu; Chad N. Hancock; Joseph W. Fischer

Collaboration


Dive into the Chad N. Hancock's collaboration.

Top Co-Authors

Avatar

James M. Phang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wei Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anne Le

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Chi V. Dang

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge