PGC1α is a transcription factor coactivator that influences a majority of cellular metabolic pathways. Abnormal expression of PGC1α is associated with several chronic diseases and, in recent years, it has been shown to be a critical controller of cancer development. PGC1α acts as a stress sensor in cancer cells and can be activated by nutrient deprivation, oxidative damage, and chemotherapy. It influences mitochondria respiration, reactive oxygen species defense system, and fatty acid metabolism by interacting with specific transcription factors. The characteristic traits of PGC1α in maintaining metabolic homeostasis promote cancer cell survival and tumor metastasis in harsh microenvironments. Not only does PGC1α act as a coactivator, but is also itself controlled by oncogenes and transcription factors. PGC1α and these molecules can form signaling axes that include PML/PGC1α/PPARα, MITF/PGC1α, and PGC1α/ERRα, which are important in regulating metabolic adaptation in specific cancer types. Some of these PGC1α-associated pathways are inherently activated in cancer cells, and others are induced by stress, which enable cancer cells to acquire resistance against therapy. Notably, certain therapeutic-resistant cancer cells are addicted to PGC1α-dependent metabolic activities. Suppression of PGC1α expression resensitizes these cells to therapeutic treatments, which implicates PGC1α as a promising target in cancer molecular classification and therapy. Mol Cancer Ther; 15(5); 774–82. ©2016 AACR.

Metabolic reprogramming is considered to be a hallmark of cancer (1). Glycolysis, mitochondrial respiration, glutaminolysis, and fatty acid metabolism are important participants in cancer development (2–6). These processes provide cancer cells with an adaptable metabolic feature and afford survival opportunities for cancer cells undergoing stress (7–9). Among the numerous regulators or mediators of cancer metabolism, PPARγ coactivator-1 alpha (PGC1α) is emerging as an essential controller of multiple metabolic pathways (10, 11).

PGC1α is strongly activated by conditions causing energy limitation, including cold, exercise, and fasting, and is particularly abundant in tissues demanding large energy consumption (12–14). Once activated, PGC1α interacts with several transcription factors and affects various biologic activities under normal physiologic conditions (15). Induced by exercise, PGC1α can promote a functional fiber switch toward more oxidative types in skeletal muscle cells by interacting with muscle-specific myocyte enhancer factor 2 family transcription factors (16, 17). PGC1α also facilitates mitochondrial oxidative phosphorylation (OxPhos) by coactivating nuclear respiratory factor 1 and estrogen-related receptors (ERR; refs.18, 19). It has been identified to stimulate fatty acid oxidation (FAO) through interactions with several transcription factors including sirtuin 1 (SIRT1), PPARs, and hepatocyte nuclear factor 4 (20–25), and PGC1α also increases autophagy and thermogenesis through transcription factor EB and uncoupling protein-1, respectively, under stress condition (26–30). In addition, it has been shown to protect cells against oxidative damage through nuclear factor erythroid 2 (Nrf2) and forkhead box O3 (31–33).

In all of these instances, PGC1α functions as a necessary adaptor for cells to maintain metabolic balance under harsh situations, and it plays a protective role in preventing chronic disease, such as skeletal muscle atrophy, heart failure, neurodegeneration, obesity, diabetes, and hepatic steatosis, and some of these diseases are predisposing factors for cancer initiation (10, 11, 34, 35). Recently, several advances have shown that PGC1α expression is tightly associated with cancer progression (10, 11). The exceptional ability of PGC1α in manipulating cellular metabolism enables cancer cells to thrive under a constantly fluctuating energy status, and highlights the importance of PGC1α in effective cancer therapy (36).

The PGC1α gene is located on chromosome 4 in human and encodes a protein containing 798 amino acids (29). When activated, PGC1α can be recruited as a transcriptional coactivator to subsequently dock or bind with transcription factors or nuclear receptors (NR; refs.15, 37). The N-terminal activation domain and LXXLL motifs of PGC1α interact with various transcription factors (19, 37, 38). Proteins, such as CREB-binding protein, p300 and steroid receptor that acetylate histones, can be sequentially recruited to the PGC1α transcriptional activator complex, to remodel the chromatin structure and provide optimal biochemical conditions for target gene transactivation (39). The C terminal of PGC-1α is believed to recruit the thyroid receptor–associated protein/vitamin D receptor–interacting protein/mediator complex that facilitates transcription initiation and interferes with RNA processing through Ser/Arg-rich or RNA-binding domains (40, 41).

Notably, PGC1α is controlled by several posttranslational modifications. Because PGC1α is markedly sensitive to cellular energy status, it is tightly regulated by stress sensors such as AMP-activated protein kinase (AMPK) and SIRT1. Both AMPK-mediated phosphorylation and SIRT1-mediated deacetylation activate PGC1α under energy deprivation conditions (42, 43). In addition, the p38 MAPK stabilizes the PGC1α protein by increasing its phosphorylation state (44). On the other hand, methylation of the arginine residues at the C-terminal region by protein arginine methyltransferase 1 (PRMT1) decreases PGC1α stability whereas phosphorylation of PGC-1α by Akt/protein kinase B and SUMOylation at the conserved lysine residue 183 attenuate PGC1α activity (44–46). These diverse posttranslational modifications direct PGC1α to different target genes. In addition to posttranslational modifications, PGC1α is also influenced by cellular calcium (Ca2+) and cyclic adenosine monophosphate signaling (47, 48). Notably, some transcription factors coactivated by PGC1α can, in turn, regulate PGC1α, comprising autoregulatory loops that augment target gene transcription (17).

PGC1α has been shown to be a promoter of carcinogenesis in chemical-induced colon and liver carcinoma mouse models (49). Ectopic expression of PGC1α has been observed in several cancer types (50–53), and it is regulated by several oncogenes and signaling pathways (Fig. 1; refs.54–57). Similar to its normal physiologic functions, PGC1α primarily regulates mitochondrial respiration and detoxification of reactive oxygen species (ROS) in cancer cells through specific signaling pathways and transcription factors (Table 1). In addition, the involvement of PGC1α in regulating FAO and glucose- or glutamine-derived lipogenesis in cancer cells has become clearer in recent years (Fig. 2; refs.49, 51, 58).

Figure 1.

Regulation mechanisms of PGC1α in cancer. Oncogenes such as MIFT, P53 enhance the transcriptional activity of PGC1α, whereas MYC and HIF suppress it. PGC1α can also be activated by posttranslational modifications. It is deacetylated by PML or SIRT1, and is phosphorylated by AMPK in cancer cells.

Figure 1.

Regulation mechanisms of PGC1α in cancer. Oncogenes such as MIFT, P53 enhance the transcriptional activity of PGC1α, whereas MYC and HIF suppress it. PGC1α can also be activated by posttranslational modifications. It is deacetylated by PML or SIRT1, and is phosphorylated by AMPK in cancer cells.

Close modal
Table 1.

A summary of PGC1α-dependent signaling in cancer

Signaling axisCancer typeMetabolic pathwaysFunctionReferences
MITF/PGC1α Melanoma OxPhos; ROS clearance Resistance to BRAF or MEK inhibitors (53, 57, 68) 
PGC1α/ERRα Breast cancer OxPhos; Glycolysis Support anchorage independent growth (70) 
PGC1α/ERRα ERBB2/Neu-induced breast cancer Glutamine-derived lipogenesis Promote cell growth in hypoxia (51) 
PML/PGC1α/PPARα Breast cancer FAO Promote cell growth and luminal filling (58) 
Androgen/AR/AMPK/PGC1α Prostate cancer OxPhos; Glucose and FAO Promote cell growth (52, 93, 94) 
p53/PGC1α Lung and liver cancer OxPhos; ROS clearance Contribute to p53 target selection; Resistance to transient starvation (96) 
SIRT1/PGC1α/Nrf2 Breast, ovarian, colon, and lung cancer ROS clearance A target axis of miR-34 and metformin (102) 
RIP1/PGC1α Lung cancer OxPhos; Glycolysis Maintain DNA integrity and promote cell growth (98) 
PGC1α/ACLY-FASN Colon and liver cancer Glucose-derived lipogenesis Promote tumor growth (49) 
MYC/PGC1αa PDAC OxPhos; glycolysis Resistance to metformin (56) 
HIF/Dec1/PGC1αa ccRCC OxPhos; ROS Promote tumor growth (55) 
Signaling axisCancer typeMetabolic pathwaysFunctionReferences
MITF/PGC1α Melanoma OxPhos; ROS clearance Resistance to BRAF or MEK inhibitors (53, 57, 68) 
PGC1α/ERRα Breast cancer OxPhos; Glycolysis Support anchorage independent growth (70) 
PGC1α/ERRα ERBB2/Neu-induced breast cancer Glutamine-derived lipogenesis Promote cell growth in hypoxia (51) 
PML/PGC1α/PPARα Breast cancer FAO Promote cell growth and luminal filling (58) 
Androgen/AR/AMPK/PGC1α Prostate cancer OxPhos; Glucose and FAO Promote cell growth (52, 93, 94) 
p53/PGC1α Lung and liver cancer OxPhos; ROS clearance Contribute to p53 target selection; Resistance to transient starvation (96) 
SIRT1/PGC1α/Nrf2 Breast, ovarian, colon, and lung cancer ROS clearance A target axis of miR-34 and metformin (102) 
RIP1/PGC1α Lung cancer OxPhos; Glycolysis Maintain DNA integrity and promote cell growth (98) 
PGC1α/ACLY-FASN Colon and liver cancer Glucose-derived lipogenesis Promote tumor growth (49) 
MYC/PGC1αa PDAC OxPhos; glycolysis Resistance to metformin (56) 
HIF/Dec1/PGC1αa ccRCC OxPhos; ROS Promote tumor growth (55) 

aPGC1α is suppressed by the oncogene or signaling pathway in this context.

Figure 2.

Biologic function of PGC1α in cancer cells. Once induced by stress, PGC1α interacts with specific transcription factors and promotes mitochondrial respiration, fatty acid metabolism, or ROS detoxification, which facilitates cancer metabolic adaptation.

Figure 2.

Biologic function of PGC1α in cancer cells. Once induced by stress, PGC1α interacts with specific transcription factors and promotes mitochondrial respiration, fatty acid metabolism, or ROS detoxification, which facilitates cancer metabolic adaptation.

Close modal

MITF/PGC1α Axis in Melanoma

Among all its functions, PGC1α-dependent regulation of OxPhos is best studied in cancer, especially in melanoma. On the basis of PGC1α expression levels, melanomas have been defined into two subsets with different biologic phenotypes (53, 59, 60). The PGC1α-positive cells exhibit elevated mitochondrial oxidative metabolism and substantial ROS detoxifying capacities. In contrast, the PGC1α-negative cells are dependent on glycolysis for survival and are more sensitive to ROS-induced apoptosis (53). In this context, PGC1α is transactivated by the oncogenic melanocyte lineage-specification transcription factor (MITF), and the PGC1α-negative cells seldom express MITF (57, 61). These findings indicate that melanomas classified by the expression of MITF/PGC1α have different metabolic capacities, resulting in distinct destinies following ROS-inducing treatments (53, 57).

The V600E BRAF mutation plays a critical role in melanomagenesis by constitutively activating the MAPK signaling pathways (62, 63). Although the BRAF inhibitor, vemurafenib, and the MEK inhibitor, selumetinib, have achieved superior clinical effects to treat BRAF V600E–positive individuals, de novo and acquired resistance are still prevalent (64–67). PGC1α and MITF are induced in both melanoma cell lines and patient biopsies following BRAF or MEK inhibition (57). MITF/PGC1α signaling promotes OxPhos and protects drug-resistant cells from BRAF inhibitor–induced apoptosis. Encouragingly, a genome-wide siRNA screen and mRNA expression profile of resistant melanoma cell lines showed that resistance can be abrogated by inhibiting mTORC1/2 (68). Inhibition of mTORC1/2 decreases MITF nuclear localization and PGC1α expression. The combination inhibition of MEK and mTORC1/2 markedly reduces xenograft tumor proliferation in MEK inhibitor–resistant mice (Fig. 3). This finding supports the idea that PGC1α could be a candidate biomarker to determine therapeutic strategies. At the very least, blocking mitochondria respiration genes or mTORC1/2 signaling could resensitize cancer cells to BRAF or MEK inhibition in melanomas harboring activated MITF/PGC1α/OxPhos signaling.

Figure 3.

Involvement of the MITF/PGC1α signaling in melanoma therapy. A, mechanism of the MITF/PGC1α signaling axis in melanoma therapeutic resistance. MITF can be activated by BRAF or MEK inhibition and then transactivate PGC1α, which promotes mitochondrial respiration and ROS detoxification, leading to drug tolerance of BRAF/MEK inhibition. B, combinatory suppression of BRAF/MEK and mTORC1/2 contributes to melanoma cell death. mTORC1/2 inhibition can decrease nuclear localization of MITF, then suppress PGC1α expression and mitochondrial respiration, which resensitizes melanoma to BRAF/MEK inhibition.

Figure 3.

Involvement of the MITF/PGC1α signaling in melanoma therapy. A, mechanism of the MITF/PGC1α signaling axis in melanoma therapeutic resistance. MITF can be activated by BRAF or MEK inhibition and then transactivate PGC1α, which promotes mitochondrial respiration and ROS detoxification, leading to drug tolerance of BRAF/MEK inhibition. B, combinatory suppression of BRAF/MEK and mTORC1/2 contributes to melanoma cell death. mTORC1/2 inhibition can decrease nuclear localization of MITF, then suppress PGC1α expression and mitochondrial respiration, which resensitizes melanoma to BRAF/MEK inhibition.

Close modal

PGC1α/ERRα axis in breast cancer

ERRα is a ligand-independent NR that regulates a number of metabolic genes in the presence of a coactivator, such as PGC1α (69). PGC1α and ERRα are both downstream proteins of the kinase suppressor of RAS1 (KSR1), a molecular supporting Ras-induced transformation in breast cancer (70). The interaction of PGC1α and ERRα is required to promote OxPhos and glycolysis in an H-RasV12/KSR1-dependent manner to support anchorage-independent growth. Besides glucose use, glutamine utilization is also a characteristic feature of cancer metabolism (71), and the PGC1α/ERRα axis was reported to regulate glutaminolysis in the ErbB2/Neu–induced breast cancer model (51). This axis increases the expression of glutamine metabolism genes and manipulates glutamine-derived carbon flux into lipogenesis, which favors the proliferation of breast cancer cells in hypoxia. On the other hand, the axis was also reported to increase angiogenesis by promoting VEGF secretion in the same model (72). Indeed, the hypoxia-inducible factor 1 (HIF1)-independent regulation of VEGF by PGC1α/ERRα signaling was already reported in muscle cells under nutrient-deprivation conditions (73). These observations indicated that this signaling axis is responsive to microenvironmental stress and can be a promoter of cancer metastasis.

Another group demonstrated that PGC1α and ERRα are overexpressed in brain metastatic breast cancer cells, with enhanced mitochondria respiration, FAO, glycolysis, and elimination of ROS (74). These changes give brain metastatic cells a considerable growth advantage, which is accompanied by resistance to therapeutic drugs. Recently, a study also confirmed that PGC1α expression is markedly upregulated in circulating cancer cells (CCC) in a breast cancer metastatic mouse model (75). These cells relied on PGC1α to maintain mitochondria activity during their transition to target organs. Clinical analysis in this research also indicated that PGC1α is enriched in invasive ductal breast cancer patients with bone marrow dissemination, and illustrated a significant negative correlation between PGC1α expression and patient outcome. Even though this study did not identify the target transcription factor of PGC1α in CCCs, the data illustrated that PGC1α is essential in cancer metastasis.

Overall, the PGC1α/ERRα signaling axis is responsible for altering cellular metabolic status and guarantees cancer cell survival under limited nutrient supply and high-energy demanding microenvironments (76). Disruption of PGC1α, ERRα, or OxPhos genes could abrogate these malignant properties of cancer providing valuable strategies to achieve better, more effective, therapeutic effects.

MYC/PGC1α axis in pancreatic ductal adenocarcinoma

MYC-driven metabolic reprogramming is critical for cancer cell survival and proliferation (77, 78). Most recently, MYC has been confirmed to be a direct regulator of PGC1α in pancreatic ductal adenocarcinoma (PDAC; ref. 56). It binds to the promoter of PGC1α and has an inhibitory effect on PGC1α at transcriptional level. The MYC/PGC1α ratio was reported to be a main controller of metabolic phenotype in PDAC cells. In this study, high PGC1α expression was found in pancreatic cancer stem cells, which was allowed by MYC suppression (56). Although the MYClowPGC1αhigh status is essential to maintain mitochondrial respiration, stemness, and tumorigenicity of pancreatic CSCs, it also makes CSCs more vulnerable to metformin treatment than more differentiated PDAC cells. With low MYC expression, CSCs are unable to activate glycolysis under metabolic stress (56). Even so, a subset of metformin-resistant pancreatic CSCs can emerge during treatment. The resistant cells display an intermediate metabolic phenotype with reduced OxPhos but enhanced glycolysis, as a result of increased MYC/PGC1α ratio, compared with metformin-sensitive CSCs (56), and MYC inhibition increases the sensitivity to metformin in resistant CSCs. These data indicated MYC/PGC1α balance determines the metabolic plasticity and sensitivity to metformin of pancreatic CSCs.

Another research study performed in a mouse model of pancreatic tumor also highlighted the importance of PGC1α in PDAC therapeutic resistance. Tumor cells which survive the genetic or pharmacologic ablation of KRAS, overexpress PGC1α (79). Although PGC1α expression is closely related to therapeutic resistance of PDAC in both models, mechanisms of how PGC1α influences tumor relapse are different. The former study emphasized the regulation of MYC on PGC1α in a KRas-independent manner (56). It is the increased MYC/PGC1α ratio reduces the reliance of CSCs on OxPhos, leading to metformin resistance. While, in KRas ablation-resistant mouse model, it is the strong reliance on OxPhos ensures resistant cells survival (79), and this phenomenon might be restricted to KRas-ablated dormant pancreatic tumor cells.

PML/PGC1α/PPARα axis in breast cancer

Even though mitochondrial respiration has been considered as the main biologic function of PGC1α, the crucial role of PGC1α in regulating FAO has received more attention in recent years. FAO is activated in several type of cancers, and the transcription factor, PPARα, is the main regulator of this process (80–83). PPARα can be activated by endogenous fatty acid, fatty acid derivatives or fibrates, and is coregulated by PGC1α (84, 85). In breast cancer cells, PGC1α is controlled by the promyelocytic leukemia (PML) gene which is enhanced in a subset of breast cancers and especially enriched in triple-negative cases (58, 86). PGC1α is deacetylated by PML, leading to transactivation of PPARα and FAO activation. The PML/PGC1α/PPARα/FAO signaling pathway provides ATP to promote cell survival and luminal filling in breast cancer (58). Furthermore, this pathway is also involved in hematopoietic stem cell maintenance (87, 88). Although only a small number of studies reported the involvement of PGC1α in cancer stem cells, the metabolic profile of hematopoietic stem cells and cancer stem cells could have some common traits in stemness maintenance (79, 89–91). These studies provided evidence indicating that PML/PGC1α/PPARα signaling is essential for maintaining cellular metabolic homeostasis with potential therapeutic implications.

AR/PGC1α axis in prostate cancer

The androgen receptor (AR) is a ligand-activated transcription factor that is substantially modulated by coregulators (92). Among the coactivators, PGC1α interacts with AR to orchestrate central metabolism and cell survival in prostate cancer (93). PGC1α can bind to the N-terminal domain of the AR and promote the formation of AR homodimer, which increases the transcription of AR target genes, such as PSA (52). Inhibition of PGC1α causes cell-cycle arrest at the G1-phase and suppresses growth of either AR-positive or castration-resistant prostate cancer cells. These findings indicated that disruption of PGC1α expression could be useful for treating AR-positive prostate cancer and also might be more efficient in castration-resistant prostate cancer, which depends more on AR signaling (52).

More than a coactivator of AR, PGC1α can be simultaneously regulated by AR signaling. Both mRNA and protein levels of PGC1α can be induced by androgens in prostate cancer in an AMPK-dependent manner (94). The androgen/AR/AMPK/PGC1α signaling axis increases mitochondria biogenesis, glucose oxidation, and FAO to generate building blocks and ATP for cancer cell growth. The positive feedback regulation provides a great advantage to sustain PGC1α expression and augment its influence on cancer metabolism.

p53/PGC1α axis in cell fate determination

P53 can either be a tumor suppressor or an oncogene in cancer. Its biologic function is tightly regulated by translational modifications and interaction with other proteins (95). PGC1α acts as a contributor to wild-type p53 target selection in cancer cells in response to glucose deprivation (96). When PGC1α is induced by transient starvation and binds to p53, Lys120 acetylation of p53 is prevented, and p53 preferentially transactivates genes associated with ROS clearance and mitochondrial metabolism. However, in prolonged or chronic starvation, PGC1α undergoes degradation by ring finger protein 2-mediated ubiquitin–proteasome pathway. This process facilitates Lys120 acetylation of p53, leading to p53-dependent apoptosis (96). In this case, p53 has both cytoprotective and cytotoxic functions under nutrient-limited conditions. PGC1α performs as an essential switch in modulating stress-dependent transcription of p53, and directs the stress response of p53 toward prosurviving outcomes (97).

In addition to the interaction of PGC1α and wild-type p53, other studies suggested that PGC1α expression can be influenced by wide-type p53. Gene expression analysis in 28 human lung adenocarcinoma cell lines with different p53 mutational status showed that the mRNA level of PGC1α is higher in p53 wild-type cell lines compared with cell lines with p53 loss or missense mutation (50). Suppression of PGC1α inhibits the growth of p53 wild-type lung adenocarcinoma cells, implicating that PGC1α might be a potential target of wild-type p53 in lung adenocarcinoma, and the direct transcription regulation of wild-type p53 on PGC1α has been demonstrated in neuroblastoma cells and myoblasts (54). Similar to the AR/PGC1α axis, an autoregulatory loop might also exist between p53 and PGC1α.

As well as the direct regulatory mechanism between PGC1α and p53, the receptor-interacting protein 1 (RIP1)/PGC1α signaling axis indirectly affects the control of p53-dependent cell proliferation (98). This signaling pathway maintains metabolic homeostasis in lung cancer cells. Loss of RIP1 suppresses PGC1α expression and OxPhos, resulting in accelerated glycolysis. However, excessive glycolysis decreases cellular NAD+ levels and impairs DNA repair, which activates p53-mediated cell growth inhibition (98, 99). These data provide additional evidence supporting a reciprocal regulation between metabolic adaptation and wild-type p53 in cancer cell fate determination.

SIRT1/PGC1α axis in stress adaptation

SIRT1 is the main regulator of PGC1α and activates PGC1α through the deacetylation of PGC1α at specific lysine residues (20). SIRT1/PGC1α signaling is required to transactivate FAO genes under nutrient restriction in skeletal muscle cells (100). The switch from glucose to fatty acid utilization benefits cell survival under stress conditions, and SIRT1/PGC1α signaling also mediates Nrf2 expression to antagonize oxidative damage (101). It was identified as a targeted pathway of metformin in p53 wild-type cancer cells (102). Metformin suppresses this signaling through miR-34a in a p53-dependent manner and then sensitizes cancer cells to oxidative stress. These research findings provide new evidence for the effectiveness of metformin in cancer therapy (103).

As noted above, cells with high expression of PGC1α have an advantage to survive under metabolic stresses, including oxidative damage, energy deprivation, and even cancer therapy. The therapeutic resistant cancer cells especially have a strong reliance on metabolic activities mediated by PGC1α. On the basis of the evidence, PGC1α is believed to have an emerging role in cancer therapeutic resistance (57, 68, 75).

Whereas specific inhibitors of PGC1α are not yet available, the major methods to disrupt PGC1α signaling have been focused on suppressing key enzymes of PGC1α-dependent metabolic pathways or targeting-related transcription factors. OxPhos, FAO, or ERRα inhibitors were all observed to reduce cell growth in PGC1α-positive cancer cells (51, 68, 75, 87).

Metabolic intervention significantly limits adaptive responses in cancer therapy as discussed above. However, suppressing a single metabolic node might not be fatal to certain cancer cells, because alternative metabolic paths still could be engaged to compensate for the deficiencies. In melanoma, suppressing PGC1α expression or OxPhos resensitize therapeutic-resistant cells to oxidative damage (53). However, these treatments simultaneously activate alternate metabolic fluxes in a small set of cancer cells (104). The increased ROS production caused by PGC1α inhibition stabilizes HIF1α protein, and HIF1α induces a metabolic switch from OxPhos to glycolysis which promotes cell survival (105). Surprisingly, even a combinatory suppression of PGC1α and HIF1α could not eradicate tumors in mice. Instead, this process promotes glutamine utilization to offset the energy crisis. Only blocking all of these pathways can maximize tumor-suppressing efficiency. Another study also indicated that robust mitochondria respiration activity strongly relied on autophagy and FAO, which provide nutrients to mitochondria (79). Suppression of the compensatory fluxes could greatly increase the sensitivity of melanoma cells to OxPhos inhibition (79, 105). These data highlight the metabolic flexibility of cancer and emphasize the demand for a multitargeted therapeutic strategy to reduce or avert the treatment resistance.

In contrast to most studies, a few studies reported that PGC1α was a tumor suppressor in some cancer types (55, 106–109). For instance, low expression of PGC1α is associated with poor survival in VHL-deficient clear cell renal cell carcinoma (ccRCC) and breast cancer (55, 110). In VHL-deficient ccRCC, PGC1α is suppressed by HIF-dependent activation of transcriptional repressor Dec1 (55). Overexpression of PGC1α protects mouse intestinal epithelium against colon cancer formation (111). Mechanically, induction of cell death is the major cause associated with PGC1α-mediated tumor suppression. PGC1α acts as a stabilizer of a tumor suppressor mitostatin, which triggers mitophagy in breast cancer (112). It also enhances Bax-mediated apoptosis in colorectal and ovarian epithelial carcinoma cells (109, 113).

The paradoxical role of PGC1α in cancer is caused by several factors. As PGC1α is highly sensitive to environmental stimulation, thus methods include gene interference and the discrepancy between in vitro cell culture and tumor microenvironments technically can influence the expression of PGC1α (11). As a transcriptional coactivator, the tissue specificity of corresponding transcription factors can significantly contribute to the variability of PGC1α expression (35). The diverse upstream signals of PGC1α also correspond to its dynamic role in tumor development based on activation by different oncogenes and changes in the metabolic mode.

According to the abovementioned discussion, PGC1α facilitates a flexible metabolic profile in cancer cells and contributes to cancer survival and therapeutic resistance. (15, 114, 115). Even though mitochondrial respiration is identified as a predominant biologic function of PGC1α in cancer, a few studies also underscore PGC1α as a promoter of FAO and glucose- or glutamine-derived lipogenesis in recent years (49, 51, 58). The discoveries indicated that PGC1α can not only enhance ATP production, but also affect carbon flux. On the basis of a deeper understanding of FAO and autophagy in cancer therapy, more attention should be paid on the role of PGC1α in the regulation of lipid metabolism in the future (116–119).

Despite PGC1α can interact with various transcription factors under physiologic conditions, research studies focusing on PGC1α-targeted transcription factors in cancer cells are still limited, and mechanisms of the interactions are also elusive. Moreover, except for PML and MYC, few oncogenes have been reported conclusively to regulate PGC1α (56, 58). Whether changes in PGC1α expression are due to oncogene control or a nongenetic metabolic adaptation might depend on cellular context. Elucidating these issues would be very helpful in understanding the function of PGC1α in cancer.

For the reason that certain chemotherapeutically resistant cancer cells are extremely addicted to PGC1α-dependent metabolic activities, targeting PGC1α-associated pathways could be a desirable means to resensitize cancer cell to therapy. However, even more than chemotherapy, radiotherapy is also a necessary strategy against some tumor types. The relationship between radiotherapy and PGC1α needs to be further studied. According to the evidence discussed earlier, disruption of PGC1α target genes and the potential compensatory pathways, combined with traditional chemotherapy/radiotherapy should be the best strategies to achieve maximized therapeutic efficiency in the future.

No potential conflicts of interest were disclosed.

Y. Cao is a recipient of the National Basic Research Program of China, grant no. 2011CB504305 and the National High Technology Research and Development Program of China, grant no. 2012AA02A501. X.J. Luo is a recipient of the National Science Foundation of China, grant no. 81573014. M. Tang is a recipient of the National Science Foundation of China, grant no. 81402250.

1.
Boroughs
LK
,
DeBerardinis
RJ
. 
Metabolic pathways promoting cancer cell survival and growth
.
Nat Cell Biol
2015
;
17
:
351
9
.
2.
Ward
PS
,
Thompson
CB
. 
Metabolic reprogramming: a cancer hallmark even Warburg did not anticipate
.
Cancer Cell
2012
;
21
:
297
308
.
3.
Hensley
CT
,
Wasti
AT
,
DeBerardinis
RJ
. 
Glutamine and cancer: cell biology, physiology, and clinical opportunities
.
J Clin Invest
2013
;
123
:
3678
84
.
4.
Currie
E
,
Schulze
A
,
Zechner
R
,
Walther
TC
,
Farese
RV
 Jr
Cellular fatty acid metabolism and cancer
.
Cell Metab
2013
;
18
:
153
61
.
5.
Locasale
JW
. 
Serine, glycine and one-carbon units: cancer metabolism in full circle
.
Nat Rev Cancer
2013
;
13
:
572
83
.
6.
Weinberg
SE
,
Chandel
NS
. 
Targeting mitochondria metabolism for cancer therapy
.
Nat Chem Biol
2015
;
11
:
9
15
.
7.
Seyfried
TN
,
Flores
RE
,
Poff
AM
,
D'Agostino
DP
. 
Cancer as a metabolic disease: implications for novel therapeutics
.
Carcinogenesis
2014
;
35
:
515
27
.
8.
Bobrovnikova-Marjon
E
,
Hurov
JB
. 
Targeting metabolic changes in cancer: novel therapeutic approaches
.
Annu Rev Med
2014
;
65
:
157
70
.
9.
Staubert
C
,
Bhuiyan
H
,
Lindahl
A
,
Broom
OJ
,
Zhu
Y
,
Islam
S
, et al
Rewired metabolism in drug-resistant leukemia cells: a metabolic switch hallmarked by reduced dependence on exogenous glutamine
.
J Biol Chem
2015
;
290
:
8348
59
.
10.
Jones
AW
,
Yao
Z
,
Vicencio
JM
,
Karkucinska-Wieckowska
A
,
Szabadkai
G
. 
PGC-1 family coactivators and cell fate: roles in cancer, neurodegeneration, cardiovascular disease and retrograde mitochondria-nucleus signalling
.
Mitochondrion
2012
;
12
:
86
99
.
11.
Girnun
GD
. 
The diverse role of the PPARgamma coactivator 1 family of transcriptional coactivators in cancer
.
Semin Cell Dev Biol
2012
;
23
:
381
8
.
12.
Finck
BN
,
Kelly
DP
. 
PGC-1 coactivators: inducible regulators of energy metabolism in health and disease
.
J Clin Invest
2006
;
116
:
615
22
.
13.
Handschin
C
,
Spiegelman
BM
. 
The role of exercise and PGC1alpha in inflammation and chronic disease
.
Nature
2008
;
454
:
463
9
.
14.
Liu
C
,
Lin
JD
. 
PGC-1 coactivators in the control of energy metabolism
.
Acta Biochim Biophys Sin
2011
;
43
:
248
57
.
15.
Lin
J
,
Handschin
C
,
Spiegelman
BM
. 
Metabolic control through the PGC-1 family of transcription coactivators
.
Cell Metab
2005
;
1
:
361
70
.
16.
Lin
J
,
Wu
H
,
Tarr
PT
,
Zhang
CY
,
Wu
Z
,
Boss
O
, et al
Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres
.
Nature
2002
;
418
:
797
801
.
17.
Handschin
C
,
Rhee
J
,
Lin
J
,
Tarr
PT
,
Spiegelman
BM
. 
An autoregulatory loop controls peroxisome proliferator-activated receptor gamma coactivator 1alpha expression in muscle
.
Proc Natl Acad Sci U S A
2003
;
100
:
7111
6
.
18.
Patti
ME
,
Butte
AJ
,
Crunkhorn
S
,
Cusi
K
,
Berria
R
,
Kashyap
S
, et al
Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: Potential role of PGC1 and NRF1
.
Proc Natl Acad Sci USA
2003
;
100
:
8466
71
.
19.
Mootha
VK
,
Handschin
C
,
Arlow
D
,
Xie
X
,
St Pierre
J
,
Sihag
S
, et al
Erralpha and Gabpa/b specify PGC-1alpha-dependent oxidative phosphorylation gene expression that is altered in diabetic muscle
.
Proc Natl Acad Sci USA
2004
;
101
:
6570
5
.
20.
Nemoto
S
,
Fergusson
MM
,
Finkel
T
. 
SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1(alpha)
.
J Biol Chem
2005
;
280
:
16456
60
.
21.
Amat
R
,
Planavila
A
,
Chen
SL
,
Iglesias
R
,
Giralt
M
,
Villarroya
F
. 
SIRT1 controls the transcription of the peroxisome proliferator-activated receptor-gamma Co-activator-1alpha (PGC-1alpha) gene in skeletal muscle through the PGC-1alpha autoregulatory loop and interaction with MyoD
.
J Biol Chem
2009
;
284
:
21872
80
.
22.
Haemmerle
G
,
Moustafa
T
,
Woelkart
G
,
Buttner
S
,
Schmidt
A
,
van de Weijer
T
, et al
ATGL-mediated fat catabolism regulates cardiac mitochondrial function via PPAR-alpha and PGC-1
.
Nat Med
2011
;
17
:
1076
85
.
23.
Khan
SA
,
Sathyanarayan
A
,
Mashek
MT
,
Ong
KT
,
Wollaston-Hayden
EE
,
Mashek
DG
. 
ATGL-catalyzed lipolysis regulates SIRT1 to control PGC-1alpha/PPAR-alpha signaling
.
Diabetes
2015
;
64
:
418
26
.
24.
Louet
JF
,
Hayhurst
G
,
Gonzalez
FJ
,
Girard
J
,
Decaux
JF
. 
The coactivator PGC-1 is involved in the regulation of the liver carnitine palmitoyltransferase I gene expression by cAMP in combination with HNF4 alpha and cAMP-response element-binding protein (CREB)
.
J Biol Chem
2002
;
277
:
37991
8000
.
25.
Zhang
Y
,
Ma
K
,
Song
S
,
Elam
MB
,
Cook
GA
,
Park
EA
. 
Peroxisomal proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1 alpha) enhances the thyroid hormone induction of carnitine palmitoyltransferase I (CPT-I alpha)
.
J Biol Chem
2004
;
279
:
53963
71
.
26.
La Spada
AR
. 
PPARGC1A/PGC-1alpha, TFEB and enhanced proteostasis in Huntington disease: defining regulatory linkages between energy production and protein-organelle quality control
.
Autophagy
2012
;
8
:
1845
7
.
27.
Tsunemi
T
,
Ashe
TD
,
Morrison
BE
,
Soriano
KR
,
Au
J
,
Roque
RA
, et al
PGC-1alpha rescues Huntington's disease proteotoxicity by preventing oxidative stress and promoting TFEB function
.
Sci Transl Med
2012
;
4
:
142ra97
.
28.
Wu
Z
,
Puigserver
P
,
Andersson
U
,
Zhang
C
,
Adelmant
G
,
Mootha
V
, et al
Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1
.
Cell
1999
;
98
:
115
24
.
29.
Puigserver
P
,
Wu
Z
,
Park
CW
,
Graves
R
,
Wright
M
,
Spiegelman
BM
. 
A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis
.
Cell
1998
;
92
:
829
39
.
30.
Bostrom
P
,
Wu
J
,
Jedrychowski
MP
,
Korde
A
,
Ye
L
,
Lo
JC
, et al
A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis
.
Nature
2012
;
481
:
463
8
.
31.
Olmos
Y
,
Valle
I
,
Borniquel
S
,
Tierrez
A
,
Soria
E
,
Lamas
S
, et al
Mutual dependence of Foxo3a and PGC-1alpha in the induction of oxidative stress genes
.
J Biol Chem
2009
;
284
:
14476
84
.
32.
Abdanipour
A
,
Tiraihi
T
,
Noori-Zadeh
A
,
Majdi
A
,
Gosaili
R
. 
Evaluation of lovastatin effects on expression of anti-apoptotic Nrf2 and PGC-1alpha genes in neural stem cells treated with hydrogen peroxide
.
Mol Neurobiol
2014
;
49
:
1364
72
.
33.
St-Pierre
J
,
Drori
S
,
Uldry
M
,
Silvaggi
JM
,
Rhee
J
,
Jager
S
, et al
Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators
.
Cell
2006
;
127
:
397
408
.
34.
Austin
S
,
St-Pierre
J
. 
PGC1alpha and mitochondrial metabolism–emerging concepts and relevance in ageing and neurodegenerative disorders
.
J Cell Sci
2012
;
125
:
4963
71
.
35.
Puigserver
P.
Tissue-specific regulation of metabolic pathways through the transcriptional coactivator PGC1-alpha
.
Int J Obes
2005
;
29
:
S5
9
.
36.
Wolf
DA.
Is reliance on mitochondrial respiration a "chink in the armor" of therapy-resistant cancer?
Cancer Cell
2014
;
26
:
788
95
.
37.
Puigserver
P
,
Adelmant
G
,
Wu
Z
,
Fan
M
,
Xu
J
,
O'Malley
B
, et al
Activation of PPARgamma coactivator-1 through transcription factor docking
.
Science
1999
;
286
:
1368
71
.
38.
Puigserver
P
,
Rhee
J
,
Donovan
J
,
Walkey
CJ
,
Yoon
JC
,
Oriente
F
, et al
Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction
.
Nature
2003
;
423
:
550
5
.
39.
Knutti
D
,
Kaul
A
,
Kralli
A
. 
A tissue-specific coactivator of steroid receptors, identified in a functional genetic screen
.
Mol Cell Biol
2000
;
20
:
2411
22
.
40.
Wallberg
AE
,
Yamamura
S
,
Malik
S
,
Spiegelman
BM
,
Roeder
RG
. 
Coordination of p300-mediated chromatin remodeling and TRAP/mediator function through coactivator PGC-1alpha
.
Mol Cell
2003
;
12
:
1137
49
.
41.
Monsalve
M
,
Wu
Z
,
Adelmant
G
,
Puigserver
P
,
Fan
M
,
Spiegelman
BM
. 
Direct coupling of transcription and mRNA processing through the thermogenic coactivator PGC-1
.
Mol Cell
2000
;
6
:
307
16
.
42.
Jager
S
,
Handschin
C
,
St-Pierre
J
,
Spiegelman
BM
. 
AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha
.
Proc Natl Acad Sci U S A
2007
;
104
:
12017
22
.
43.
Rodgers
JT
,
Lerin
C
,
Haas
W
,
Gygi
SP
,
Spiegelman
BM
,
Puigserver
P
. 
Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1
.
Nature
2005
;
434
:
113
8
.
44.
Knutti
D
,
Kressler
D
,
Kralli
A
. 
Regulation of the transcriptional coactivator PGC-1 via MAPK-sensitive interaction with a repressor
.
Proc Natl Acad Sci U S A
2001
;
98
:
9713
8
.
45.
Li
X
,
Monks
B
,
Ge
Q
,
Birnbaum
MJ
. 
Akt/PKB regulates hepatic metabolism by directly inhibiting PGC-1alpha transcription coactivator
.
Nature
2007
;
447
:
1012
6
.
46.
Rytinki
MM
,
Palvimo
JJ
. 
SUMOylation attenuates the function of PGC-1alpha
.
J Biol Chem
2009
;
284
:
26184
93
.
47.
Herzig
S
,
Long
F
,
Jhala
US
,
Hedrick
S
,
Quinn
R
,
Bauer
A
, et al
CREB regulates hepatic gluconeogenesis through the coactivator PGC-1
.
Nature
2001
;
413
:
179
83
.
48.
Bianchi
K
,
Vandecasteele
G
,
Carli
C
,
Romagnoli
A
,
Szabadkai
G
,
Rizzuto
R
. 
Regulation of Ca2+ signalling and Ca2+-mediated cell death by the transcriptional coactivator PGC-1alpha
.
Cell Death Differ
2006
;
13
:
586
96
.
49.
Bhalla
K
,
Hwang
BJ
,
Dewi
RE
,
Ou
L
,
Twaddel
W
,
Fang
HB
, et al
PGC1alpha promotes tumor growth by inducing gene expression programs supporting lipogenesis
.
Cancer Res
2011
;
71
:
6888
98
.
50.
Taguchi
A
,
Delgado
O
,
Celiktas
M
,
Katayama
H
,
Wang
H
,
Gazdar
AF
, et al
Proteomic signatures associated with p53 mutational status in lung adenocarcinoma
.
Proteomics
2014
;
14
:
2750
9
.
51.
McGuirk
S
,
Gravel
SP
,
Deblois
G
,
Papadopoli
DJ
,
Faubert
B
,
Wegner
A
, et al
PGC-1alpha supports glutamine metabolism in breast cancer
.
Cancer Metab
2013
;
1
:
22
.
52.
Shiota
M
,
Yokomizo
A
,
Tada
Y
,
Inokuchi
J
,
Tatsugami
K
,
Kuroiwa
K
, et al
Peroxisome proliferator-activated receptor gamma coactivator-1alpha interacts with the androgen receptor (AR) and promotes prostate cancer cell growth by activating the AR
.
Mol Endocrinol
2010
;
24
:
114
27
.
53.
Vazquez
F
,
Lim
JH
,
Chim
H
,
Bhalla
K
,
Girnun
G
,
Pierce
K
, et al
PGC1alpha expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress
.
Cancer Cell
2013
;
23
:
287
301
.
54.
Aquilano
K
,
Baldelli
S
,
Pagliei
B
,
Cannata
SM
,
Rotilio
G
,
Ciriolo
MR
. 
p53 orchestrates the PGC-1alpha-mediated antioxidant response upon mild redox and metabolic imbalance
.
Antioxid Redox Signal
2013
;
18
:
386
99
.
55.
LaGory
EL
,
Wu
C
,
Taniguchi
CM
,
Ding
CK
,
Chi
JT
,
von Eyben
R
, et al
Suppression of PGC-1α is critical for reprogramming oxidative metabolism in renal cell carcinoma
.
Cell Rep
2015
;
12
:
116
27
.
56.
Sancho
P
,
Burgos-Ramos
E
,
Tavera
A
,
Bou Kheir
T
,
Jagust
P
,
Schoenhals
M
, et al
MYC/PGC-1α balance determines the metabolic phenotype and plasticity of pancreatic cancer stem cells
.
Cell Metab
2015
;
22
:
590
605
.
57.
Haq
R
,
Shoag
J
,
Andreu-Perez
P
,
Yokoyama
S
,
Edelman
H
,
Rowe
GC
, et al
Oncogenic BRAF regulates oxidative metabolism via PGC1alpha and MITF
.
Cancer Cell
2013
;
23
:
302
15
.
58.
Carracedo
A
,
Weiss
D
,
Leliaert
AK
,
Bhasin
M
,
de Boer
VC
,
Laurent
G
, et al
A metabolic prosurvival role for PML in breast cancer
.
J Clin Invest
2012
;
122
:
3088
100
.
59.
Riker
AI
,
Enkemann
SA
,
Fodstad
O
,
Liu
S
,
Ren
S
,
Morris
C
, et al
The gene expression profiles of primary and metastatic melanoma yields a transition point of tumor progression and metastasis
.
BMC Med Genomics
2008
;
1
:
13
.
60.
Bogunovic
D
,
O'Neill
DW
,
Belitskaya-Levy
I
,
Vacic
V
,
Yu
YL
,
Adams
S
, et al
Immune profile and mitotic index of metastatic melanoma lesions enhance clinical staging in predicting patient survival
.
Proc Natl Acad Sci U S A
2009
;
106
:
20429
34
.
61.
Ronai
Z.
The masters talk: the PGC-1alpha-MITF axis as a melanoma energizer
.
Pigment Cell Melanoma Res
2013
;
26
:
294
295
.
62.
Davies
H
,
Bignell
GR
,
Cox
C
,
Stephens
P
,
Edkins
S
,
Clegg
S
, et al
Mutations of the BRAF gene in human cancer
.
Nature
2002
;
417
:
949
54
.
63.
Wan
PT
,
Garnett
MJ
,
Roe
SM
,
Lee
S
,
Niculescu-Duvaz
D
,
Good
VM
, et al
Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF
.
Cell
2004
;
116
:
855
67
.
64.
Poulikakos
PI
,
Rosen
N
. 
Mutant BRAF melanomas–dependence and resistance
.
Cancer Cell
2011
;
19
:
11
5
.
65.
Hernandez-Davies
JE
,
Tran
TQ
,
Reid
MA
,
Rosales
KR
,
Lowman
XH
,
Pan
M
, et al
Vemurafenib resistance reprograms melanoma cells towards glutamine dependence
.
J Transl Med
2015
;
13
:
210
.
66.
Chapman
PB
,
Hauschild
A
,
Robert
C
,
Haanen
JB
,
Ascierto
P
,
Larkin
J
, et al
Improved survival with vemurafenib in melanoma with BRAF V600E mutation
.
N Engl J Med
2011
;
364
:
2507
16
.
67.
Flaherty
KT
,
Robert
C
,
Hersey
P
,
Nathan
P
,
Garbe
C
,
Milhem
M
, et al
Improved survival with MEK inhibition in BRAF-mutated melanoma
.
N Engl J Med
2012
;
367
:
107
14
.
68.
Gopal
YN
,
Rizos
H
,
Chen
G
,
Deng
W
,
Frederick
DT
,
Cooper
ZA
, et al
Inhibition of mTORC1/2 overcomes resistance to MAPK pathway inhibitors mediated by PGC1α and oxidative phosphorylation in melanoma
.
Cancer Res
2014
;
74
:
7037
47
.
69.
Huss
JM
,
Garbacz
WG
,
Xie
W
. 
Constitutive activities of estrogen-related receptors: transcriptional regulation of metabolism by the ERR pathways in health and disease
.
Biochim Biophys Acta
2015
;
1852
:
1912
27
.
70.
Fisher
KW
,
Das
B
,
Kortum
RL
,
Chaika
OV
,
Lewis
RE
. 
Kinase suppressor of ras 1 (KSR1) regulates PGC1alpha and estrogen-related receptor alpha to promote oncogenic Ras-dependent anchorage-independent growth
.
Mol Cell Biol
2011
;
31
:
2453
61
.
71.
Sun
RC
,
Denko
NC
. 
Hypoxic regulation of glutamine metabolism through HIF1 and SIAH2 supports lipid synthesis that is necessary for tumor growth
.
Cell Metab
2014
;
19
:
285
92
.
72.
Klimcakova
E
,
Chenard
V
,
McGuirk
S
,
Germain
D
,
Avizonis
D
,
Muller
WJ
, et al
PGC-1alpha promotes the growth of ErbB2/Neu-induced mammary tumors by regulating nutrient supply
.
Cancer Res
2012
;
72
:
1538
46
.
73.
Arany
Z
,
Foo
SY
,
Ma
Y
,
Ruas
JL
,
Bommi-Reddy
A
,
Girnun
G
, et al
HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1alpha
.
Nature
2008
;
451
:
1008
12
.
74.
Chen
EI
,
Hewel
J
,
Krueger
JS
,
Tiraby
C
,
Weber
MR
,
Kralli
A
, et al
Adaptation of energy metabolism in breast cancer brain metastases
.
Cancer Res
2007
;
67
:
1472
86
.
75.
LeBleu
VS
,
O'Connell
JT
,
Gonzalez Herrera
KN
,
Wikman
H
,
Pantel
K
,
Haigis
MC
, et al
PGC-1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis
.
Nat Cell Biol
2014
;
16
:
992
1003
,
1
15
.
76.
Deblois
G
,
St-Pierre
J
,
Giguere
V
. 
The PGC-1/ERR signaling axis in cancer
.
Oncogene
2013
;
32
:
3483
90
.
77.
Hsieh
AL
,
Walton
ZE
,
Altman
BJ
,
Stine
ZE
,
Dang
CV
. 
MYC and metabolism on the path to cancer
.
Semin Cell Dev Biol
2015
.
78.
Stine
ZE
,
Walton
ZE
,
Altman
BJ
,
Hsieh
AL
,
Dang
CV
. 
MYC, metabolism, and cancer
.
Cancer Discov
2015
;
5
:
1024
39
.
79.
Viale
A
,
Pettazzoni
P
,
Lyssiotis
CA
,
Ying
H
,
Sanchez
N
,
Marchesini
M
, et al
Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function
.
Nature
2014
;
514
:
628
32
.
80.
Samudio
I
,
Harmancey
R
,
Fiegl
M
,
Kantarjian
H
,
Konopleva
M
,
Korchin
B
, et al
Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction
.
J Clin Invest
2010
;
120
:
142
56
.
81.
Caro
P
,
Kishan
AU
,
Norberg
E
,
Stanley
IA
,
Chapuy
B
,
Ficarro
SB
, et al
Metabolic signatures uncover distinct targets in molecular subsets of diffuse large B cell lymphoma
.
Cancer Cell
2012
;
22
:
547
60
.
82.
Schlaepfer
IR
,
Rider
L
,
Rodrigues
LU
,
Gijon
MA
,
Pac
CT
,
Romero
L
, et al
Lipid catabolism via CPT1 as a therapeutic target for prostate cancer
.
Mol Cancer Ther
2014
;
13
:
2361
71
.
83.
Zaugg
K
,
Yao
Y
,
Reilly
PT
,
Kannan
K
,
Kiarash
R
,
Mason
J
, et al
Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress
.
Genes Dev
2011
;
25
:
1041
51
.
84.
Vega
RB
,
Huss
JM
,
Kelly
DP
. 
The coactivator PGC-1 cooperates with peroxisome proliferator-activated receptor alpha in transcriptional control of nuclear genes encoding mitochondrial fatty acid oxidation enzymes
.
Mol Cell Biol
2000
;
20
:
1868
76
.
85.
Peters
JM
,
Shah
YM
,
Gonzalez
FJ
. 
The role of peroxisome proliferator-activated receptors in carcinogenesis and chemoprevention
.
Nat Rev Cancer
2012
;
12
:
181
95
.
86.
van't Veer
LJ
,
Dai
H
,
van de Vijver
MJ
,
He
YD
,
Hart
AA
,
Mao
M
, et al
Gene expression profiling predicts clinical outcome of breast cancer
.
Nature
2002
;
415
:
530
6
.
87.
Ito
K
,
Carracedo
A
,
Weiss
D
,
Arai
F
,
Ala
U
,
Avigan
DE
, et al
A PML-PPAR-delta pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance
.
Nat Med
2012
;
18
:
1350
8
.
88.
Gan
B
,
Hu
J
,
Jiang
S
,
Liu
Y
,
Sahin
E
,
Zhuang
L
, et al
Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells
.
Nature
2010
;
468
:
701
4
.
89.
Morrison
SJ
,
Kimble
J
. 
Asymmetric and symmetric stem-cell divisions in development and cancer
.
Nature
2006
;
441
:
1068
74
.
90.
Ito
K
,
Bernardi
R
,
Pandolfi
PP
. 
A novel signaling network as a critical rheostat for the biology and maintenance of the normal stem cell and the cancer-initiating cell
.
Curr Opin Genet Dev
2009
;
19
:
51
9
.
91.
De Luca
A
,
Fiorillo
M
,
Peiris-Pages
M
,
Ozsvari
B
,
Smith
DL
,
Sanchez-Alvarez
R
, et al
Mitochondrial biogenesis is required for the anchorage-independent survival and propagation of stem-like cancer cells
.
Oncotarget
2015
;
6
:
14777
95
.
92.
Rahman
M
,
Miyamoto
H
,
Chang
C
. 
Androgen receptor coregulators in prostate cancer: mechanisms and clinical implications
.
Clin Cancer Res
2004
;
10
:
2208
19
.
93.
Massie
CE
,
Lynch
A
,
Ramos-Montoya
A
,
Boren
J
,
Stark
R
,
Fazli
L
, et al
The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis
.
EMBO J
2011
;
30
:
2719
33
.
94.
Tennakoon
JB
,
Shi
Y
,
Han
JJ
,
Tsouko
E
,
White
MA
,
Burns
AR
, et al
Androgens regulate prostate cancer cell growth via an AMPK-PGC-1alpha-mediated metabolic switch
.
Oncogene
2014
;
33
:
5251
61
.
95.
Smeenk
L
,
Lohrum
M
. 
Behind the scenes: unravelling the molecular mechanisms of p53 target gene selectivity (Review)
.
Int J Oncol
2010
;
37
:
1061
70
.
96.
Sen
N
,
Satija
YK
,
Das
S
. 
PGC-1α, a key modulator of p53, promotes cell survival upon metabolic stress
.
Mol Cell
2011
;
44
:
621
34
.
97.
Cioce
M
,
Blandino
G
. 
PGC1α confers specificity-metabolic stress and p53-dependent transcription
.
Mol Cell
2011
;
44
:
515
6
.
98.
Chen
W
,
Wang
Q
,
Bai
L
,
Chen
W
,
Wang
X
,
Tellez
CS
, et al
RIP1 maintains DNA integrity and cell proliferation by regulating PGC-1α-mediated mitochondrial oxidative phosphorylation and glycolysis
.
Cell Death Differ
2014
;
21
:
1061
70
.
99.
Chiarugi
A
,
Dolle
C
,
Felici
R
,
Ziegler
M
. 
The NAD metabolome–a key determinant of cancer cell biology
.
Nat Rev Cancer
2012
;
12
:
741
52
.
100.
Gerhart-Hines
Z
,
Rodgers
JT
,
Bare
O
,
Lerin
C
,
Kim
SH
,
Mostoslavsky
R
, et al
Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha
.
EMBO J
2007
;
26
:
1913
23
.
101.
Kulkarni
SR
,
Donepudi
AC
,
Xu
J
,
Wei
W
,
Cheng
QC
,
Driscoll
MV
, et al
Fasting induces nuclear factor E2-related factor 2 and ATP-binding Cassette transporters via protein kinase A and Sirtuin-1 in mouse and human
.
Antioxid Redox Signal
2014
;
20
:
15
30
.
102.
Do
MT
,
Kim
HG
,
Choi
JH
,
Jeong
HG
. 
Metformin induces microRNA-34a to downregulate the Sirt1/Pgc-1alpha/Nrf2 pathway, leading to increased susceptibility of wild-type p53 cancer cells to oxidative stress and therapeutic agents
.
Free Radic Biol Med
2014
;
74
:
21
34
.
103.
Sadeghi
N
,
Abbruzzese
JL
,
Yeung
SC
,
Hassan
M
,
Li
D
. 
Metformin use is associated with better survival of diabetic patients with pancreatic cancer
.
Clin Cancer Res
2012
;
18
:
2905
12
.
104.
Dang
CV.
Links between metabolism and cancer
.
Genes Dev
2012
;
26
:
877
90
.
105.
Lim
JH
,
Luo
C
,
Vazquez
F
,
Puigserver
P
. 
Targeting mitochondrial oxidative metabolism in melanoma causes metabolic compensation through glucose and glutamine utilization
.
Cancer Res
2014
;
74
:
3535
45
.
106.
Watkins
G
,
Douglas-Jones
A
,
Mansel
RE
,
Jiang
WG
. 
The localisation and reduction of nuclear staining of PPARgamma and PGC-1 in human breast cancer
.
Oncol Rep
2004
;
12
:
483
8
.
107.
Feilchenfeldt
J
,
Brundler
MA
,
Soravia
C
,
Totsch
M
,
Meier
CA
. 
Peroxisome proliferator-activated receptors (PPARs) and associated transcription factors in colon cancer: reduced expression of PPARgamma-coactivator 1 (PGC-1)
.
Cancer Lett
2004
;
203
:
25
33
.
108.
Lee
HJ
,
Su
Y
,
Yin
PH
,
Lee
HC
,
Chi
CW
. 
PPAR(gamma)/PGC-1(alpha) pathway in E-cadherin expression and motility of HepG2 cells
.
Anticancer Res
2009
;
29
:
5057
63
.
109.
Zhang
Y
,
Ba
Y
,
Liu
C
,
Sun
G
,
Ding
L
,
Gao
S
, et al
PGC-1alpha induces apoptosis in human epithelial ovarian cancer cells through a PPARgamma-dependent pathway
.
Cell Res
2007
;
17
:
363
73
.
110.
Jiang
WG
,
Douglas-Jones
A
,
Mansel
RE
. 
Expression of peroxisome-proliferator activated receptor-gamma (PPARgamma) and the PPARgamma co-activator, PGC-1, in human breast cancer correlates with clinical outcomes
.
Int J Cancer
2003
;
106
:
752
7
.
111.
D'Errico
I
,
Salvatore
L
,
Murzilli
S
,
Lo Sasso
G
,
Latorre
D
,
Martelli
N
, et al
Peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1alpha) is a metabolic regulator of intestinal epithelial cell fate
.
Proc Natl Acad Sci U S A
2011
;
108
:
6603
8
.
112.
Neill
T
,
Torres
A
,
Buraschi
S
,
Owens
RT
,
Hoek
JB
,
Baffa
R
, et al
Decorin induces mitophagy in breast carcinoma cells via peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and mitostatin
.
J Biol Chem
2014
;
289
:
4952
68
.
113.
D'Errico
I
,
Lo Sasso
G
,
Salvatore
L
,
Murzilli
S
,
Martelli
N
,
Cristofaro
M
, et al
Bax is necessary for PGC1alpha pro-apoptotic effect in colorectal cancer cells
.
Cell Cycle
2011
;
10
:
2937
45
.
114.
Li
X
,
Lu
Y
,
Lu
H
,
Luo
J
,
Hong
Y
,
Fan
Z
. 
AMPK-mediated energy homeostasis and associated metabolic effects on cancer cell response and resistance to cetuximab
.
Oncotarget
2015
;
6
:
11507
18
.
115.
Zhang
JG
,
Hong
DF
,
Zhang
CW
,
Sun
XD
,
Wang
ZF
,
Shi
Y
, et al
Sirtuin 1 facilitates chemoresistance of pancreatic cancer cells by regulating adaptive response to chemotherapy-induced stress
.
Cancer Sci
2014
;
105
:
445
54
.
116.
Egan
DF
,
Chun
MG
,
Vamos
M
,
Zou
H
,
Rong
J
,
Miller
CJ
, et al
Small molecule inhibition of the autophagy kinase ULK1 and identification of ULK1 substrates
.
Mol Cell
2015
;
59
:
285
97
.
117.
Galluzzi
L
,
Pietrocola
F
,
Bravo-San Pedro
JM
,
Amaravadi
RK
,
Baehrecke
EH
,
Cecconi
F
, et al
Autophagy in malignant transformation and cancer progression
.
EMBO J
2015
;
34
:
856
80
.
118.
Rodriguez-Enriquez
S
,
Hernandez-Esquivel
L
,
Marin-Hernandez
A
,
El Hafidi
M
,
Gallardo-Perez
JC
,
Hernandez-Resendiz
I
, et al
Mitochondrial free fatty acid beta-oxidation supports oxidative phosphorylation and proliferation in cancer cells
.
Int J Biochem Cell Biol
2015
;
65
:
209
21
.
119.
Hossain
F
,
Al-Khami
AA
,
Wyczechowska
D
,
Hernandez
C
,
Zheng
L
,
Reiss
K
, et al
Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances cancer therapies
.
Cancer Immunol Res
2015
;
3
:
1236
47
.