Mol. Cells 2022; 45(6): 413-424
Published online June 3, 2022
https://doi.org/10.14348/molcells.2022.0067
© The Korean Society for Molecular and Cellular Biology
Correspondence to : ykeeshin@snu.ac.kr
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
Suppressor of mothers against decapentaplegic homolog (SMAD) 4 is a pluripotent signaling mediator that regulates myriad cellular functions, including cell growth, cell division, angiogenesis, apoptosis, cell invasion, and metastasis, through transforming growth factor β (TGF-β)-dependent and -independent pathways. SMAD4 is a critical modulator in signal transduction and functions primarily as a transcription factor or cofactor. Apart from being a DNA-binding factor, the additional SMAD4 mechanisms in tumor suppression remain elusive. We previously identified methyl malonyl aciduria cobalamin deficiency B type (MMAB) as a critical SMAD4 binding protein using a proto array analysis. This study confirmed the interaction between SMAD4 and MMAB using bimolecular fluorescence complementation (BiFC) assay, proximity ligation assay (PLA), and conventional immunoprecipitation. We found that transient SMAD4 overexpression down-regulates MMAB expression via a proteasome-dependent pathway. SMAD4-MMAB interaction was independent of TGF-β signaling. Finally, we determined the effect of MMAB downregulation on cancer cells. siRNA-mediated knockdown of MMAB affected cancer cell metabolism in HeLa cells by decreasing ATP production and glucose consumption as well as inducing apoptosis. These findings suggest that SMAD4 controls cancer cell metabolism by regulating MMAB.
Keywords methyl malonyl aciduria cobalamin deficiency B type, mitochondrial energy production, proteasomal pathway, suppressor of mothers against decapentaplegic homolog 4
SMAD4, an SMAD (suppressor of mothers against decapentaplegic homolog) family member, is a central mediator in transforming growth factor β (TGF-β) signaling and transduces extracellular signals to the nucleus (Massague, 1998; Massague and Wotton, 2000). TGF-β signaling activates transmembrane serine-threonine receptor kinases, which phosphorylate SMAD2/3. The phosphorylated SMAD2/3 and their co-mediator SMAD4 translocate to the nucleus, positively or negatively regulating the target gene transcription (Lee et al., 2021).
MMAB is located at the 12q24 and encodes adenosyltransferase (ATR), which is involved in the intracellular vitamin B12 metabolism.
The HeLa cervical, A549 lung, and SW480 colon cancer cell lines were purchased from the Korean Cell Line Bank (Korea). They were cultured in an RPMI 1640 medium (Thermo Fisher Scientific, USA) supplemented with 10% fetal bovine serum (Thermo Fisher Scientific) at 37°C in a 5% CO2 atmosphere. Cells were routinely monitored for mycoplasma contamination and authenticated using short tandem repeat DNA technology. Cells were transfected with 0.5-4 µg of each DNA plasmid using FuGENE® HD (Promega, USA) or with 10-40 nM siRNA using siLentFect (Bio-Rad Laboratories, USA) following the manufacturer’s protocol.
BiFC constructs using fragments derived from newly engineered fluorescent protein-Venus were kindly provided by Professor Chang-Deng Hu (Department of Medicinal Chemistry and Molecular Pharmacology and Purdue Cancer Center, Purdue University, West Lafayette, IN, USA). cDNAs encoding
HeLa cells were transiently transfected with
PLA experiments were conducted using cultured HeLa cells following the manufacturer’s protocol (O-LINK Bioscience, Sweden). HeLa cells were fixed on four-well cell culture slides (SPL Life Sciences, Korea) using 4% paraformaldehyde. After blocking with 5% non-fat milk for 60 min, cells were co-incubated with the primary antibodies, MMAB and SMAD4, at 1:100 dilutions overnight at 4°C. Then, cells were washed twice with TBS-T for five minutes, incubated with PLA probe solutions for 60 min at 37°C, washed twice with TBS-T for five minutes, and incubated with ligase solution for 30 min at 37°C for ligation. After ligation, samples were washed twice with TBS-T for two minutes and incubated with polymerase solution for 100 min at 37°C for amplification. Finally, the samples were washed with SSC buffers (prepared according to the manufacturer’s protocol) and ethanol, and subsequently mounted on slides. Slides were imaged using an LSM 700 ZEISS laser scanning confocal microscope. CellLightTM Mitochondria-GFP (Invitrogen) was used to stain HeLa cell mitochondria.
HeLa cells were fixed in 4% paraformaldehyde on an eight-well cell culture slide (SPL Life Sciences) for double immunofluorescence staining. After fixation, cells were blocked in 5% non-fat milk for 60 min, followed by incubation with primary antibodies against MMAB and SMAD4 at 1:100 dilutions overnight at 4°C. Then, cells were incubated in the secondary antibodies, Alexa Fluor 488-conjugated goat anti-mouse IgG (1:100; Molecular Probes, USA) and Alexa Fluor 546-conjugated goat anti-rabbit IgG (1:100; Molecular Probes), for one hour at room temperature (RT). Finally, samples were washed in phosphate-buffered saline (PBS)/1% Triton X-100 and mounted. Cell nuclei were stained with Hoechst 33342 (Invitrogen). Slides were imaged using an LSM 700 ZEISS laser scanning confocal microscope (Carl Zeiss).
Cells were collected and washed with PBS. Then, they were extracted in RIPA buffer (150-mM NaCl, 50-mM Tris pH 7.6, 10% Glycerol, 0.1% SDS, and 1% NP40, 0.5% deoxycholic acid) for Western blotting or in IP buffer (25-mM HEPES pH 7.5, 150-mM NaCl, 0.2% NP-40, 10% glycerol, protease and phosphatase inhibitor cocktails) for IP. The supernatant was collected for IP after cell lysis on ice for 20 min and centrifugation at 13,000 ×
Immunoprecipitated samples were subjected to SDS-PAGE (10%-12%) and transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, USA). The membranes were blocked in 5% non-fat milk in TBS-T buffer for 60 min. Primary antibodies against MMAB, HA (3F10 clone; Roche Diagnostics GmbH, Germany), or FLAG (M2 clone; Sigma-Aldrich, USA) were used at 1:1,000 dilutions in 1% non-fat milk in TBS-T buffer for 90 min, followed by washing in TBS-T buffer. Membranes were incubated with an anti-mouse secondary antibody (1:5,000) in TBS-T buffer with 1% non-fat milk for 60 min before washing again in TBS-T buffer for 30 min. Both ACTB (C4 clone; Santa Cruz Biotechnology) and GAPDH (FL335 clone; Santa Cruz Biotechnology) were used as loading controls. The results were visualized with horseradish peroxidase-conjugated secondary antibodies (Thermo Fisher Scientific). Western blot images were developed on photographic film using enhanced chemiluminescence (ECL) reagents (GE Healthcare, USA).
HeLa cells transiently transfected with
Three siRNAs (
For viable cell counting, 5 × 104 HeLa cells were seeded on a six-well microplate in glucose (conventional RPMI) or pyruvate media, both of which were supplemented with 10% fetal bovine serum and 1% antibiotic for 24 h. The pyruvate medium consisted of RPMI with 10-mM pyruvate (rather than glucose) and 5-mM HEPES (Invitrogen). Then, HeLa cells were transfected with siRNA targeting
Cellular ATP levels were determined using a CellTiter-Glo® luminescent cell viability assay kit (Promega). Briefly, 4 × 103 HeLa cells were seeded on an opaque-walled 96-well microplate and transfected for 120 h at 37°C with non-targeting or
A glucose assay kit (Sigma-Aldrich) was used to measure glucose consumption. Briefly, 1 × 105
Briefly,
The metabolic phenotype of
IBM SPSS Statistics software (ver. 21; IBM, USA) was used for statistical analyses. A Mann–Whitney test was used to compare cell viability, ATP production, glucose consumption, reactive oxygen species generation, apoptosis, and senescence data between the two groups. Data were expressed as the mean ± SD or standard error of the mean, calculated from three or more independent experiments. Additional methods are given in Supplementary Materials and Methods.
We conducted a eukaryotic-expressed human protein array (ProtoArray) with purified SMAD4, identifying the mitochondrial protein, MMAB, as a novel SMAD4 binding protein (Rajasekaran et al., 2021). The SMAD4 and MMAB interaction was assessed using a BiFC assay, allowing the detection of the subcellular localization of the SMAD4-MMAB complex in live cells. We observed the cytosolic interaction between SMAD4 and MMAB; however, the BiFC signals from the SMAD4-MMAB complex did not overlap with mitochondria, as confirmed by MitoTracker® Red FM staining (Fig. 1A). The specificity of the SMAD4 and MMAB interaction was checked in the presence of the
To identify the protein binding domains responsible for the SMAD4-MMAB interaction, we designed
SMAD4 is a major component of TGF-β signaling. Hence, we investigated whether the SMAD4-MMAB interaction was TGF-β-dependent or not. HeLa cells transfected with BiFC constructs (
Subsequently, we investigated the signaling downstream of the SMAD4-MMAB interaction. SMAD4 expression decreased both exogenous and endogenous
To determine the effects of MMAB downregulation, siRNA targeting
The role of MMAB in cancer is incompletely understood. MMAB is involved in converting L-methylmalonyl-CoA to succinyl-CoA, a TCA cycle intermediate. We postulated that the SMAD4-mediated downregulation of MMAB inhibits the production of succinyl-CoA, thereby inhibiting the TCA cycle. Thus, we investigated whether MMAB depletion decreased ATP production and compromised energy metabolism. MMAB depletion significantly decreased ATP production and glucose consumption in HeLa cells (Figs. 6A and 6B). Since glucose is a component of normal cell culture medium, ATP could be produced by both glycolysis and the TCA cycle under these conditions. Thus, to determine whether MMAB downregulation affected the TCA cycle, we examined the effect of
The role of SMAD4 as a tumor suppressor has been extensively studied (Schutte, 1999). SMAD4 inactivation is mediated by LOH, loss-of-function mutations, downregulation via promoter hypermethylation, and ubiquitin proteosome-mediated degradation. The deregulation of the nuclear-cytoplasm SMAD4 shuttle is implicated in the development and progression of different cancers, including pancreatic, gastrointestinal, and colorectal cancers (Bardeesy et al., 2006; Blackford et al., 2009; Papageorgis et al., 2011; Yan et al., 2016). The anti-tumorigenic mechanisms of SMAD4 are mediated by its DNA-binding functions, where SMAD4 acts as a transcription factor or cofactor (Johnson et al., 1999; McCarthy and Chetty, 2018). Here we describe a novel mechanism underlying the anti-tumor effects of SMAD4.
Our previous protein array study identified MMAB as a novel SMAD4 binding partner (Rajasekaran et al., 2021). Although the inhibitory effects of SMAD4 on cancer progression are well-known, the regulatory mechanisms independent of its transcriptional function are poorly understood. Additionally, studies on how MMAB is involved in cancer cells’ metabolism remain infant (Chan et al., 2015; Sorin et al., 2021; Gomes et al., 2022). Here, to the best of our knowledge, we identified for the first time that SMAD4 suppresses tumorigenesis by decreasing MMAB and disrupting cancer cell energy metabolism by inhibiting the TCA cycle.
Firstly, we demonstrated that cytosolic SMAD4-MMAB binding is TGF-β-independent. The SMAD4-linker domain specifically interacts with the MMAB catalytic domain. SMAD4-MMAB binding decreased MMAB protein levels by inducing proteasomal MMAB degradation. SMAD4-mediated decrease in MMAB levels was mimicked using
MMAB is involved in AdoCbl synthesis, an activated form of vitamin B12 that is a required enzymatic cofactor of methylmalonyl-CoA mutase, converting methylmalonyl-CoA to succinyl-CoA (Dobson et al., 2002). The loss of enzyme activity or cofactors for this reaction as well as consequent methylmalonic acid accumulation, can disrupt normal glucose metabolism (Rosenberg et al., 1968a; 1968b). Anaplerotic deficiencies of the TCA cycle are frequently observed in patients with methylmalonyl aciduria, a disease characterized by methylmalonic acid accumulation (Anzmann et al., 2019). Genome-wide association studies have identified that single-nucleotide polymorphisms in the
Here, based on the role of MMAB in providing substrates for the TCA cycle, we postulated that MMAB could be related to mitochondrial energy metabolism, promoting the TCA cycle. Through the downregulation of MMAB by interaction with SMAD4, the suppression of methylmalonyl-CoA mutase expression decreased the succinyl-CoA synthesis. Thus, the supply of succinyl-CoA ultimately becomes insufficient to sustain the mitochondrial energy production via the TCA cycle. Therefore, the decreased succinyl-CoA production interferes with the TCA cycle function. We observed that cell viability significantly decreased 72 h after
The depletion of cellular ATP is a crucial event that mediates cell death. Notably, previous studies have demonstrated that apoptosis is an ATP-dependent process and that decreased ATP levels promote cell death (Ferrari et al., 1998; Tsujimoto, 1997; Zamaraeva et al., 2005). Decreased ATP levels could occur due to the direct role of succinyl-CoA in ATP production via the TCA cycle, or an indirect effect due to TCA cycle inhibition (Williamson and Cooper, 1980). Therefore, proteasomal MMAB degradation regulated by the SMAD4-MMAB interaction primarily inhibits mitochondrial ATP synthesis due to alterations in the levels of TCA cycle intermediates, consequently activating caspase 3-mediated apoptosis. Further in-depth studies are essential to understand the input of other TCA cycle intermediates and anaplerotic substrates in inhibiting ATP production.
In contrast to the conventional belief that cancer cells bypass the TCA cycle and rely primarily on glycolysis (Pavlova and Thompson, 2016), recent studies have demonstrated that cancer cells with defective tumor suppressor mechanisms and oncogenic mutations depend on the TCA cycle for sufficient ATP production and survival (Anderson et al., 2018). Exploiting these unique metabolic dependencies opens a new horizon for targeted cancer therapies (Lee and Kim, 2016; Pathania et al., 2009). Several components of the TCA cycle, including α-ketoglutarate dehydrogenase (KGDHC), isocitrate dehydrogenase (IDH), and pyruvate dehydrogenase (PDH), are emerging as compelling anti-cancer therapeutic targets (Allen et al., 2016; Dang et al., 2009; Zachar et al., 2011). Recently, CPI-613 (Devimistat), a molecule that simultaneously inhibits KGDHC and PDH, is being investigated in clinical trials as a stand-alone therapy or along with chemotherapy (Lycan et al., 2016; Pardee et al., 2014). Small molecule inhibitors of IDH; AG-221 and AG-881, are also involved in clinical development (Yen et al., 2017). However, the dependency of normal cells on the TCA cycle for ATP production is likely to induce off-target cytotoxic effects, posing a significant barrier to this approach. Recent studies also demonstrated that cancer cells use the TCA cycle differently than normal cells, thereby increasing their sensitivity to TCA cycle inhibitors (Grassian et al., 2014; Kishton and Rathmell, 2016; Yuneva et al., 2012). In this study, we identified MMAB as a novel SMAD4 binding partner, and identified that SMAD4-MMAB interaction decreases MMAB protein levels in cancer cells, primarily inducing cell death via TCA cycle inhibition. Although the role of MMAB in cancer has not been widely investigated, public databases, such as the human protein atlas, intriguingly revealed increased MMAB protein levels in various cancerous tissue samples.
This prompted us to propose the previously untested hypothesis that normal cells could transform into cancer cells
Authors would like to extend their thanks to Professor Chang-Deng Hu (Department of Medicinal Chemistry and Molecular Pharmacology and Purdue Cancer Center, Purdue University, West Lafayette, IN) for kindly gifting them the BiFC constructs using fragments derived from newly engineered fluorescent protein-Venus.
K.S., H.S.L., and Y.K.S. conceived and conceptualized the manuscript. Y.K.S. supervised and administered the project, and acquired the fund. K.S., H.S.L., C.C., N.R., and L.J. performed the experiments and acquired the data. K.S., H.S.L., and C.C. wrote the original draft. K.S., C.C., and Y.K.S. wrote, reviewed, and edited the final manuscript. All authors have read and agreed to the published version of this manuscript.
The authors have no potential conflicts of interest to disclose.
Mol. Cells 2022; 45(6): 413-424
Published online June 30, 2022 https://doi.org/10.14348/molcells.2022.0067
Copyright © The Korean Society for Molecular and Cellular Biology.
Kyoung Song1,6 , Hun Seok Lee2,6
, Lina Jia3
, Chaithanya Chelakkot4
, Nirmal Rajasekaran5
, and Young Kee Shin2,4,5,*
1College of Pharmacy, Duksung Women’s University, Seoul 01366, Korea, 2Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul 08826, Korea, 3Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China, 4Bio-MAX Institute, Seoul National University, Seoul 08826, Korea, 5Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea, 6These authors contributed equally to this work.
Correspondence to:ykeeshin@snu.ac.kr
This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.
Suppressor of mothers against decapentaplegic homolog (SMAD) 4 is a pluripotent signaling mediator that regulates myriad cellular functions, including cell growth, cell division, angiogenesis, apoptosis, cell invasion, and metastasis, through transforming growth factor β (TGF-β)-dependent and -independent pathways. SMAD4 is a critical modulator in signal transduction and functions primarily as a transcription factor or cofactor. Apart from being a DNA-binding factor, the additional SMAD4 mechanisms in tumor suppression remain elusive. We previously identified methyl malonyl aciduria cobalamin deficiency B type (MMAB) as a critical SMAD4 binding protein using a proto array analysis. This study confirmed the interaction between SMAD4 and MMAB using bimolecular fluorescence complementation (BiFC) assay, proximity ligation assay (PLA), and conventional immunoprecipitation. We found that transient SMAD4 overexpression down-regulates MMAB expression via a proteasome-dependent pathway. SMAD4-MMAB interaction was independent of TGF-β signaling. Finally, we determined the effect of MMAB downregulation on cancer cells. siRNA-mediated knockdown of MMAB affected cancer cell metabolism in HeLa cells by decreasing ATP production and glucose consumption as well as inducing apoptosis. These findings suggest that SMAD4 controls cancer cell metabolism by regulating MMAB.
Keywords: methyl malonyl aciduria cobalamin deficiency B type, mitochondrial energy production, proteasomal pathway, suppressor of mothers against decapentaplegic homolog 4
SMAD4, an SMAD (suppressor of mothers against decapentaplegic homolog) family member, is a central mediator in transforming growth factor β (TGF-β) signaling and transduces extracellular signals to the nucleus (Massague, 1998; Massague and Wotton, 2000). TGF-β signaling activates transmembrane serine-threonine receptor kinases, which phosphorylate SMAD2/3. The phosphorylated SMAD2/3 and their co-mediator SMAD4 translocate to the nucleus, positively or negatively regulating the target gene transcription (Lee et al., 2021).
MMAB is located at the 12q24 and encodes adenosyltransferase (ATR), which is involved in the intracellular vitamin B12 metabolism.
The HeLa cervical, A549 lung, and SW480 colon cancer cell lines were purchased from the Korean Cell Line Bank (Korea). They were cultured in an RPMI 1640 medium (Thermo Fisher Scientific, USA) supplemented with 10% fetal bovine serum (Thermo Fisher Scientific) at 37°C in a 5% CO2 atmosphere. Cells were routinely monitored for mycoplasma contamination and authenticated using short tandem repeat DNA technology. Cells were transfected with 0.5-4 µg of each DNA plasmid using FuGENE® HD (Promega, USA) or with 10-40 nM siRNA using siLentFect (Bio-Rad Laboratories, USA) following the manufacturer’s protocol.
BiFC constructs using fragments derived from newly engineered fluorescent protein-Venus were kindly provided by Professor Chang-Deng Hu (Department of Medicinal Chemistry and Molecular Pharmacology and Purdue Cancer Center, Purdue University, West Lafayette, IN, USA). cDNAs encoding
HeLa cells were transiently transfected with
PLA experiments were conducted using cultured HeLa cells following the manufacturer’s protocol (O-LINK Bioscience, Sweden). HeLa cells were fixed on four-well cell culture slides (SPL Life Sciences, Korea) using 4% paraformaldehyde. After blocking with 5% non-fat milk for 60 min, cells were co-incubated with the primary antibodies, MMAB and SMAD4, at 1:100 dilutions overnight at 4°C. Then, cells were washed twice with TBS-T for five minutes, incubated with PLA probe solutions for 60 min at 37°C, washed twice with TBS-T for five minutes, and incubated with ligase solution for 30 min at 37°C for ligation. After ligation, samples were washed twice with TBS-T for two minutes and incubated with polymerase solution for 100 min at 37°C for amplification. Finally, the samples were washed with SSC buffers (prepared according to the manufacturer’s protocol) and ethanol, and subsequently mounted on slides. Slides were imaged using an LSM 700 ZEISS laser scanning confocal microscope. CellLightTM Mitochondria-GFP (Invitrogen) was used to stain HeLa cell mitochondria.
HeLa cells were fixed in 4% paraformaldehyde on an eight-well cell culture slide (SPL Life Sciences) for double immunofluorescence staining. After fixation, cells were blocked in 5% non-fat milk for 60 min, followed by incubation with primary antibodies against MMAB and SMAD4 at 1:100 dilutions overnight at 4°C. Then, cells were incubated in the secondary antibodies, Alexa Fluor 488-conjugated goat anti-mouse IgG (1:100; Molecular Probes, USA) and Alexa Fluor 546-conjugated goat anti-rabbit IgG (1:100; Molecular Probes), for one hour at room temperature (RT). Finally, samples were washed in phosphate-buffered saline (PBS)/1% Triton X-100 and mounted. Cell nuclei were stained with Hoechst 33342 (Invitrogen). Slides were imaged using an LSM 700 ZEISS laser scanning confocal microscope (Carl Zeiss).
Cells were collected and washed with PBS. Then, they were extracted in RIPA buffer (150-mM NaCl, 50-mM Tris pH 7.6, 10% Glycerol, 0.1% SDS, and 1% NP40, 0.5% deoxycholic acid) for Western blotting or in IP buffer (25-mM HEPES pH 7.5, 150-mM NaCl, 0.2% NP-40, 10% glycerol, protease and phosphatase inhibitor cocktails) for IP. The supernatant was collected for IP after cell lysis on ice for 20 min and centrifugation at 13,000 ×
Immunoprecipitated samples were subjected to SDS-PAGE (10%-12%) and transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, USA). The membranes were blocked in 5% non-fat milk in TBS-T buffer for 60 min. Primary antibodies against MMAB, HA (3F10 clone; Roche Diagnostics GmbH, Germany), or FLAG (M2 clone; Sigma-Aldrich, USA) were used at 1:1,000 dilutions in 1% non-fat milk in TBS-T buffer for 90 min, followed by washing in TBS-T buffer. Membranes were incubated with an anti-mouse secondary antibody (1:5,000) in TBS-T buffer with 1% non-fat milk for 60 min before washing again in TBS-T buffer for 30 min. Both ACTB (C4 clone; Santa Cruz Biotechnology) and GAPDH (FL335 clone; Santa Cruz Biotechnology) were used as loading controls. The results were visualized with horseradish peroxidase-conjugated secondary antibodies (Thermo Fisher Scientific). Western blot images were developed on photographic film using enhanced chemiluminescence (ECL) reagents (GE Healthcare, USA).
HeLa cells transiently transfected with
Three siRNAs (
For viable cell counting, 5 × 104 HeLa cells were seeded on a six-well microplate in glucose (conventional RPMI) or pyruvate media, both of which were supplemented with 10% fetal bovine serum and 1% antibiotic for 24 h. The pyruvate medium consisted of RPMI with 10-mM pyruvate (rather than glucose) and 5-mM HEPES (Invitrogen). Then, HeLa cells were transfected with siRNA targeting
Cellular ATP levels were determined using a CellTiter-Glo® luminescent cell viability assay kit (Promega). Briefly, 4 × 103 HeLa cells were seeded on an opaque-walled 96-well microplate and transfected for 120 h at 37°C with non-targeting or
A glucose assay kit (Sigma-Aldrich) was used to measure glucose consumption. Briefly, 1 × 105
Briefly,
The metabolic phenotype of
IBM SPSS Statistics software (ver. 21; IBM, USA) was used for statistical analyses. A Mann–Whitney test was used to compare cell viability, ATP production, glucose consumption, reactive oxygen species generation, apoptosis, and senescence data between the two groups. Data were expressed as the mean ± SD or standard error of the mean, calculated from three or more independent experiments. Additional methods are given in Supplementary Materials and Methods.
We conducted a eukaryotic-expressed human protein array (ProtoArray) with purified SMAD4, identifying the mitochondrial protein, MMAB, as a novel SMAD4 binding protein (Rajasekaran et al., 2021). The SMAD4 and MMAB interaction was assessed using a BiFC assay, allowing the detection of the subcellular localization of the SMAD4-MMAB complex in live cells. We observed the cytosolic interaction between SMAD4 and MMAB; however, the BiFC signals from the SMAD4-MMAB complex did not overlap with mitochondria, as confirmed by MitoTracker® Red FM staining (Fig. 1A). The specificity of the SMAD4 and MMAB interaction was checked in the presence of the
To identify the protein binding domains responsible for the SMAD4-MMAB interaction, we designed
SMAD4 is a major component of TGF-β signaling. Hence, we investigated whether the SMAD4-MMAB interaction was TGF-β-dependent or not. HeLa cells transfected with BiFC constructs (
Subsequently, we investigated the signaling downstream of the SMAD4-MMAB interaction. SMAD4 expression decreased both exogenous and endogenous
To determine the effects of MMAB downregulation, siRNA targeting
The role of MMAB in cancer is incompletely understood. MMAB is involved in converting L-methylmalonyl-CoA to succinyl-CoA, a TCA cycle intermediate. We postulated that the SMAD4-mediated downregulation of MMAB inhibits the production of succinyl-CoA, thereby inhibiting the TCA cycle. Thus, we investigated whether MMAB depletion decreased ATP production and compromised energy metabolism. MMAB depletion significantly decreased ATP production and glucose consumption in HeLa cells (Figs. 6A and 6B). Since glucose is a component of normal cell culture medium, ATP could be produced by both glycolysis and the TCA cycle under these conditions. Thus, to determine whether MMAB downregulation affected the TCA cycle, we examined the effect of
The role of SMAD4 as a tumor suppressor has been extensively studied (Schutte, 1999). SMAD4 inactivation is mediated by LOH, loss-of-function mutations, downregulation via promoter hypermethylation, and ubiquitin proteosome-mediated degradation. The deregulation of the nuclear-cytoplasm SMAD4 shuttle is implicated in the development and progression of different cancers, including pancreatic, gastrointestinal, and colorectal cancers (Bardeesy et al., 2006; Blackford et al., 2009; Papageorgis et al., 2011; Yan et al., 2016). The anti-tumorigenic mechanisms of SMAD4 are mediated by its DNA-binding functions, where SMAD4 acts as a transcription factor or cofactor (Johnson et al., 1999; McCarthy and Chetty, 2018). Here we describe a novel mechanism underlying the anti-tumor effects of SMAD4.
Our previous protein array study identified MMAB as a novel SMAD4 binding partner (Rajasekaran et al., 2021). Although the inhibitory effects of SMAD4 on cancer progression are well-known, the regulatory mechanisms independent of its transcriptional function are poorly understood. Additionally, studies on how MMAB is involved in cancer cells’ metabolism remain infant (Chan et al., 2015; Sorin et al., 2021; Gomes et al., 2022). Here, to the best of our knowledge, we identified for the first time that SMAD4 suppresses tumorigenesis by decreasing MMAB and disrupting cancer cell energy metabolism by inhibiting the TCA cycle.
Firstly, we demonstrated that cytosolic SMAD4-MMAB binding is TGF-β-independent. The SMAD4-linker domain specifically interacts with the MMAB catalytic domain. SMAD4-MMAB binding decreased MMAB protein levels by inducing proteasomal MMAB degradation. SMAD4-mediated decrease in MMAB levels was mimicked using
MMAB is involved in AdoCbl synthesis, an activated form of vitamin B12 that is a required enzymatic cofactor of methylmalonyl-CoA mutase, converting methylmalonyl-CoA to succinyl-CoA (Dobson et al., 2002). The loss of enzyme activity or cofactors for this reaction as well as consequent methylmalonic acid accumulation, can disrupt normal glucose metabolism (Rosenberg et al., 1968a; 1968b). Anaplerotic deficiencies of the TCA cycle are frequently observed in patients with methylmalonyl aciduria, a disease characterized by methylmalonic acid accumulation (Anzmann et al., 2019). Genome-wide association studies have identified that single-nucleotide polymorphisms in the
Here, based on the role of MMAB in providing substrates for the TCA cycle, we postulated that MMAB could be related to mitochondrial energy metabolism, promoting the TCA cycle. Through the downregulation of MMAB by interaction with SMAD4, the suppression of methylmalonyl-CoA mutase expression decreased the succinyl-CoA synthesis. Thus, the supply of succinyl-CoA ultimately becomes insufficient to sustain the mitochondrial energy production via the TCA cycle. Therefore, the decreased succinyl-CoA production interferes with the TCA cycle function. We observed that cell viability significantly decreased 72 h after
The depletion of cellular ATP is a crucial event that mediates cell death. Notably, previous studies have demonstrated that apoptosis is an ATP-dependent process and that decreased ATP levels promote cell death (Ferrari et al., 1998; Tsujimoto, 1997; Zamaraeva et al., 2005). Decreased ATP levels could occur due to the direct role of succinyl-CoA in ATP production via the TCA cycle, or an indirect effect due to TCA cycle inhibition (Williamson and Cooper, 1980). Therefore, proteasomal MMAB degradation regulated by the SMAD4-MMAB interaction primarily inhibits mitochondrial ATP synthesis due to alterations in the levels of TCA cycle intermediates, consequently activating caspase 3-mediated apoptosis. Further in-depth studies are essential to understand the input of other TCA cycle intermediates and anaplerotic substrates in inhibiting ATP production.
In contrast to the conventional belief that cancer cells bypass the TCA cycle and rely primarily on glycolysis (Pavlova and Thompson, 2016), recent studies have demonstrated that cancer cells with defective tumor suppressor mechanisms and oncogenic mutations depend on the TCA cycle for sufficient ATP production and survival (Anderson et al., 2018). Exploiting these unique metabolic dependencies opens a new horizon for targeted cancer therapies (Lee and Kim, 2016; Pathania et al., 2009). Several components of the TCA cycle, including α-ketoglutarate dehydrogenase (KGDHC), isocitrate dehydrogenase (IDH), and pyruvate dehydrogenase (PDH), are emerging as compelling anti-cancer therapeutic targets (Allen et al., 2016; Dang et al., 2009; Zachar et al., 2011). Recently, CPI-613 (Devimistat), a molecule that simultaneously inhibits KGDHC and PDH, is being investigated in clinical trials as a stand-alone therapy or along with chemotherapy (Lycan et al., 2016; Pardee et al., 2014). Small molecule inhibitors of IDH; AG-221 and AG-881, are also involved in clinical development (Yen et al., 2017). However, the dependency of normal cells on the TCA cycle for ATP production is likely to induce off-target cytotoxic effects, posing a significant barrier to this approach. Recent studies also demonstrated that cancer cells use the TCA cycle differently than normal cells, thereby increasing their sensitivity to TCA cycle inhibitors (Grassian et al., 2014; Kishton and Rathmell, 2016; Yuneva et al., 2012). In this study, we identified MMAB as a novel SMAD4 binding partner, and identified that SMAD4-MMAB interaction decreases MMAB protein levels in cancer cells, primarily inducing cell death via TCA cycle inhibition. Although the role of MMAB in cancer has not been widely investigated, public databases, such as the human protein atlas, intriguingly revealed increased MMAB protein levels in various cancerous tissue samples.
This prompted us to propose the previously untested hypothesis that normal cells could transform into cancer cells
Authors would like to extend their thanks to Professor Chang-Deng Hu (Department of Medicinal Chemistry and Molecular Pharmacology and Purdue Cancer Center, Purdue University, West Lafayette, IN) for kindly gifting them the BiFC constructs using fragments derived from newly engineered fluorescent protein-Venus.
K.S., H.S.L., and Y.K.S. conceived and conceptualized the manuscript. Y.K.S. supervised and administered the project, and acquired the fund. K.S., H.S.L., C.C., N.R., and L.J. performed the experiments and acquired the data. K.S., H.S.L., and C.C. wrote the original draft. K.S., C.C., and Y.K.S. wrote, reviewed, and edited the final manuscript. All authors have read and agreed to the published version of this manuscript.
The authors have no potential conflicts of interest to disclose.