Pub Date : 2025-02-13DOI: 10.1016/j.jbc.2025.108316
Sharan Paul, Warunee Dansithong, Karla P Figueroa, Mandi Gandelman, Pravin Hivare, Daniel R Scoles, Stefan M Pulst
Staufen2 (STAU2) is an RNA binding protein that controls mRNA trafficking and expression. Previously, we showed that its paralog Staufen1 (STAU1) was overabundant in cellular and mouse models of neurodegenerative diseases and amyotrophic lateral sclerosis (ALS) patient spinal cord. Here we investigated features of STAU2 that might parallel STAU1. STAU2 protein, but not mRNA, was overabundant in spinocerebellar ataxia type 2 (SCA2), ALS/frontotemporal dementia (FTD) patient fibroblasts, ALS patient spinal cord tissues, and in central nervous system (CNS) tissues from SCA2 and ALS animal models. Exogenous expression of STAU2 in HEK293 cells activated mechanistic target of rapamycin (mTOR) and stress granule formation. Targeting STAU2 by RNAi normalized mTOR in SCA2 and C9ORF72 cellular models. The microRNA miR-217, previously identified as downregulated in SCA2 mice, targets the STAU2 3'-UTR. We now demonstrate that exogenous expression of miR-217 significantly reduced STAU2 and mTOR levels in cellular models of neurodegenerative disease. These results suggest a functional link between STAU2 and mTOR signaling and identify a major role for miR-217 that could be exploited in therapeutic development.
{"title":"Staufen2 dysregulation in neurodegenerative disease.","authors":"Sharan Paul, Warunee Dansithong, Karla P Figueroa, Mandi Gandelman, Pravin Hivare, Daniel R Scoles, Stefan M Pulst","doi":"10.1016/j.jbc.2025.108316","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108316","url":null,"abstract":"<p><p>Staufen2 (STAU2) is an RNA binding protein that controls mRNA trafficking and expression. Previously, we showed that its paralog Staufen1 (STAU1) was overabundant in cellular and mouse models of neurodegenerative diseases and amyotrophic lateral sclerosis (ALS) patient spinal cord. Here we investigated features of STAU2 that might parallel STAU1. STAU2 protein, but not mRNA, was overabundant in spinocerebellar ataxia type 2 (SCA2), ALS/frontotemporal dementia (FTD) patient fibroblasts, ALS patient spinal cord tissues, and in central nervous system (CNS) tissues from SCA2 and ALS animal models. Exogenous expression of STAU2 in HEK293 cells activated mechanistic target of rapamycin (mTOR) and stress granule formation. Targeting STAU2 by RNAi normalized mTOR in SCA2 and C9ORF72 cellular models. The microRNA miR-217, previously identified as downregulated in SCA2 mice, targets the STAU2 3'-UTR. We now demonstrate that exogenous expression of miR-217 significantly reduced STAU2 and mTOR levels in cellular models of neurodegenerative disease. These results suggest a functional link between STAU2 and mTOR signaling and identify a major role for miR-217 that could be exploited in therapeutic development.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108316"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143424802","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13DOI: 10.1016/j.jbc.2025.108315
Yue Dai, Dennis J Stuehr
Nitric oxide (NO) signaling often relies on it activating cGMP production by the heterodimeric enzyme soluble guanylyl cyclase (sGC). To mature to function an sGCβ subunit must first incorporate heme and then form a heterodimer with a partner α subunit. Our previous studies in cells showed that glyceraldehyde 3-phosphate dehydrogenase (GAPDH) supplies heme to the apo-sGCβ subunit, which is complexed with the cell chaperone Hsp90. Through its ATP hydrolysis, Hsp90 then promotes heme insertion into apo-sGCβ and consequent formation of a functional heterodimer. NO at physiologic levels somehow stimulates cell heme allocation into apo-sGCβ by this process. To gain insight we utilized purified apo-sGCβ and GAPDH reporter proteins whose heme contents can be followed by fluorescence and determined the impact of Hsp90 and NO on heme transfer between them. Results show that heme transfer out of GAPDH and into apo-sGCβ is tightly coupled in all circumstances and is limited by the ability of the apo-sGCβ to incorporate the heme, which in turn relies on a ferric to ferrous heme transition taking place inside the sGCβ. Hsp90 can influence the heme transfer kinetics in a negative or positive manner through its conformational effects on apo-sGCβ, while NO speeds heme transfer by binding to the heme iron and thus speeding heme dissociation from GAPDH. Our findings provide new mechanistic understanding of sGC maturation and how Hsp90 and NO combine to dynamically regulate heme incorporation for sGC heterodimer formation and consequent cGMP production in biological settings.
{"title":"JBC-D-24-03305 revision 1 Heme Delivery into Soluble Guanylyl Cyclase Requires a Heme Redox Change and is Regulated by NO and Hsp90 by Distinct Mechanisms.","authors":"Yue Dai, Dennis J Stuehr","doi":"10.1016/j.jbc.2025.108315","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108315","url":null,"abstract":"<p><p>Nitric oxide (NO) signaling often relies on it activating cGMP production by the heterodimeric enzyme soluble guanylyl cyclase (sGC). To mature to function an sGCβ subunit must first incorporate heme and then form a heterodimer with a partner α subunit. Our previous studies in cells showed that glyceraldehyde 3-phosphate dehydrogenase (GAPDH) supplies heme to the apo-sGCβ subunit, which is complexed with the cell chaperone Hsp90. Through its ATP hydrolysis, Hsp90 then promotes heme insertion into apo-sGCβ and consequent formation of a functional heterodimer. NO at physiologic levels somehow stimulates cell heme allocation into apo-sGCβ by this process. To gain insight we utilized purified apo-sGCβ and GAPDH reporter proteins whose heme contents can be followed by fluorescence and determined the impact of Hsp90 and NO on heme transfer between them. Results show that heme transfer out of GAPDH and into apo-sGCβ is tightly coupled in all circumstances and is limited by the ability of the apo-sGCβ to incorporate the heme, which in turn relies on a ferric to ferrous heme transition taking place inside the sGCβ. Hsp90 can influence the heme transfer kinetics in a negative or positive manner through its conformational effects on apo-sGCβ, while NO speeds heme transfer by binding to the heme iron and thus speeding heme dissociation from GAPDH. Our findings provide new mechanistic understanding of sGC maturation and how Hsp90 and NO combine to dynamically regulate heme incorporation for sGC heterodimer formation and consequent cGMP production in biological settings.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108315"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143425446","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13DOI: 10.1016/j.jbc.2025.108310
John E McLaughlin, Michael J Rudolph, Arkajyoti Dutta, Xiao-Ping Li, Anastasiia M Tsymbal, Yang Chen, Shibani Bhattacharya, Benjamin Algava, Michael Goger, Jacques Y Roberge, Nilgun E Tumer
Ricin is a Category B agent for bioterrorism and Shiga toxins are the primary virulence factors of Shiga toxin (Stx) producing E. coli (STEC). Ricin and Stxs bind the ribosomal P-stalk proteins to depurinate the sarcin/ricin loop (SRL) on the eukaryotic ribosome and inhibit translation. Both toxins are prime targets for therapeutic intervention because no effective therapy exists for ricin intoxication or STEC infection. Binding of ricin toxin A subunit (RTA) to the ribosomal P-stalk stimulates depurination of the SRL by an unknown mechanism. We previously identified compounds that bind the P-stalk pocket of RTA and inhibit catalytic activity. Here we characterize a second-generation lead compound, which binds the P-stalk pocket of RTA with over 30-fold improved affinity relative to the original compound and inhibits the cytotoxicity of ricin holotoxin in Vero cells with no apparent cellular toxicity by itself. This compound also shows protection against Stx2A1. X-ray crystal structure of RTA-inhibitor complexes suggests that the orientation of the carboxylic acid influences the inhibitor contacts at the P-stalk site of RTA and contributes to inhibitor potency. The structural changes triggered at the P-stalk site of RTA were validated by solution NMR based chemical shift perturbation analysis. A key finding by NMR is that binding induced conformational changes extend beyond the P-stalk site to residues in the active site cleft of RTA. Collectively, these results provide valuable new insight into the conformational flexibility in the C-terminal domain of RTA and its potential role in mediating the remarkable catalytic activity of ricin.
蓖麻毒素是生物恐怖主义的 B 类制剂,志贺毒素是产生志贺毒素(Stx)的大肠杆菌(STEC)的主要致病因子。蓖麻毒素和志贺毒素与核糖体 P-茎蛋白结合,使真核核糖体上的沙丁鱼毒素/蓖麻毒素环(SRL)去嘌呤化,从而抑制翻译。这两种毒素都是治疗干预的主要目标,因为目前还没有针对蓖麻毒素中毒或 STEC 感染的有效疗法。蓖麻毒素 A 亚基(RTA)与核糖体 P-茎结合会刺激 SRL 的去urination,其机制不明。我们之前发现了能与 RTA 的 P-stalk 袋结合并抑制催化活性的化合物。在这里,我们对第二代先导化合物进行了表征,该化合物与 RTA 的 P-stalk 袋结合的亲和力比原始化合物提高了 30 多倍,并能抑制蓖麻毒素全毒素在 Vero 细胞中的细胞毒性,其本身没有明显的细胞毒性。该化合物还对 Stx2A1 具有保护作用。RTA - 抑制剂复合物的 X 射线晶体结构表明,羧酸的取向会影响 RTA P-stalk 位点的抑制剂接触,并有助于提高抑制剂的效力。基于溶液核磁共振的化学位移扰动分析验证了在 RTA P-stalk 位点引发的结构变化。核磁共振的一个重要发现是,结合诱导的构象变化超出了 P-stalk 位点,延伸到了 RTA 活性位点裂隙中的残基。总之,这些结果为深入了解 RTA C 端结构域的构象灵活性及其在介导蓖麻毒素显著催化活性方面的潜在作用提供了宝贵的新见解。
{"title":"Binding of small molecules at the P-stalk site of ricin A subunit trigger conformational changes that extend into the active site.","authors":"John E McLaughlin, Michael J Rudolph, Arkajyoti Dutta, Xiao-Ping Li, Anastasiia M Tsymbal, Yang Chen, Shibani Bhattacharya, Benjamin Algava, Michael Goger, Jacques Y Roberge, Nilgun E Tumer","doi":"10.1016/j.jbc.2025.108310","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108310","url":null,"abstract":"<p><p>Ricin is a Category B agent for bioterrorism and Shiga toxins are the primary virulence factors of Shiga toxin (Stx) producing E. coli (STEC). Ricin and Stxs bind the ribosomal P-stalk proteins to depurinate the sarcin/ricin loop (SRL) on the eukaryotic ribosome and inhibit translation. Both toxins are prime targets for therapeutic intervention because no effective therapy exists for ricin intoxication or STEC infection. Binding of ricin toxin A subunit (RTA) to the ribosomal P-stalk stimulates depurination of the SRL by an unknown mechanism. We previously identified compounds that bind the P-stalk pocket of RTA and inhibit catalytic activity. Here we characterize a second-generation lead compound, which binds the P-stalk pocket of RTA with over 30-fold improved affinity relative to the original compound and inhibits the cytotoxicity of ricin holotoxin in Vero cells with no apparent cellular toxicity by itself. This compound also shows protection against Stx2A1. X-ray crystal structure of RTA-inhibitor complexes suggests that the orientation of the carboxylic acid influences the inhibitor contacts at the P-stalk site of RTA and contributes to inhibitor potency. The structural changes triggered at the P-stalk site of RTA were validated by solution NMR based chemical shift perturbation analysis. A key finding by NMR is that binding induced conformational changes extend beyond the P-stalk site to residues in the active site cleft of RTA. Collectively, these results provide valuable new insight into the conformational flexibility in the C-terminal domain of RTA and its potential role in mediating the remarkable catalytic activity of ricin.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108310"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143425366","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13DOI: 10.1016/j.jbc.2025.108311
Amanda Smart, Kevin Singewald, Zikri Hasanbasri, R David Britt, Glenn L Millhauser
The trafficking and aggregation of neurodegenerative proteins often involves the interaction between intrinsically disordered domains, stabilized by the inclusion of physiologic metal ions such as copper or zinc. Characterizing the metal ion coordination environment is critical for assessing the stability and organization of these relevant protein-protein interactions but is challenging given the lack of regular molecular order or global structure. The cellular prion protein (PrPC) binds both monomers and aggregates of Alzheimer's amyloid-beta (Aβ), promoting Aβ internalization and aberrant signaling, respectively. Both proteins bind Cu2+ with high affinity, opening the potential for copper to form an intermolecular bridge. We describe here a novel approach utilizing multiple EPR experiments to investigate the simultaneous Cu2+ coordination of PrPC and Aβ in a 1:1:1 mixture. Uniformly 15N-labeled PrPC is used in conjunction with natural abundance 14N Aβ, the combination of which leads to distinct energy manifolds for paramagnetic Cu2+ and resolved by the pulsed EPR experiments ESEEM and HYSCORE. We develop acquisition parameters to simultaneously optimize 14N (I = 1) and 15N (I = ½) pulsed EPR signals and we also advance the theory of ESEEM and HYSCORE to quantitatively describe multiple 15N imidazole coordination. This unique approach provides compelling evidence of a copper-stabilized ternary complex, with equatorial Cu2+ coordination formed by one histidine imidazole from Aβ and three from PrP. Moreover, the methodologies developed here provide a framework for assessing the copper environment in other interacting neurodegenerative proteins.
{"title":"Identifying the Copper Coordination Environment Between Interacting Neurodegenerative Proteins: A New Approach Using Pulsed EPR with <sup>14</sup>N/<sup>15</sup>N Isotopic Labelling.","authors":"Amanda Smart, Kevin Singewald, Zikri Hasanbasri, R David Britt, Glenn L Millhauser","doi":"10.1016/j.jbc.2025.108311","DOIUrl":"10.1016/j.jbc.2025.108311","url":null,"abstract":"<p><p>The trafficking and aggregation of neurodegenerative proteins often involves the interaction between intrinsically disordered domains, stabilized by the inclusion of physiologic metal ions such as copper or zinc. Characterizing the metal ion coordination environment is critical for assessing the stability and organization of these relevant protein-protein interactions but is challenging given the lack of regular molecular order or global structure. The cellular prion protein (PrP<sup>C</sup>) binds both monomers and aggregates of Alzheimer's amyloid-beta (Aβ), promoting Aβ internalization and aberrant signaling, respectively. Both proteins bind Cu<sup>2+</sup> with high affinity, opening the potential for copper to form an intermolecular bridge. We describe here a novel approach utilizing multiple EPR experiments to investigate the simultaneous Cu<sup>2+</sup> coordination of PrP<sup>C</sup> and Aβ in a 1:1:1 mixture. Uniformly <sup>15</sup>N-labeled PrP<sup>C</sup> is used in conjunction with natural abundance <sup>14</sup>N Aβ, the combination of which leads to distinct energy manifolds for paramagnetic Cu<sup>2+</sup> and resolved by the pulsed EPR experiments ESEEM and HYSCORE. We develop acquisition parameters to simultaneously optimize <sup>14</sup>N (I = 1) and <sup>15</sup>N (I = ½) pulsed EPR signals and we also advance the theory of ESEEM and HYSCORE to quantitatively describe multiple <sup>15</sup>N imidazole coordination. This unique approach provides compelling evidence of a copper-stabilized ternary complex, with equatorial Cu<sup>2+</sup> coordination formed by one histidine imidazole from Aβ and three from PrP. Moreover, the methodologies developed here provide a framework for assessing the copper environment in other interacting neurodegenerative proteins.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108311"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143425387","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13DOI: 10.1016/j.jbc.2025.108312
Xiaojuan Mi, Junjie Li, Ziqi Feng, Yanbo Liu, Chun Zhang, Yu Shao, Ting Wang, Zhilun Yang, Haowen Lv, Juan Liu
Microglial activation is the initial pathological event that occurs in demyelination, a prevalent feature in various neurological diseases. G protein-coupled estrogen receptor (GPER1), which is highly expressed in microglia, has been reported to reduce myelin damage. However, the precise molecular mechanisms involved remain unclear. In this study, the cuprizone (CPZ) -induced demyelination model was used to investigate the relationship between GPER1 and myelin sheath injury and its mechanism. The results demonstrated that GPER1 deficiency exacerbated cognitive impairment in mice. Along with more severe myelin damage as well as fewer oligodendrocytes. Moreover, GPER1 deficiency not only directly reduced the number of microglia in CPZ mice, but also caused iron ions overload in microglia of myelin debris induced in vitro. Transcriptomic, molecular biological, and morphological analyses revealed that microglial ferroptosis caused by GPER1 deficiency contributes to the reduction of microglia number. In summary, these findings revealed that GPER1 can regulate demyelination through ferroptosis of microglia.
{"title":"G protein-coupled estrogen receptor deficiency exacerbates demyelination through microglial ferroptosis.","authors":"Xiaojuan Mi, Junjie Li, Ziqi Feng, Yanbo Liu, Chun Zhang, Yu Shao, Ting Wang, Zhilun Yang, Haowen Lv, Juan Liu","doi":"10.1016/j.jbc.2025.108312","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108312","url":null,"abstract":"<p><p>Microglial activation is the initial pathological event that occurs in demyelination, a prevalent feature in various neurological diseases. G protein-coupled estrogen receptor (GPER1), which is highly expressed in microglia, has been reported to reduce myelin damage. However, the precise molecular mechanisms involved remain unclear. In this study, the cuprizone (CPZ) -induced demyelination model was used to investigate the relationship between GPER1 and myelin sheath injury and its mechanism. The results demonstrated that GPER1 deficiency exacerbated cognitive impairment in mice. Along with more severe myelin damage as well as fewer oligodendrocytes. Moreover, GPER1 deficiency not only directly reduced the number of microglia in CPZ mice, but also caused iron ions overload in microglia of myelin debris induced in vitro. Transcriptomic, molecular biological, and morphological analyses revealed that microglial ferroptosis caused by GPER1 deficiency contributes to the reduction of microglia number. In summary, these findings revealed that GPER1 can regulate demyelination through ferroptosis of microglia.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108312"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143425367","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13DOI: 10.1016/j.jbc.2025.108308
Matheus Brandemarte Severino, Ana Paula Morelli, Isadora Carolina Betim Pavan, Mariana Camargo Silva Mancini, Mariana Marcela Góis, Rafael Junqueira Borges, Renata Rosseto Braga, Luiz Guilherme Salvino da Silva, Nathalia Quintero-Ruiz, Maíra Maftoum Costa, Wesley de Lima Oliveira, Rosângela Maria Neves Bezerra, Eduardo Rochete Ropelle, Fernando Moreira Simabuco
Lung cancer presents the highest mortality rate in the world when compared to other cancer types and often presents chemotherapy resistance to cisplatin. The A549 non-small cell lung cancer (NSCLC) line is widely used as a model for lung adenocarcinoma studies since it presents a high proliferative rate and a nonsense mutation in the STK11 gene. The LKB1 protein, encoded by the STK11 gene, is one of the major regulators of cellular metabolism through AMPK activation under nutrient deprivation. Mutation in the STK11 gene in A549 cells potentiates cancer hallmarks, such as deregulation of cellular metabolism, aside from the Warburg effect, mTOR activation, autophagy inhibition, and NRF2 and redox activation. In this study, we investigated the integration of these pathways associated with the metabolism regulation by LKB1/AMPK to improve cisplatin response in the A549 cell line. We first used the CRISPR/Cas9 system to generate cell lines with a CRISPR-edited LKB1 isoform (called Super LKB1), achieved through the introduction of a +1 adenine insertion in the first exon of the STK11 gene after NHEJ mediated repair. This insertion led to the expression of a higher molecular weight protein containing an alternative exon described in the Peutz-Jeghers Syndrome (PJS). Through metabolic regulation by Super LKB1 expression and AMPK activation, we found an increase in autophagy flux (LC3 GFP/RFP p<0.05), as well as a reduction in the phosphorylation of mTORC1 downstream targets (S6K2 phospho-Serine 423; p<0.05; and S6 ribosomal protein phospho-Serine 240/244; p<0.03). The NRF2 protein exhibited increased levels and more nuclear localization in A549 WT (wild-type) cells compared to the edited cells (p<0.01). We also observed lower levels of H2O2 in the WT A549 cells, as a possible result of NRF2 activation, and a higher requirement of cisplatin to achieve the IC50 (WT: 10 μM; c2SL+: 5.5 μM; c3SL+: 6 μM). The data presented here suggests that the regulation of molecular pathways by the novel Super LKB1 in A549 cells related to metabolism, mTORC1, and autophagy promotes a better response of lung cancer cells to cisplatin. This NHEJ-CRISPR-based approach may be potentially used for lung cancer gene therapy.
{"title":"A CRISPR-edited isoform of the AMPK kinase LKB1 improves the response to cisplatin in A549 lung cancer cells.","authors":"Matheus Brandemarte Severino, Ana Paula Morelli, Isadora Carolina Betim Pavan, Mariana Camargo Silva Mancini, Mariana Marcela Góis, Rafael Junqueira Borges, Renata Rosseto Braga, Luiz Guilherme Salvino da Silva, Nathalia Quintero-Ruiz, Maíra Maftoum Costa, Wesley de Lima Oliveira, Rosângela Maria Neves Bezerra, Eduardo Rochete Ropelle, Fernando Moreira Simabuco","doi":"10.1016/j.jbc.2025.108308","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108308","url":null,"abstract":"<p><p>Lung cancer presents the highest mortality rate in the world when compared to other cancer types and often presents chemotherapy resistance to cisplatin. The A549 non-small cell lung cancer (NSCLC) line is widely used as a model for lung adenocarcinoma studies since it presents a high proliferative rate and a nonsense mutation in the STK11 gene. The LKB1 protein, encoded by the STK11 gene, is one of the major regulators of cellular metabolism through AMPK activation under nutrient deprivation. Mutation in the STK11 gene in A549 cells potentiates cancer hallmarks, such as deregulation of cellular metabolism, aside from the Warburg effect, mTOR activation, autophagy inhibition, and NRF2 and redox activation. In this study, we investigated the integration of these pathways associated with the metabolism regulation by LKB1/AMPK to improve cisplatin response in the A549 cell line. We first used the CRISPR/Cas9 system to generate cell lines with a CRISPR-edited LKB1 isoform (called Super LKB1), achieved through the introduction of a +1 adenine insertion in the first exon of the STK11 gene after NHEJ mediated repair. This insertion led to the expression of a higher molecular weight protein containing an alternative exon described in the Peutz-Jeghers Syndrome (PJS). Through metabolic regulation by Super LKB1 expression and AMPK activation, we found an increase in autophagy flux (LC3 GFP/RFP p<0.05), as well as a reduction in the phosphorylation of mTORC1 downstream targets (S6K2 phospho-Serine 423; p<0.05; and S6 ribosomal protein phospho-Serine 240/244; p<0.03). The NRF2 protein exhibited increased levels and more nuclear localization in A549 WT (wild-type) cells compared to the edited cells (p<0.01). We also observed lower levels of H<sub>2</sub>O<sub>2</sub> in the WT A549 cells, as a possible result of NRF2 activation, and a higher requirement of cisplatin to achieve the IC<sub>50</sub> (WT: 10 μM; c2SL+: 5.5 μM; c3SL+: 6 μM). The data presented here suggests that the regulation of molecular pathways by the novel Super LKB1 in A549 cells related to metabolism, mTORC1, and autophagy promotes a better response of lung cancer cells to cisplatin. This NHEJ-CRISPR-based approach may be potentially used for lung cancer gene therapy.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108308"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143425348","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13DOI: 10.1016/j.jbc.2025.108309
Lin Yi, Tiepeng Liao, Man Yuan, Qi Chen, Wei Xiong, Hongying Zhu
Metabolic alterations in the somatosensory cortex play a crucial role in neuropathic pain development, as evidenced by magnetic resonance spectroscopy and mass spectrometry analyses of brain homogenates. However, investigating metabolic changes in specific neuronal subtypes during neuropathic pain development remains challenging. Here, utilizing a recently developed technique called single-cell mass spectrometry (SCMS), we investigated metabolomic alterations within excitatory glutamatergic neurons located in the primary somatosensory cortex (S1) during various stages of neuropathic pain. Specifically, we induced neuropathic pain in mice using a spared nerve injury (SNI) model and observed activation of glutamatergic neurons in layer II/III of S1 through c-Fos staining and electrophysiology. We profiled metabolic changes and performed pathway enrichment analysis in these neurons by SCMS during both acute and sub-chronic phases of SNI. Further analyses revealed metabolites whose alterations significantly correlated with changes in pain thresholds, as well as distinct temporal patterns of metabolite expression during pain progression. From these analyses, we identified several key metabolites (homogentisic acid, phosphatidylcholine, phosphorylcholine and rhein) and validated their causal roles in pain modulation via pharmacological interventions. Thus, our study provides a valuable resource for elucidating the neurometabolic regulatory mechanisms underlying neuropathic pain from a single-cell perspective.
{"title":"Single-cell Metabolomics Profiling of Somatosensory Neurons in Various stages of Neuropathic Pain.","authors":"Lin Yi, Tiepeng Liao, Man Yuan, Qi Chen, Wei Xiong, Hongying Zhu","doi":"10.1016/j.jbc.2025.108309","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108309","url":null,"abstract":"<p><p>Metabolic alterations in the somatosensory cortex play a crucial role in neuropathic pain development, as evidenced by magnetic resonance spectroscopy and mass spectrometry analyses of brain homogenates. However, investigating metabolic changes in specific neuronal subtypes during neuropathic pain development remains challenging. Here, utilizing a recently developed technique called single-cell mass spectrometry (SCMS), we investigated metabolomic alterations within excitatory glutamatergic neurons located in the primary somatosensory cortex (S1) during various stages of neuropathic pain. Specifically, we induced neuropathic pain in mice using a spared nerve injury (SNI) model and observed activation of glutamatergic neurons in layer II/III of S1 through c-Fos staining and electrophysiology. We profiled metabolic changes and performed pathway enrichment analysis in these neurons by SCMS during both acute and sub-chronic phases of SNI. Further analyses revealed metabolites whose alterations significantly correlated with changes in pain thresholds, as well as distinct temporal patterns of metabolite expression during pain progression. From these analyses, we identified several key metabolites (homogentisic acid, phosphatidylcholine, phosphorylcholine and rhein) and validated their causal roles in pain modulation via pharmacological interventions. Thus, our study provides a valuable resource for elucidating the neurometabolic regulatory mechanisms underlying neuropathic pain from a single-cell perspective.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108309"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143424499","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-12DOI: 10.1016/j.jbc.2025.108306
Jessie Lee Cunningham, Hsing-Yin Liu, Jamie Francisco, Karla K Frietze, J Jose Corbalan, Joseph T Nickels
Evidence suggests that ARV1 regulates sterol movement within the cell. Saccharomyces cerevisiae cells lacking ScArv1 have defects in sterol trafficking, distribution, and biosynthesis. HepG2 cells treated with hARV1 anti-sense oligonucleotides accumulate cholesterol in the endoplasmic reticulum. Mice lacking Arv1 have a lean phenotype when fed a high fat diet and show no signs of liver triglyceride or cholesterol accumulation, suggesting a role for Arv1 in lipid transport. Here, we explored the direct lipid binding activity of recombinant human ARV1 using in vitro lipid binding assays. ARV1 lipid binding activity was observed within the first N-terminal 98 amino acids containing the conserved ARV1 homology domain (AHD). The zinc-binding domain and conserved cysteine clusters within the AHD were necessary for lipid binding. Both full-length ARV1 and the AHD bound cholesterol, several phospholipids, and phosphoinositides with high affinity. The AHD showed the highest binding affinity for monophosphorylated phosphoinositides. Several conserved amino acids within the AHD were necessary for phospholipid binding. Biochemical studies suggested that ARV1 exists as a dimer in cells, with oligomerization being critical for ARV1 function, as amino acid mutations predicted to have a negative effect on dimerization cause weakened or complete loss of lipid binding. Our results show for the first time that human ARV1 can directly bind cholesterol and phospholipids. How this activity may function to regulate lipid binding and maintain proper lipid trafficking and/or transport in cells requires further studies.
{"title":"The sterol-regulating human ARV1 binds cholesterol and phospholipids through its conserved ARV1 homology domain.","authors":"Jessie Lee Cunningham, Hsing-Yin Liu, Jamie Francisco, Karla K Frietze, J Jose Corbalan, Joseph T Nickels","doi":"10.1016/j.jbc.2025.108306","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108306","url":null,"abstract":"<p><p>Evidence suggests that ARV1 regulates sterol movement within the cell. Saccharomyces cerevisiae cells lacking ScArv1 have defects in sterol trafficking, distribution, and biosynthesis. HepG2 cells treated with hARV1 anti-sense oligonucleotides accumulate cholesterol in the endoplasmic reticulum. Mice lacking Arv1 have a lean phenotype when fed a high fat diet and show no signs of liver triglyceride or cholesterol accumulation, suggesting a role for Arv1 in lipid transport. Here, we explored the direct lipid binding activity of recombinant human ARV1 using in vitro lipid binding assays. ARV1 lipid binding activity was observed within the first N-terminal 98 amino acids containing the conserved ARV1 homology domain (AHD). The zinc-binding domain and conserved cysteine clusters within the AHD were necessary for lipid binding. Both full-length ARV1 and the AHD bound cholesterol, several phospholipids, and phosphoinositides with high affinity. The AHD showed the highest binding affinity for monophosphorylated phosphoinositides. Several conserved amino acids within the AHD were necessary for phospholipid binding. Biochemical studies suggested that ARV1 exists as a dimer in cells, with oligomerization being critical for ARV1 function, as amino acid mutations predicted to have a negative effect on dimerization cause weakened or complete loss of lipid binding. Our results show for the first time that human ARV1 can directly bind cholesterol and phospholipids. How this activity may function to regulate lipid binding and maintain proper lipid trafficking and/or transport in cells requires further studies.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108306"},"PeriodicalIF":4.0,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143424888","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-12DOI: 10.1016/j.jbc.2025.108292
Runchuan Yan, Yan Zhang, Hui Zhang, Jiyan Ma
Secretory protein expression in mammalian cells is widely used in various fields, including biomedical research and biopharmaceutical production. However, achieving high-level expression of certain secretory proteins / peptides can be challenging. The naturally occurring N1 fragment of the prion protein is one of these difficult-to-produce secretory proteins, which hinders our understanding of its biological functions and limits its potential as a therapeutic molecule. To improve N1 production, we screened several well-folded protein domains and found that fusing N1 with a camelid nanobody (Nb) improved its translocation into the endoplasmic reticulum and significantly enhanced its secretion. Nb-fusion does not alter the translocation mechanism, which remains dependent on the Sec61/Sec62/Sec63 complex. This approach also resulted in a significant increase in N1 production in the mouse brain using recombinant adeno-associated virus. Furthermore, fusing Nb to another unstructured protein, Shadoo (without GPI anchor), or a peptide hormone, somatostatin, also greatly increased their production, demonstrating the applicability of this approach to other proteins and peptides. The enhancement of N1 production is comparable or better than Fc fusion, and the effect is observed with all tested camelid Nb, but not with a shark Nb and to a lesser extent, with a human immunoglobulin heavy chain variable region. Importantly, the Nb in the fusion protein retained its antigen-binding capability, paving the way for the development of a dual-functional protein. Collectively, we present a novel strategy for enhancing the production of secretory proteins, which holds great promise in creating functional biological molecules for a wide range of applications.
在哺乳动物细胞中表达分泌蛋白被广泛应用于各个领域,包括生物医学研究和生物制药生产。然而,实现某些分泌蛋白/肽的高水平表达具有挑战性。天然存在的朊病毒蛋白 N1 片段就是这些难以生产的分泌蛋白之一,这阻碍了我们对其生物功能的了解,并限制了其作为治疗分子的潜力。为了提高 N1 的产量,我们筛选了几个折叠良好的蛋白结构域,发现将 N1 与驼科纳米抗体(Nb)融合可改善其在内质网中的转位,并显著增强其分泌能力。Nb融合不会改变转运机制,该机制仍然依赖于Sec61/Sec62/Sec63复合物。这种方法还导致使用重组腺相关病毒的小鼠大脑中的 N1 产量显著增加。此外,将 Nb 与另一种非结构化蛋白质 Shadoo(不含 GPI 锚)或肽类激素 somatostatin 融合,也大大提高了它们的产量,这表明这种方法适用于其他蛋白质和肽类。N1 产量的提高效果与 Fc 融合效果相当或更好,所有测试过的驼科 Nb 都能观察到这种效果,但鲨鱼 Nb 和人类免疫球蛋白重链可变区的效果较差。重要的是,融合蛋白中的 Nb 保留了其抗原结合能力,为开发双功能蛋白铺平了道路。总之,我们提出了一种提高分泌蛋白产量的新策略,它为创造功能性生物分子的广泛应用带来了巨大希望。
{"title":"Nanobody fusion enhances production of difficult-to-produce secretory proteins.","authors":"Runchuan Yan, Yan Zhang, Hui Zhang, Jiyan Ma","doi":"10.1016/j.jbc.2025.108292","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108292","url":null,"abstract":"<p><p>Secretory protein expression in mammalian cells is widely used in various fields, including biomedical research and biopharmaceutical production. However, achieving high-level expression of certain secretory proteins / peptides can be challenging. The naturally occurring N1 fragment of the prion protein is one of these difficult-to-produce secretory proteins, which hinders our understanding of its biological functions and limits its potential as a therapeutic molecule. To improve N1 production, we screened several well-folded protein domains and found that fusing N1 with a camelid nanobody (Nb) improved its translocation into the endoplasmic reticulum and significantly enhanced its secretion. Nb-fusion does not alter the translocation mechanism, which remains dependent on the Sec61/Sec62/Sec63 complex. This approach also resulted in a significant increase in N1 production in the mouse brain using recombinant adeno-associated virus. Furthermore, fusing Nb to another unstructured protein, Shadoo (without GPI anchor), or a peptide hormone, somatostatin, also greatly increased their production, demonstrating the applicability of this approach to other proteins and peptides. The enhancement of N1 production is comparable or better than Fc fusion, and the effect is observed with all tested camelid Nb, but not with a shark Nb and to a lesser extent, with a human immunoglobulin heavy chain variable region. Importantly, the Nb in the fusion protein retained its antigen-binding capability, paving the way for the development of a dual-functional protein. Collectively, we present a novel strategy for enhancing the production of secretory proteins, which holds great promise in creating functional biological molecules for a wide range of applications.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108292"},"PeriodicalIF":4.0,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143425450","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Vitiligo, an autoimmune disease caused by environmental and genetic factors, is characterized by the specific loss of epidermal melanocytes (MCs). IFN-γ, predominantly derived from MC-targeting CD8+ T cells, plays a key role in vitiligo pathogenesis. Previously, we found that glycoprotein nonmetastatic melanoma protein B (GPNMB) is specifically lost in the basal epidermal layer of vitiligo lesions and downregulated by IFN-γ in normal human epidermal keratinocytes (KCs) (NHEKs). This study aimed to determine the role of KC GPNMB in normal and vitiligo epidermis and demonstrated that GPNMB plays a protective role against H2O2-induced oxidative stress due to its extracellular domain. In contrast, the NRF2/KEEP1 system was not involved in the anti-oxidative response in NHEKs but was active in MCs. GPNMB knockdown reduced the phosphorylation levels of AKTT308 and AKTS473 after H2O2 treatment, accompanied by reduced Dickkopf-1 (DKK1) mRNA and protein production and decreased FOXM1 mRNA expression. These results suggested that GPNMB protects KCs from H2O2-induced cell death through enhanced PI3K/AKT signaling, and WNT/β-catenin/FOXM1 and DKK1/CKAP4/AKT pathways. Furthermore, a significant increase in thioredoxin-interacting protein (TXNIP) following GPNMB knockdown was observed, indicating the enhanced phosphorylation of JNK and p38 and suppression of WNT/β-catenin signaling. These results suggest that the decreased expression of epidermal GPNMB in vitiligo lesions triggers increased sensitivity to H2O2-induced oxidative stress and decreased WNT/β-catenin signaling, consistent with the pathological features of the vitiligo epidermis. These findings may enhance our understanding of vitiligo pathogenesis, provide insights into the reduced risk of epidermal cancers, and highlight novel targets for treatment.
{"title":"The glycoprotein GPNMB protects against oxidative stress through enhanced PI3K/Akt signaling in epidermal keratinocytes.","authors":"Natsuki Nishida, Mariko Otsu, Yukiko Mizutani, Asako Ishitsuka, Yoichi Mizukami, Shintaro Inoue","doi":"10.1016/j.jbc.2025.108299","DOIUrl":"https://doi.org/10.1016/j.jbc.2025.108299","url":null,"abstract":"<p><p>Vitiligo, an autoimmune disease caused by environmental and genetic factors, is characterized by the specific loss of epidermal melanocytes (MCs). IFN-γ, predominantly derived from MC-targeting CD8<sup>+</sup> T cells, plays a key role in vitiligo pathogenesis. Previously, we found that glycoprotein nonmetastatic melanoma protein B (GPNMB) is specifically lost in the basal epidermal layer of vitiligo lesions and downregulated by IFN-γ in normal human epidermal keratinocytes (KCs) (NHEKs). This study aimed to determine the role of KC GPNMB in normal and vitiligo epidermis and demonstrated that GPNMB plays a protective role against H<sub>2</sub>O<sub>2</sub>-induced oxidative stress due to its extracellular domain. In contrast, the NRF2/KEEP1 system was not involved in the anti-oxidative response in NHEKs but was active in MCs. GPNMB knockdown reduced the phosphorylation levels of AKT<sup>T308</sup> and AKT<sup>S473</sup> after H<sub>2</sub>O<sub>2</sub> treatment, accompanied by reduced Dickkopf-1 (DKK1) mRNA and protein production and decreased FOXM1 mRNA expression. These results suggested that GPNMB protects KCs from H<sub>2</sub>O<sub>2</sub>-induced cell death through enhanced PI3K/AKT signaling, and WNT/β-catenin/FOXM1 and DKK1/CKAP4/AKT pathways. Furthermore, a significant increase in thioredoxin-interacting protein (TXNIP) following GPNMB knockdown was observed, indicating the enhanced phosphorylation of JNK and p38 and suppression of WNT/β-catenin signaling. These results suggest that the decreased expression of epidermal GPNMB in vitiligo lesions triggers increased sensitivity to H<sub>2</sub>O<sub>2</sub>-induced oxidative stress and decreased WNT/β-catenin signaling, consistent with the pathological features of the vitiligo epidermis. These findings may enhance our understanding of vitiligo pathogenesis, provide insights into the reduced risk of epidermal cancers, and highlight novel targets for treatment.</p>","PeriodicalId":15140,"journal":{"name":"Journal of Biological Chemistry","volume":" ","pages":"108299"},"PeriodicalIF":4.0,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143414358","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}