1
|
Wang D, Chu WK, Yam JCS, Pang CP, Leung YC, Shum ASW, Chan SO. GCN2-SLC7A11 axis coordinates autophagy, cell cycle and apoptosis and regulates cell growth in retinoblastoma upon arginine deprivation. Cancer Metab 2024; 12:31. [PMID: 39462426 PMCID: PMC11515237 DOI: 10.1186/s40170-024-00361-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Arginine deprivation was previously shown to inhibit retinoblastoma cell proliferation and induce cell death in vitro. However, the mechanisms by which retinoblastoma cells respond to arginine deprivation remain to be elucidated. METHODS The human-derived retinoblastoma cell lines Y79 and WERI-Rb-1 were subjected to arginine depletion, and the effects on inhibiting cell growth and survival were evaluated. This study investigated potential mechanisms, including autophagy, cell cycle arrest and apoptosis. Moreover, the roles of the general control nonderepressible 2 (GCN2) and mechanistic target of rapamycin complex 1 (mTORC1) signaling pathways in these processes were examined. RESULTS We demonstrated that arginine deprivation effectively inhibited the growth of retinoblastoma cells in vitro. This treatment caused an increase in the autophagic response. Additionally, prolonged arginine deprivation induced G2 cell cycle arrest and was accompanied by an increase in early apoptotic cells. Importantly, arginine depletion also induced the activation of GCN2 and the inhibition of mTOR signaling. We also discovered that the activation of SLC7A11 was regulated by GCN2 upon arginine deprivation. Knockdown of SLC7A11 rendered retinoblastoma cells partially resistant to arginine deprivation. Furthermore, we found that knockdown of GCN2 led to a decrease in the autophagic response in WERI-Rb-1 cells and arrested more cells in S phase, which was accompanied by fewer apoptotic cells. Moreover, knockdown of GCN2 induced the constant expression of ATF4 and the phosphorylation of 70S6K and 4E-BP1 regardless of arginine deprivation. CONCLUSIONS Collectively, our findings suggest that the GCN2‒SLC7A11 axis regulates cell growth and survival upon arginine deprivation through coordinating autophagy, cell cycle arrest, and apoptosis in retinoblastoma cells. This work paves the way for the development of a novel treatment for retinoblastoma.
Collapse
Affiliation(s)
- Dan Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wai Kit Chu
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Hub of Pediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jason Cheuk Sing Yam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Hub of Pediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology, Hong Kong Children's Hospital, Hong Kong SAR, China
- Hong Kong Eye Hospital, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Hub of Pediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yun Chung Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Alisa Sau Wun Shum
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Sun-On Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
2
|
Canè S, Geiger R, Bronte V. The roles of arginases and arginine in immunity. Nat Rev Immunol 2024:10.1038/s41577-024-01098-2. [PMID: 39420221 DOI: 10.1038/s41577-024-01098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Arginase activity and arginine metabolism in immune cells have important consequences for health and disease. Their dysregulation is commonly observed in cancer, autoimmune disorders and infectious diseases. Following the initial description of a role for arginase in the dysfunction of T cells mounting an antitumour response, numerous studies have broadened our understanding of the regulation and expression of arginases and their integration with other metabolic pathways. Here, we highlight the differences in arginase compartmentalization and storage between humans and rodents that should be taken into consideration when assessing the effects of arginase activity. We detail the roles of arginases, arginine and its metabolites in immune cells and their effects in the context of cancer, autoimmunity and infectious disease. Finally, we explore potential therapeutic strategies targeting arginases and arginine.
Collapse
Affiliation(s)
- Stefania Canè
- The Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Roger Geiger
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Università della Svizzera italiana, Bellinzona, Switzerland
| | | |
Collapse
|
3
|
Pavlova NN, Thompson CB. Oncogenic Control of Metabolism. Cold Spring Harb Perspect Med 2024; 14:a041531. [PMID: 38565265 PMCID: PMC11444253 DOI: 10.1101/cshperspect.a041531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
A cell committed to proliferation must reshape its metabolism to enable robust yet balanced production of building blocks for the assembly of proteins, lipids, nucleic acids, and other macromolecules, from which two functional daughter cells can be produced. The metabolic remodeling associated with proliferation is orchestrated by a number of pro-proliferative signaling nodes, which include phosphatidylinositol-3 kinase (PI3K), the RAS family of small GTPases, and transcription factor c-myc In metazoan cells, these signals are activated in a paracrine manner via growth factor-mediated activation of receptor (or receptor-associated) tyrosine kinases. Such stimuli are limited in duration and therefore allow the metabolism of target cells to return to the resting state once the proliferation demands have been satisfied. Cancer cells acquire activating genetic alterations within common pro-proliferative signaling nodes. These alterations lock cellular nutrient uptake and utilization into a perpetual progrowth state, leading to the aberrant accumulation and spread of cancer cells.
Collapse
Affiliation(s)
- Natalya N Pavlova
- Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
4
|
Groessl S, Palm W. Nutrient Levels and Nutrient Sources in Pancreatic Tumors. Cancer Res 2024; 84:2947-2949. [PMID: 39279381 DOI: 10.1158/0008-5472.can-24-2447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 09/18/2024]
Abstract
It has been known that poor tumor perfusion and dysregulated cancer cell metabolism give rise to tumor microenvironments with unphysiologic nutrient levels, but the precise alterations in metabolite abundance are not well defined. In a 2015 study in Cancer Research, Kamphorst and colleagues published a detailed comparison of the metabolome from human pancreatic tumors and benign tissues. Tumors were depleted in glucose and various nonessential amino acids but, surprisingly, enriched in essential amino acids. The authors attributed these nutrient imbalances to macropinocytosis of extracellular proteins, a RAS-driven amino acid acquisition pathway that was found to be increased in human tumors and supports pancreatic cancer cell growth during amino acid starvation. These findings substantially contributed to the understanding of altered nutrient levels in tumors and extracellular proteins as noncanonical nutrients. Intratumoral nutrient levels in different cancer contexts and signaling pathways that regulate nutrient acquisition by cancer cells remain a focus of current research. See related article by Kamphorst and colleagues, Cancer Res 2015;75:544-53.
Collapse
Affiliation(s)
- Sven Groessl
- Division of Cell Signaling and Metabolism, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Wilhelm Palm
- Division of Cell Signaling and Metabolism, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
5
|
Pan H, Sun Y, Qian LH, Liao YN, Gai YZ, Huo YM, Li ZQ, Nie HZ. A Nutrient-Deficient Microenvironment Facilitates Ferroptosis Resistance via the FAM60A-PPAR Axis in Pancreatic Ductal Adenocarcinoma. RESEARCH (WASHINGTON, D.C.) 2024; 7:0300. [PMID: 38314086 PMCID: PMC10836236 DOI: 10.34133/research.0300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024]
Abstract
Ferroptosis, a nonapoptotic form of cell death, is an emerging potential therapeutic target for various diseases, including cancer. However, the role of ferroptosis in pancreatic cancer remains poorly understood. Pancreatic ductal adenocarcinoma (PDAC) is characterized by a poor prognosis and chemotherapy resistance, attributed to its high Kirsten rats arcomaviral oncogene homolog mutation rate and severe nutritional deficits resulting from a dense stroma. Several studies have linked rat sarcoma (RAS) mutations to ferroptosis, suggesting that inducing ferroptosis may be an effective strategy against oncogenic RAS-bearing tumors. We investigated the role of Family With Sequence Similarity 60 Member A (FAM60A) in this study, a protein closely associated with a poor prognosis and highly expressed in PDAC and tumor tissue from KrasG12D/+;Trp53R172H/+; Pdx1-Cre mice, in regulating ferroptosis, tumor growth, and gemcitabine sensitivity in vitro and in vivo. Our results demonstrate that FAM60A regulates 3 essential metabolic enzymes, ACSL1/4 and GPX4, to protect PDAC cells from ferroptosis. Furthermore, we found that YY1 transcriptionally regulates FAM60A expression by promoting its transcription, and the Hippo-YY1 pathway is restricted in the low-amino-acid milieu in the context of nutrient deprivation, leading to downstream suppression of peroxisome proliferator-activated receptor and ACSL1/4 and activation of GPX4 pathways. Importantly, FAM60A knockdown sensitized PDAC cells to gemcitabine treatment. A new understanding of FAM60A transcriptional regulation pattern in PDAC and its dual function in ferroptosis reliever and chemotherapy resistance is provided by our study. Targeting FAM60A may therefore offer a promising therapeutic approach for PDAC by simultaneously addressing 2 major features of the disease (high RAS mutation rate and tumor microenvironment nutrient deficiency) and preventing tumor cell metabolic adaptation.
Collapse
Affiliation(s)
- Hong Pan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yue Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Li-Heng Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying-Na Liao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan-Zhi Gai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan-Miao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zuo-Qing Li
- Innomodels Biotechnology Co., Ltd., 51 Xinpei Road, Jiading District, Shanghai, China
| | - Hui-Zhen Nie
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
6
|
Lambies G, Lee SW, Duong-Polk K, Aza-Blanc P, Maganti S, Dawson DW, Commisso C. Cell polarity proteins promote macropinocytosis in response to metabolic stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575943. [PMID: 38293142 PMCID: PMC10827152 DOI: 10.1101/2024.01.16.575943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Macropinocytosis has emerged as a nutrient-scavenging pathway that cancer cells exploit to survive the nutrient-deprived conditions of the tumor microenvironment. Cancer cells are especially reliant on glutamine for their survival, and in pancreatic ductal adenocarcinoma (PDAC) cells, glutamine deficiency can enhance the stimulation of macropinocytosis, allowing the cells to escape metabolic stress through the production of extracellular-protein-derived amino acids. Here, we identify the atypical protein kinase C (aPKC) enzymes, PKCζ and PKCι as novel regulators of macropinocytosis. In normal epithelial cells, aPKCs are known to regulate cell polarity in association with the scaffold proteins Par3 and Par6, controlling the function of several targets, including the Par1 kinases. In PDAC cells, we identify that each of these cell polarity proteins are required for glutamine stress-induced macropinocytosis. Mechanistically, we find that the aPKCs are regulated by EGFR signaling or by the transcription factor CREM to promote the relocation of Par3 to microtubules, facilitating macropinocytosis in a dynein-dependent manner. Importantly, we determine that cell fitness impairment caused by aPKC depletion is rescued by the restoration of macropinocytosis and that aPKCs support PDAC growth in vivo. These results identify a previously unappreciated role for cell polarity proteins in the regulation of macropinocytosis and provide a better understanding of the mechanistic underpinnings that control macropinocytic uptake in the context of metabolic stress.
Collapse
|
7
|
Nofi CP, Tan C, Ma G, Kobritz M, Prince JM, Wang H, Aziz M, Wang P. A novel opsonic eCIRP inhibitor for lethal sepsis. J Leukoc Biol 2024; 115:385-400. [PMID: 37774691 PMCID: PMC10799304 DOI: 10.1093/jleuko/qiad119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
Sepsis is a life-threatening inflammatory condition partly orchestrated by the release of various damage-associated molecular patterns such as extracellular cold-inducible RNA-binding protein (eCIRP). Despite advances in understanding the pathogenic role of eCIRP in inflammatory diseases, novel therapeutic strategies to prevent its excessive inflammatory response are lacking. Milk fat globule-epidermal growth factor-VIII (MFG-E8) is critical for the opsonic clearance of apoptotic cells, but its potential involvement in the removal of eCIRP was previously unknown. Here, we report that MFG-E8 can strongly bind eCIRP to facilitate αvβ3-integrin-dependent internalization and lysosome-dependent degradation of MFG-E8/eCIRP complexes, thereby attenuating excessive inflammation. Genetic disruption of MFG-E8 expression exaggerated sepsis-induced systemic accumulation of eCIRP and other cytokines, and consequently exacerbated sepsis-associated acute lung injury. In contrast, MFG-E8-derived oligopeptide recapitulated its eCIRP binding properties, and significantly attenuated eCIRP-induced inflammation to confer protection against sepsis. Our findings suggest a novel therapeutic approach to attenuate eCIRP-induced inflammation to improve outcomes of lethal sepsis.
Collapse
Affiliation(s)
- Colleen P Nofi
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Chuyi Tan
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Gaifeng Ma
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Molly Kobritz
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Jose M Prince
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Haichao Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| | - Ping Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, United States
| |
Collapse
|
8
|
Nazemi M, Yanes B, Martinez ML, Walker HJ, Pham K, Collins MO, Bard F, Rainero E. The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism. PLoS Biol 2024; 22:e3002406. [PMID: 38227562 PMCID: PMC10791009 DOI: 10.1371/journal.pbio.3002406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/26/2023] [Indexed: 01/18/2024] Open
Abstract
Breast tumours are embedded in a collagen I-rich extracellular matrix (ECM) network, where nutrients are scarce due to limited blood flow and elevated tumour growth. Metabolic adaptation is required for cancer cells to endure these conditions. Here, we demonstrated that the presence of ECM supported the growth of invasive breast cancer cells, but not non-transformed mammary epithelial cells, under amino acid starvation, through a mechanism that required macropinocytosis-dependent ECM uptake. Importantly, we showed that this behaviour was acquired during carcinoma progression. ECM internalisation, followed by lysosomal degradation, contributed to the up-regulation of the intracellular levels of several amino acids, most notably tyrosine and phenylalanine. This resulted in elevated tyrosine catabolism on ECM under starvation, leading to increased fumarate levels, potentially feeding into the tricarboxylic acid (TCA) cycle. Interestingly, this pathway was required for ECM-dependent cell growth and invasive cell migration under amino acid starvation, as the knockdown of p-hydroxyphenylpyruvate hydroxylase-like protein (HPDL), the third enzyme of the pathway, opposed cell growth and motility on ECM in both 2D and 3D systems, without affecting cell proliferation on plastic. Finally, high HPDL expression correlated with poor prognosis in breast cancer patients. Collectively, our results highlight that the ECM in the tumour microenvironment (TME) represents an alternative source of nutrients to support cancer cell growth by regulating phenylalanine and tyrosine metabolism.
Collapse
Affiliation(s)
- Mona Nazemi
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Bian Yanes
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Montserrat Llanses Martinez
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Institute of Molecular and Cell Biology, Singapore
| | - Heather J. Walker
- biOMICS Facility, School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Khoa Pham
- biOMICS Facility, School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Mark O. Collins
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- biOMICS Facility, School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Frederic Bard
- Institute of Molecular and Cell Biology, Singapore
- Centre de Recherche en Cancerologie de Marseille, CRCM, Marseille, France
| | - Elena Rainero
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
9
|
Wang T, Zhang Y, Liu Y, Huang Y, Wang W. Amino Acid-Starved Cancer Cells Utilize Macropinocytosis and Ubiquitin-Proteasome System for Nutrient Acquisition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304791. [PMID: 37983609 PMCID: PMC10767443 DOI: 10.1002/advs.202304791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/06/2023] [Indexed: 11/22/2023]
Abstract
To grow in nutrient-deprived tumor microenvironment, cancer cells often internalize and degrade extracellular proteins to refuel intracellular amino acids. However, the nutrient acquisition routes reported by previous studies are mainly restricted in autophagy-lysosomal pathway. It remains largely unknown if other protein degradation systems also contribute to the utilization of extracellular nutrients. Herein, it is demonstrated that under amino acid starvation, extracellular protein internalization through macropinocytosis and protein degradation through ubiquitin-proteasome system are activated as a nutrient supply route, sensitizing cancer cells to proteasome inhibition. By inhibiting both macropinocytosis and ubiquitin-proteasome system, an innovative approach to intensify amino acid starvation for cancer therapy is presented. To maximize therapeutic efficacy and minimize systemic side effects, a pH-responsive polymersome nanocarrier is developed to deliver therapeutic agents specifically to tumor tissues. This nanoparticle system provides an approach to exacerbate amino acid starvation for cancer therapy, which represents a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Tianyi Wang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Yaming Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Yuwei Liu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Yi Huang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| |
Collapse
|
10
|
Zhang C, Shafaq-Zadah M, Pawling J, Hesketh GG, Dransart E, Pacholczyk K, Longo J, Gingras AC, Penn LZ, Johannes L, Dennis JW. SLC3A2 N-glycosylation and Golgi remodeling regulate SLC7A amino acid exchangers and stress mitigation. J Biol Chem 2023; 299:105416. [PMID: 37918808 PMCID: PMC10698284 DOI: 10.1016/j.jbc.2023.105416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023] Open
Abstract
Proteostasis requires oxidative metabolism (ATP) and mitigation of the associated damage by glutathione, in an increasingly dysfunctional relationship with aging. SLC3A2 (4F2hc, CD98) plays a role as a disulfide-linked adaptor to the SLC7A5 and SLC7A11 exchangers which import essential amino acids and cystine while exporting Gln and Glu, respectively. The positions of N-glycosylation sites on SLC3A2 have evolved with the emergence of primates, presumably in synchrony with metabolism. Herein, we report that each of the four sites in SLC3A2 has distinct profiles of Golgi-modified N-glycans. N-glycans at the primate-derived site N381 stabilized SLC3A2 in the galectin-3 lattice against coated-pit endocytosis, while N365, the site nearest the membrane promoted glycolipid-galectin-3 (GL-Lect)-driven endocytosis. Our results indicate that surface retention and endocytosis are precisely balanced by the number, position, and remodeling of N-glycans on SLC3A2. Furthermore, proteomics and functional assays revealed an N-glycan-dependent clustering of the SLC3A2∗SLC7A5 heterodimer with amino-acid/Na+ symporters (SLC1A4, SLC1A5) that balances branched-chain amino acids and Gln levels, at the expense of ATP to maintain the Na+/K+ gradient. In replete conditions, SLC3A2 interactions require Golgi-modified N-glycans at N365D and N381D, whereas reducing N-glycosylation in the endoplasmic reticulum by fluvastatin treatment promoted the recruitment of CD44 and transporters needed to mitigate stress. Thus, SLC3A2 N-glycosylation and Golgi remodeling of the N-glycans have distinct roles in amino acids import for growth, maintenance, and metabolic stresses.
Collapse
Affiliation(s)
- Cunjie Zhang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto Ontario, Canada
| | - Massiullah Shafaq-Zadah
- Cellular and Chemical Biology Unit, Institut Curie, INSERM U1143, CNRS UMR3666, PSL Research University, Paris, France
| | - Judy Pawling
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto Ontario, Canada
| | - Geoffrey G Hesketh
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto Ontario, Canada
| | - Estelle Dransart
- Cellular and Chemical Biology Unit, Institut Curie, INSERM U1143, CNRS UMR3666, PSL Research University, Paris, France
| | - Karina Pacholczyk
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto Ontario, Canada
| | - Joseph Longo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Linda Z Penn
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Ludger Johannes
- Cellular and Chemical Biology Unit, Institut Curie, INSERM U1143, CNRS UMR3666, PSL Research University, Paris, France
| | - James W Dennis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Qu W, Zhou X, Jiang X, Xie X, Xu K, Gu X, Na R, Piao M, Xi X, Sun N, Wang X, Peng X, Xu J, Tian J, Zhang J, Guo J, Zhang M, Zhang Y, Pan Z, Wang K, Yu B, Sun B, Li S, Tian J. Long Noncoding RNA Gpr137b-ps Promotes Advanced Atherosclerosis via the Regulation of Autophagy in Macrophages. Arterioscler Thromb Vasc Biol 2023; 43:e468-e489. [PMID: 37767704 DOI: 10.1161/atvbaha.123.319037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Current therapies cannot completely reverse advanced atherosclerosis. High levels of amino acids, induced by Western diet, stimulate mTORC1 (mammalian target of rapamycin complex 1)-autophagy defects in macrophages, accelerating atherosclerotic plaque progression. In addition, autophagy-lysosomal dysfunction contributes to plaque necrotic core enlargement and lipid accumulation. Therefore, it is essential to investigate the novel mechanism and molecules to reverse amino acid-mTORC1-autophagy signaling dysfunction in macrophages of patients with advanced atherosclerosis. METHODS We observed that Gpr137b-ps (G-protein-coupled receptor 137B, pseudogene) was upregulated in advanced atherosclerotic plaques. The effect of Gpr137b-ps on the progression of atherosclerosis was studied by generating advanced plaques in ApoE-/- mice with cardiac-specific knockout of Gpr137b-ps. Bone marrow-derived macrophages and mouse mononuclear macrophage cell line RAW264.7 cells were subjected to starvation or amino acid stimulation to study amino acid-mTORC1-autophagy signaling. Using both gain- and loss-of-function approaches, we explored the mechanism of Gpr137b-ps-regulated autophagy. RESULTS Our results demonstrated that Gpr137b-ps deficiency led to enhanced autophagy in macrophages and reduced atherosclerotic lesions, characterized by fewer necrotic cores and less lipid accumulation. Knockdown of Gpr137b-ps increased autophagy and prevented amino acid-induced mTORC1 signaling activation. As the downstream binding protein of Gpr137b-ps, HSC70 (heat shock cognate 70) rescued the impaired autophagy induced by Gpr137b-ps. Furthermore, Gpr137b-ps interfered with the HSC70 binding to G3BP (Ras GTPase-activating protein-binding protein), which tethers the TSC (tuberous sclerosis complex) complex to lysosomes and suppresses mTORC1 signaling. In addition to verifying that the NTF2 (nuclear transport factor 2) domain of G3BP binds to HSC70 by in vitro protein synthesis, we further demonstrated that HSC70 binds to the NTF2 domain of G3BP through its W90-F92 motif by using computational modeling. CONCLUSIONS These findings reveal that Gpr137b-ps plays an essential role in the regulation of macrophage autophagy, which is crucial for the progression of advanced atherosclerosis. Gpr137b-ps impairs the interaction of HSC70 with G3BP to regulate amino acid-mTORC1-autophagy signaling, and these results provide a new potential therapeutic direction for the treatment of advanced atherosclerosis.
Collapse
Affiliation(s)
- Wenbo Qu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xin Zhou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xinjian Jiang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xianwei Xie
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China (X. Xie)
| | - Kaijian Xu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xia Gu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Ruisi Na
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, China (R.N.)
| | - Minghui Piao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xiangwen Xi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Na Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xueyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xiang Peng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Junyan Xu
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China (J.X.)
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China (J.X., J.G.)
| | - Jiangtian Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Jian Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology (J.Z.)
| | - Junli Guo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China (J.X., J.G.)
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Yao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Zhenwei Pan
- College of Pharmacy (Z.P., B.S.), Harbin Medical University, China
| | - Kun Wang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (K.W.)
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Bin Sun
- College of Pharmacy (Z.P., B.S.), Harbin Medical University, China
| | - Shuijie Li
- Department of Biopharmaceutical Sciences, College of Pharmacy (S.L.), Harbin Medical University, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases Harbin Medical University, China (S.L.)
- Department of Biopharmaceutical Sciences, College of Pharmacy Harbin Medical University, China (S.L.)
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| |
Collapse
|
12
|
Sniegowski T, Rajasekaran D, Sennoune SR, Sunitha S, Chen F, Fokar M, Kshirsagar S, Reddy PH, Korac K, Mahmud Syed M, Sharker T, Ganapathy V, Bhutia YD. Amino acid transporter SLC38A5 is a tumor promoter and a novel therapeutic target for pancreatic cancer. Sci Rep 2023; 13:16863. [PMID: 37803043 PMCID: PMC10558479 DOI: 10.1038/s41598-023-43983-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/01/2023] [Indexed: 10/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) cells have a great demand for nutrients in the form of sugars, amino acids, and lipids. Particularly, amino acids are critical for cancer growth and, as intermediates, connect glucose, lipid and nucleotide metabolism. PDAC cells meet these requirements by upregulating selective amino acid transporters. Here we show that SLC38A5 (SN2/SNAT5), a neutral amino acid transporter is highly upregulated and functional in PDAC cells. Using CRISPR/Cas9-mediated knockout of SLC38A5, we show its tumor promoting role in an in vitro cell line model as well as in a subcutaneous xenograft mouse model. Using metabolomics and RNA sequencing, we show significant reduction in many amino acid substrates of SLC38A5 as well as OXPHOS inactivation in response to SLC38A5 deletion. Experimental validation demonstrates inhibition of mTORC1, glycolysis and mitochondrial respiration in KO cells, suggesting a serious metabolic crisis associated with SLC38A5 deletion. Since many SLC38A5 substrates are activators of mTORC1 as well as TCA cycle intermediates/precursors, we speculate amino acid insufficiency as a possible link between SLC38A5 deletion and inactivation of mTORC1, glycolysis and mitochondrial respiration, and the underlying mechanism for PDAC attenuation. Overall, we show that SLC38A5 promotes PDAC, thereby identifying a novel, hitherto unknown, therapeutic target for PDAC.
Collapse
Affiliation(s)
- Tyler Sniegowski
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Devaraja Rajasekaran
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Souad R Sennoune
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Sukumaran Sunitha
- Center for Biotechnology & Genomics, Texas Tech University, Lubbock, TX, 79409, USA
| | - Fang Chen
- Center for Biotechnology & Genomics, Texas Tech University, Lubbock, TX, 79409, USA
| | - Mohamed Fokar
- Center for Biotechnology & Genomics, Texas Tech University, Lubbock, TX, 79409, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Ksenija Korac
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Mosharaf Mahmud Syed
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Tanima Sharker
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Yangzom D Bhutia
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
13
|
Hu H, Ng TS, Kang M, Scott E, Li R, Quintana JM, Matvey D, Vantaku VR, Weissleder R, Parangi S, Miller MA. Thyroid Cancers Exhibit Oncogene-Enhanced Macropinocytosis that Is Restrained by IGF1R and Promote Albumin-Drug Conjugate Response. Clin Cancer Res 2023; 29:3457-3470. [PMID: 37289199 PMCID: PMC10527034 DOI: 10.1158/1078-0432.ccr-22-2976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/22/2022] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
PURPOSE Oncogene-driven macropinocytosis fuels nutrient scavenging in some cancer types, yet whether this occurs in thyroid cancers with prominent MAPK-ERK and PI3K pathway mutations remains unclear. We hypothesized that understanding links between thyroid cancer signaling and macropinocytosis might uncover new therapeutic strategies. EXPERIMENTAL DESIGN Macropinocytosis was assessed across cells derived from papillary thyroid cancer (PTC), follicular thyroid cancer (FTC), non-malignant follicular thyroid, and aggressive anaplastic thyroid cancer (ATC), by imaging fluorescent dextran and serum albumin. The impacts of ectopic BRAFV600E and mutant RAS, genetic PTEN silencing, and inhibitors targeting RET, BRAF, and MEK kinases were quantified. BrafV600E p53-/- ATC tumors in immunocompetent mice were used to measure efficacy of an albumin-drug conjugate comprising microtubule-destabilizing monomethyl auristatin E (MMAE) linked to serum albumin via a cathepsin-cleavable peptide (Alb-vc-MMAE). RESULTS FTC and ATC cells showed greater macropinocytosis than non-malignant and PTC cells. ATC tumors accumulated albumin at 8.8% injected dose per gram tissue. Alb-vc-MMAE, but not MMAE alone, reduced tumor size by >90% (P < 0.01). ATC macropinocytosis depended on MAPK/ERK activity and nutrient signaling, and increased by up to 230% with metformin, phenformin, or inhibition of IGF1Ri in monoculture but not in vivo. Macrophages also accumulated albumin and express the cognate IGF1R ligand, IGF1, which reduced ATC responsiveness to IGF1Ri. CONCLUSIONS These findings identify regulated oncogene-driven macropinocytosis in thyroid cancers and demonstrate the potential of designing albumin-bound drugs to efficiently treat them.
Collapse
Affiliation(s)
- Huiyu Hu
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
- Department of General Surgery, Xiangya Hospital, Central South University, China
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Mikyung Kang
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Ella Scott
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Jeremy M. Quintana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| | - Dylan Matvey
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
| | - Venkata R. Vantaku
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
- Department of Systems Biology, Harvard Medical School, United States
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, United States
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, United States
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, United States
| |
Collapse
|
14
|
Vidal CM, Ouyang C, Qi Y, Mendez-Dorantes C, Coblentz A, Alva-Ornelas JA, Stark JM, Seewaldt VL, Ann DK. Arginine regulates HSPA5/BiP translation through ribosome pausing in triple-negative breast cancer cells. Br J Cancer 2023; 129:444-454. [PMID: 37386138 PMCID: PMC10403569 DOI: 10.1038/s41416-023-02322-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/17/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with a high mortality rate due to a lack of therapeutic targets. Many TNBC cells are reliant on extracellular arginine for survival and express high levels of binding immunoglobin protein (BiP), a marker of metastasis and endoplasmic reticulum (ER) stress response. METHODS In this study, the effect of arginine shortage on BiP expression in the TNBC cell line MDA-MB-231 was evaluated. Two stable cell lines were generated in MDA-MB-231 cells: the first expressed wild-type BiP, and the second expressed a mutated BiP free of the two arginine pause-site codons, CCU and CGU, termed G-BiP. RESULTS The results showed that arginine shortage induced a non-canonical ER stress response by inhibiting BiP translation via ribosome pausing. Overexpression of G-BiP in MDA-MB-231 cells promoted cell resistance to arginine shortage compared to cells overexpressing wild-type BiP. Additionally, limiting arginine led to decreased levels of the spliced XBP1 in the G-BiP overexpressing cells, potentially contributing to their improved survival compared to the parental WT BiP overexpressing cells. CONCLUSION In conclusion, these findings suggest that the downregulation of BiP disrupts proteostasis during arginine shortage-induced non-canonical ER stress and plays a key role in cell growth inhibition, indicating BiP as a target of codon-specific ribosome pausing upon arginine shortage.
Collapse
Affiliation(s)
- Christina M Vidal
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Ching Ouyang
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Yue Qi
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Carlos Mendez-Dorantes
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Alaysia Coblentz
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Jackelyn A Alva-Ornelas
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Jeremy M Stark
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Victoria L Seewaldt
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - David K Ann
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
15
|
Wang M, Zhang J, Wu Y. Tumor metabolism rewiring in epithelial ovarian cancer. J Ovarian Res 2023; 16:108. [PMID: 37277821 DOI: 10.1186/s13048-023-01196-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 05/29/2023] [Indexed: 06/07/2023] Open
Abstract
The mortality rate of epithelial ovarian cancer (EOC) remains the first in malignant tumors of the female reproductive system. The characteristics of rapid proliferation, extensive implanted metastasis, and treatment resistance of cancer cells require an extensive metabolism rewiring during the progression of cancer development. EOC cells satisfy their rapid proliferation through the rewiring of perception, uptake, utilization, and regulation of glucose, lipids, and amino acids. Further, complete implanted metastasis by acquiring a superior advantage in microenvironment nutrients competing. Lastly, success evolves under the treatment stress of chemotherapy and targets therapy. Understanding the above metabolic characteristics of EOCs helps to find new methods of its treatment.
Collapse
Affiliation(s)
- Ming Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Jingjing Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China
| | - Yumei Wu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 Qihelou St, Dongcheng District, Beijing, 100006, China.
| |
Collapse
|
16
|
Huang Z, Chen CW, Buj R, Tangudu NK, Fang RS, Leon KE, Dahl ES, Varner EL, von Krusenstiern E, Cole AR, Snyder NW, Aird KM. ATM inhibition drives metabolic adaptation via induction of macropinocytosis. J Cell Biol 2023; 222:e202007026. [PMID: 36399181 PMCID: PMC9679964 DOI: 10.1083/jcb.202007026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 05/30/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022] Open
Abstract
Macropinocytosis is a nonspecific endocytic process that may enhance cancer cell survival under nutrient-poor conditions. Ataxia-Telangiectasia mutated (ATM) is a tumor suppressor that has been previously shown to play a role in cellular metabolic reprogramming. We report that the suppression of ATM increases macropinocytosis to promote cancer cell survival in nutrient-poor conditions. Combined inhibition of ATM and macropinocytosis suppressed proliferation and induced cell death both in vitro and in vivo. Supplementation of ATM-inhibited cells with amino acids, branched-chain amino acids (BCAAs) in particular, abrogated macropinocytosis. Analysis of ATM-inhibited cells in vitro demonstrated increased BCAA uptake, and metabolomics of ascites and interstitial fluid from tumors indicated decreased BCAAs in the microenvironment of ATM-inhibited tumors. These data reveal a novel basis of ATM-mediated tumor suppression whereby loss of ATM stimulates protumorigenic uptake of nutrients in part via macropinocytosis to promote cancer cell survival and reveal a potential metabolic vulnerability of ATM-inhibited cells.
Collapse
Affiliation(s)
- Zhentai Huang
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Chi-Wei Chen
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Raquel Buj
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Naveen Kumar Tangudu
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Richard S. Fang
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kelly E. Leon
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Biomedical Sciences Graduate Program, Penn State College of Medicine, Hershey, PA
| | - Erika S. Dahl
- Biomedical Sciences Graduate Program, Penn State College of Medicine, Hershey, PA
| | - Erika L. Varner
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA
| | - Eliana von Krusenstiern
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA
| | - Aidan R. Cole
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Nathaniel W. Snyder
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University, Philadelphia, PA
| | - Katherine M. Aird
- Department of Pharmacology & Chemical Biology and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
17
|
Armenta DA, Laqtom NN, Alchemy G, Dong W, Morrow D, Poltorack CD, Nathanson DA, Abu-Remalieh M, Dixon SJ. Ferroptosis inhibition by lysosome-dependent catabolism of extracellular protein. Cell Chem Biol 2022; 29:1588-1600.e7. [PMID: 36306785 PMCID: PMC9762237 DOI: 10.1016/j.chembiol.2022.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/01/2022] [Accepted: 10/05/2022] [Indexed: 01/31/2023]
Abstract
Cancer cells need a steady supply of nutrients to evade cell death and proliferate. Depriving cancer cells of the amino acid cystine can trigger the non-apoptotic cell death process of ferroptosis. Here, we report that cancer cells can evade cystine deprivation-induced ferroptosis by uptake and catabolism of the cysteine-rich extracellular protein albumin. This protective mechanism is enhanced by mTORC1 inhibition and involves albumin degradation in the lysosome, predominantly by cathepsin B (CTSB). CTSB-dependent albumin breakdown followed by export of cystine from the lysosome via the transporter cystinosin fuels the synthesis of glutathione, which suppresses lethal lipid peroxidation. When cancer cells are grown under non-adherent conditions as spheroids, mTORC1 pathway activity is reduced, and albumin supplementation alone affords considerable protection against ferroptosis. These results identify the catabolism of extracellular protein within the lysosome as a mechanism that can inhibit ferroptosis in cancer cells.
Collapse
Affiliation(s)
- David A Armenta
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Nouf N Laqtom
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering, & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Grace Alchemy
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Wentao Dong
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering, & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Danielle Morrow
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | - David A Nathanson
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Monther Abu-Remalieh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; The Institute for Chemistry, Engineering, & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA.
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
18
|
Brüggenthies JB, Fiore A, Russier M, Bitsina C, Brötzmann J, Kordes S, Menninger S, Wolf A, Conti E, Eickhoff JE, Murray PJ. A cell-based chemical-genetic screen for amino acid stress response inhibitors reveals torins reverse stress kinase GCN2 signaling. J Biol Chem 2022; 298:102629. [PMID: 36273589 PMCID: PMC9668732 DOI: 10.1016/j.jbc.2022.102629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
mTORC1 and GCN2 are serine/threonine kinases that control how cells adapt to amino acid availability. mTORC1 responds to amino acids to promote translation and cell growth while GCN2 senses limiting amino acids to hinder translation via eIF2α phosphorylation. GCN2 is an appealing target for cancer therapies because malignant cells can harness the GCN2 pathway to temper the rate of translation during rapid amino acid consumption. To isolate new GCN2 inhibitors, we created cell-based, amino acid limitation reporters via genetic manipulation of Ddit3 (encoding the transcription factor CHOP). CHOP is strongly induced by limiting amino acids and in this context, GCN2-dependent. Using leucine starvation as a model for essential amino acid sensing, we unexpectedly discovered ATP-competitive PI3 kinase-related kinase inhibitors, including ATR and mTOR inhibitors like torins, completely reversed GCN2 activation in a time-dependent way. Mechanistically, via inhibiting mTORC1-dependent translation, torins increased intracellular leucine, which was sufficient to reverse GCN2 activation and the downstream integrated stress response including stress-induced transcriptional factor ATF4 expression. Strikingly, we found that general translation inhibitors mirrored the effects of torins. Therefore, we propose that mTOR kinase inhibitors concurrently inhibit different branches of amino acid sensing by a dual mechanism involving direct inhibition of mTOR and indirect suppression of GCN2 that are connected by effects on the translation machinery. Collectively, our results highlight distinct ways of regulating GCN2 activity.
Collapse
Affiliation(s)
| | | | - Marion Russier
- Max Planck Institute for Biochemistry, Martinsried, Germany
| | | | | | | | | | | | - Elena Conti
- Max Planck Institute for Biochemistry, Martinsried, Germany
| | | | - Peter J. Murray
- Max Planck Institute for Biochemistry, Martinsried, Germany,For correspondence: Peter J. Murray
| |
Collapse
|
19
|
Liu X, Oh S, Kirschner MW. The uniformity and stability of cellular mass density in mammalian cell culture. Front Cell Dev Biol 2022; 10:1017499. [PMID: 36313562 PMCID: PMC9597509 DOI: 10.3389/fcell.2022.1017499] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/23/2022] [Indexed: 12/04/2022] Open
Abstract
Cell dry mass is principally determined by the sum of biosynthesis and degradation. Measurable change in dry mass occurs on a time scale of hours. By contrast, cell volume can change in minutes by altering the osmotic conditions. How changes in dry mass and volume are coupled is a fundamental question in cell size control. If cell volume were proportional to cell dry mass during growth, the cell would always maintain the same cellular mass density, defined as cell dry mass dividing by cell volume. The accuracy and stability against perturbation of this proportionality has never been stringently tested. Normalized Raman Imaging (NoRI), can measure both protein and lipid dry mass density directly. Using this new technique, we have been able to investigate the stability of mass density in response to pharmaceutical and physiological perturbations in three cultured mammalian cell lines. We find a remarkably narrow mass density distribution within cells, that is, significantly tighter than the variability of mass or volume distribution. The measured mass density is independent of the cell cycle. We find that mass density can be modulated directly by extracellular osmolytes or by disruptions of the cytoskeleton. Yet, mass density is surprisingly resistant to pharmacological perturbations of protein synthesis or protein degradation, suggesting there must be some form of feedback control to maintain the homeostasis of mass density when mass is altered. By contrast, physiological perturbations such as starvation or senescence induce significant shifts in mass density. We have begun to shed light on how and why cell mass density remains fixed against some perturbations and yet is sensitive during transitions in physiological state.
Collapse
Affiliation(s)
| | | | - Marc W. Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
20
|
Zhang L, Li F, Guo Q, Duan Y, Wang W, Yang Y, Yin Y, Gong S, Han M, Yin Y. Balanced branched-chain amino acids modulate meat quality by adjusting muscle fiber type conversion and intramuscular fat deposition in finishing pigs. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:3796-3807. [PMID: 34921408 DOI: 10.1002/jsfa.11728] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/13/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Pork is an important food for humans and improving the quality of pork is closely related to human health. This study was designed to investigate the effects of balanced branched-chain amino acid (BCAA)-supplemented protein-restricted diets on meat quality, muscle fiber types, and intramuscular fat (IMF) in finishing pigs. RESULTS The results showed that, compared with the normal protein diet (160 g kg-1 crude protein), the reduced-protein diet (120 g kg-1 crude protein) supplemented with BCAAs to the ratio of 2:1:2 not only had higher average daily gain (P < 0.05) and carcass weight (P < 0.05) but also improved meat tenderness and juiciness by decreasing shear force (P < 0.05) and increasing water-holding capacity (P < 0.05). In particular, this treatment showed higher (P < 0.05) levels of phospho-acetyl-CoA carboxylase (P-ACC) and peroxisome proliferation-activated receptor-γ (PPARγ), and lower (P < 0.05) levels of P-adenosine 5'-monophosphate (AMP)-activated protein kinase (P-AMPK), increasing the composition of IMF and MyHC I (P < 0.05) in the longissimus dorsi muscle (LDM). In terms of health, this group increased eicosapentaenoic acid (EPA) (P < 0.01) and desirable hypocholesterolemic fatty acids (DHFA) (P < 0.05), and decreased atherogenicity (AI) (P < 0.01) and hypercholesterolemic saturated fatty acids (HSFA) (P < 0.05). CONCLUSION Our findings suggest a novel role for a balanced BCAA-supplemented restricted protein (RP) diet in the epigenetic regulation of more tender and healthier pork by increasing IMF deposition and fiber type conversion, providing a cross-regulatory molecular basis for revealing the nutritional regulation network of meat quality. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Lingyu Zhang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Wenlong Wang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Yuhuan Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yunju Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Saiming Gong
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Mengmeng Han
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process; Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| |
Collapse
|
21
|
Abstract
Macropinocytosis is an evolutionarily conserved endocytic pathway that mediates the nonselective acquisition of extracellular material via large endocytic vesicles known as macropinosomes. In addition to other functions, this uptake pathway supports cancer cell metabolism through the uptake of nutrients. Cells harboring oncogene or tumor suppressor mutations are known to display heightened macropinocytosis, which confers to the cancer cells the ability to survive and proliferate despite the nutrient-scarce conditions of the tumor microenvironment. Thus, macropinocytosis is associated with cancer malignancy. Macropinocytic uptake can be induced in cancer cells by different stress stimuli, acting as an adaptive mechanism for the cells to resist stresses in the tumor milieu. Here, we review the cellular stresses that are known to promote macropinocytosis, as well as the underlying molecular mechanisms that drive this process.
Collapse
Affiliation(s)
- Guillem Lambies
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Cosimo Commisso
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
22
|
Pavlova NN, Zhu J, Thompson CB. The hallmarks of cancer metabolism: Still emerging. Cell Metab 2022; 34:355-377. [PMID: 35123658 PMCID: PMC8891094 DOI: 10.1016/j.cmet.2022.01.007] [Citation(s) in RCA: 509] [Impact Index Per Article: 254.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Metabolism of cancer cells is geared toward biomass production and proliferation. Since the metabolic resources within the local tissue are finite, this can lead to nutrient depletion and accumulation of metabolic waste. To maintain growth in these conditions, cancer cells employ a variety of metabolic adaptations, the nature of which is collectively determined by the physiology of their cell of origin, the identity of transforming lesions, and the tissue in which cancer cells reside. Furthermore, select metabolites not only serve as substrates for energy and biomass generation, but can also regulate gene and protein expression and influence the behavior of non-transformed cells in the tumor vicinity. As they grow and metastasize, tumors can also affect and be affected by the nutrient distribution within the body. In this hallmark update, recent advances are incorporated into a conceptual framework that may help guide further research efforts in exploring cancer cell metabolism.
Collapse
Affiliation(s)
- Natalya N Pavlova
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jiajun Zhu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
23
|
Hypoxia-induced macropinocytosis represents a metabolic route for liver cancer. Nat Commun 2022; 13:954. [PMID: 35177645 PMCID: PMC8854584 DOI: 10.1038/s41467-022-28618-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 01/28/2022] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) invariably exhibits inadequate O2 (hypoxia) and nutrient supply. Hypoxia-inducible factor (HIF) mediates cascades of molecular events that enable cancer cells to adapt and propagate. Macropinocytosis is an endocytic process initiated by membrane ruffling, causing the engulfment of extracellular fluids (proteins), protein digestion and subsequent incorporation into the biomass. We show that macropinocytosis occurs universally in HCC under hypoxia. HIF-1 activates the transcription of a membrane ruffling protein, EH domain-containing protein 2 (EHD2), to initiate macropinocytosis. Knockout of HIF-1 or EHD2 represses hypoxia-induced macropinocytosis and prevents hypoxic HCC cells from scavenging protein that support cell growth. Germline or somatic deletion of Ehd2 suppresses macropinocytosis and HCC development in mice. Intriguingly, EHD2 is overexpressed in HCC. Consistently, HIF-1 or macropinocytosis inhibitor suppresses macropinocytosis and HCC development. Thus, we show that hypoxia induces macropinocytosis through the HIF/EHD2 pathway in HCC cells, harnessing extracellular protein as a nutrient to survive. Cancer cells rely on macropinocytosis to scavenge extracellular proteins for growth. Here the authors show that macropinocytosis supports the survival of hypoxic hepatocellular carcinoma cells and this is dependent on HIF-1, which in turns activates the transcription of a membrane ruffling protein, EH domain-containing protein 2.
Collapse
|
24
|
Means N, Elechalawar CK, Chen WR, Bhattacharya R, Mukherjee P. Revealing macropinocytosis using nanoparticles. Mol Aspects Med 2022; 83:100993. [PMID: 34281720 PMCID: PMC8761201 DOI: 10.1016/j.mam.2021.100993] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/12/2021] [Indexed: 02/03/2023]
Abstract
Endocytosis mechanisms are one of the methods that cells use to interact with their environments. Endocytosis mechanisms vary from the clathrin-mediated endocytosis to the receptor independent macropinocytosis. Macropinocytosis is a niche of endocytosis that is quickly becoming more relevant in various fields of research since its discovery in the 1930s. Macropinocytosis has several distinguishing factors from other receptor-mediated forms of endocytosis, including: types of extracellular material for uptake, signaling cascade, and niche uses between cell types. Nanoparticles (NPs) are an important tool for various applications, including drug delivery and disease treatment. However, surface engineering of NPs could be tailored to target them inside the cells exploiting different endocytosis pathways, such as endocytosis versus macropinocytosis. Such surface engineering of NPs mainly, size, charge, shape and the core material will allow identification of new adapter molecules regulating different endocytosis process and provide further insight into how cells tweak these pathways to meet their physiological need. In this review, we focus on the description of macropinocytosis, a lesser studied endocytosis mechanism than the conventional receptor mediated endocytosis. Additionally, we will discuss nanoparticle endocytosis (including macropinocytosis), and how the physio-chemical properties of the NP (size, charge, and surface coating) affect their intracellular uptake and exploiting them as tools to identify new adapter molecules regulating these processes.
Collapse
Affiliation(s)
- Nicolas Means
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Wei R Chen
- Stephenson School of Biomedical Engineering, Gallogly College of Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Resham Bhattacharya
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
25
|
Zhang YF, Li Q, Huang PQ, Su T, Jiang SH, Hu LP, Zhang XL, Sun Y, Pan H, Yang XM, Li J, Gai YZ, Zhu L, Yao LL, Li DX, Sun YW, Zhang ZG, Liu DJ, Zhang YL, Nie HZ. A low amino acid environment promotes cell macropinocytosis through the YY1-FGD6 axis in Ras-mutant pancreatic ductal adenocarcinoma. Oncogene 2022; 41:1203-1215. [PMID: 35082383 DOI: 10.1038/s41388-021-02159-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 11/09/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), cancer with a high mortality rate and the highest rate of KRAS mutation, reportedly internalizes proteins via macropinocytosis to adapt to low amino acid levels in the tumor microenvironment. Here, we aimed to identify a key regulator of macropinocytosis for the survival of tumor cells in a low amino acid environment in PDAC. FYVE, RhoGEF, and PH domain-containing protein 6 (FGD6) were identified as key regulators of macropinocytosis. FGD6 promoted PDAC cell proliferation, macropinocytosis, and tumor growth both in vitro and in vivo. The macropinocytosis level was decreased with FGD6 knockdown in PDAC cell lines. Moreover, FGD6 promoted macropinocytosis by participating in the trans-Golgi network and enhancing the membrane localization of growth factor receptors, especially the TGF-beta receptor. TGF-beta enhanced macropinocytosis in PDAC cells. Additionally, YAP nuclear translocation induced by a low amino acid tumor environment initiated FGD6 expression by coactivation with YY1. Clinical data analysis based on TCGA and GEO datasets showed that FGD6 expression was upregulated in PDAC tissue, and high FGD6 expression was correlated with poor prognosis in patients with PDAC. In tumor tissue from KrasG12D/+/Trp53R172H/-/Pdx1-Cre (KPC) mice, FGD6 expression escalated during PDAC development. Our results uncover a previously unappreciated mechanism of macropinocytosis in PDAC. Strategies to target FGD6 and growth factors membrane localization might be developed for the treatment of PDAC.
Collapse
Affiliation(s)
- Yi-Fan Zhang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Pei-Qi Huang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tong Su
- Department of Gynecology and Obstetrics, Shanghai Key Laboratory of Gynecology and Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yue Sun
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hong Pan
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiao-Mei Yang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan-Zhi Gai
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lei Zhu
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lin-Li Yao
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Dong-Xue Li
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong-Wei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - De-Jun Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Yan-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Hui-Zhen Nie
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
26
|
Han Y, Liang C, Manthari RK, Yu Y, Zhang J, Wang J, Cao J. Distribution characteristics and regulation of amino acids and fatty acids in muscle and adipose tissues of sheep grown in natural grazing environment. Anim Sci J 2022; 93:e13769. [PMID: 36127314 DOI: 10.1111/asj.13769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 07/26/2022] [Accepted: 08/18/2022] [Indexed: 12/01/2022]
Abstract
The composition of amino acid and fatty acid has a vital function on meat quality and animal health. However, the underlying mechanism of amino acid and fatty acid metabolism in sheep during different grazing periods is still unclear. In this study, a total of 12 sheep were employed in different grazing periods. Our results showed that the composition of amino acids and fatty acids in muscle and adipose tissues was significantly altered between dry grass (DG) period and green grass (GG) period. Changes in the activities of the metabolism-related enzymes including BCKD, BCAT2, ACC, SCD, HSL, GSK3β, p-GSK3β, and FABP4 were observed in muscle and adipose during different grazing periods. In addition, the mRNA expression levels of ACC, FAS, SCD, HSL, LPL, and DGAT1 in muscle and adipose tissue were changed markedly in different grazing periods. Furthermore, the expression levels of mTOR and β-catenin/PPARγ/C/EBPα pathway-related proteins were predominantly altered in muscle and adipose among DG and GG. Taken together, all investigations simplified the process of amino acid and fatty acid metabolism disorders caused by different grazing periods, and the mTOR and β-catenin/PPARγ/C/EBPα play the essential role in this process, which provided an underlying mechanism of metabolism and meat quality.
Collapse
Affiliation(s)
- Yongli Han
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Chen Liang
- College of Animal Science, Shanxi Agricultural University, Taigu, China
| | - Ram Kumar Manthari
- Department of Biotechnology, GITAM Institute of Science, Gandhi Institute of Technology and Management, Visakhapatnam, India
| | - Yuxiang Yu
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Jianhai Zhang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Jundong Wang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Jinling Cao
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China.,College of Food Science and Engineering, Shanxi Agricultural University, Taigu, China
| |
Collapse
|
27
|
Zinecker H, Simon AK. Autophagy takes it all - autophagy inducers target immune aging. Dis Model Mech 2022; 15:dmm049345. [PMID: 35098310 PMCID: PMC8822217 DOI: 10.1242/dmm.049345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Autophagy, as the key nutrient recycling pathway, enables eukaryotic cells to adapt to surging cellular stress during aging and, thereby, delays age-associated deterioration. Autophagic flux declines with age and, in turn, decreases in autophagy contribute to the aging process itself and promote senescence. Here, we outline how autophagy regulates immune aging and discuss autophagy-inducing interventions that target senescent immune cells, which are major drivers of systemic aging. We examine how cutting-edge technologies, such as single-cell omics methods hold the promise to capture the complexity of molecular and cellular phenotypes associated with aging, driving the development of suitable putative biomarkers and clinical bioassays. Finally, we debate the urgency to initiate large-scale human clinical trials. We give special preference to small molecule probes and to dietary interventions that can extend healthy lifespan and are affordable for most of the world's population.
Collapse
Affiliation(s)
- Heidi Zinecker
- Turkish-German University, Department of Molecular Biotechnology, 34820, Beykoz/Istanbul, Turkey
| | - Anna Katharina Simon
- NDORMS, The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| |
Collapse
|
28
|
Li Z, Zhou S, Yang X, Li X, Yang GX, Chant J, Snyder M, Wang X. Broad Anti-Cancer Activity Produced by Targeted Nutrients Deprivation (TND) of Multiple Non-Essential Amino Acids. Nutr Cancer 2021; 74:2607-2621. [PMID: 34905997 DOI: 10.1080/01635581.2021.2013904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
It has been known for close to 100 years that the metabolism of cancer cells is altered and different than that of healthy cells in the body. On that basis, we have developed an entirely novel approach to managing cancer, termed Targeted Nutrients Deprivation (TND). TND employs a formulated diet depleted of multiple non-essential amino acids (NEAAs) that are required by tumor cells but not by normal cells. Cancer cells specifically require those NEAAs due to their heightened and rewired metabolism. We demonstrated that our first proprietary formulated TND diet-FTN203-significantly reduced the growth of multiple human tumor xenografts in mouse. In combination with chemotherapy and immunotherapy, FTN203 further enhanced therapeutic efficacy. Reliance on FTN203 as the sole nutrition source was shown to be safe without causing detrimental body-weight loss or internal organ damage. Our findings indicate that TND is a novel and safe approach to managing cancer.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.2013904 .
Collapse
Affiliation(s)
- Zehui Li
- Filtricine, Inc, Santa Clara, California, USA
| | - Shuang Zhou
- Filtricine, Inc, Santa Clara, California, USA
| | | | - Xiyan Li
- Filtricine, Inc, Santa Clara, California, USA
| | | | - John Chant
- Filtricine, Inc, Santa Clara, California, USA
| | | | - Xin Wang
- Filtricine, Inc, Santa Clara, California, USA
| |
Collapse
|
29
|
Amino acid deprivation induces AKT activation by inducing GCN2/ATF4/REDD1 axis. Cell Death Dis 2021; 12:1127. [PMID: 34862383 PMCID: PMC8642548 DOI: 10.1038/s41419-021-04417-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 11/17/2022]
Abstract
Amino acid availability is sensed by various signaling molecules, including general control nonderepressible 2 (GCN2) and mechanistic target of rapamycin complex 1 (mTORC1). However, it is unclear how these sensors are associated with cancer cell survival under low amino acid availability. In the present study, we investigated AKT activation in non-small cell lung cancer (NSCLC) cells deprived of each one of 20 amino acids. Among the 20 amino acids, deprivation of glutamine, arginine, methionine, and lysine induced AKT activation. AKT activation was induced by GCN2/ATF4/REDD1 axis-mediated mTORC2 activation under amino acid deprivation. In CRISPR-Cas9-mediated REDD1-knockout cells, AKT activation was not induced by amino acid deprivation, indicating that REDD1 plays a major role in AKT activation under amino acid deprivation. Knockout of REDD1 sensitized cells cultured under glutamine deprivation conditions to radiotherapy. Taken together, GCN2/ATF4/REDD1 axis induced by amino acid deprivation promotes cell survival signal, which might be a potential target for cancer therapy.
Collapse
|
30
|
Rossmeislová L, Gojda J, Smolková K. Pancreatic cancer: branched-chain amino acids as putative key metabolic regulators? Cancer Metastasis Rev 2021; 40:1115-1139. [PMID: 34962613 DOI: 10.1007/s10555-021-10016-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/18/2021] [Indexed: 02/06/2023]
Abstract
Branched-chain amino acids (BCAA) are essential amino acids utilized in anabolic and catabolic metabolism. While extensively studied in obesity and diabetes, recent evidence suggests an important role for BCAA metabolism in cancer. Elevated plasma levels of BCAA are associated with an increased risk of developing pancreatic cancer, namely pancreatic ductal adenocarcinoma (PDAC), a tumor with one of the highest 1-year mortality rates. The dreadful prognosis for PDAC patients could be attributable also to the early and frequent development of cancer cachexia, a fatal host metabolic reprogramming leading to muscle and adipose wasting. We propose that BCAA dysmetabolism is a unifying component of several pathological conditions, i.e., obesity, insulin resistance, and PDAC. These conditions are mutually dependent since PDAC ranks among cancers tightly associated with obesity and insulin resistance. It is also well-established that PDAC itself can trigger insulin resistance and new-onset diabetes. However, the exact link between BCAA metabolism, development of PDAC, and tissue wasting is still unclear. Although tissue-specific intracellular and systemic metabolism of BCAA is being intensively studied, unresolved questions related to PDAC and cancer cachexia remain, namely, whether elevated circulating BCAA contribute to PDAC etiology, what is the biological background of BCAA elevation, and what is the role of adipose tissue relative to BCAA metabolism during cancer cachexia. To cover those issues, we provide our view on BCAA metabolism at the intracellular, tissue, and whole-body level, with special emphasis on different metabolic links to BCAA intermediates and the role of insulin in substrate handling.
Collapse
Affiliation(s)
- Lenka Rossmeislová
- Department of Pathophysiology, Center for Research On Nutrition, Metabolism, and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research On Obesity, Third Faculty of Medicine, Prague, Czech Republic
| | - Jan Gojda
- Franco-Czech Laboratory for Clinical Research On Obesity, Third Faculty of Medicine, Prague, Czech Republic
- Department of Internal Medicine, Královské Vinohrady University Hospital and Third Faculty of Medicine, Prague, Czech Republic
| | - Katarína Smolková
- Laboratory of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
31
|
Targeting lysosomes in human disease: from basic research to clinical applications. Signal Transduct Target Ther 2021; 6:379. [PMID: 34744168 PMCID: PMC8572923 DOI: 10.1038/s41392-021-00778-y] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/26/2021] [Indexed: 01/18/2023] Open
Abstract
In recent years, accumulating evidence has elucidated the role of lysosomes in dynamically regulating cellular and organismal homeostasis. Lysosomal changes and dysfunction have been correlated with the development of numerous diseases. In this review, we interpreted the key biological functions of lysosomes in four areas: cellular metabolism, cell proliferation and differentiation, immunity, and cell death. More importantly, we actively sought to determine the characteristic changes and dysfunction of lysosomes in cells affected by these diseases, the causes of these changes and dysfunction, and their significance to the development and treatment of human disease. Furthermore, we outlined currently available targeting strategies: (1) targeting lysosomal acidification; (2) targeting lysosomal cathepsins; (3) targeting lysosomal membrane permeability and integrity; (4) targeting lysosomal calcium signaling; (5) targeting mTOR signaling; and (6) emerging potential targeting strategies. Moreover, we systematically summarized the corresponding drugs and their application in clinical trials. By integrating basic research with clinical findings, we discussed the current opportunities and challenges of targeting lysosomes in human disease.
Collapse
|
32
|
Nofal M, Wang T, Yang L, Jankowski CSR, Hsin-Jung Li S, Han S, Parsons L, Frese AN, Gitai Z, Anthony TG, Wühr M, Sabatini DM, Rabinowitz JD. GCN2 adapts protein synthesis to scavenging-dependent growth. Cell Syst 2021; 13:158-172.e9. [PMID: 34706266 DOI: 10.1016/j.cels.2021.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/22/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer cells with limited access to free amino acids can grow by scavenging extracellular protein. In a murine model of pancreatic cancer, we performed a genome-wide CRISPR screen for genes required for scavenging-dependent growth. The screen identified key mediators of macropinocytosis, peripheral lysosome positioning, endosome-lysosome fusion, lysosomal protein catabolism, and translational control. The top hit was GCN2, a kinase that suppresses translation initiation upon amino acid depletion. Using isotope tracers, we show that GCN2 is not required for protein scavenging. Instead, GCN2 prevents ribosome stalling but without slowing protein synthesis; cells still use all of the limiting amino acids as they emerge from lysosomes. GCN2 also adapts gene expression to the nutrient-poor environment, reorienting protein synthesis away from ribosomes and toward lysosomal hydrolases, such as cathepsin L. GCN2, cathepsin L, and the other genes identified in the screen are potential therapeutic targets in pancreatic cancer.
Collapse
Affiliation(s)
- Michel Nofal
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Tim Wang
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lifeng Yang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Connor S R Jankowski
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Sophia Hsin-Jung Li
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Seunghun Han
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Lance Parsons
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Alexander N Frese
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Zemer Gitai
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Tracy G Anthony
- Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ 08901, USA
| | - Martin Wühr
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton University, Princeton, NJ 08540, USA.
| |
Collapse
|
33
|
Papalazarou V, Maddocks ODK. Supply and demand: Cellular nutrient uptake and exchange in cancer. Mol Cell 2021; 81:3731-3748. [PMID: 34547236 DOI: 10.1016/j.molcel.2021.08.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/27/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022]
Abstract
Nutrient supply and demand delineate cell behavior in health and disease. Mammalian cells have developed multiple strategies to secure the necessary nutrients that fuel their metabolic needs. This is more evident upon disruption of homeostasis in conditions such as cancer, when cells display high proliferation rates in energetically challenging conditions where nutritional sources may be scarce. Here, we summarize the main routes of nutrient acquisition that fuel mammalian cells and their implications in tumorigenesis. We argue that the molecular mechanisms of nutrient acquisition not only tip the balance between nutrient supply and demand but also determine cell behavior upon nutrient limitation and energetic stress and contribute to nutrient partitioning and metabolic coordination between different cell types in inflamed or tumorigenic environments.
Collapse
Affiliation(s)
- Vasileios Papalazarou
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1QH, UK
| | - Oliver D K Maddocks
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1QH, UK.
| |
Collapse
|
34
|
Colombero C, Remy D, Antoine‐Bally S, Macé A, Monteiro P, ElKhatib N, Fournier M, Dahmani A, Montaudon E, Montagnac G, Marangoni E, Chavrier P. mTOR Repression in Response to Amino Acid Starvation Promotes ECM Degradation Through MT1-MMP Endocytosis Arrest. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101614. [PMID: 34250755 PMCID: PMC8425857 DOI: 10.1002/advs.202101614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/07/2021] [Indexed: 05/02/2023]
Abstract
Under conditions of starvation, normal and tumor epithelial cells can rewire their metabolism toward the consumption of extracellular proteins, including extracellular matrix-derived components as nutrient sources. The mechanism of pericellular matrix degradation by starved cells has been largely overlooked. Here it is shown that matrix degradation by breast and pancreatic tumor cells and patient-derived xenograft explants increases by one order of magnitude upon amino acid and growth factor deprivation. In addition, it is found that collagenolysis requires the invadopodia components, TKS5, and the transmembrane metalloproteinase, MT1-MMP, which are key to the tumor invasion program. Increased collagenolysis is controlled by mTOR repression upon nutrient depletion or pharmacological inhibition by rapamycin. The results reveal that starvation hampers clathrin-mediated endocytosis, resulting in MT1-MMP accumulation in arrested clathrin-coated pits. The study uncovers a new mechanism whereby mTOR repression in starved cells leads to the repurposing of abundant plasma membrane clathrin-coated pits into robust ECM-degradative assemblies.
Collapse
Affiliation(s)
| | - David Remy
- Institut CuriePSL Research UniversityCNRS UMR 144Paris75005France
| | | | - Anne‐Sophie Macé
- Institut CuriePSL Research UniversityCNRS UMR 144Paris75005France
- Cell and Tissue Imaging Facility (PICT‐IBiSA)Institut CuriePSL Research UniversityParis75005France
| | - Pedro Monteiro
- Institut CuriePSL Research UniversityCNRS UMR 144Paris75005France
| | - Nadia ElKhatib
- Gustave Roussy InstituteUniversité Paris‐SaclayINSERM U1279Villejuif94805France
| | - Margot Fournier
- Institut CuriePSL Research UniversityCNRS UMR 144Paris75005France
| | - Ahmed Dahmani
- Translational Research DepartmentInstitut CuriePSL Research UniversityParis75005France
| | - Elodie Montaudon
- Translational Research DepartmentInstitut CuriePSL Research UniversityParis75005France
| | - Guillaume Montagnac
- Gustave Roussy InstituteUniversité Paris‐SaclayINSERM U1279Villejuif94805France
| | - Elisabetta Marangoni
- Translational Research DepartmentInstitut CuriePSL Research UniversityParis75005France
| | | |
Collapse
|
35
|
Lei G, Zhuang L, Gan B. mTORC1 and ferroptosis: Regulatory mechanisms and therapeutic potential. Bioessays 2021; 43:e2100093. [PMID: 34121197 DOI: 10.1002/bies.202100093] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/29/2021] [Accepted: 06/02/2021] [Indexed: 12/31/2022]
Abstract
Ferroptosis, a form of regulated cell death triggered by lipid hydroperoxide accumulation, has an important role in a variety of diseases and pathological conditions, such as cancer. Targeting ferroptosis is emerging as a promising means of therapeutic intervention in cancer treatment. Polyunsaturated fatty acids, reactive oxygen species, and labile iron constitute the major underlying triggers for ferroptosis. Other regulators of ferroptosis have also been discovered recently, among them the mechanistic target of rapamycin complex 1 (mTORC1), a central controller of cell growth and metabolism. Inhibitors of mTORC1 have been used in treating diverse diseases, including cancer. In this review, we discuss recent findings linking mTORC1 to ferroptosis, dissect mechanisms underlying the establishment of mTORC1 as a key ferroptosis modulator, and highlight the potential of co-targeting mTORC1 and ferroptosis in cancer treatment. This review will provide valuable insights for future investigations of ferroptosis and mTORC1 in fundamental biology and cancer therapy.
Collapse
Affiliation(s)
- Guang Lei
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
36
|
A compendium of kinetic modulatory profiles identifies ferroptosis regulators. Nat Chem Biol 2021; 17:665-674. [PMID: 33686292 PMCID: PMC8159879 DOI: 10.1038/s41589-021-00751-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/27/2021] [Indexed: 01/31/2023]
Abstract
Cell death can be executed by regulated apoptotic and nonapoptotic pathways, including the iron-dependent process of ferroptosis. Small molecules are essential tools for studying the regulation of cell death. Using time-lapse imaging and a library of 1,833 bioactive compounds, we assembled a large compendium of kinetic cell death modulatory profiles for inducers of apoptosis and ferroptosis. From this dataset we identify dozens of ferroptosis suppressors, including numerous compounds that appear to act via cryptic off-target antioxidant or iron chelating activities. We show that the FDA-approved drug bazedoxifene acts as a potent radical trapping antioxidant inhibitor of ferroptosis both in vitro and in vivo. ATP-competitive mechanistic target of rapamycin (mTOR) inhibitors, by contrast, are on-target ferroptosis inhibitors. Further investigation revealed both mTOR-dependent and mTOR-independent mechanisms that link amino acid metabolism to ferroptosis sensitivity. These results highlight kinetic modulatory profiling as a useful tool to investigate cell death regulation.
Collapse
|
37
|
Placental trophoblast syncytialization potentiates macropinocytosis via mTOR signaling to adapt to reduced amino acid supply. Proc Natl Acad Sci U S A 2021; 118:2017092118. [PMID: 33402432 DOI: 10.1073/pnas.2017092118] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
During pregnancy, the appropriate allocation of nutrients between the mother and the fetus is dominated by maternal-fetal interactions, which is primarily governed by the placenta. The syncytiotrophoblast (STB) lining at the outer surface of the placental villi is directly bathed in maternal blood and controls feto-maternal exchange. The STB is the largest multinucleated cell type in the human body, and is formed through syncytialization of the mononucleated cytotrophoblast. However, the physiological advantage of forming such an extensively multinucleated cellular structure remains poorly understood. Here, we discover that the STB uniquely adapts to nutrient stress by inducing the macropinocytosis machinery through repression of mammalian target of rapamycin (mTOR) signaling. In primary human trophoblasts and in trophoblast cell lines, differentiation toward a syncytium triggers macropinocytosis, which is greatly enhanced during amino acid shortage, induced by inhibiting mTOR signaling. Moreover, inhibiting mTOR in pregnant mice markedly stimulates macropinocytosis in the syncytium. Blocking macropinocytosis worsens the phenotypes of fetal growth restriction caused by mTOR-inhibition. Consistently, placentas derived from fetal growth restriction patients display: 1) Repressed mTOR signaling, 2) increased syncytialization, and 3) enhanced macropinocytosis. Together, our findings suggest that the unique ability of STB to undergo macropinocytosis serves as an essential adaptation to the cellular nutrient status, and support fetal survival and growth under nutrient deprivation.
Collapse
|
38
|
Zhang Y, Recouvreux MV, Jung M, Galenkamp KMO, Li Y, Zagnitko O, Scott DA, Lowy AM, Commisso C. Macropinocytosis in Cancer-Associated Fibroblasts Is Dependent on CaMKK2/ARHGEF2 Signaling and Functions to Support Tumor and Stromal Cell Fitness. Cancer Discov 2021; 11:1808-1825. [PMID: 33653692 DOI: 10.1158/2159-8290.cd-20-0119] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 01/15/2021] [Accepted: 02/26/2021] [Indexed: 12/27/2022]
Abstract
Although pancreatic ductal adenocarcinoma (PDAC) cells are exposed to a nutrient-depleted tumor microenvironment, they can acquire nutrients via macropinocytosis, an endocytic form of protein scavenging that functions to support cancer metabolism. Here, we provide evidence that macropinocytosis is also operational in the pancreatic tumor stroma. We find that glutamine deficiency triggers macropinocytic uptake in pancreatic cancer-associated fibroblasts (CAF). Mechanistically, we decipher that stromal macropinocytosis is potentiated via the enhancement of cytosolic Ca2+ and dependent on ARHGEF2 and CaMKK2-AMPK signaling. We elucidate that macropinocytosis has a dual function in CAFs-it serves as a source of intracellular amino acids that sustain CAF cell fitness and function, and it provides secreted amino acids that promote tumor cell survival. Importantly, we demonstrate that stromal macropinocytosis supports PDAC tumor growth. These results highlight the functional role of macropinocytosis in the tumor stroma and provide a mechanistic understanding of how nutrient deficiency can control stromal protein scavenging. SIGNIFICANCE: Glutamine deprivation drives stromal macropinocytosis to support CAF cell fitness and provide amino acids that sustain PDAC cell survival. Selective disruption of macropinocytosis in CAFs suppresses PDAC tumor growth.This article is highlighted in the In This Issue feature, p. 1601.
Collapse
Affiliation(s)
- Yijuan Zhang
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - M Victoria Recouvreux
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Michael Jung
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Koen M O Galenkamp
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Yunbo Li
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California
| | - Olga Zagnitko
- Cancer Metabolism Core Resource, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - David A Scott
- Cancer Metabolism Core Resource, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Andrew M Lowy
- Moores Cancer Center, University of California, San Diego, La Jolla, California.,Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, California
| | - Cosimo Commisso
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.
| |
Collapse
|
39
|
Pathria G, Verma S, Yin J, Scott DA, Ronai ZA. MAPK signaling regulates c-MYC for melanoma cell adaptation to asparagine restriction. EMBO Rep 2021; 22:e51436. [PMID: 33554439 DOI: 10.15252/embr.202051436] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 01/07/2023] Open
Abstract
Amino acid restriction is among promising potential cancer treatment strategies. However, cancer cells employ a multitude of mechanisms to mount resistance to amino acid restriction, which impede the latter's clinical development. Here we show that MAPK signaling activation in asparagine-restricted melanoma cells impairs GSK3-β-mediated c-MYC degradation. In turn, elevated c-MYC supports ATF4 translational induction by enhancing the expression of the amino acid transporter SLC7A5, increasing the uptake of essential amino acids, and the subsequent maintenance of mTORC1 activity in asparagine-restricted melanoma cells. Blocking the MAPK-c-MYC-SLC7A5 signaling axis cooperates with asparagine restriction to effectively suppress melanoma cell proliferation. This work reveals a previously unknown axis of cancer cell adaptation to asparagine restriction and informs mechanisms that may be targeted for enhanced therapeutic efficacy of asparagine limiting strategies.
Collapse
Affiliation(s)
- Gaurav Pathria
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sachin Verma
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jun Yin
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - David A Scott
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ze'ev A Ronai
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
40
|
Alanio A. Dormancy in Cryptococcus neoformans: 60 years of accumulating evidence. J Clin Invest 2021; 130:3353-3360. [PMID: 32484459 DOI: 10.1172/jci136223] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cryptococcus neoformans is an opportunistic yeast that is present worldwide and interacts with various organisms. In humans, it is responsible for cryptococcosis, a deadly invasive fungal infection that represents around 220,000 cases per year worldwide. Starting from the natural history of the disease in humans, there is accumulating evidence on the capacity of this organism to enter dormancy. In response to the harsh host environment, the yeast is able to adapt dramatically and escape the vigilance of the host's immune cells to survive. Indeed, the yeast exposed to the host takes on pleiotropic phenotypes, enabling the generation of populations in heterogeneous states, including dormancy, to eventually survive at low metabolic cost and revive in favorable conditions. The concept of dormancy has been validated in C. neoformans from both epidemiological and genotyping data, and more recently from the biological point of view with the characterization of dormancy through the description of viable but nonculturable cells.
Collapse
Affiliation(s)
- Alexandre Alanio
- Laboratoire de Parasitologie-Mycologie, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand-Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Molecular Mycology Unit, CNRS UMR 2000, National Reference Center for Invasive Mycoses and Antifungals (NRCMA), Institut Pasteur, Paris, France.,Université de Paris, Paris, France
| |
Collapse
|
41
|
Andreasson C, Ansari D, Ekbom F, Andersson R. Macropinocytosis: the Achilles' heel of pancreatic cancer? Scand J Gastroenterol 2021; 56:177-179. [PMID: 33280476 DOI: 10.1080/00365521.2020.1855471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Carl Andreasson
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund SE -221 85, Sweden
| | - Daniel Ansari
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund SE -221 85, Sweden
| | - Fredrik Ekbom
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund SE -221 85, Sweden
| | - Roland Andersson
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund SE -221 85, Sweden
| |
Collapse
|
42
|
Harnessing the Co-vulnerabilities of Amino Acid-Restricted Cancers. Cell Metab 2021; 33:9-20. [PMID: 33406406 PMCID: PMC7837405 DOI: 10.1016/j.cmet.2020.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022]
Abstract
Sustained proliferative potential of cancer cells creates heightened energetic and biosynthetic demands. The resulting overt dependence of cancer cells on unperturbed nutrient supply has prompted a widespread interest in amino acid restriction strategies as potential cancer therapeutics. However, owing to rapid signaling and metabolic reprogramming in cancer cells, the prospects for success of amino acid restriction approaches remain unclear. We thus recognize that the identification of co-vulnerabilities of amino acid-restricted cancers may inform actionable targets for effective combined interventions. In this perspective, we outline the current state of key cellular mechanisms underlying adaptation to amino acid restriction and discuss the role of signal transduction pathways governing cancer cell resistance to amino acid restriction, with potential ramifications for the design of future therapeutic efforts.
Collapse
|
43
|
Meador JP, Bettcher LF, Ellenberger MC, Senn TD. Metabolomic profiling for juvenile Chinook salmon exposed to contaminants of emerging concern. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 747:141097. [PMID: 32781313 DOI: 10.1016/j.scitotenv.2020.141097] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 06/11/2023]
Abstract
Both targeted and non-targeted metabolomic analyses were conducted on juvenile ocean-type fall Chinook salmon (Oncorhynchus tshawytscha) residing in two estuaries receiving wastewater treatment plant (WWTP) effluent and one reference estuary. The data show that the metabolome patterns for fish from the two WWTP-receiving estuaries were more similar to each other compared to that for the reference site fish. Also, a comparison of the metabolome for fish from the reference site and fish from a hatchery upstream of one of the effluent-receiving estuaries indicated no differences, implying that residency for fish in the contaminated estuary resulted in major changes to the metabolome. Based on general health parameters including whole-body lipid content and condition factor, plus the availability of prey for these fish, we conclude that juvenile Chinook salmon in these contaminated estuaries may have been experiencing metabolic disruption without any overt signs of impairment. Additionally, a non-targeted analysis was performed on hatchery summer Chinook salmon from a laboratory study where fish were dosed for 32 days with feed containing 16 of the most common contaminants of emerging concern (CECs) detected in wild fish. In the laboratory experiment a relationship was observed between dose and the number of liver metabolites that were different between control and treatment fish. Laboratory fish were exposed to only 16 CECs, but are generally exposed to hundreds of these compounds in contaminated aquatic environments. These results have implications for the health of juvenile Chinook salmon and the likelihood of a successful life cycle when exposed to effluent-related chemicals.
Collapse
Affiliation(s)
- James P Meador
- Ecotoxicology Program, Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd E, Seattle, WA 98112, USA; Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA 98195, USA.
| | - Lisa F Bettcher
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Mathew C Ellenberger
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, School of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Taurence D Senn
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
44
|
Hesketh GG, Papazotos F, Pawling J, Rajendran D, Knight JDR, Martinez S, Taipale M, Schramek D, Dennis JW, Gingras AC. The GATOR–Rag GTPase pathway inhibits mTORC1 activation by
lysosome-derived amino acids. Science 2020; 370:351-356. [DOI: 10.1126/science.aaz0863] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 04/18/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) couples nutrient
sufficiency to cell growth. mTORC1 is activated by exogenously acquired
amino acids sensed through the GATOR–Rag guanosine triphosphatase (GTPase)
pathway, or by amino acids derived through lysosomal degradation of protein
by a poorly defined mechanism. Here, we revealed that amino acids derived
from the degradation of protein (acquired through oncogenic Ras-driven
macropinocytosis) activate mTORC1 by a Rag GTPase–independent mechanism.
mTORC1 stimulation through this pathway required the HOPS complex and was
negatively regulated by activation of the GATOR-Rag GTPase pathway.
Therefore, distinct but functionally coordinated pathways control mTORC1
activity on late endocytic organelles in response to distinct sources of
amino acids.
Collapse
Affiliation(s)
- Geoffrey G. Hesketh
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Fotini Papazotos
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Judy Pawling
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Dushyandi Rajendran
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - James D. R. Knight
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Sebastien Martinez
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Mikko Taipale
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada
| | - Daniel Schramek
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - James W. Dennis
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
45
|
Xu R, Yang J, Ren B, Wang H, Yang G, Chen Y, You L, Zhao Y. Reprogramming of Amino Acid Metabolism in Pancreatic Cancer: Recent Advances and Therapeutic Strategies. Front Oncol 2020; 10:572722. [PMID: 33117704 PMCID: PMC7550743 DOI: 10.3389/fonc.2020.572722] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/31/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal malignancies with an extremely poor prognosis. Energy metabolism reprogramming, an emerging hallmark of cancer, has been implicated in the tumorigenesis and development of pancreatic cancer. In addition to well-elaborated enhanced glycolysis, investigating the role of reprogramming of amino acid metabolism has sparked great interests in recent years. The rewiring amino acid metabolism orchestrated by genetic alterations contributes to pancreatic cancer malignant characteristics including cell proliferation, invasion, metastasis, angiogenesis and redox balance. In the unique hypoperfused and nutrient-deficient tumor microenvironment (TME), the interactions between cancer cells and stromal components and salvaging processes including autophagy and macropinocytosis play critical roles in fulfilling the metabolic requirements and supporting growth of PDAC. In this review, we elucidate the recent advances in the amino acid metabolism reprogramming in pancreatic cancer and the mechanisms of amino acid metabolism regulating PDAC progression, which will provide opportunities to develop promising therapeutic strategies.
Collapse
Affiliation(s)
- Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jinshou Yang
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Huanyu Wang
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
46
|
King B, Araki J, Palm W, Thompson CB. Yap/Taz promote the scavenging of extracellular nutrients through macropinocytosis. Genes Dev 2020; 34:1345-1358. [PMID: 32912902 PMCID: PMC7528706 DOI: 10.1101/gad.340661.120] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022]
Abstract
The uptake of macromolecules and cellular debris through macropinocytosis has emerged as an important nutrient acquisition strategy of cancer cells. Genetic alterations commonly found in human cancers (e.g. mutations in KRAS or loss of PTEN) have been shown to increase macropinocytosis. To identify additional effectors that enable cell growth dependent on the uptake of extracellular proteins, pancreatic ductal adenocarcinoma (PDA) cells were selected for growth in medium where extracellular albumin was the obligate source of the essential amino acid leucine. Analysis of global changes in chromatin availability and gene expression revealed that PDA cells selected under these conditions exhibited elevated activity of the transcriptional activators Yap/Taz. Knockout of Yap/Taz prevented growth of PDA cells in leucine-deficient medium, but not in complete medium. Furthermore, constitutively active forms of Yap or Taz were sufficient to stimulate macropinocytosis of extracellular protein. In addition to promoting the uptake of plasma proteins, Yap/Taz also promoted the scavenging of apoptotic cell bodies and necrotic debris by PDA cells. The Yap/Taz transcriptional target Axl was found to be essential for cell growth dependent on the uptake of dead cells and cell debris. Together, these studies suggest that the Hippo pathway effectors Yap and Taz are important transcriptional regulators of endocytic nutrient uptake.
Collapse
Affiliation(s)
- Bryan King
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Jingwen Araki
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Wilhelm Palm
- Cell Biology and Tumor Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
47
|
Vadlakonda L, Indracanti M, Kalangi SK, Gayatri BM, Naidu NG, Reddy ABM. The Role of Pi, Glutamine and the Essential Amino Acids in Modulating the Metabolism in Diabetes and Cancer. J Diabetes Metab Disord 2020; 19:1731-1775. [PMID: 33520860 DOI: 10.1007/s40200-020-00566-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 06/04/2020] [Indexed: 02/07/2023]
Abstract
Purpose Re-examine the current metabolic models. Methods Review of literature and gene networks. Results Insulin activates Pi uptake, glutamine metabolism to stabilise lipid membranes. Tissue turnover maintains the metabolic health. Current model of intermediary metabolism (IM) suggests glucose is the source of energy, and anaplerotic entry of fatty acids and amino acids into mitochondria increases the oxidative capacity of the TCA cycle to produce the energy (ATP). The reduced cofactors, NADH and FADH2, have different roles in regulating the oxidation of nutrients, membrane potentials and biosynthesis. Trans-hydrogenation of NADH to NADPH activates the biosynthesis. FADH2 sustains the membrane potential during the cell transformations. Glycolytic enzymes assume the non-canonical moonlighting functions, enter the nucleus to remodel the genetic programmes to affect the tissue turnover for efficient use of nutrients. Glycosylation of the CD98 (4F2HC) stabilises the nutrient transporters and regulates the entry of cysteine, glutamine and BCAA into the cells. A reciprocal relationship between the leucine and glutamine entry into cells regulates the cholesterol and fatty acid synthesis and homeostasis in cells. Insulin promotes the Pi transport from the blood to tissues, activates the mitochondrial respiratory activity, and glutamine metabolism, which activates the synthesis of cholesterol and the de novo fatty acids for reorganising and stabilising the lipid membranes for nutrient transport and signal transduction in response to fluctuations in the microenvironmental cues. Fatty acids provide the lipid metabolites, activate the second messengers and protein kinases. Insulin resistance suppresses the lipid raft formation and the mitotic slippage activates the fibrosis and slow death pathways.
Collapse
Affiliation(s)
| | - Meera Indracanti
- Institute of Biotechnology, University of Gondar, Gondar, Ethiopia
| | - Suresh K Kalangi
- Amity Stem Cell Institute, Amity University Haryana, Amity Education Valley Pachgaon, Manesar, Gurugram, HR 122413 India
| | - B Meher Gayatri
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046 India
| | - Navya G Naidu
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046 India
| | - Aramati B M Reddy
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046 India
| |
Collapse
|
48
|
Jarrell ZR, Smith MR, Hu X, Orr M, Liu KH, Quyyumi AA, Jones DP, Go YM. Plasma acylcarnitine levels increase with healthy aging. Aging (Albany NY) 2020; 12:13555-13570. [PMID: 32554854 PMCID: PMC7377890 DOI: 10.18632/aging.103462] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/27/2020] [Indexed: 05/27/2023]
Abstract
Acylcarnitines transport fatty acids into mitochondria and are essential for β-oxidation and energy metabolism. Decreased mitochondrial activity results in increased plasma acylcarnitines, and increased acylcarnitines activate proinflammatory signaling and associate with age-related disease. Changes in acylcarnitines associated with healthy aging, however, are not well characterized. In the present study, we examined the associations of plasma acylcarnitines with age (range: 20-90) in 163 healthy, non-diseased individuals from the predictive medicine research cohort (NCT00336570) and tested for gender-specific differences. The results show that long-chain and very long-chain acylcarnitines increased with age, while many odd-chain acylcarnitines decreased with age. Gender-specific differences were observed for several acylcarnitines, e.g., eicosadienoylcarnitine varied with age in males, and hydroxystearoylcarnitine varied in females. Metabolome-wide association study (MWAS) of age-associated acylcarnitines with all untargeted metabolic features showed little overlap between genders. These results show that plasma concentrations of acylcarnitines vary with age and gender in individuals selected for criteria of health. Whether these variations reflect mitochondrial dysfunction with aging, mitochondrial reprogramming in response to chronic environmental exposures, early pre-disease change, or an adaptive response to healthy aging, is unclear. The results highlight a potential utility for untargeted metabolomics research to elucidate gender-specific mechanisms of aging and age-related disease.
Collapse
Affiliation(s)
- Zachery R. Jarrell
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta, GA 30322, USA
| | - M. Ryan Smith
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta, GA 30322, USA
| | - Xin Hu
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta, GA 30322, USA
| | - Michael Orr
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta, GA 30322, USA
| | - Ken H. Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta, GA 30322, USA
| | - Arshed A. Quyyumi
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dean P. Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta, GA 30322, USA
| | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
49
|
Xu J, Martien J, Gilbertson C, Ma J, Amador-Noguez D, Park JO. Metabolic flux analysis and fluxomics-driven determination of reaction free energy using multiple isotopes. Curr Opin Biotechnol 2020; 64:151-160. [PMID: 32304936 DOI: 10.1016/j.copbio.2020.02.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 02/08/2020] [Accepted: 02/25/2020] [Indexed: 10/24/2022]
Abstract
Metabolite concentrations, fluxes, and free energies constitute the basis for understanding and controlling metabolism. Mass spectrometry and stable isotopes are integral tools in quantifying these metabolic features. For absolute metabolite concentration and flux measurement, 13C internal standards and tracers have been the gold standard. In contrast, no established methods exist for comprehensive thermodynamic quantitation under physiological environments. Recently, using high-resolution mass spectrometry and multi-isotope tracing, flux quantitation has been increasingly adopted in broader metabolism. The improved flux quantitation led to determination of Gibbs free energy of reaction (ΔG) in central carbon metabolism using a relationship between reaction reversibility and thermodynamic driving force. Here we highlight recent advances in multi-isotope tracing for metabolic flux and free energy analysis.
Collapse
Affiliation(s)
- Jimmy Xu
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Julia Martien
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cole Gilbertson
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Junyu Ma
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel Amador-Noguez
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Junyoung O Park
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
50
|
He J, Ma J, Ren B, Liu A. Advances in systemic lupus erythematosus pathogenesis via mTOR signaling pathway. Semin Arthritis Rheum 2020; 50:314-320. [DOI: 10.1016/j.semarthrit.2019.09.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/31/2019] [Accepted: 09/25/2019] [Indexed: 12/16/2022]
|