1
|
Solomon RN, Pittner NA, McCoy JR, Warwick PA, McBride JW. Cell signaling in Ehrlichia infection and cancer: Parallels in pathogenesis. Front Cell Infect Microbiol 2025; 15:1539847. [PMID: 40028182 PMCID: PMC11868041 DOI: 10.3389/fcimb.2025.1539847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Ehrlichia chaffeensis (E. chaffeensis) has recently emerged as an intracellular bacterial pathogen with sophisticated survival mechanisms that include repurposing evolutionarily conserved eukaryotic cell signaling pathways for immune evasion. E. chaffeensis exploits four major developmental signaling pathways (Wnt, Notch, Hedgehog, and Hippo) using short linear motif (SLiM) ligand mimicry to initiate signaling cascades. Dysregulation of these major signaling pathways leading to unchecked cell survival is implicated in various diseases, most notably cancer. E. chaffeensis exploits Wnt, Notch, Hedgehog and Hippo signaling pathways to inhibit apoptosis and co-opt other cellular functions to promote infection. This review will explore the signaling pathways exploited during Ehrlichia infection and the new discoveries that have illuminated this interesting example of the cell signaling convergence in cellular infection and cancer biology.
Collapse
Affiliation(s)
- Regina N. Solomon
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nicholas A. Pittner
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jaclyn R. McCoy
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Paityn A. Warwick
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
2
|
Li Z, Ngu R, Naik AA, Trinh K, Paharkova V, Liao H, Liu Y, Zhuang C, Le D, Pei H, Asante I, Mittelman SD, Louie S. Adipocyte maturation impacts daunorubicin disposition and metabolism. Eur J Clin Invest 2024; 54:e14307. [PMID: 39254480 DOI: 10.1111/eci.14307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Acute lymphoblastic leukaemia (ALL) is the most common type of childhood leukaemia with effective chemotherapeutic treatment. However, obesity has been associated with higher ALL chemoresistance rates and lower event-free survival rates. The molecular mechanism of how obesity promotes chemotherapy resistance is not well delineated. OBJECTIVES This study evaluated the effect of adipocyte maturation on sequestration and metabolism of chemotherapeutic drug daunorubicin (DNR). METHODS Using targeted LC-MS/MS multi-analyte assay, DNR sequestration and metabolism were studied in human preadipocyte and adipocyte cell lines, where expressions of DNR-metabolizing enzymes aldo-keto reductases (AKR) and carbonyl reductases (CBR) were also evaluated. In addition, to identify the most DNR-metabolizing AKR/CBR isoforms, recombinant human AKR and CBR enzymes were subject to DNR metabolism. The results were further validated by AKR-, CBR-specific inhibitors. RESULTS This report shows that adipocyte maturation upregulates expressions of AKR and CBR enzymes (by 4- to 60- folds, p < .05), which is positively associated with enhanced sequestration and metabolism of DNR in adipocytes compared to preadipocytes (by ~30%, p < .05). In particular, adipocyte maturation upregulates AKR1C3 and CBR1, which are the predominate metabolic enzyme isoforms responsible for DNR biotransformation to its metabolites. CONCLUSION Fat is an expandable tissue that can sequester and detoxify DNR when stimulated by obesity, likely through the upregulation of DNR-metabolizing enzymes AKR1C3 and CBR1. Our data partially explains why obese ALL patients may be more likely to become chemoresistant towards DNR, and provides evidence for potential clinical investigation targeting obesity to reduce DNR chemoresistance.
Collapse
Affiliation(s)
- Zeyang Li
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Rachael Ngu
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Aditya Anil Naik
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Kaitlyn Trinh
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Vladislava Paharkova
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, California, USA
| | - Hanyue Liao
- College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Yulu Liu
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Cindy Zhuang
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Danh Le
- Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Hua Pei
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Isaac Asante
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Steven D Mittelman
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, California, USA
| | - Stan Louie
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
3
|
Ehm PAH, Horn S, Hoffer K, Kriegs M, Horn M, Giehler S, Nalaskowski M, Rehbach C, Horstmann MA, Jücker M. Ikaros sets the threshold for negative B-cell selection by regulation of the signaling strength of the AKT pathway. Cell Commun Signal 2024; 22:360. [PMID: 38992657 PMCID: PMC11241878 DOI: 10.1186/s12964-024-01732-5] [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: 03/01/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
Inhibitory phosphatases, such as the inositol-5-phosphatase SHIP1 could potentially contribute to B-cell acute lymphoblastic leukemia (B-ALL) by raising the threshold for activation of the autoimmunity checkpoint, allowing malignant cells with strong oncogenic B-cell receptor signaling to escape negative selection. Here, we show that SHIP1 is differentially expressed across B-ALL subtypes and that high versus low SHIP1 expression is associated with specific B-ALL subgroups. In particular, we found high SHIP1 expression in both, Philadelphia chromosome (Ph)-positive and ETV6-RUNX1-rearranged B-ALL cells. As demonstrated by targeted knockdown of SHIP1 by RNA interference, proliferation of B-ALL cells in vitro and their tumorigenic spread in vivo depended in part on SHIP1 expression. We investigated the regulation of SHIP1, as an important antagonist of the AKT signaling pathway, by the B-cell-specific transcription factor Ikaros. Targeted restoration of Ikaros and pharmacological inhibition of the antagonistic casein kinase 2, led to a strong reduction in SHIP1 expression and at the same time to a significant inhibition of AKT activation and cell growth. Importantly, the tumor suppressive function of Ikaros was enhanced by a SHIP1-dependent additive effect. Furthermore, our study shows that all three AKT isoforms contribute to the pro-mitogenic and anti-apoptotic signaling in B-ALL cells. Conversely, hyperactivation of a single AKT isoform is sufficient to induce negative selection by increased oxidative stress. In summary, our study demonstrates the regulatory function of Ikaros on SHIP1 expression in B-ALL and highlights the relevance of sustained SHIP1 expression to prevent cells with hyperactivated PI3K/AKT/mTOR signaling from undergoing negative selection.
Collapse
Affiliation(s)
- Patrick A H Ehm
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany.
- Department of Pediatric Oncology and Hematology, Research Institute Children's Cancer Center Hamburg, University Medical Center, Hamburg, 20246, Germany.
| | - Stefan Horn
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Konstantin Hoffer
- UCCH Kinomics Core Facility, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Malte Kriegs
- UCCH Kinomics Core Facility, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Michael Horn
- University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
- Mildred Scheel Cancer Career Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Susanne Giehler
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Marcus Nalaskowski
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Christoph Rehbach
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
- Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Martin A Horstmann
- Department of Pediatric Oncology and Hematology, Research Institute Children's Cancer Center Hamburg, University Medical Center, Hamburg, 20246, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| |
Collapse
|
4
|
Peeters R, Jellusova J. Lipid metabolism in B cell biology. Mol Oncol 2024; 18:1795-1813. [PMID: 38013654 PMCID: PMC11223608 DOI: 10.1002/1878-0261.13560] [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: 08/03/2023] [Revised: 10/30/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023] Open
Abstract
In recent years, the field of immunometabolism has solidified its position as a prominent area of investigation within the realm of immunological research. An expanding body of scientific literature has unveiled the intricate interplay between energy homeostasis, signalling molecules, and metabolites in relation to fundamental aspects of our immune cells. It is now widely accepted that disruptions in metabolic equilibrium can give rise to a myriad of pathological conditions, ranging from autoimmune disorders to cancer. Emerging evidence, although sometimes fragmented and anecdotal, has highlighted the indispensable role of lipids in modulating the behaviour of immune cells, including B cells. In light of these findings, this review aims to provide a comprehensive overview of the current state of knowledge regarding lipid metabolism in the context of B cell biology.
Collapse
Affiliation(s)
- Rens Peeters
- School of Medicine and Health, Institute of Clinical Chemistry and PathobiochemistryTechnical University of MunichGermany
- TranslaTUM, Center for Translational Cancer ResearchTechnical University of MunichGermany
| | - Julia Jellusova
- School of Medicine and Health, Institute of Clinical Chemistry and PathobiochemistryTechnical University of MunichGermany
- TranslaTUM, Center for Translational Cancer ResearchTechnical University of MunichGermany
| |
Collapse
|
5
|
Simon‐Molas H, Del Prete R, Kabanova A. Glucose metabolism in B cell malignancies: a focus on glycolysis branching pathways. Mol Oncol 2024; 18:1777-1794. [PMID: 38115544 PMCID: PMC11223612 DOI: 10.1002/1878-0261.13570] [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: 07/17/2023] [Revised: 10/13/2023] [Accepted: 12/15/2023] [Indexed: 12/21/2023] Open
Abstract
Glucose catabolism, one of the essential pathways sustaining cellular bioenergetics, has been widely studied in the context of tumors. Nevertheless, the function of various branches of glucose metabolism that stem from 'classical' glycolysis have only been partially explored. This review focuses on discussing general mechanisms and pathological implications of glycolysis and its branching pathways in the biology of B cell malignancies. We summarize here what is known regarding pentose phosphate, hexosamine, serine biosynthesis, and glycogen synthesis pathways in this group of tumors. Despite most findings have been based on malignant B cells themselves, we also discuss the role of glucose metabolism in the tumor microenvironment, with a focus on T cells. Understanding the contribution of glycolysis branching pathways and how they are hijacked in B cell malignancies will help to dissect the role they have in sustaining the dissemination and proliferation of tumor B cells and regulating immune responses within these tumors. Ultimately, this should lead to deciphering associated vulnerabilities and improve current therapeutic schedules.
Collapse
Affiliation(s)
- Helga Simon‐Molas
- Departments of Experimental Immunology and HematologyAmsterdam UMC location University of AmsterdamThe Netherlands
- Cancer ImmunologyCancer Center AmsterdamThe Netherlands
| | | | - Anna Kabanova
- Fondazione Toscana Life Sciences FoundationSienaItaly
| |
Collapse
|
6
|
Tsilingiris D, Vallianou NG, Spyrou N, Kounatidis D, Christodoulatos GS, Karampela I, Dalamaga M. Obesity and Leukemia: Biological Mechanisms, Perspectives, and Challenges. Curr Obes Rep 2024; 13:1-34. [PMID: 38159164 PMCID: PMC10933194 DOI: 10.1007/s13679-023-00542-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW To examine the epidemiological data on obesity and leukemia; evaluate the effect of obesity on leukemia outcomes in childhood acute lymphoblastic leukemia (ALL) survivors; assess the potential mechanisms through which obesity may increase the risk of leukemia; and provide the effects of obesity management on leukemia. Preventive (diet, physical exercise, obesity pharmacotherapy, bariatric surgery) measures, repurposing drugs, candidate therapeutic agents targeting oncogenic pathways of obesity and insulin resistance in leukemia as well as challenges of the COVID-19 pandemic are also discussed. RECENT FINDINGS Obesity has been implicated in the development of 13 cancers, such as breast, endometrial, colon, renal, esophageal cancers, and multiple myeloma. Leukemia is estimated to account for approximately 2.5% and 3.1% of all new cancer incidence and mortality, respectively, while it represents the most frequent cancer in children younger than 5 years. Current evidence indicates that obesity may have an impact on the risk of leukemia. Increased birthweight may be associated with the development of childhood leukemia. Obesity is also associated with worse outcomes and increased mortality in leukemic patients. However, there are several limitations and challenges in meta-analyses and epidemiological studies. In addition, weight gain may occur in a substantial number of childhood ALL survivors while the majority of studies have documented an increased risk of relapse and mortality among patients with childhood ALL and obesity. The main pathophysiological pathways linking obesity to leukemia include bone marrow adipose tissue; hormones such as insulin and the insulin-like growth factor system as well as sex hormones; pro-inflammatory cytokines, such as IL-6 and TNF-α; adipocytokines, such as adiponectin, leptin, resistin, and visfatin; dyslipidemia and lipid signaling; chronic low-grade inflammation and oxidative stress; and other emerging mechanisms. Obesity represents a risk factor for leukemia, being among the only known risk factors that could be prevented or modified through weight loss, healthy diet, and physical exercise. Pharmacological interventions, repurposing drugs used for cardiometabolic comorbidities, and bariatric surgery may be recommended for leukemia and obesity-related cancer prevention.
Collapse
Affiliation(s)
- Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, 68100, Alexandroupolis, Greece
| | - Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Nikolaos Spyrou
- Tisch Cancer Institute Icahn School of Medicine at Mount Sinai, 1190 One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Dimitris Kounatidis
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini Str, 12462, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, 11527, Athens, Greece.
| |
Collapse
|
7
|
Phan UTT, Nguyen HD, Nguyen TKO, Tran TH, Le TH, Tran TTP. Anti-inflammatory effect of Piper longum L. fruit methanolic extract on lipopolysaccharide-treated RAW 264.7 murine macrophages. Heliyon 2024; 10:e26174. [PMID: 38404825 PMCID: PMC10884859 DOI: 10.1016/j.heliyon.2024.e26174] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
Context The Piper species was studied several potential properties such as anti-tumor, anti-inflammatory and antioxidant activity. However, the specific anti-inflammatory activity of the extract from the fruits of P. longum L. has not been investigated. Objectives Our study want to examine the anti-inflammatory effects of P. longum L. fruit methanolic extracts (PLE) on lipopolysachharide (LPS)-stimulated RAW 264.7 murine macrophages to understand the mechanism of this effect. Method This study examined the chemical profiling of PLE by LC-HRMS analysis and measured the presence of nitric oxide (NO), interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) in the supernatant using the Griess reagent assay and enzyme-linked immunosorbent assay (ELISA), respectively. The mRNA expression of IL-6, TNF-α, cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) were evaluated by using real-time quantitative polymerase chain reaction (RT-qPCR). Furthermore, the protein expression of COX-2, iNOS and the phosphorylation of MAPK family, c-Jun N-terminal kinase (JNK), p38 in protein level were observed by western blotting. Result PLE have detected 66 compounds which belong to different classes such as alkaloids, flavonoids, terpenoids, phenolics, lactones, and organic acids inhibited nitric oxide products with the IC50 = 28.5 ± 0.91 μg/mL. Moreover, PLE at 10-100 μg/mL up-regulate HO-1 protein expression from 3 to 10 folds at 3 h. It also downregulated the mRNA and protein expression of iNOS, COX-2, decreased IL-6 and TNF-α secretion by modulating the mitogen-activated protein kinase (MAPK) signaling pathway, specifically by decreasing the phosphorylation of p38 and JNK. Conclusion These results shown chemical profiling of PLE and demonstrated that PLE exhibits anti-inflammatory effects by regulating the MAPK family and could be a potential candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Uyen Thi Tu Phan
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| | - Hai Dang Nguyen
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| | - Thi Kieu Oanh Nguyen
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi, 12116, Viet Nam
| | - Thanh Huong Le
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| | - Thi Thu Phuong Tran
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| |
Collapse
|
8
|
Brookens SK, Cho SH, Paik Y, Meyer K, Raybuck AL, Park C, Greenwood DL, Rathmell JC, Boothby MR. Plasma Cell Differentiation, Antibody Quality, and Initial Germinal Center B Cell Population Depend on Glucose Influx Rate. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:43-56. [PMID: 37955416 PMCID: PMC10841396 DOI: 10.4049/jimmunol.2200756] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/19/2023] [Indexed: 11/14/2023]
Abstract
Serum Ab concentrations, selection for higher affinity BCRs, and generation of higher Ab affinities are important elements of immune response optimization and functions of germinal center (GC) reactions. B cell proliferation requires nutrients to support the anabolism inherent in clonal expansion. Glucose usage by mouse GC B cells has been reported to contribute little to their energy needs, with questions raised as to whether glucose uptake or glycolysis increases in GC B cells compared with their naive precursors. Indeed, metabolism can be highly flexible, such that supply shortage along one pathway may be compensated by increased flux on others. We now show that reduction of the glucose transporter GLUT1 in mice after establishment of a preimmune B cell repertoire, even after initiation of the GC B cell gene expression program, decreased initial GC B cell population numbers, affinity maturation, and plasma cell outputs. Glucose oxidation was heightened in GC B cells, but this hexose flowed more into the pentose phosphate pathway, whose activity was important in controlling reactive oxygen species (ROS) and Ab-secreting cell production. In modeling how glucose usage by B cells promotes the Ab response, the control of ROS appeared insufficient. Surprisingly, the combination of galactose, which mitigated ROS, with provision of mannose, an efficient precursor to glycosylation, supported robust production of and normal Ab secretion by Ab-secreting cells under glucose-free conditions. Collectively, the findings indicate that GCs depend on normal glucose influx, especially in plasma cell production, but reveal an unexpected metabolic flexibility in hexose requirements.
Collapse
Affiliation(s)
- Shawna K. Brookens
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Cancer Biology Program, Vanderbilt University
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104
| | - Sung Hoon Cho
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Inflammation, & Immunology
| | - Yeeun Paik
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Kaylor Meyer
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Ariel L. Raybuck
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Chloe Park
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Dalton L. Greenwood
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jeffrey C. Rathmell
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Cancer Biology Program, Vanderbilt University
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Inflammation, & Immunology
| | - Mark R. Boothby
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Cancer Biology Program, Vanderbilt University
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Inflammation, & Immunology
| |
Collapse
|
9
|
Byerly CD, Patterson LL, Pittner NA, Solomon RN, Patel JG, Rogan MR, McBride JW. Ehrlichia Wnt SLiM ligand mimic deactivates the Hippo pathway to engage the anti-apoptotic Yap-GLUT1-BCL-xL axis. Infect Immun 2023; 91:e0008523. [PMID: 37530530 PMCID: PMC10501218 DOI: 10.1128/iai.00085-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/03/2023] [Indexed: 08/03/2023] Open
Abstract
Ehrlichia chaffeensis TRP120 effector has evolved short linear motif (SLiM) ligand mimicry to repurpose multiple evolutionarily conserved cellular signaling pathways, including Wnt, Notch, and Hedgehog. In this investigation, we demonstrate that E. chaffeensis and recombinant TRP120 deactivate Hippo signaling, resulting in the activation of Hippo transcription coactivator Yes-associated protein (Yap). Moreover, a homologous 6 amino acid (QDVASH) SLiM shared by TRP120 and Wnt3a/5a ligands phenocopied Yap and β-catenin activation induced by E. chaffeensis, rTRP120, and Wnt5a. Similar Hippo gene expression profiles were also stimulated by E. chaffeensis, rTRP120, SLiM, and Wnt5a. Single siRNA knockdown of Hippo transcription co-activator/factors, Yap, and transcriptional enhanced associate domain (TEAD) significantly decreased E. chaffeensis infection. Yap activation was abolished in THP-1 Wnt Frizzled-5 (Fzd5) receptor knockout cells (KO), demonstrating Fzd5 receptor dependence. In addition, the TRP120-Wnt-SLiM antibody blocked Hippo deactivation (Yap activation). Expression of anti-apoptotic Hippo target gene SLC2A1 (encodes glucose transporter 1; GLUT1) was upregulated by E. chaffeensis and corresponded to increased levels of GLUT1. Conversely, siRNA knockdown of SLC2A1 significantly inhibited infection. Higher GLUT1 levels correlated with increased B cell lymphoma-extra large (BCL-xL) and decreased BCL2-associated X, apoptosis regulator (Bax) levels. Moreover, blocking Yap activation with the inhibitor Verteporfin induced apoptosis that corresponded to significant reductions in GLUT1 and BCL-xL levels and activation of Bax and Caspase-3 and -9. This study identifies a novel shared Wnt/Hippo SLiM ligand mimic and demonstrates that E. chaffeensis deactivates the Hippo pathway to engage the anti-apoptotic Yap-GLUT1-BCL-xL axis.
Collapse
Affiliation(s)
- Caitlan D. Byerly
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - LaNisha L. Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Nicholas A. Pittner
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Regina N. Solomon
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jignesh G. Patel
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Madison R. Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
10
|
Bahremani M, Rashtchizadeh N, Sabzichi M, Vatankhah AM, Danaiyan S, Poursistany H, Mohammadian J, Ghorbanihaghjo A. Enhanced chemotherapeutic efficacy of docetaxel in human lung cancer cell line via GLUT1 inhibitor. J Biochem Mol Toxicol 2023; 37:e23348. [PMID: 36999407 DOI: 10.1002/jbt.23348] [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: 06/02/2022] [Revised: 02/01/2023] [Accepted: 03/20/2023] [Indexed: 04/01/2023]
Abstract
The dose-dependent adverse effects of anticancer agents need new methods with lesser toxicity. The objective of the current research was to evaluate the efficacy of GLUT1 inhibitor, as an inhibitor of glucose consumption in cancer cells, in augmenting the efficiency of docetaxel with respect to cytotoxicity and apoptosis. Cell cytotoxicity was assessed by using methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. Annexin V/PI double staining was employed to evaluate apoptosis percentage. Quantitative real-time polymerase chain reaction (RT-PCR) analysis was accomplished to detect the expression of genes involved in the apoptosis pathway. The IC50 values for docetaxel and BAY-876 were 3.7 ± 0.81 and 34.1 ± 3.4 nM, respectively. The severity of synergistic mutual effects of these agents on each other was calculated by synergy finder application. It showed that the percentage of apoptotic cells following co-administration of docetaxel and BAY-876 increased to 48.1 ± 2.8%. In comparison without GLUT1 co-administration, the combined therapy decreased significantly the transcriptome levels of the Bcl-2 and Ki-67 and a remarkable increase in the level of the Bax as proapoptotic protein(p < 0.05). Co-treatment of BAY-876 and docetaxel depicted a synergistic effect which was calculated using the synergy finder highest single agent (HSA) method (HSA synergy score: 28.055). These findings recommend that the combination of GLUT-1 inhibitor and docetaxel can be considered as a promising therapeutic approach for the treatment of patients with lung cancer.
Collapse
Affiliation(s)
- Mona Bahremani
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadereh Rashtchizadeh
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Sabzichi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sepideh Danaiyan
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haniyeh Poursistany
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jamal Mohammadian
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghorbanihaghjo
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Byerly CD, Patterson LL, Pittner NA, Solomon RN, Patel JG, Rogan MR, McBride JW. Ehrlichia Wnt short linear motif ligand mimetic deactivates the Hippo pathway to engage the anti-apoptotic Yap-GLUT1-BCL-xL axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531456. [PMID: 36945589 PMCID: PMC10028901 DOI: 10.1101/2023.03.06.531456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Ehrlichia chaffeensis TRP120 effector has evolved short linear motif (SLiM) ligand mimicry to repurpose multiple evolutionarily conserved cellular signaling pathways including Wnt, Notch and Hedgehog. In this investigation, we demonstrate that E. chaffeensis and recombinant TRP120 deactivate Hippo signaling resulting in activation of Hippo transcription coactivator Yap and target gene expression. Moreover, a homologous 6 amino acid (QDVASH) SLiM shared by TRP120 and Wnt3a/5a ligands phenocopied Yap and β-catenin activation induced by E. chaffeensis, rTRP120 and Wnt5a. Similar Hippo gene expression profiles were also stimulated by E. chaffeensis, rTRP120, SLiM and Wnt5a. Single siRNA knockdown of Hippo transcription co-activator/factors (Yap and TEAD) significantly decreased E. chaffeensis infection. Yap activation was abolished in THP-1 Wnt Frizzled-5 (Fzd5) receptor knockout cells (KO), demonstrating Fzd5 receptor dependence. In addition, TRP120 Wnt-SLiM antibody blocked Hippo deactivation (Yap activation). Expression of anti-apoptotic Hippo target gene SLC2A1 (encodes glucose transporter 1; GLUT1) was upregulated by E. chaffeensis and corresponded to increased levels of GLUT1. Conversely, siRNA knockdown of SLC2A1 significantly inhibited infection. Higher GLUT1 levels correlated with increased BCL-xL and decreased Bax levels. Moreover, blocking Yap activation with the inhibitor Verteporfin induced apoptosis that corresponded to significant reductions in levels of GLUT1 and BCL-xL, and activation of Bax and Caspase-3 and -9. This study identifies a novel shared Wnt/Hippo SLiM ligand mimetic and demonstrates that E. chaffeensis deactivates the Hippo pathway to engage the anti-apoptotic Yap-GLUT1-BCL-xL axis.
Collapse
Affiliation(s)
- Caitlan D. Byerly
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - LaNisha L. Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Nicholas A. Pittner
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Regina N. Solomon
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jignesh G. Patel
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Madison R. Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Department Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
12
|
Jung J, Gokhale S, Xie P. TRAF3: A novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes. Front Oncol 2023; 13:1081253. [PMID: 36776285 PMCID: PMC9911533 DOI: 10.3389/fonc.2023.1081253] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Mitochondria, the organelle critical for cell survival and metabolism, are exploited by cancer cells and provide an important therapeutic target in cancers. Mitochondria dynamically undergo fission and fusion to maintain their diverse functions. Proteins controlling mitochondrial fission and fusion have been recognized as essential regulators of mitochondrial functions, mitochondrial quality control, and cell survival. In a recent proteomic study, we identified the key mitochondrial fission factor, MFF, as a new interacting protein of TRAF3, a known tumor suppressor of multiple myeloma and other B cell malignancies. This interaction recruits the majority of cytoplasmic TRAF3 to mitochondria, allowing TRAF3 to regulate mitochondrial morphology, mitochondrial functions, and mitochondria-dependent apoptosis in resting B lymphocytes. Interestingly, recent transcriptomic, metabolic and lipidomic studies have revealed that TRAF3 also vitally regulates multiple metabolic pathways in B cells, including phospholipid metabolism, glucose metabolism, and ribonucleotide metabolism. Thus, TRAF3 emerges as a novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes and B cell malignancies. Here we review current knowledge in this area and discuss relevant clinical implications.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
13
|
Hu YZ, Li Q, Wang PF, Li XP, Hu ZL. Multiple functions and regulatory network of miR-150 in B lymphocyte-related diseases. Front Oncol 2023; 13:1140813. [PMID: 37182123 PMCID: PMC10172652 DOI: 10.3389/fonc.2023.1140813] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/06/2023] [Indexed: 05/16/2023] Open
Abstract
MicroRNAs (miRNAs) play vital roles in the post-transcriptional regulation of gene expression. Previous studies have shown that miR-150 is a crucial regulator of B cell proliferation, differentiation, metabolism, and apoptosis. miR-150 regulates the immune homeostasis during the development of obesity and is aberrantly expressed in multiple B-cell-related malignant tumors. Additionally, the altered expression of MIR-150 is a diagnostic biomarker of various autoimmune diseases. Furthermore, exosome-derived miR-150 is considered as prognostic tool in B cell lymphoma, autoimmune diseases and immune-mediated disorders, suggesting miR-150 plays a vital role in disease onset and progression. In this review, we summarized the miR-150-dependent regulation of B cell function in B cell-related immune diseases.
Collapse
Affiliation(s)
- Yue-Zi Hu
- Clinical Laboratory, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Qiao Li
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
| | - Peng-Fei Wang
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
| | - Xue-Ping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Affiliated Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhao-Lan Hu,
| |
Collapse
|
14
|
Wang K, Yang T, Zhang Y, Gao X, Tao L. The opportunities and challenges for nutritional intervention in childhood cancers. Front Nutr 2023; 10:1091067. [PMID: 36925958 PMCID: PMC10012036 DOI: 10.3389/fnut.2023.1091067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Diet dictates nutrient availability in the tumor microenvironment, thus affecting tumor metabolic activity and growth. Intrinsically, tumors develop unique metabolic features and are sensitive to environmental nutrient concentrations. Tumor-driven nutrient dependencies provide opportunities to control tumor growth by nutritional restriction or supplementation. This review summarized the existing data on nutrition and pediatric cancers after systematically searching articles up to 2023 from four databases (PubMed, Web of Science, Scopus, and Ovid MEDLINE). Epidemiological studies linked malnutrition with advanced disease stages and poor clinical outcomes in pediatric cancer patients. Experimental studies identified several nutrient dependencies (i.e., amino acids, lipids, vitamins, etc.) in major pediatric cancer types. Dietary modifications such as calorie restriction, ketogenic diet, and nutrient restriction/supplementation supported pediatric cancer treatment, but studies remain limited. Future research should expand epidemiological studies through data sharing and multi-institutional collaborations and continue to discover critical and novel nutrient dependencies to find optimal nutritional approaches for pediatric cancer patients.
Collapse
Affiliation(s)
- Kaiyue Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| | - Tianyou Yang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yubin Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| | - Xiang Gao
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| | - Ling Tao
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Chen Y, Yang H, Chen S, Lu Z, Li B, Jiang T, Xuan M, Ye R, Liang H, Liu X, Liu Q, Tang H. SIRT1 regulated hexokinase-2 promoting glycolysis is involved in hydroquinone-enhanced malignant progression in human lymphoblastoid TK6 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113757. [PMID: 35714482 DOI: 10.1016/j.ecoenv.2022.113757] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 06/15/2023]
Abstract
Reprogramming of cellular metabolism is a vital event during tumorigenesis. The role of glycolysis in malignant progression promoted by hydroquinone (HQ), one of the metabolic products of benzene, remains to be understood. Recently, we reported the overexpression of sirtuin 1 (SIRT1) in HQ-enhanced malignant progression of TK6 cells and hypothesized that SIRT1 might contribute to glycolysis and favor tumorigenesis. Our data showed that acute exposure of TK6 cells to HQ for 48 h inhibited glycolysis, as indicated by reduction in glucose consumption, lactate production, hexokinase activity, and the expression of SIRT1 and glycolytic enzymes, including HIF-1α, hexokinase-2 (HK-2), ENO-1, glucose transporter 1 (Glut-1), and lactic dehydrogenase A (LDHA). Knockdown of SIRT1 or inhibition of glycolysis using the glycolytic inhibitor 2-deoxy-D-glucose (2-DG) downregulated the levels of SIRT1 and glycolytic enzymes and significantly enhanced HQ-induced cell apoptosis, although knockdown of SIRT1 or 2-DG alone had little effect on apoptosis. Furthermore, immunofluorescence and Co-IP assays demonstrated that SIRT1 regulated the expression of HK-2, and HQ treatment caused a decrease in SIRT1 and HK-2 binding to mitochondria. Importantly, we found that glycolysis was promoted with increasing HQ treatment weeks. Long-term HQ exposure increased the expression of SIRT1 and several glycolytic enzymes and promoted malignant cell progression. Moreover, compared with the PBS group, glucose consumption and lactate production increased after 10 weeks of HQ exposure, and the protein levels of SIRT1 and HK-2 were increased after 15 weeks of HQ exposure, while those of Glut-1, ENO-1, and LDHA were elevated. In addition, SIRT1 knockdown HQ 19 cells exhibited decreased lactate production, glucose consumption, glycolytic enzymes expression, cell growth, and tumor formation in nude mice. Our findings identify the high expression of SIRT1 as a strong oncogenic driver that positively regulates HK-2 and promotes glycolysis in HQ-accelerated malignant progression of TK6 cells.
Collapse
Affiliation(s)
- Yuting Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Hui Yang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Shaoyun Chen
- Department of Obstetrics, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen 518102, China
| | - Zhaohong Lu
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Boxin Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Tikeng Jiang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Mei Xuan
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Ruifang Ye
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Hairong Liang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Xiaoshan Liu
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China
| | - Qizhan Liu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Huanwen Tang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Guangdong 523808, China.
| |
Collapse
|
16
|
Chakraborty S, Khamaru P, Bhattacharyya A. Regulation of immune cell metabolism in health and disease: Special focus on T and B cell subsets. Cell Biol Int 2022; 46:1729-1746. [PMID: 35900141 DOI: 10.1002/cbin.11867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 04/03/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022]
Abstract
Metabolism is a dynamic process and keeps changing from time to time according to the demand of a particular cell to meet its bio-energetic requirement. Different immune cells rely on distinct metabolic programs which allow the cell to balance its requirements for energy, molecular biosynthesis, and effector activity. In the aspect of infection and cancer immunology, effector T and B cells get exhausted and help tumor cells to evade immunosurveillance. On the other hand, T cells become hyperresponsive in the scenario of autoimmune diseases. In this article, we have explored the uniqueness and distinct metabolic features of key CD4+ T and B helper cell subsets, CD4+ T, B regulatory cell subsets and CD8+ T cells regarding health and disease. Th1 cells rely on glycolysis and glutaminolysis; inhibition of these metabolic pathways promotes Th1 cells in Treg population. However, Th2 cells are also dependent on glycolysis but an abundance of lactate within TME shifts their metabolic dependency to fatty acid metabolism. Th17 cells depend on HIF-1α mediated glycolysis, ablation of HIF-1α reduces Th17 cells but enhance Treg population. In contrast to effector T cells which are largely dependent on glycolysis for their differentiation and function, Treg cells mainly rely on FAO for their function. Therefore, it is of utmost importance to understand the metabolic fates of immune cells and how it facilitates their differentiation and function for different disease models. Targeting metabolic pathways to restore the functionality of immune cells in diseased conditions can lead to potent therapeutic measures.
Collapse
Affiliation(s)
- Sayan Chakraborty
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Poulomi Khamaru
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| |
Collapse
|
17
|
Zuo F, Yu J, He X. Single-Cell Metabolomics in Hematopoiesis and Hematological Malignancies. Front Oncol 2022; 12:931393. [PMID: 35912231 PMCID: PMC9326066 DOI: 10.3389/fonc.2022.931393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Aberrant metabolism contributes to tumor initiation, progression, metastasis, and drug resistance. Metabolic dysregulation has emerged as a hallmark of several hematologic malignancies. Decoding the molecular mechanism underlying metabolic rewiring in hematological malignancies would provide promising avenues for novel therapeutic interventions. Single-cell metabolic analysis can directly offer a meaningful readout of the cellular phenotype, allowing us to comprehensively dissect cellular states and access biological information unobtainable from bulk analysis. In this review, we first highlight the unique metabolic properties of hematologic malignancies and underscore potential metabolic vulnerabilities. We then emphasize the emerging single-cell metabolomics techniques, aiming to provide a guide to interrogating metabolism at single-cell resolution. Furthermore, we summarize recent studies demonstrating the power of single-cell metabolomics to uncover the roles of metabolic rewiring in tumor biology, cellular heterogeneity, immunometabolism, and therapeutic resistance. Meanwhile, we describe a practical view of the potential applications of single-cell metabolomics in hematopoiesis and hematological malignancies. Finally, we present the challenges and perspectives of single-cell metabolomics development.
Collapse
|
18
|
Patel SB, Nemkov T, D'Alessandro A, Welner RS. Deciphering Metabolic Adaptability of Leukemic Stem Cells. Front Oncol 2022; 12:846149. [PMID: 35756656 PMCID: PMC9213881 DOI: 10.3389/fonc.2022.846149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Therapeutic targeting of leukemic stem cells is widely studied to control leukemia. An emerging approach gaining popularity is altering metabolism as a potential therapeutic opportunity. Studies have been carried out on hematopoietic and leukemic stem cells to identify vulnerable pathways without impacting the non-transformed, healthy counterparts. While many metabolic studies have been conducted using stem cells, most have been carried out in vitro or on a larger population of progenitor cells due to challenges imposed by the low frequency of stem cells found in vivo. This creates artifacts in the studies carried out, making it difficult to interpret and correlate the findings to stem cells directly. This review discusses the metabolic difference seen between hematopoietic stem cells and leukemic stem cells across different leukemic models. Moreover, we also shed light on the advancements of metabolic techniques and current limitations and areas for additional research of the field to study stem cell metabolism.
Collapse
Affiliation(s)
- Sweta B Patel
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at, Birmingham, AL, United States.,Divison of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Robert S Welner
- Department of Medicine, Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at, Birmingham, AL, United States
| |
Collapse
|
19
|
Piktel D, Nair RR, Rellick SL, Geldenhuys WJ, Martin KH, Craig MD, Gibson LF. Pitavastatin Is Anti-Leukemic in a Bone Marrow Microenvironment Model of B-Lineage Acute Lymphoblastic Leukemia. Cancers (Basel) 2022; 14:cancers14112681. [PMID: 35681662 PMCID: PMC9179467 DOI: 10.3390/cancers14112681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Chemoresistance after chemotherapy is a negative prognostic indicator for B-cell acute lymphoblastic leukemia (ALL), necessitating the search for novel therapies. By growing ALL cells together with bone marrow stromal cells, we developed a chemoresistant ALL model. Using this model, we found that the lipid lowering drug pitavastatin had antileukemic activity in this chemoresistant co-culture model. Our data suggests that pitavastatin may be a novel treatment option for repurposing in chemoresistant, relapse ALL. Abstract The lack of complete therapeutic success in the treatment of B-cell acute lymphoblastic leukemia (ALL) has been attributed, in part, to a subset of cells within the bone marrow microenvironment that are drug resistant. Recently, the cholesterol synthesis inhibitor, pitavastatin (PIT), was shown to be active in acute myeloid leukemia, prompting us to evaluate it in our in vitro co-culture model, which supports a chemo-resistant ALL population. We used phospho-protein profiling to evaluate the use of lipid metabolic active compounds in these chemo-resistant cells, due to the up-regulation of multiple active survival signals. In a co-culture with stromal cells, a shift towards anabolic processes occurred, which was further confirmed by assays showing increased lipid content. The treatment of REH leukemia cells with pitavastatin in the co-culture model resulted in significantly higher leukemic cell death than exposure to the standard-of-care chemotherapeutic agent, cytarabine (Ara-C). Our data demonstrates the use of pitavastatin as a possible alternative treatment strategy to improve patient outcomes in chemo-resistant, relapsed ALL.
Collapse
Affiliation(s)
- Debbie Piktel
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
| | - Rajesh R. Nair
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
| | - Stephanie L. Rellick
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
| | - Werner J. Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA;
| | - Karen H. Martin
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | | | - Laura F. Gibson
- Robert C. Byrd Health Sciences Center, West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA; (D.P.); (R.R.N.); (S.L.R.); (K.H.M.)
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Correspondence: ; Tel.: +1-304-293-7206
| |
Collapse
|
20
|
Kaumeyer BA, Fidai SS, Thakral B, Wang SA, Arber DA, Cheng JX, Gurbuxani S, Venkataraman G. GLUT1 Immunohistochemistry Is a Highly Sensitive and Relatively Specific Marker for Erythroid Lineage in Benign and Malignant Hematopoietic Tissues. Am J Clin Pathol 2022; 158:228-234. [PMID: 35311938 DOI: 10.1093/ajcp/aqac034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/21/2022] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Glucose transporter 1 (GLUT1), a glucose transporter, is an abundant protein in erythrocytes with expression beginning early in erythropoiesis. We sought to evaluate the utility of GLUT1 immunohistochemistry (IHC) as a diagnostic marker for identifying erythroid differentiation in hematopoietic tissues, including neoplastic erythroid proliferations. METHODS A variety of benign and neoplastic bone marrow biopsy specimens containing variable proportions of erythroid precursors were selected (n = 46, including 36 cases of leukemia). GLUT1 IHC was performed using a commercially available polyclonal antibody. Each case was evaluated for staining of erythroid precursors, nonerythroid hematopoietic cells, and blasts. A GATA1/GLUT1 double stain was performed on one case to confirm coexpression of GLUT1 on early erythroid precursors. Staining was compared with other erythroid markers, including glycophorin C. RESULTS GLUT1 demonstrated strong membranous staining in erythroid precursors of all cases, which was restricted largely to the erythroid lineage. Of the 36 leukemia cases, all 6 cases of pure erythroid leukemia and both cases of therapy-related acute myeloid leukemia with erythroid differentiation showed positive GLUT1 staining in blasts. Otherwise, only lymphoblasts in B-lymphoblastic leukemia showed weak to moderate granular cytoplasmic staining (four of five cases). CONCLUSIONS GLUT1 IHC is a highly sensitive and relatively specific marker for erythroid lineage in benign and neoplastic bone marrow biopsy specimens.
Collapse
Affiliation(s)
| | - Shiraz S Fidai
- Department of Pathology, University of Chicago, Chicago, IL, USA
- Department of Pathology and Laboratory Medicine, John H. Stroger Hospital of Cook County, Chicago, IL, USA
| | - Beenu Thakral
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sa A Wang
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Jason X Cheng
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | | | |
Collapse
|
21
|
Sharma ND, Keewan E, Matlawska-Wasowska K. Metabolic Reprogramming and Cell Adhesion in Acute Leukemia Adaptation to the CNS Niche. Front Cell Dev Biol 2021; 9:767510. [PMID: 34957100 PMCID: PMC8703109 DOI: 10.3389/fcell.2021.767510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Involvement of the Central Nervous System (CNS) in acute leukemia confers poor prognosis and lower overall survival. Existing CNS-directed therapies are associated with a significant risk of short- or long-term toxicities. Leukemic cells can metabolically adapt and survive in the microenvironment of the CNS. The supporting role of the CNS microenvironment in leukemia progression and dissemination has not received sufficient attention. Understanding the mechanism by which leukemic cells survive in the nutrient-poor and oxygen-deprived CNS microenvironment will lead to the development of more specific and less toxic therapies. Here, we review the current literature regarding the roles of metabolic reprogramming in leukemic cell adhesion and survival in the CNS.
Collapse
Affiliation(s)
- Nitesh D Sharma
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, United States
| | - Esra'a Keewan
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, United States
| | - Ksenia Matlawska-Wasowska
- Department of Pediatrics, Division of Hematology-Oncology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
22
|
Hui PY, Chen YH, Qin J, Jiang XH. PON2 blockade overcomes dexamethasone resistance in acute lymphoblastic leukemia. Hematology 2021; 27:32-42. [PMID: 34957927 DOI: 10.1080/16078454.2021.2009643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVES The high frequency of chemotherapy resistance is ultimately responsible for clinical relapse in acute lymphoblastic leukemia (ALL). Nevertheless, the molecular mechanism relevant to glucocorticoid (GC) resistance remains ambiguous. METHODS Quantitative real-time polymerase chain reaction and Western blot were performed to detect the expressions of paraoxonase 2 (PON2), Bcl-2 and Bax. shRNA was used to knockdown PON2 expression in SUP-B15 and REH cell. CCK-8 and flow cytometry assay were conducted to monitor the changes of proliferation and apoptosis in ALL cells. The growth of ALL REH cells in vivo was determined using transplanted tumor model. RESULTS This study was designed to identify GC resistance-associated genes by means of the transcriptome chip from the public Gene Expression Omnibus database, and preliminarily investigation of dexamethasone (DEX)-resistance mechanism in ALL. We disclosed that PON2 expression was elevated in ALL patients and especially higher in DEX-resistance ALL patients. Then, cell apoptosis assay suggested that silencing of PON2 dramatically promoted in DEX-resistant ALL cells apoptosis and the activity of Caspase 3 induced by DEX administration. In xenograft tumor model, PON2 knockdown significantly reduced DEX-resistant ALL cells growth in immunodeficient mice. CONCLUSIONS Collectively, inhibition of PON2 may represent a novel method to restore the sensitivity of treatment-resistant ALL to GC-induced cell death.
Collapse
Affiliation(s)
- Pei-Ye Hui
- Pharmacy Department, Shandong Weifang Maternal and Child Health Hospital, Weifang, People's Republic of China
| | - Yan-Hua Chen
- Pharmacy Department, Rizhao people's Hospital, Rizhao, People's Republic of China
| | - Jing Qin
- Pharmacy Department, Rizhao people's Hospital, Rizhao, People's Republic of China
| | - Xiao-Hua Jiang
- Department of Pediatrics, 970 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Yantai, People's Republic of China
| |
Collapse
|
23
|
Soltani M, Zhao Y, Xia Z, Ganjalikhani Hakemi M, Bazhin AV. The Importance of Cellular Metabolic Pathways in Pathogenesis and Selective Treatments of Hematological Malignancies. Front Oncol 2021; 11:767026. [PMID: 34868994 PMCID: PMC8636012 DOI: 10.3389/fonc.2021.767026] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/20/2021] [Indexed: 02/05/2023] Open
Abstract
Despite recent advancements in the treatment of hematologic malignancies and the emergence of newer and more sophisticated therapeutic approaches such as immunotherapy, long-term overall survival remains unsatisfactory. Metabolic alteration, as an important hallmark of cancer cells, not only contributes to the malignant transformation of cells, but also promotes tumor progression and metastasis. As an immune-escape mechanism, the metabolic adaptation of the bone marrow microenvironment and leukemic cells is a major player in the suppression of anti-leukemia immune responses. Therefore, metabolic rewiring in leukemia would provide promising opportunities for newer therapeutic interventions. Several therapeutic agents which affect essential bioenergetic pathways in cancer cells including glycolysis, β-oxidation of fatty acids and Krebs cycle, or anabolic pathways such as lipid biosynthesis and pentose phosphate pathway, are being tested in various types of cancers. So far, numerous preclinical or clinical trial studies using such metabolic agents alone or in combination with other remedies such as immunotherapy are in progress and have demonstrated promising outcomes. In this review, we aim to argue the importance of metabolic alterations and bioenergetic pathways in different types of leukemia and their vital roles in disease development. Designing treatments based on targeting leukemic cells vulnerabilities, particularly in nonresponsive leukemia patients, should be warranted.
Collapse
Affiliation(s)
- Mojdeh Soltani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yue Zhao
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Alexandr V. Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
24
|
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: 4.8] [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
|
25
|
PON2 subverts metabolic gatekeeper functions in B cells to promote leukemogenesis. Proc Natl Acad Sci U S A 2021; 118:2016553118. [PMID: 33531346 DOI: 10.1073/pnas.2016553118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Unlike other cell types, developing B cells undergo multiple rounds of somatic recombination and hypermutation to evolve high-affinity antibodies. Reflecting the high frequency of DNA double-strand breaks, adaptive immune protection by B cells comes with an increased risk of malignant transformation. B lymphoid transcription factors (e.g., IKZF1 and PAX5) serve as metabolic gatekeepers by limiting glucose to levels insufficient to fuel transformation. We here identified aberrant expression of the lactonase PON2 in B cell acute lymphoblastic leukemia (B-ALL) as a mechanism to bypass metabolic gatekeeper functions. Compared to normal pre-B cells, PON2 expression was elevated in patient-derived B-ALL samples and correlated with poor clinical outcomes in pediatric and adult cohorts. Genetic deletion of Pon2 had no measurable impact on normal B cell development. However, in mouse models for BCR-ABL1 and NRASG12D-driven B-ALL, deletion of Pon2 compromised proliferation, colony formation, and leukemia initiation in transplant recipient mice. Compromised leukemogenesis resulted from defective glucose uptake and adenosine triphosphate (ATP) production in PON2-deficient murine and human B-ALL cells. Mechanistically, PON2 enabled glucose uptake by releasing the glucose-transporter GLUT1 from its inhibitor stomatin (STOM) and genetic deletion of STOM largely rescued PON2 deficiency. While not required for glucose transport, the PON2 lactonase moiety hydrolyzes the lactone-prodrug 3OC12 to form a cytotoxic intermediate. Mirroring PON2 expression levels in B-ALL, 3OC12 selectively killed patient-derived B-ALL cells but was well tolerated in transplant recipient mice. Hence, while B-ALL cells critically depend on aberrant PON2 expression to evade metabolic gatekeeper functions, PON2 lactonase activity can be leveraged as synthetic lethality to overcome drug resistance in refractory B-ALL.
Collapse
|
26
|
Metabolic determinants of B-cell selection. Biochem Soc Trans 2021; 49:1467-1478. [PMID: 34196360 DOI: 10.1042/bst20201316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 11/17/2022]
Abstract
B-cells are antibody-producing cells of the adaptive immune system. Approximately 75% of all newly generated B-cells in the bone marrow are autoreactive and express potentially harmful autoantibodies. To prevent autoimmune disease, the immune system has evolved a powerful mechanism to eliminate autoreactive B-cells, termed negative B-cell selection. While designed to remove autoreactive clones during early B-cell development, our laboratory recently discovered that transformed B-cells in leukemia and lymphoma are also subject to negative selection. Indeed, besides the risk of developing autoimmune disease, B-cells are inherently prone to malignant transformation: to produce high-affinity antibodies, B-cells undergo multiple rounds of somatic immunoglobulin gene recombination and hypermutation. Reflecting high frequencies of DNA-breaks, adaptive immune protection by B-cells comes with a dramatically increased risk of development of leukemia and lymphoma. Of note, B-cells exist under conditions of chronic restriction of energy metabolism. Here we discuss how these metabolic gatekeeper functions during B-cell development provide a common mechanism for the removal of autoreactive and premalignant B-cells to safeguard against both autoimmune diseases and B-cell malignancies.
Collapse
|
27
|
Sadras T, Chan LN, Xiao G, Müschen M. Metabolic Gatekeepers of Pathological B Cell Activation. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:323-349. [DOI: 10.1146/annurev-pathol-061020-050135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Unlike other cell types, B cells undergo multiple rounds of V(D)J recombination and hypermutation to evolve high-affinity antibodies. Reflecting high frequencies of DNA double-strand breaks, adaptive immune protection by B cells comes with an increased risk of malignant transformation. In addition, the vast majority of newly generated B cells express an autoreactive B cell receptor (BCR). Thus, B cells are under intense selective pressure to remove autoreactive and premalignant clones. Despite stringent negative selection, B cells frequently give rise to autoimmune disease and B cell malignancies. In this review, we discuss mechanisms that we term metabolic gatekeepers to eliminate pathogenic B cell clones on the basis of energy depletion. Chronic activation signals from autoreactive BCRs or transforming oncogenes increase energy demands in autoreactive and premalignant B cells. Thus, metabolic gatekeepers limit energy supply to levels that are insufficient to fuel either a transforming oncogene or hyperactive signaling from an autoreactive BCR.
Collapse
Affiliation(s)
- Teresa Sadras
- Center of Molecular and Cellular Oncology, Yale Cancer Center, and Department of Immunobiology, Yale University, New Haven, Connecticut 06520, USA
| | - Lai N. Chan
- Center of Molecular and Cellular Oncology, Yale Cancer Center, and Department of Immunobiology, Yale University, New Haven, Connecticut 06520, USA
| | - Gang Xiao
- Current affiliation: Department of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Markus Müschen
- Center of Molecular and Cellular Oncology, Yale Cancer Center, and Department of Immunobiology, Yale University, New Haven, Connecticut 06520, USA
| |
Collapse
|
28
|
Selenium nanoparticles reduce glucose metabolism and promote apoptosis of glioma cells through reactive oxygen species-dependent manner. Neuroreport 2021; 31:226-234. [PMID: 31876687 DOI: 10.1097/wnr.0000000000001386] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gliomas are the most common, malignant, and lethal tumors in adults. Furthermore, gliomas are highly resistant to current chemotherapeutic drugs. Thus, new effective anticancer drugs for glioma are urgently needed. Selenium nanoparticles have been reported to have potent anti-tumor activity, although the specific mechanism is not fully understood. This study aimed to test the anti-tumor effect of selenium nanoparticles and its mechanism. We used selenium nanoparticles to treat commercial glioma cell lines, and patient-derived glioma cells, and then used the MTT assay to determine selenium nanoparticles effect against these. Apoptotic cell death was determined by annexin V-Fluos staining kit. Glucose uptake, lactate, and adenosine triphosphate production, together with hexokinase 2 and pyruvate kinase activities were measured to determine the glucose metabolism level. Reactive oxygen species production was tested using 2',7'-dichlorodihydrofluorescein diacetate. Our results showed that selenium nanoparticles had a potent cytotoxic effect in glioma cells, regardless of whether they were drug-resistant or not, whereas it showed less toxic effect in normal healthy cells. Further tests showed that selenium nanoparticles treatment leads to apoptotic cell death enhancement and glucose metabolism reduction, and this process was in a reactive oxygen species pathway-dependent manner. These results may provide a novel direction for glioma therapy in the future.
Collapse
|
29
|
Dushnicky MJ, Nazarali S, Mir A, Portwine C, Samaan MC. Is There A Causal Relationship between Childhood Obesity and Acute Lymphoblastic Leukemia? A Review. Cancers (Basel) 2020; 12:E3082. [PMID: 33105727 PMCID: PMC7690432 DOI: 10.3390/cancers12113082] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/20/2022] Open
Abstract
Childhood obesity is a growing epidemic with numerous global health implications. Over the past few years, novel insights have emerged about the contribution of adult obesity to cancer risk, but the evidence base is far more limited in children. While pediatric patients with acute lymphoblastic leukemia (ALL) are at risk of obesity, it is unclear if there are potential causal mechanisms by which obesity leads to ALL development. This review explores the endocrine, metabolic and immune dysregulation triggered by obesity and its potential role in pediatric ALL's genesis. We describe possible mechanisms, including adipose tissue attraction and protection of lymphoblasts, and their impact on ALL chemotherapies' pharmacokinetics. We also explore the potential contribution of cytokines, growth factors, natural killer cells and adipose stem cells to ALL initiation and propagation. While there are no current definite causal links between obesity and ALL, critical questions persist as to whether the adipose tissue microenvironment and endocrine actions can play a causal role in childhood ALL, and there is a need for more research to address these questions.
Collapse
Affiliation(s)
- Molly J. Dushnicky
- Department of Pediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.J.D.); (S.N.); (A.M.); (C.P.)
- Division of Pediatric Endocrinology, McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
| | - Samina Nazarali
- Department of Pediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.J.D.); (S.N.); (A.M.); (C.P.)
- Division of Pediatric Endocrinology, McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
- Michael G. De Groote School of Medicine, McMaster University, Hamilton, ON L8S4L8, Canada
| | - Adhora Mir
- Department of Pediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.J.D.); (S.N.); (A.M.); (C.P.)
- Division of Pediatric Endocrinology, McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
- Michael G. De Groote School of Medicine, McMaster University, Hamilton, ON L8S4L8, Canada
| | - Carol Portwine
- Department of Pediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.J.D.); (S.N.); (A.M.); (C.P.)
- Division of Pediatric Hematology/Oncology, McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
| | - Muder Constantine Samaan
- Department of Pediatrics, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.J.D.); (S.N.); (A.M.); (C.P.)
- Division of Pediatric Endocrinology, McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
- Michael G. De Groote School of Medicine, McMaster University, Hamilton, ON L8S4L8, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
30
|
Orgel E, Sea JL, Mittelman SD. Mechanisms by Which Obesity Impacts Survival from Acute Lymphoblastic Leukemia. J Natl Cancer Inst Monogr 2020; 2019:152-156. [PMID: 31532535 DOI: 10.1093/jncimonographs/lgz020] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/22/2019] [Accepted: 07/01/2019] [Indexed: 01/29/2023] Open
Abstract
The prevalence of obesity has steadily risen over the past decades, even doubling in more than 70 countries. High levels of body fat (adiposity) and obesity are associated with endocrine and hormonal dysregulation, cardiovascular compromise, hepatic dysfunction, pancreatitis, changes in drug metabolism and clearance, inflammation, and metabolic stress. It is thus unsurprising that obesity can affect the development of and survival from a wide variety of malignancies. This review focuses on acute lymphoblastic leukemia, the most common malignancy in children, to explore the multiple mechanisms connecting acute lymphoblastic leukemia, obesity, and adipocytes, and the implications for leukemia therapy.
Collapse
Affiliation(s)
- Etan Orgel
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA Department of Pediatrics, Keck School of Medicine, University of Southern California
| | - Jessica L Sea
- Division of Pediatric Endocrinology, UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, CA
| | - Steven D Mittelman
- Division of Pediatric Endocrinology, UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, CA
| |
Collapse
|
31
|
Liu M, Zhou P, Li J, Jiang Y. Nicotinamide Inhibits Glycolysis of HL-60 Cells by Modulating Sirtuin 1 (SIRT1)/Peroxisome Proliferator-Activated Receptor γ Coactivator 1α (PGC-1α)/Hypoxia-Inducible Factor-2α (HIF2α) Signaling Pathway. Med Sci Monit 2020; 26:e920810. [PMID: 32469848 PMCID: PMC7282349 DOI: 10.12659/msm.920810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background Nicotinamide can affect differentiation and proliferation of leukemia cells. This research aimed to explore the regulatory effect of nicotinamide on glycolysis metabolism of leukemia cells and to clarify the associated mechanisms. Material/Methods HL-60 cells were treated with nicotinamide and divided into 0.1, 1, and 10 μmol/l groups. HL-60 cells without any administration were assigned as negative control (CT group). Glucolytic activity was evaluated by detecting lactic acid production, and glucose level was measured using glucose consumption assay. Apoptosis of HL-60 was examined using flow cytometry assay, when cells were cultured for 24 h. Expressions of sirtuin 1 (SIRT1), peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), and hypoxia-inducible factor-2α (HIF2α) were evaluated using a reverse transcription PCR assay and Western blotting assay, respectively. Results Nicotinamide remarkably decreased lactic acid production and glucose levels in leukemia cells compared with that of the CT group (p<0.05). Nicotinamide significantly induced the apoptosis of HL-60 cells compared to that of the negative control group (p<0.05). Nicotinamide significantly inhibited the SIRT1/PGC-1α/HIF2α signaling pathway mRNAs compared to that of the CT group (p<0.05). Nicotinamide remarkably reduced mitochondrial regulatory factors SIRT1/PGC-1α expression compared to that in the CT group (p<0.05). Nicotinamide obviously downregulated HIF2α compared with that of the CT group (p<0.05). Moreover, all of the above nicotinamide-induced effects, including glycolytic activity, apoptosis, and expression of SIRT1/PGC-1α/HIF2α, were changed in a dose-dependent manner. Conclusions Nicotinamide can inhibit glycolysis of HL-60 cells by inhibiting the mitochondrial regulatory factor SIRT1/PGC-1α and suppressing transcription factor HIF2α.
Collapse
Affiliation(s)
- Miao Liu
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Pan Zhou
- Hubei Medical Devices, Quality Supervision and Test Institute, Wuhan, Hubei, China (mainland)
| | - Jiaojiao Li
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Yi Jiang
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
32
|
mTOR Regulation of Metabolism in Hematologic Malignancies. Cells 2020; 9:cells9020404. [PMID: 32053876 PMCID: PMC7072383 DOI: 10.3390/cells9020404] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/02/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023] Open
Abstract
Neoplastic cells rewire their metabolism, acquiring a selective advantage over normal cells and a protection from therapeutic agents. The mammalian Target of Rapamycin (mTOR) is a serine/threonine kinase involved in a variety of cellular activities, including the control of metabolic processes. mTOR is hyperactivated in a large number of tumor types, and among them, in many hematologic malignancies. In this article, we summarized the evidence from the literature that describes a central role for mTOR in the acquisition of new metabolic phenotypes for different hematologic malignancies, in concert with other metabolic modulators (AMPK, HIF1α) and microenvironmental stimuli, and shows how these features can be targeted for therapeutic purposes.
Collapse
|
33
|
Abstract
In this Review, Rashkovan et al. discuss the role of cancer metabolic circuitries feeding anabolism and redox potential in leukemia development and recent progress in translating these important findings to the clinic. Leukemia cell proliferation requires up-regulation and rewiring of metabolic pathways to feed anabolic cell growth. Oncogenic drivers directly and indirectly regulate metabolic pathways, and aberrant metabolism is central not only for leukemia proliferation and survival, but also mediates oncogene addiction with significant implications for the development of targeted therapies. This review explores leukemia metabolic circuitries feeding anabolism, redox potential, and energy required for tumor propagation with an emphasis on emerging therapeutic opportunities.
Collapse
Affiliation(s)
- Marissa Rashkovan
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA.,Department of Pediatrics, Columbia University, New York, NY 10032, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
34
|
Liu T, Peng XC, Li B. The Metabolic Profiles in Hematological Malignancies. Indian J Hematol Blood Transfus 2019; 35:625-634. [PMID: 31741613 DOI: 10.1007/s12288-019-01107-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/25/2019] [Indexed: 11/24/2022] Open
Abstract
Leukemia is one of the most aggressive hematological malignancies. Leukemia stem cells account for the poor prognosis and relapse of the disease. Decades of investigations have been performed to figure out how to eradicate the leukemia stem cells. It has also been known that cancer cells especially solid cancer cells use energy differently than most of the cell types. The same thing happens to leukemia. Since there are metabolic differences between the hematopoietic stem cells and their immediate descendants, we aim at manipulating the energy sources with which that could have an effect on leukemia stem cells while sparing the normal blood cells. In this review we summarize the metabolic characteristics of distinct leukemias such as acute myeloid leukemia, chronic myeloid leukemia, T cell lymphoblastic leukemia, B-cell lymphoblastic leukemia, chronic lymphocytic leukemia and other leukemia associated hematological malignancies such as multiple myeloma and myelodysplastic syndrome. A better understanding of the metabolic profiles in distinct leukemias might provide novel perspectives and shed light on novel metabolic targeting strategies towards the clinical treatment of leukemias.
Collapse
Affiliation(s)
- Tao Liu
- Department of Pathology, People's Hospital of Longhua, Shenzhen, 518131 People's Republic of China
| | - Xing-Chun Peng
- Department of Pathology, People's Hospital of Longhua, Shenzhen, 518131 People's Republic of China
| | - Bin Li
- 2Department of Pathology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clinical Center, CAS, Huaihai Road 966, Shanghai City, 200031 Shanghai People's Republic of China
| |
Collapse
|
35
|
Castro I, Sampaio-Marques B, Ludovico P. Targeting Metabolic Reprogramming in Acute Myeloid Leukemia. Cells 2019; 8:cells8090967. [PMID: 31450562 PMCID: PMC6770240 DOI: 10.3390/cells8090967] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/13/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022] Open
Abstract
The cancer metabolic reprogramming allows the maintenance of tumor proliferation, expansion and survival by altering key bioenergetics, biosynthetic and redox functions to meet the higher demands of tumor cells. In addition, several metabolites are also needed to perform signaling functions that further promote tumor growth and progression. These metabolic alterations have been exploited in different cancers, including acute myeloid leukemia, as novel therapeutic strategies both in preclinical models and clinical trials. Here, we review the complexity of acute myeloid leukemia (AML) metabolism and discuss how therapies targeting different aspects of cellular metabolism have demonstrated efficacy and how they provide a therapeutic window that should be explored to target the metabolic requirements of AML cells.
Collapse
Affiliation(s)
- Isabel Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal.
| |
Collapse
|
36
|
Lee HJ, Li CF, Ruan D, He J, Montal ED, Lorenz S, Girnun GD, Chan CH. Non-proteolytic ubiquitination of Hexokinase 2 by HectH9 controls tumor metabolism and cancer stem cell expansion. Nat Commun 2019; 10:2625. [PMID: 31201299 PMCID: PMC6573064 DOI: 10.1038/s41467-019-10374-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 05/09/2019] [Indexed: 12/28/2022] Open
Abstract
Enormous efforts have been made to target metabolic dependencies of cancer cells for developing new therapies. However, the therapeutic efficacy of glycolysis inhibitors is limited due to their inability to elicit cell death. Hexokinase 2 (HK2), via its mitochondrial localization, functions as a central nexus integrating glycolysis activation and apoptosis resilience. Here we identify that K63-linked ubiquitination by HectH9 regulates the mitochondrial localization and function of HK2. Through stable isotope tracer approach and functional metabolic analyses, we show that HectH9 deficiency impedes tumor glucose metabolism and growth by HK2 inhibition. The HectH9/HK2 pathway regulates cancer stem cell (CSC) expansion and CSC-associated chemoresistance. Histological analyses show that HectH9 expression is upregulated and correlated with disease progression in prostate cancer. This work uncovers that HectH9 is a novel regulator of HK2 and cancer metabolism. Targeting HectH9 represents an effective strategy to achieve long-term tumor remission by concomitantly disrupting glycolysis and inducing apoptosis.
Collapse
Affiliation(s)
- Hong-Jen Lee
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Chien-Feng Li
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan
- Department of Pathology, Chi-Mei Foundational Medical Center, Tainan, 710, Taiwan
| | - Diane Ruan
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Jiabei He
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Emily D Montal
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
- Department of Pathology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sonja Lorenz
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Josef- Schneider-Strasse 2, D-97080, Würzburg, Germany
| | - Geoffrey D Girnun
- Department of Pathology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Chia-Hsin Chan
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA.
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
37
|
Xiong T, Li Z, Huang X, Lu K, Xie W, Zhou Z, Tu J. TO901317 inhibits the development of hepatocellular carcinoma by LXRα/Glut1 decreasing glycometabolism. Am J Physiol Gastrointest Liver Physiol 2019; 316:G598-G607. [PMID: 30817182 DOI: 10.1152/ajpgi.00061.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This study was conducted to observe the effect and possible mechanism of TO901317 in vivo and in vitro to provide a new basis for the targeted therapy of hepatocellular carcinoma (HCC). The expressions of liver X receptor (LXR)-α, glucose transporter (Glut)-1, proliferating cell nuclear antigen (PCNA), and matrix metalloproteinase (MMP)-9 were analyzed from HCC public database (NCBI PubMed database). The result showed that LXRα was downregulated, whereas Glut1, PCNA, and MMP9 were upregulated in human HCC compared with normal liver. Furthermore, LXRα mRNA was negatively correlated with Glut1 mRNA. At the same time, HCC cells were cultivated in vitro and axillary injected in nude mice to establish the xenograft model. The xenograft in the TO901317-treated group was slower and smaller than the control group. The protein expression of LXRα, Glut1, and MMP9 could be detected by Western blot and glucose level. As a result, TO901317 could inhibit the cell proliferation of HCC in a dose-dependent manner by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. With the increase of TO901317 concentration, the cellular glucose concentration and ATP level were gradually decreased. Western blot results showed TO901317 could upregulate LXRα expression but downregulate MMP9 and Glut1 expression. Transwell and wound-healing analysis confirmed that, by increasing the concentration of TO901317, the cell invasion and migration were both decreased. LXRα small-interfering RNA (siRNA) could relieve the suppression effect of TO901317 on the cell invasion and migration and the expression of LXRα, Glut1, and MMP9. The glucose concentration was also raised. TO901317 could repress the progress of HCC cells by reducing the glucose concentration, upregulating LXRα expression, but downregulating the expression of Glut1 and MMP9. NEW & NOTEWORTHY This subject confirmed that TO901317, a specific liver X receptor agonist, could inhibit the progression of liver cancer through upregulating liver X receptor-α, downregulating the expression of glucose transporter-1 and matrix metalloproteinase-9, and decreasing the glucose content in SMMC-7721 and HepG2 cells.
Collapse
Affiliation(s)
- Ting Xiong
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan, People's Republic of China.,School of Pharmacy, Changsha Medical University, Hunan, People's Republic of China
| | - Zihan Li
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan, People's Republic of China
| | - Xuelong Huang
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan, People's Republic of China
| | - Kaiqiang Lu
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan, People's Republic of China
| | - Weiquan Xie
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan, People's Republic of China
| | - Zhigang Zhou
- Department of Anesthesia, the First Affiliated Hospital, University of South China, Hengyang, Hunan, People's Republic of China
| | - Jian Tu
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan, People's Republic of China
| |
Collapse
|
38
|
Müschen M. Metabolic gatekeepers to safeguard against autoimmunity and oncogenic B cell transformation. Nat Rev Immunol 2019; 19:337-348. [DOI: 10.1038/s41577-019-0154-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
39
|
Targeting mTOR in Acute Lymphoblastic Leukemia. Cells 2019; 8:cells8020190. [PMID: 30795552 PMCID: PMC6406494 DOI: 10.3390/cells8020190] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/12/2019] [Accepted: 02/16/2019] [Indexed: 12/12/2022] Open
Abstract
Acute Lymphoblastic Leukemia (ALL) is an aggressive hematologic disorder and constitutes approximately 25% of cancer diagnoses among children and teenagers. Pediatric patients have a favourable prognosis, with 5-years overall survival rates near 90%, while adult ALL still correlates with poorer survival. However, during the past few decades, the therapeutic outcome of adult ALL was significantly ameliorated, mainly due to intensive pediatric-based protocols of chemotherapy. Mammalian (or mechanistic) target of rapamycin (mTOR) is a conserved serine/threonine kinase belonging to the phosphatidylinositol 3-kinase (PI3K)-related kinase family (PIKK) and resides in two distinct signalling complexes named mTORC1, involved in mRNA translation and protein synthesis and mTORC2 that controls cell survival and migration. Moreover, both complexes are remarkably involved in metabolism regulation. Growing evidence reports that mTOR dysregulation is related to metastatic potential, cell proliferation and angiogenesis and given that PI3K/Akt/mTOR network activation is often associated with poor prognosis and chemoresistance in ALL, there is a constant need to discover novel inhibitors for ALL treatment. Here, the current knowledge of mTOR signalling and the development of anti-mTOR compounds are documented, reporting the most relevant results from both preclinical and clinical studies in ALL that have contributed significantly into their efficacy or failure.
Collapse
|
40
|
Freemerman AJ, Zhao L, Pingili AK, Teng B, Cozzo AJ, Fuller AM, Johnson AR, Milner JJ, Lim MF, Galanko JA, Beck MA, Bear JE, Rotty JD, Bezavada L, Smallwood HS, Puchowicz MA, Liu J, Locasale JW, Lee DP, Bennett BJ, Abel ED, Rathmell JC, Makowski L. Myeloid Slc2a1-Deficient Murine Model Revealed Macrophage Activation and Metabolic Phenotype Are Fueled by GLUT1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:1265-1286. [PMID: 30659108 PMCID: PMC6360258 DOI: 10.4049/jimmunol.1800002] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 12/03/2018] [Indexed: 12/15/2022]
Abstract
Macrophages (MΦs) are heterogeneous and metabolically flexible, with metabolism strongly affecting immune activation. A classic response to proinflammatory activation is increased flux through glycolysis with a downregulation of oxidative metabolism, whereas alternative activation is primarily oxidative, which begs the question of whether targeting glucose metabolism is a viable approach to control MΦ activation. We created a murine model of myeloid-specific glucose transporter GLUT1 (Slc2a1) deletion. Bone marrow-derived MΦs (BMDM) from Slc2a1M-/- mice failed to uptake glucose and demonstrated reduced glycolysis and pentose phosphate pathway activity. Activated BMDMs displayed elevated metabolism of oleate and glutamine, yet maximal respiratory capacity was blunted in MΦ lacking GLUT1, demonstrating an incomplete metabolic reprogramming. Slc2a1M-/- BMDMs displayed a mixed inflammatory phenotype with reductions of the classically activated pro- and anti-inflammatory markers, yet less oxidative stress. Slc2a1M-/- BMDMs had reduced proinflammatory metabolites, whereas metabolites indicative of alternative activation-such as ornithine and polyamines-were greatly elevated in the absence of GLUT1. Adipose tissue MΦs of lean Slc2a1M-/- mice had increased alternative M2-like activation marker mannose receptor CD206, yet lack of GLUT1 was not a critical mediator in the development of obesity-associated metabolic dysregulation. However, Ldlr-/- mice lacking myeloid GLUT1 developed unstable atherosclerotic lesions. Defective phagocytic capacity in Slc2a1M-/- BMDMs may have contributed to unstable atheroma formation. Together, our findings suggest that although lack of GLUT1 blunted glycolysis and the pentose phosphate pathway, MΦ were metabolically flexible enough that inflammatory cytokine release was not dramatically regulated, yet phagocytic defects hindered MΦ function in chronic diseases.
Collapse
Affiliation(s)
- Alex J Freemerman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Liyang Zhao
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Ajeeth K Pingili
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Bin Teng
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Alyssa J Cozzo
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Ashley M Fuller
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Amy R Johnson
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - J Justin Milner
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Maili F Lim
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - Joseph A Galanko
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Melinda A Beck
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jeremy D Rotty
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Lavanya Bezavada
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27710
| | | | - Brian J Bennett
- U.S. Department of Agriculture Western Human Nutrition Research Center, Davis, CA 95616
| | - E Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City, IA 52242
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242; and
| | - Jeff C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN 37232
| | - Liza Makowski
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 25799;
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| |
Collapse
|
41
|
Han TJ, Xu HZ, Li JS, Geng LY, Li XY, Zhou XX, Wang X. Leptin and its receptor in glucose metabolism of T-cell lymphoma. Oncol Lett 2018; 16:5838-5846. [PMID: 30333864 PMCID: PMC6176461 DOI: 10.3892/ol.2018.9356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 02/23/2018] [Indexed: 01/08/2023] Open
Abstract
T-cell lymphoma (TCL) is a group of heterogeneous disorders with a poor response to conventional treatment. In order to identify novel therapeutic targets, the present study investigated the effect of leptin and its receptor on glucose metabolism in TCL. The expression of the leptin receptor (ObR), and glucose transporter (Glut)1 and 4 was detected in TCL and reactive lymphoid hyperplasia (RLH) tissues by immunohistochemical analysis. A higher level of ObR expression was observed in the TCL tissues than in the RLH tissues (58.3 vs. 22.2%; P=0.012), and ObR overexpression was associated with high expression of Glut1 (P=0.007). In vitro analysis using the human TCL MOLT-3 cell line demonstrated that leptin stimulated cell glucose uptake via promoting recruitment and expression of Glut1, effects which were abolished by ObR-specific small interfering RNA (siRNA). Additionally, MOLT-3 cell viability was also increased following leptin treatment. ObR-specific siRNA abolished these responses. In conclusion, these results suggested that leptin serves a critical role in TCL glucose uptake via the ObR.
Collapse
Affiliation(s)
- Tian-Jie Han
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China.,Department of Hematology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Hong-Zhi Xu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jun-Shan Li
- Department of Gastroenterology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Ling-Yun Geng
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xin-Yu Li
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xiang-Xiang Zhou
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
42
|
Irigoyen M, García-Ruiz JC, Berra E. The hypoxia signalling pathway in haematological malignancies. Oncotarget 2018; 8:36832-36844. [PMID: 28415662 PMCID: PMC5482702 DOI: 10.18632/oncotarget.15981] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/27/2017] [Indexed: 12/25/2022] Open
Abstract
Haematological malignancies are tumours that affect the haematopoietic and the lymphatic systems. Despite the huge efforts to eradicate these tumours, the percentage of patients suffering resistance to therapies and relapse still remains significant. The tumour environment favours drug resistance of cancer cells, and particularly of cancer stem/initiating cells. Hypoxia promotes aggressiveness, metastatic spread and relapse in most of the solid tumours. Furthermore, hypoxia is associated with worse prognosis and resistance to conventional treatments through activation of the hypoxia-inducible factors. Haematological malignancies are not considered solid tumours, and therefore, the role of hypoxia in these diseases was initially presumed to be inconsequential. However, hypoxia is a hallmark of the haematopoietic niche. Here, we will review the current understanding of the role of both hypoxia and hypoxia-inducible factors in different haematological tumours.
Collapse
Affiliation(s)
- Marta Irigoyen
- Centro de Investigación Cooperativa en Biociencias CIC bioGUNE, Derio, Spain
| | - Juan Carlos García-Ruiz
- Servicio de Hematología y Hemoterapia, BioCruces Health Research Institute, Hospital Universitario Cruces, Spain
| | - Edurne Berra
- Centro de Investigación Cooperativa en Biociencias CIC bioGUNE, Derio, Spain
| |
Collapse
|
43
|
Starkova J, Hermanova I, Hlozkova K, Hararova A, Trka J. Altered Metabolism of Leukemic Cells: New Therapeutic Opportunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 336:93-147. [PMID: 29413894 DOI: 10.1016/bs.ircmb.2017.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The cancer metabolic program alters bioenergetic processes to meet the higher demands of tumor cells for biomass production, nucleotide synthesis, and NADPH-balancing redox homeostasis. It is widely accepted that cancer cells mostly utilize glycolysis, as opposed to normal cells, in which oxidative phosphorylation is the most employed bioenergetic process. Still, studies examining cancer metabolism had been overlooked for many decades, and it was only recently discovered that metabolic alterations affect both the oncogenic potential and therapeutic response. Since most of the published works concern solid tumors, in this comprehensive review, we aim to summarize knowledge about the metabolism of leukemia cells. Leukemia is a malignant disease that ranks first and fifth in cancer-related deaths in children and adults, respectively. Current treatment has reached its limits due to toxicity, and there has been a need for new therapeutic approaches. One of the possible scenarios is improved use of established drugs and another is to introduce new druggable targets. Herein, we aim to describe the complexity of leukemia metabolism and highlight cellular processes that could be targeted therapeutically and enhance the effectiveness of current treatments.
Collapse
Affiliation(s)
- Julia Starkova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Ivana Hermanova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Katerina Hlozkova
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alzbeta Hararova
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Trka
- CLIP-Childhood Leukaemia Investigation Prague, Charles University, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic; University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
44
|
Xu JY, Luo JM. [Association between BIM gene and glucocorticoid resistance in children with acute lymphoblastic leukemia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:945-949. [PMID: 28774373 PMCID: PMC7390050 DOI: 10.7499/j.issn.1008-8830.2017.08.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 06/16/2017] [Indexed: 06/07/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignant hematological disease in childhood. Glucocorticoids are frequently used in the chemoradiotherapy regimen for ALL and can induce the apoptosis of ALL cells through several signaling pathways, but about 10% of ALL children have poor response to glucocorticoids. Studies have revealed that glucocorticoids induce the apoptosis of ALL cells by upregulating the expression of BIM gene, and BIM gene is associated with glucocorticoid resistance in childhood ALL. This article reviews the recent studies on glucocorticoid resistance in childhood ALL, especially the role of BIM and its expression products in this process.
Collapse
Affiliation(s)
- Jin-Yun Xu
- Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | | |
Collapse
|
45
|
Barwe SP, Quagliano A, Gopalakrishnapillai A. Eviction from the sanctuary: Development of targeted therapy against cell adhesion molecules in acute lymphoblastic leukemia. Semin Oncol 2017; 44:101-112. [PMID: 28923207 DOI: 10.1053/j.seminoncol.2017.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/10/2017] [Accepted: 06/29/2017] [Indexed: 02/04/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is a malignant hematological disease afflicting hematopoiesis in the bone marrow. While 80%-90% of patients diagnosed with ALL will achieve complete remission at some point during treatment, ALL is associated with high relapse rate, with a 5-year overall survival rate of 68%. The initial remission failure and the high rate of relapse can be attributed to intrinsic chemoprotective mechanisms that allow persistence of ALL cells despite therapy. These mechanisms are mediated, at least in part, through the engagement of cell adhesion molecules (CAMs) within the bone marrow microenvironment. This review assembles CAMs implicated in protection of leukemic cells from chemotherapy. Such studies are limited in ALL. Therefore, CAMs that are associated with poor outcomes or are overexpressed in ALL and have been shown to be involved in chemoprotection in other hematological cancers are also included. It is likely that these molecules play parallel roles in ALL because the CAMs identified to be a factor in ALL chemoresistance also work similarly in other hematological malignancies. We review the signaling mechanisms activated by the engagement of CAMs that provide protection from chemotherapy. Development of targeted therapies against CAMs could improve outcome and raise the overall cure rate in ALL.
Collapse
Affiliation(s)
- Sonali P Barwe
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE.
| | - Anthony Quagliano
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE
| | | |
Collapse
|
46
|
Zatula A, Dikic A, Mulder C, Sharma A, Vågbø CB, Sousa MML, Waage A, Slupphaug G. Proteome alterations associated with transformation of multiple myeloma to secondary plasma cell leukemia. Oncotarget 2017; 8:19427-19442. [PMID: 28038447 PMCID: PMC5386695 DOI: 10.18632/oncotarget.14294] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/30/2016] [Indexed: 01/22/2023] Open
Abstract
Plasma cell leukemia is a rare and aggressive plasma cell neoplasm that may either originate de novo (primary PCL) or by leukemic transformation of multiple myeloma (MM) to secondary PCL (sPCL). The prognosis of sPCL is very poor, and currently no standard treatment is available due to lack of prospective clinical studies. In an attempt to elucidate factors contributing to transformation, we have performed super-SILAC quantitative proteome profiling of malignant plasma cells collected from the same patient at both the MM and sPCL stages of the disease. 795 proteins were found to be differentially expressed in the MM and sPCL samples. Gene ontology analysis indicated a metabolic shift towards aerobic glycolysis in sPCL as well as marked down-regulation of enzymes involved in glycan synthesis, potentially mediating altered glycosylation of surface receptors. There was no significant change in overall genomic 5-methylcytosine or 5-hydroxymethylcytosine at the two stages, indicating that epigenetic dysregulation was not a major driver of transformation to sPCL. The present study constitutes the first attempt to provide a comprehensive map of the altered protein expression profile accompanying transformation of MM to sPCL in a single patient, identifying several candidate proteins that can be targeted by currently available small molecule drugs. Our dataset furthermore constitutes a reference dataset for further proteomic analysis of sPCL transformation.
Collapse
Affiliation(s)
- Alexey Zatula
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Aida Dikic
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Celine Mulder
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Present address: University of Utrecht, Utrecht, Holland
| | - Animesh Sharma
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Cathrine B Vågbø
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Mirta M L Sousa
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Anders Waage
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Department of Hematology, Department of Medicine, St. Olav's Hospital, Trondheim, Norway
| | - Geir Slupphaug
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| |
Collapse
|
47
|
Mambetsariev N, Lin WW, Wallis AM, Stunz LL, Bishop GA. TRAF3 deficiency promotes metabolic reprogramming in B cells. Sci Rep 2016; 6:35349. [PMID: 27752131 PMCID: PMC5082756 DOI: 10.1038/srep35349] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/27/2016] [Indexed: 12/23/2022] Open
Abstract
The adaptor protein TNF receptor-associated factor 3 (TRAF3) is a critical regulator of B lymphocyte survival. B cell-specific TRAF3 deficiency results in enhanced viability and is associated with development of lymphoma and multiple myeloma. We show that TRAF3 deficiency led to induction of two proteins important for glucose metabolism, Glut1 and Hexokinase 2 (HXK2). This was associated with increased glucose uptake. In the absence of TRAF3, anaerobic glycolysis and oxidative phosphorylation were increased in B cells without changes in mitochondrial mass or reactive oxygen species. Chemical inhibition of glucose metabolism or glucose deprivation substantially attenuated the enhanced survival of TRAF3-deficient B cells, with a decrease in the pro-survival protein Mcl-1. Changes in Glut1 and Mcl-1 levels, glucose uptake and B cell number in the absence of TRAF3 were all dependent upon NF-κB inducing kinase (NIK). These results indicate that TRAF3 deficiency suffices to metabolically reprogram B cells, a finding that improves our understanding of the role of TRAF3 as a tumor suppressor, and suggests potential therapeutic strategies.
Collapse
Affiliation(s)
- Nurbek Mambetsariev
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Medical Scientist Training Program, The University of Iowa, Carver College of Medicine, 2206 MERF, Iowa City, IA 52242-2600, USA
- Immunology Graduate Program, 357 Medical Research Center, Iowa City, IA 52242-1182, USA
| | - Wai W. Lin
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Immunology Graduate Program, 357 Medical Research Center, Iowa City, IA 52242-1182, USA
| | - Alicia M. Wallis
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Immunology Graduate Program, 357 Medical Research Center, Iowa City, IA 52242-1182, USA
| | - Laura L. Stunz
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Gail A. Bishop
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Medical Scientist Training Program, The University of Iowa, Carver College of Medicine, 2206 MERF, Iowa City, IA 52242-2600, USA
- Immunology Graduate Program, 357 Medical Research Center, Iowa City, IA 52242-1182, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
- University of Iowa and DVA Medical Center, 601 Highway 6 West, Iowa City, IA 52246, USA
- Internal Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, USA
| |
Collapse
|
48
|
Abstract
B cell growth and proliferation is tightly regulated by signaling through the B cell receptor and by other membrane bound receptors responding to different cytokines. The PI3K signaling pathway has been shown to play a crucial role in B cell activation, differentiation and survival. Activated B cells undergo metabolic reprograming in response to changing energetic and biosynthetic demands. B cells also need to be able to coordinate metabolic activity and proliferation with nutrient availability. The PI3K signaling network has been implicated in regulating nutrient acquisition, utilization and biosynthesis, thus integrating receptor-mediated signaling with cell metabolism. In this review, we discuss the current knowledge about metabolic changes induced in activated B cells, strategies to adapt to metabolic stress and the role of PI3K signaling in these processes.
Collapse
Affiliation(s)
- Julia Jellusova
- a BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg , Freiburg , Germany.,b Max Planck Institute of Immunobiology and Epigenetics , Freiburg , Germany
| | - Robert C Rickert
- c Sanford Burnham Prebys Medical Discovery Institute , La Jolla , CA , USA
| |
Collapse
|
49
|
Kishton RJ, Barnes CE, Nichols AG, Cohen S, Gerriets VA, Siska PJ, Macintyre AN, Goraksha-Hicks P, de Cubas AA, Liu T, Warmoes MO, Abel ED, Yeoh AEJ, Gershon TR, Rathmell WK, Richards KL, Locasale JW, Rathmell JC. AMPK Is Essential to Balance Glycolysis and Mitochondrial Metabolism to Control T-ALL Cell Stress and Survival. Cell Metab 2016; 23:649-62. [PMID: 27076078 PMCID: PMC4832577 DOI: 10.1016/j.cmet.2016.03.008] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 12/23/2015] [Accepted: 03/24/2016] [Indexed: 01/20/2023]
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy associated with Notch pathway mutations. While both normal activated and leukemic T cells can utilize aerobic glycolysis to support proliferation, it is unclear to what extent these cell populations are metabolically similar and if differences reveal T-ALL vulnerabilities. Here we show that aerobic glycolysis is surprisingly less active in T-ALL cells than proliferating normal T cells and that T-ALL cells are metabolically distinct. Oncogenic Notch promoted glycolysis but also induced metabolic stress that activated 5' AMP-activated kinase (AMPK). Unlike stimulated T cells, AMPK actively restrained aerobic glycolysis in T-ALL cells through inhibition of mTORC1 while promoting oxidative metabolism and mitochondrial Complex I activity. Importantly, AMPK deficiency or inhibition of Complex I led to T-ALL cell death and reduced disease burden. Thus, AMPK simultaneously inhibits anabolic growth signaling and is essential to promote mitochondrial pathways that mitigate metabolic stress and apoptosis in T-ALL.
Collapse
Affiliation(s)
- Rigel J Kishton
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Carson E Barnes
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Amanda G Nichols
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Sivan Cohen
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Valerie A Gerriets
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Peter J Siska
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN 37232, USA
| | - Andrew N Macintyre
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | | | - Aguirre A de Cubas
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Tingyu Liu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Marc O Warmoes
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - E Dale Abel
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Allen Eng Juh Yeoh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 119077, Singapore; Department of Pediatrics, National University Health System, Singapore 119228, Singapore
| | - Timothy R Gershon
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - W Kimryn Rathmell
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Kristy L Richards
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA; Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Jeffrey C Rathmell
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
50
|
Selwan EM, Finicle BT, Kim SM, Edinger AL. Attacking the supply wagons to starve cancer cells to death. FEBS Lett 2016; 590:885-907. [PMID: 26938658 DOI: 10.1002/1873-3468.12121] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/10/2016] [Accepted: 02/29/2016] [Indexed: 12/14/2022]
Abstract
The constitutive anabolism of cancer cells not only supports proliferation but also addicts tumor cells to a steady influx of exogenous nutrients. Limiting access to metabolic substrates could be an effective and selective means to block cancer growth. In this review, we define the pathways by which cancer cells acquire the raw materials for anabolism, highlight the actionable proteins in each pathway, and discuss the status of therapeutic interventions that disrupt nutrient acquisition. Critical open questions to be answered before apical metabolic inhibitors can be successfully and safely deployed in the clinic are also outlined. In summary, recent studies provide strong support that substrate limitation is a powerful therapeutic strategy to effectively, and safely, starve cancer cells to death.
Collapse
Affiliation(s)
- Elizabeth M Selwan
- Department of Developmental and Cell Biology, University of California Irvine, CA, USA
| | - Brendan T Finicle
- Department of Developmental and Cell Biology, University of California Irvine, CA, USA
| | - Seong M Kim
- Department of Developmental and Cell Biology, University of California Irvine, CA, USA
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California Irvine, CA, USA
| |
Collapse
|