1
|
Pojero F, Gervasi F. Polyphenol Treatment of Peripheral Blood Mononuclear Cells from Individuals of Different Ages. Methods Mol Biol 2025; 2857:191-221. [PMID: 39348067 DOI: 10.1007/978-1-0716-4128-6_18] [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] [Indexed: 10/01/2024]
Abstract
Human peripheral blood mononuclear cells (PBMCs) have been largely utilized to assess the cytotoxic, immunomodulatory, and anti-inflammatory properties of both synthetic and natural compounds. Within the latter category, polyphenols from dietary sources have been extensively analyzed. PBMCs represent a feasible in vitro model to study polyphenol hallmarks and activity according to quantitative and qualitative differences in immune responses in individuals of different age. In this chapter, we propose a method for PBMC treatment with polyphenols and analysis designed on age-dependent qualitative and quantitative variability in immune cell performance.
Collapse
Affiliation(s)
- Fanny Pojero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Francesco Gervasi
- Specialistic Oncology Laboratory Unit, A.R.N.A.S. Hospitals Civico, Di Cristina e Benfratelli, Palermo, Italy
| |
Collapse
|
2
|
Abu El-Naga EM, Ali ME, Ali RH, Hozyen HF, Hussein HA. Cleavage and in vitro cultivation rates monitoring in culture media supplemented with energy sources, non-essential amino acids, and antioxidants in the buffalo embryos. BMC Vet Res 2024; 20:521. [PMID: 39558345 PMCID: PMC11571879 DOI: 10.1186/s12917-024-04118-4] [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: 01/01/2024] [Accepted: 06/06/2024] [Indexed: 11/20/2024] Open
Abstract
The study was designed to monitor the cleavage rate (CR) and in-vitro cultivation rate (IVC) after addition of energy sources, non-essential amino acids, and antioxidants to the Synthetic oviductal fluid (SOF) and FertiCult. After in-vitro maturation and in-vitro fertilization, presumptive zygotes were cultured in one of two culture media: FertiCult media and SOF medium, supplemented with pyruvate, glucose, and sodium lactate as energy sources, as well as 10, 20, 250, 500, and 750 mg non-essential amino acids, and antioxidants. All stages of cleavage rate (CR), and in-vitro cultivation rate (IVC) of embryonic development including morula stage (MOR) and blastocyst (BLAS) have been assessed. The findings revealed that there were no significant differences in the CR between the control and other treated groups with sources of energy when added to SOF media (P > 0.05), while there were significant differences (P < 0.05) in the IVC of embryonic development between groups (The percentages of MOR stage in the control, pyruvate, glucose and mixture of source of energy (MIX) were at 50%, 62.5%, 60%, and 63.6%, respectively). The highest percentage of the BLAS was recorded after SOF supplementation with glucose (40%). Similarly, there were no significant differences (P > 0.05) in the CR between control and FertiCult supplemented with sources of energy, while the IVC stages increased significantly (P < 0.05) in the FertiCult media supplemented with glucose, pyruvate, sodium lactate, and MIX. The percentages of the MOR stage in the control, pyruvate, glucose and mix media were at 50%, 55.6%, 55.6%, 54.5%, 57.1% respectively. The lowest percentage of the BLAS was recorded after FertiCult supplementation with pyruvate (11.1%). Replenishing the SOF maturation media with 20 mg of non-essential amino acids significantly (P < 0.05) enhanced the MOR stage (100%). There was also an improvement in the development of BLAS stage, where it reached 31.2% and 47.4% in the SOF maturation media supplemented with 10, and 750 mg non-essential amino acids, respectively. There were no significant differences (P > 0.05) in neither CR nor IVC between control and FertiCult supplemented with antioxidants. There were significant differences (P < 0.05) in the MOR stages (control, 42.9% & treated, 57.9%) and BLAS stages (control, 21.4% & treated, 42.1%) in antioxidant supplemented SOF maturation media compared to control. In conclusion, supplementation of SOF cultivation medium with energy sources, 20 mg of non-essential amino acids and antioxidant addition may improve the cleavage rate (CR) and in vitro cultivation rate (IVC) of buffalos' embryonic development.
Collapse
Affiliation(s)
- Eman M Abu El-Naga
- Department of Theriogenology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Montaser E Ali
- Animal Production Department, Faculty of Agriculture, Assiut Branch, Al-Azhar University, Assiut, Egypt
| | - Rawda H Ali
- Department of Animal Production, Faculty of Agriculture, Assiut University, Assiut, 71526, Egypt.
| | - Heba F Hozyen
- Animal Reproduction and Artificial Insemination Department, Veterinary Research Institute, National Research Center, Giza, Egypt
- Physiology and Biochemistry Department, Faculty of Veterinary Medicine, Ain Shams University, Cairo Governorate, Egypt
| | - Hassan A Hussein
- Department of Theriogenology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
- Department of Theriogenology, Faculty of Veterinary Medicine, Sphinx University, New Assiut, 71684, Egypt
| |
Collapse
|
3
|
Nujoom N, Koyakutty M, Biswas L, Rajkumar T, Nair SV. Emerging Gene-editing nano-therapeutics for Cancer. Heliyon 2024; 10:e39323. [PMID: 39524822 PMCID: PMC11550658 DOI: 10.1016/j.heliyon.2024.e39323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Remarkable progress has been made in the field of genome engineering after the discovery of CRISPR/Cas9 in 2012 by Jennifer Doudna and Emmanuelle Charpentier. Compared to any other gene-editing tools, CRISPR/Cas9 attracted the attention of the scientific community because of its simplicity, specificity, and multiplex editing possibilities for which the inventors were awarded the Nobel prize for chemistry in 2020. CRISPR/Cas9 allows targeted alteration of the genomic sequence, gene regulation, and epigenetic modifications using an RNA-guided site-specific endonuclease. Though the impact of CRISPR/Cas9 was undisputed, some of its limitations led to key modifications including the use of miniature-Cas proteins, Cas9 Retron precise Parallel Editing via homologY (CRISPEY), Cas-Clover, or development of alternative methods including retron-recombineering, Obligate Mobile Element Guided Activity(OMEGA), Fanzor, and Argonaute proteins. As cancer is caused by genetic and epigenetic alterations, gene-editing was found to be highly useful for knocking out oncogenes, editing mutations to regain the normal functioning of tumor suppressor genes, knock-out immune checkpoint blockade in CAR-T cells, producing 'off-the-shelf' CAR-T cells, identify novel tumorigenic genes and functional analysis of multiple pathways in cancer, etc. Advancements in nanoparticle-based delivery of guide-RNA and Cas9 complex to the human body further enhanced the potential of CRISPR/Cas9 for clinical translation. Several studies are reported for developing novel delivery methods to enhance the tumor-specific application of CRISPR/Cas9 for anticancer therapy. In this review, we discuss new developments in novel gene editing techniques and recent progress in nanoparticle-based CRISPR/Cas9 delivery specific to cancer applications.
Collapse
Affiliation(s)
- Najma Nujoom
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| | - Manzoor Koyakutty
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| | - Lalitha Biswas
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| | - Thangarajan Rajkumar
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| | - Shantikumar V. Nair
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| |
Collapse
|
4
|
Fox AC, Blazeck J. Applying metabolic control strategies to engineered T cell cancer therapies. Metab Eng 2024; 86:250-261. [PMID: 39490640 PMCID: PMC11611646 DOI: 10.1016/j.ymben.2024.10.009] [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: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are an engineered immunotherapy that express synthetic receptors to recognize and kill cancer cells. Despite their success in treating hematologic cancers, CAR T cells have limited efficacy against solid tumors, in part due to the altered immunometabolic profile within the tumor environment, which hinders T cell proliferation, infiltration, and anti-tumor activity. For instance, CAR T cells must compete for essential nutrients within tumors, while resisting the impacts of immunosuppressive metabolic byproducts. In this review, we will describe the altered metabolic features within solid tumors that contribute to immunosuppression of CAR T cells. We'll discuss how overexpression of key metabolic enzymes can enhance the ability of CAR T cells to resist corresponding tumoral metabolic changes or even revert the metabolic profile of a tumor to a less inhibitory state. In addition, metabolic remodeling is intrinsically linked to T cell activity, differentiation, and function, such that metabolic engineering strategies can also promote establishment of more or less efficacious CAR T cell phenotypes. Overall, we will show how applying metabolic engineering strategies holds significant promise in improving CAR T cells for the treatment of solid tumors.
Collapse
Affiliation(s)
- Andrea C Fox
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA
| | - John Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta GA 303332, USA.
| |
Collapse
|
5
|
Bouzaid H, Espírito Santo L, Ferreira DM, Machado S, Costa ASG, Dias MI, Calhelha RC, Barros L, Chater O, Rodi YK, Errachidi F, Chahdi FO, Oliveira MBPP, Alves RC. Detailed Phytochemical Composition, Cyto-/Hepatotoxicity, and Antioxidant/Anti-Inflammatory Profile of Moroccan Spices: A Study on Coriander, Caraway, and Mystical Cumin. Molecules 2024; 29:3485. [PMID: 39124890 PMCID: PMC11313914 DOI: 10.3390/molecules29153485] [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: 05/16/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Coriander, caraway, and mystical cumin are famous for their aromatic properties and widely used in Moroccan cuisine. The nutritional/phytochemical composition of their seeds (used for food flavoring and preservation) were compared. Their antioxidant, anti-inflammatory, cytotoxic and hepatotoxic effects were also explored. The fat content was similar among the samples (13%), with monounsaturated fatty acids being predominant. The coriander and mystical cumin seeds were extremely rich in C18:1n9c (81 and 85%, respectively) while, in the caraway, C18:1n12 (25%) was found together with C18:1n9c (32%). The caraway seeds also presented a higher proportion of C18:2n6c (34%) than the other seeds (13 and 8%, correspondingly). γ-Tocotrienol was the major vitamin E form in all the samples. The caraway seeds contained double the amount of protein (~18%) compared to the other seeds (~8%) but, qualitatively, the amino acid profiles among all seeds were similar. The seeds were also rich in dietary fiber (40-53%); however, differences were found in their fiber profiles. Caraway showed the highest antioxidant profile and anti-inflammatory activity and an LC-DAD-ESI/MSn analysis revealed great differences in the phenolic profiles of the samples. Cytotoxicity (NCI-H460, AGS, MCF-7, and CaCo2) and hepatotoxicity (RAW 264.7) were not observed. In sum, besides their flavoring/preservation properties, these seeds are also relevant source of bioactive compounds with health-promoting activities.
Collapse
Affiliation(s)
- Hiba Bouzaid
- Laboratory of Applied Organic Chemistry, Faculty of Sciences and Technology, University of Sidi Mohamed Ben Abdellah, B.P. 2202—Route d’Imouzzer, Fez 30000, Morocco
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, n. º 228, 4050-313 Porto, Portugal
| | - Liliana Espírito Santo
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, n. º 228, 4050-313 Porto, Portugal
| | - Diana M. Ferreira
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, n. º 228, 4050-313 Porto, Portugal
| | - Susana Machado
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, n. º 228, 4050-313 Porto, Portugal
| | - Anabela S. G. Costa
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, n. º 228, 4050-313 Porto, Portugal
| | - Maria Inês Dias
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Alameda Santa Apolónia, 5300-253 Bragança, Portugal
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Ricardo C. Calhelha
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Alameda Santa Apolónia, 5300-253 Bragança, Portugal
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Lillian Barros
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Alameda Santa Apolónia, 5300-253 Bragança, Portugal
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Oumaima Chater
- Laboratory of Functional Ecology and Environmental Engineering, Faculty of Sciences and Technology, University of Sidi Mohamed Ben Abdellah, Fez 30000, Morocco
| | - Youssef Kandri Rodi
- Laboratory of Applied Organic Chemistry, Faculty of Sciences and Technology, University of Sidi Mohamed Ben Abdellah, B.P. 2202—Route d’Imouzzer, Fez 30000, Morocco
| | - Faouzi Errachidi
- Laboratory of Functional Ecology and Environmental Engineering, Faculty of Sciences and Technology, University of Sidi Mohamed Ben Abdellah, Fez 30000, Morocco
| | - Fouad Ouazzani Chahdi
- Laboratory of Applied Organic Chemistry, Faculty of Sciences and Technology, University of Sidi Mohamed Ben Abdellah, B.P. 2202—Route d’Imouzzer, Fez 30000, Morocco
| | - Maria Beatriz P. P. Oliveira
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, n. º 228, 4050-313 Porto, Portugal
| | - Rita C. Alves
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, n. º 228, 4050-313 Porto, Portugal
| |
Collapse
|
6
|
Kośliński P, Rzepiński Ł, Koba M, Maciejek Z, Kowalewski M, Daghir-Wojtkowiak E. Comparative Analysis of Serum Amino Acid Profiles in Patients with Myasthenia Gravis and Multiple Sclerosis. J Clin Med 2024; 13:4083. [PMID: 39064122 PMCID: PMC11277976 DOI: 10.3390/jcm13144083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Background: Multiple sclerosis (MS) and myasthenia gravis (MG) are autoimmune diseases that attack the central nervous system (CNS) and the neuromuscular junction, respectively. As the common pathogenesis of both diseases is associated with an autoimmune background and the involvement of T and B lymphocytes, the overlapping of selected clinical symptoms may cause difficulties in the differential diagnosis of both diseases. Methods: The aim of the study was to use Liquid Chromatography-Electrospray Ionization-Mass Spectrometry (LC-ESI-MS/MS) in conjunction with multivariate statistical analyses to examine the changes in amino acid metabolic profiles between patients with MG, MS, and a control group. Results: Comparative analysis of amino acids (AA) between patients with MG, MS, and within the control group allowed for the identification of statistically significant differences in the amino acid profile. Comparing the patients (patients with MS and MG) with the control group, and after taking the results of multiple tests into account, it was observed that amino acids such as ARG, PRO, TRP, CIT were significantly different between the groups. When considering the comparison between the AA concentrations in MS and MG patients, we found three AAs that were significantly different in the MS and MG groups, after correcting for multiple testing (CIT, GABA, and AAA). Higher concentrations of amino acids that showed significant differences were observed in patients with myasthenia gravis. Conclusions: Our results have indicated AAs that may prove valuable for improving the diagnostics of MS and MG patients. To better assess the potential utility of these markers, their performance requires further validation in a larger study group and limitation of possible confounding factors, e.g., medications and diet.
Collapse
Affiliation(s)
- Piotr Kośliński
- Department of Toxicology and Bromatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Dr. A. Jurasza 2, 85-089 Bydgoszcz, Poland;
| | - Łukasz Rzepiński
- Department of Neurology, 10th Military Research Hospital and Polyclinic, Powstańców Warszawy 5, 85-681 Bydgoszcz, Poland; (Ł.R.); (Z.M.)
- Sanitas—Neurology Outpatient Clinic, 85-010 Bydgoszcz, Poland
| | - Marcin Koba
- Department of Toxicology and Bromatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Dr. A. Jurasza 2, 85-089 Bydgoszcz, Poland;
| | - Zdzisław Maciejek
- Department of Neurology, 10th Military Research Hospital and Polyclinic, Powstańców Warszawy 5, 85-681 Bydgoszcz, Poland; (Ł.R.); (Z.M.)
- Sanitas—Neurology Outpatient Clinic, 85-010 Bydgoszcz, Poland
| | | | | |
Collapse
|
7
|
Li N, Wang T, Zhang H, Li X, Bai H, Lu N, Lu K. Exploring the causal relationship between glutamine metabolism and leukemia risk: a Mendelian randomization and LC-MS/MS analysis. Front Immunol 2024; 15:1418738. [PMID: 39050845 PMCID: PMC11265999 DOI: 10.3389/fimmu.2024.1418738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Objective This investigation sought to delineate the causal nexus between plasma glutamine concentrations and leukemia susceptibility utilizing bidirectional Mendelian Randomization (MR) analysis and to elucidate the metabolic ramifications of asparaginase therapy on glutamine dynamics in leukemia patients. Methods A bidirectional two-sample MR framework was implemented, leveraging genetic variants as instrumental variables from extensive genome-wide association studies (GWAS) tailored to populations of European descent. Glutamine quantification was executed through a rigorously validated Liquid Chromatography-Mass Spectrometry/Mass Spectrometry (LC-MS/MS) protocol. Comparative analyses of glutamine levels were conducted across leukemia patients versus healthy controls, pre- and post-asparaginase administration. Statistical evaluations employed inverse variance weighted (IVW) models, MR-Egger regression, and sensitivity tests addressing pleiotropy and heterogeneity. Results The MR findings underscored a significant inverse association between glutamine levels and leukemia risk (IVW p = 0.03558833), positing lower glutamine levels as a contributory factor to heightened leukemia susceptibility. Conversely, the analysis disclosed no substantive causal impact of leukemia on glutamine modulation (IVW p = 0.9694758). Notably, post-asparaginase treatment, a marked decrement in plasma glutamine concentrations was observed in patients (p = 0.0068), underlining the profound metabolic influence of the therapeutic regimen. Conclusion This study corroborates the hypothesized inverse relationship between plasma glutamine levels and leukemia risk, enhancing our understanding of glutamine's role in leukemia pathophysiology. The pronounced reduction in glutamine levels following asparaginase intervention highlights the critical need for meticulous metabolic monitoring to refine therapeutic efficacy and optimize patient management in clinical oncology. These insights pave the way for more tailored and efficacious treatment modalities in the realm of personalized medicine.
Collapse
Affiliation(s)
- Na Li
- Mass Spectrometry Research Institute, Beijing Gobroad Hospital, Beijing, China
- Mass Spectrometry Research Institute, Beijing Gobroad Healthcare Group, Beijing, China
| | - Tianyi Wang
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huiying Zhang
- Mass Spectrometry Research Institute, Beijing Gobroad Hospital, Beijing, China
- Mass Spectrometry Research Institute, Beijing Gobroad Healthcare Group, Beijing, China
- Department of Laboratory Medicine, Beijing Gobroad Hospital, Beijing, China
| | - Xiao Li
- Department of Laboratory Medicine, Beijing Gobroad Hospital, Beijing, China
| | - Haochen Bai
- Mass Spectrometry Research Institute, Shanghai Liquan Hospital, Shanghai, China
| | - Ning Lu
- Department of Laboratory Medicine, Beijing Gobroad Hospital, Beijing, China
| | - Kaizhi Lu
- Mass Spectrometry Research Institute, Beijing Gobroad Hospital, Beijing, China
- Mass Spectrometry Research Institute, Beijing Gobroad Healthcare Group, Beijing, China
| |
Collapse
|
8
|
Tellier J, Nutt SL. B cell trajectories influence cancer outcomes. Science 2024; 384:510-511. [PMID: 38696586 DOI: 10.1126/science.adp1865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2024]
Abstract
Two types of B cell in the tumor microenvironment modulate antitumor immunity.
Collapse
Affiliation(s)
- Julie Tellier
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Han C, Ge M, Xing P, Xia T, Zhang C, Ma K, Ma Y, Li S, Li W, Liu X, Zhang B, Zhang L, Zhang L. Cystine deprivation triggers CD36-mediated ferroptosis and dysfunction of tumor infiltrating CD8 + T cells. Cell Death Dis 2024; 15:145. [PMID: 38360744 PMCID: PMC10869360 DOI: 10.1038/s41419-024-06503-1] [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: 11/01/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/17/2024]
Abstract
Cancer cells develop multiple strategies to evade T cell-mediated killing. On one hand, cancer cells may preferentially rely on certain amino acids for rapid growth and metastasis. On the other hand, sufficient nutrient availability and uptake are necessary for mounting an effective T cell anti-tumor response in the tumor microenvironment (TME). Here we demonstrate that tumor cells outcompete T cells for cystine uptake due to high Slc7a11 expression. This competition induces T-cell exhaustion and ferroptosis, characterized by diminished memory formation and cytokine secretion, increased PD-1 and TIM-3 expression, as well as intracellular oxidative stress and lipid-peroxide accumulation. Importantly, either Slc7a11 deletion in tumor cells or intratumoral cystine supplementation improves T cell anti-tumor immunity. Mechanistically, cystine deprivation in T cells disrupts glutathione synthesis, but promotes CD36 mediated lipid uptake due to dysregulated cystine/glutamate exchange. Moreover, enforced expression of glutamate-cysteine ligase catalytic subunit (Gclc) promotes glutathione synthesis and prevents CD36 upregulation, thus boosting T cell anti-tumor immunity. Our findings reveal cystine as an intracellular metabolic checkpoint that orchestrates T-cell survival and differentiation, and highlight Gclc as a potential therapeutic target for enhancing T cell anti-tumor function.
Collapse
Affiliation(s)
- Chenfeng Han
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
| | - Minmin Ge
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
| | - Pengfei Xing
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tian Xia
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Institute of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shaanxi, China
| | - Kaili Ma
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
| | - Yifu Ma
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shicheng Li
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenhui Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
| | - Xiaowei Liu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shaanxi, China.
| | - Liyuan Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China.
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Lianjun Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China.
- Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
10
|
Cunningham A, Oudejans LL, Geugien M, Pereira-Martins DA, Wierenga ATJ, Erdem A, Sternadt D, Huls G, Schuringa JJ. The nonessential amino acid cysteine is required to prevent ferroptosis in acute myeloid leukemia. Blood Adv 2024; 8:56-69. [PMID: 37906522 PMCID: PMC10784682 DOI: 10.1182/bloodadvances.2023010786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cysteine is a nonessential amino acid required for protein synthesis, the generation of the antioxidant glutathione, and for synthesizing the nonproteinogenic amino acid taurine. Here, we highlight the broad sensitivity of leukemic stem and progenitor cells to cysteine depletion. By CRISPR/CRISPR-associated protein 9-mediated knockout of cystathionine-γ-lyase, the cystathionine-to-cysteine converting enzyme, and by metabolite supplementation studies upstream of cysteine, we functionally prove that cysteine is not synthesized from methionine in acute myeloid leukemia (AML) cells. Therefore, although perhaps nutritionally nonessential, cysteine must be imported for survival of these specific cell types. Depletion of cyst(e)ine increased reactive oxygen species (ROS) levels, and cell death was induced predominantly as a consequence of glutathione deprivation. nicotinamide adenine dinucleotide phosphate hydrogen oxidase inhibition strongly rescued viability after cysteine depletion, highlighting this as an important source of ROS in AML. ROS-induced cell death was mediated via ferroptosis, and inhibition of glutathione peroxidase 4 (GPX4), which functions in reducing lipid peroxides, was also highly toxic. We therefore propose that GPX4 is likely key in mediating the antioxidant activity of glutathione. In line, inhibition of the ROS scavenger thioredoxin reductase with auranofin also impaired cell viability, whereby we find that oxidative phosphorylation-driven AML subtypes, in particular, are highly dependent on thioredoxin-mediated protection against ferroptosis. Although inhibition of the cystine-glutamine antiporter by sulfasalazine was ineffective as a monotherapy, its combination with L-buthionine-sulfoximine (BSO) further improved AML ferroptosis induction. We propose the combination of either sulfasalazine or antioxidant machinery inhibitors along with ROS inducers such as BSO or chemotherapy for further preclinical testing.
Collapse
Affiliation(s)
- Alan Cunningham
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lieve L. Oudejans
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjan Geugien
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Diego Antonio Pereira-Martins
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Albertus T. J. Wierenga
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ayşegül Erdem
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Dominique Sternadt
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerwin Huls
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
11
|
Zhou X, Wang Z, Yuan K. The effect of diet and nutrition on T cell function in cancer. Int J Cancer 2023; 153:1954-1966. [PMID: 37504380 DOI: 10.1002/ijc.34668] [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: 04/27/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023]
Abstract
Cancer can be considered one of the most threatening diseases to human health, and immunotherapy, especially T-cell immunotherapy, is the most promising treatment for cancers. Diet therapy is widely concerned in cancer because of its safety and fewer side effects. Many studies have shown that both the function of T cells and the progression of cancer can be affected by nutrients in the diet. In fact, it is challenging for T cells to infiltrate and eliminate cancer cells in tumor microenvironment, because of the harsh metabolic condition. The intake of different nutrients has a great influence on the proliferation, activation, differentiation and exhaustion of T cells. In this review, we summarize the effects of typical amino acids, lipids, carbohydrates and other nutritional factors on T cell functions and provide future perspectives for dietary treatment of cancer based on modifications of T cell functions.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Wang
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kefei Yuan
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Tresia GE, Anggraeny YN, Winarsih WH, Setiasih S, Rohaeni ES, Mariyono M, Pamungkas D. Nonessential amino acids in tropical ruminant feed: Investigating grass and legume forages of Indonesia. J Adv Vet Anim Res 2023; 10:820-829. [PMID: 38370889 PMCID: PMC10868679 DOI: 10.5455/javar.2023.j739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/10/2023] [Accepted: 08/20/2023] [Indexed: 02/20/2024] Open
Abstract
Objective This study aimed to examine the concentration of nonessential amino acids (NEAAs) in ruminant feed in tropical areas, with a focus on forage grasses and legumes in Indonesia. Materials and Methods A total of 11 grasses (Chrysopogon zizanioides, Brachiaria brizantha, Brachiaria humidicola, Paspalum dilatatum, Paspalum atatum, Chloris gayana, Pennisetum polystachion, Panicum maximum, Cenchrus biflorus, Andropogon canaliculatus, and Digotaria decumbens) and six legumes (Arachis hypogaea, Pueararia Javanica, Centrosema pubescens, Clitoria ternatea, and Arachis pintoi) were analyzed for NEAA content using high-performance liquid chromatography (HPLC). Results Based on the results of this research, it was found that almost all NEAA content in forage was less than 3% (Serine (Ser), Alanine (Ala), Glycine (Gly), Tyrosine (Tyr), Proline (Pro), Cysteine (Cys), and Asparagine (Asn), except for glutamic (Glu) acid and arginine (Arg) in some legumes. The Glu content in grass ranges from 0.76% to 2.61%, and the Arg content ranges from 0.92% to 2.09%. These two NEAAs were most abundant in grasses and legumes, with concentrations of 5.10% to 6.27% and 3.10% to 5.53%, respectively. Conclusion Our study concluded that Glu and Arg were the most abundant NEAAs in tropical forages in Indonesia, with legumes having a higher concentration of NEAAs compared to grasses. Among the legumes, A. hypogaea had the highest NEAA content (23.40%), while among the forages, C. zizanioides had the highest NEAA content (12.37%). However, it was observed that neither legumes nor grasses could fulfill the metabolizable TNEAAs requirements for gaining cattle (250 kg of empty body weight gain), unlike commercial concentrates, which were found to meet the requirements, especially for Arg, Glu, and thyronine. The provision of concentrate is necessary to supplement forage to meet the NEAA needs for cattle.
Collapse
Affiliation(s)
- Gresy Eva Tresia
- Research Center for Animal Husbandry, Research Organization for Agriculture and Food, National Research and Innovation Agency Cibinong Science Center, Cibinong Science Center, Cibinong, Indonesia
| | - Yenny Nur Anggraeny
- Research Center for Animal Husbandry, Research Organization for Agriculture and Food, National Research and Innovation Agency Cibinong Science Center, Cibinong Science Center, Cibinong, Indonesia
| | - Wiwik Heny Winarsih
- East Java Regional Research and Innovation Agency, Surabaya, East Java, Indonesia
| | - Setiasih Setiasih
- Research Center for Animal Husbandry, Research Organization for Agriculture and Food, National Research and Innovation Agency Cibinong Science Center, Cibinong Science Center, Cibinong, Indonesia
| | - Eni Siti Rohaeni
- Research Center for Animal Husbandry, Research Organization for Agriculture and Food, National Research and Innovation Agency Cibinong Science Center, Cibinong Science Center, Cibinong, Indonesia
| | - Mariyono Mariyono
- Research Center for Animal Husbandry, Research Organization for Agriculture and Food, National Research and Innovation Agency Cibinong Science Center, Cibinong Science Center, Cibinong, Indonesia
| | - Dicky Pamungkas
- Research Center for Animal Husbandry, Research Organization for Agriculture and Food, National Research and Innovation Agency Cibinong Science Center, Cibinong Science Center, Cibinong, Indonesia
| |
Collapse
|
13
|
Foskolou IP, Cunha PP, Sánchez-López E, Minogue EA, Nicolet BP, Guislain A, Jorgensen C, Kostidis S, Zandhuis ND, Barbieri L, Bargiela D, Nathanael D, Tyrakis PA, Palazon A, Giera M, Wolkers MC, Johnson RS. The two enantiomers of 2-hydroxyglutarate differentially regulate cytotoxic T cell function. Cell Rep 2023; 42:113013. [PMID: 37632752 DOI: 10.1016/j.celrep.2023.113013] [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: 09/19/2022] [Revised: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 08/28/2023] Open
Abstract
2-Hydroxyglutarate (2HG) is a byproduct of the tricarboxylic acid (TCA) cycle and is readily detected in the tissues of healthy individuals. 2HG is found in two enantiomeric forms: S-2HG and R-2HG. Here, we investigate the differential roles of these two enantiomers in cluster of differentiation (CD)8+ T cell biology, where we find they have highly divergent effects on proliferation, differentiation, and T cell function. We show here an analysis of structural determinants that likely underlie these differential effects on specific α-ketoglutarate (αKG)-dependent enzymes. Treatment of CD8+ T cells with exogenous S-2HG, but not R-2HG, increased CD8+ T cell fitness in vivo and enhanced anti-tumor activity. These data show that S-2HG and R-2HG should be considered as two distinct and important actors in the regulation of T cell function.
Collapse
Affiliation(s)
- Iosifina P Foskolou
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK; Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden; Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory Amsterdam University Medical Center, University of Amsterdam, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands.
| | - Pedro P Cunha
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK; Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Elena Sánchez-López
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Albinusdreef 2, 2333ZA Leiden, the Netherlands
| | - Eleanor A Minogue
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK
| | - Benoît P Nicolet
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory Amsterdam University Medical Center, University of Amsterdam, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Aurélie Guislain
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory Amsterdam University Medical Center, University of Amsterdam, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Christian Jorgensen
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000 Aarhus C, Denmark
| | - Sarantos Kostidis
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Albinusdreef 2, 2333ZA Leiden, the Netherlands
| | - Nordin D Zandhuis
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory Amsterdam University Medical Center, University of Amsterdam, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Laura Barbieri
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK
| | - David Bargiela
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK
| | - Demitris Nathanael
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK
| | - Petros A Tyrakis
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK
| | - Asis Palazon
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Albinusdreef 2, 2333ZA Leiden, the Netherlands
| | - Monika C Wolkers
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory Amsterdam University Medical Center, University of Amsterdam, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Randall S Johnson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK; Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden.
| |
Collapse
|
14
|
Huang Y, Qin G, Cui T, Zhao C, Ren J, Qu X. A bimetallic nanoplatform for STING activation and CRISPR/Cas mediated depletion of the methionine transporter in cancer cells restores anti-tumor immune responses. Nat Commun 2023; 14:4647. [PMID: 37532731 PMCID: PMC10397352 DOI: 10.1038/s41467-023-40345-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Lack of sufficient cytotoxic T lymphocytes (CD8+ T cells) infiltration and dysfunctional state of CD8+ T cells are considered enormous obstacles to antitumor immunity. Herein, we construct a synergistic nanoplatform to promote CD8+ T cell infiltration in tumors while restoring T cell function by regulating methionine metabolism and activating the STING innate immune pathway. The CRISPR/Cas9 system down-regulates the methionine transporter SLC43A2 and restricts the methionine uptake by tumor cells, thereby relieving the methionine competition pressure of T cells; simultaneously, the released nutrition metal ions activate the cGAS/STING pathway. In this work, the described nanoplatform can enhance the effect of immunotherapy in preclinical cancer models in female mice, enhancing STING pathway mediated immunity and facilitating the development of amino acid metabolic intervention-based cancer therapy.
Collapse
Affiliation(s)
- Ying Huang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Geng Qin
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
| | - TingTing Cui
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
| |
Collapse
|
15
|
Xiao C, Xiong W, Xu Y, Zou J, Zeng Y, Liu J, Peng Y, Hu C, Wu F. Immunometabolism: a new dimension in immunotherapy resistance. Front Med 2023; 17:585-616. [PMID: 37725232 DOI: 10.1007/s11684-023-1012-z] [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: 12/26/2022] [Accepted: 05/19/2023] [Indexed: 09/21/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated unparalleled clinical responses and revolutionized the paradigm of tumor treatment, while substantial patients remain unresponsive or develop resistance to ICIs as a single agent, which is traceable to cellular metabolic dysfunction. Although dysregulated metabolism has long been adjudged as a hallmark of tumor, it is now increasingly accepted that metabolic reprogramming is not exclusive to tumor cells but is also characteristic of immunocytes. Correspondingly, people used to pay more attention to the effect of tumor cell metabolism on immunocytes, but in practice immunocytes interact intimately with their own metabolic function in a way that has never been realized before during their activation and differentiation, which opens up a whole new frontier called immunometabolism. The metabolic intervention for tumor-infiltrating immunocytes could offer fresh opportunities to break the resistance and ameliorate existing ICI immunotherapy, whose crux might be to ascertain synergistic combinations of metabolic intervention with ICIs to reap synergic benefits and facilitate an adjusted anti-tumor immune response. Herein, we elaborate potential mechanisms underlying immunotherapy resistance from a novel dimension of metabolic reprogramming in diverse tumor-infiltrating immunocytes, and related metabolic intervention in the hope of offering a reference for targeting metabolic vulnerabilities to circumvent immunotherapeutic resistance.
Collapse
Affiliation(s)
- Chaoyue Xiao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Yiting Xu
- Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Ji'an Zou
- Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Yue Zeng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Junqi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yurong Peng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chunhong Hu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Changsha, 410011, China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Changsha, 410011, China.
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
16
|
Congrave-Wilson Z, Kim M, Sutherland A, Jumarang J, Lee Y, Del Valle J, Cheng WA, da Silva Antunes R, Pannaraj PS. Effect of wash media type during PBMC isolation on downstream characterization of SARS-CoV-2-specific T cells. J Immunol Methods 2023; 519:113520. [PMID: 37390890 PMCID: PMC10306416 DOI: 10.1016/j.jim.2023.113520] [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: 03/19/2023] [Revised: 06/13/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Protocols for the isolation of peripheral blood mononuclear cells (PBMCs) from whole blood vary greatly between laboratories, especially in published studies of SARS-CoV-2-specific T cell responses following infection and vaccination. Research on the effects of different wash media types or centrifugation speeds and brake usage during the PBMC isolation process on downstream T cell activation and functionality is limited. Blood samples from 26 COVID-19-vaccinated participants were processed with different PBMC isolation methods using either PBS or RPMI as the wash media with high centrifugation speed and brakes or RPMI as the wash media with low speed and brakes (RPMI+ method). SARS-CoV-2 spike-specific T cells were quantified and characterized via a flow cytometry-based activation induced markers (AIM) assay and an interferon-γ (IFNγ) FluoroSpot assay and responses were compared between processing methods. Samples washed with RPMI showed higher AIM+ CD4 T cell responses than those washed with PBS and showed a shift away from naïve and towards an effector memory phenotype. The activation marker OX40 showed higher SARS-CoV-2 spike-induced upregulation on RPMI-washed CD4 T cells, while differences in CD137 upregulation were minimal between processing methods. The magnitude of the AIM+ CD8 T cell response was similar between processing methods but showed higher stimulation indices. Background frequencies of CD69+ CD8 T cells were increased in PBS-washed samples and were associated with higher baseline numbers of IFNγ-producing cells in the FluoroSpot assay. Slower braking in the RPMI+ method did not improve detection of SARS-CoV-2-specific T cells and caused longer processing times. Thus, the use of RPMI media with full centrifugation brakes during the wash steps of PBMC isolation was found to be most effective and efficient. Further studies are needed to elucidate the pathways involved in RPMI-mediated preservation of downstream T cell activity.
Collapse
Affiliation(s)
- Zion Congrave-Wilson
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Minjun Kim
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States; Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Aaron Sutherland
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Jaycee Jumarang
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States; Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Yesun Lee
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States; Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Jennifer Del Valle
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Wesley A Cheng
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States; Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Pia S Pannaraj
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States; Department of Pediatrics and Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States; Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
17
|
Ping Y, Shan J, Liu Y, Liu F, Wang L, Liu Z, Li J, Yue D, Wang L, Chen X, Zhang Y. Taurine enhances the antitumor efficacy of PD-1 antibody by boosting CD8 + T cell function. Cancer Immunol Immunother 2023; 72:1015-1027. [PMID: 36261540 DOI: 10.1007/s00262-022-03308-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/10/2022] [Indexed: 03/20/2023]
Abstract
The functional state of CD8+ T cells determines the therapeutic efficacy of PD-1 blockade antibodies in tumors. Amino acids are key nutrients for maintaining T cell antitumor immunity. In this study, we used samples from lung cancer patients treated with PD-1 blockade antibodies to assay the amino acids in their serum by mass spectrometry. We found that lung cancer patients with high serum taurine levels generally responded to PD-1 blockade antibody therapy, in parallel with the secretion of high levels of cytotoxic cytokines (IFN-γ and TNF-α). CD8+ T cells cultured with exogenous taurine exhibited decreased apoptosis, enhanced proliferation, and increased secretion of cytotoxic cytokines. High SLC6A6 expression in CD8+ T cells was positively associated with an effector T cell signature. SLC6A6 knockdown limited the function and proliferation of CD8+ T cells. RNA sequencing revealed that SLC6A6 knockdown altered the calcium signaling pathway, oxidative phosphorylation, and T cell receptor signaling in CD8+ T cells. Furthermore, taurine enhanced T cell proliferation and function in vitro by stimulation of PLCγ1-mediated calcium and MAPK signaling. Taurine plus immune checkpoint blockade antibody significantly attenuated tumor growth and markedly improved the function and proliferation of CD8+ T cells in a mouse tumor model. Thus, our findings indicate that taurine is an important driver for improving CD8+ T cell immune responses and could serve as a potential therapeutic agent for cancer patients.
Collapse
Affiliation(s)
- Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiqi Shan
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaqing Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fengsen Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liuya Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhangnan Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jieyao Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongli Yue
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan, China.
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
18
|
van Gelderen TA, Ladisa C, Salazar-Moscoso M, Folgado C, Habibi HR, Ribas L. Metabolomic and transcriptomic profiles after immune stimulation in the zebrafish testes. Genomics 2023; 115:110581. [PMID: 36796654 DOI: 10.1016/j.ygeno.2023.110581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023]
Abstract
Fish farms are prone to disease outbreaks and stress due to high-density rearing conditions in tanks and sea cages, adversely affecting growth, reproduction, and metabolism. To understand the molecular mechanisms affected in the gonads of breeder fish after an immune challenge, we investigated the metabolome and the transcriptome profiles in zebrafish testes after inducing an immune response. After 48 h of the immune challenge, ultra-high-performance liquid chromatography (LC-MS) and transcriptomic analysis by RNA-seq (Illumina) resulted in 20 different released metabolites and 80 differentially expressed genes. Among these, glutamine and succinic acid were the most abundant metabolites released and 27,5% of the genes belong to either the immune or reproduction systems. Pathway analysis based on metabolomic and transcriptomic crosstalk identified cad and iars genes that act simultaneously with succinate metabolite. This study deciphers interactions between reproduction and immune systems and provides a basis to improve protocols in generating more resistant broodstock.
Collapse
Affiliation(s)
- T A van Gelderen
- Institut de Ciències del Mar - Consejo Superior de Investigaciones Científicas (ICM-CSIC), Department of Renewable Marine Resources, 08003 Barcelona, Spain
| | - C Ladisa
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - M Salazar-Moscoso
- Institut de Ciències del Mar - Consejo Superior de Investigaciones Científicas (ICM-CSIC), Department of Renewable Marine Resources, 08003 Barcelona, Spain
| | - C Folgado
- Institut de Ciències del Mar - Consejo Superior de Investigaciones Científicas (ICM-CSIC), Department of Renewable Marine Resources, 08003 Barcelona, Spain
| | - H R Habibi
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - L Ribas
- Institut de Ciències del Mar - Consejo Superior de Investigaciones Científicas (ICM-CSIC), Department of Renewable Marine Resources, 08003 Barcelona, Spain.
| |
Collapse
|
19
|
Abstract
Significance: Cancer immunotherapy has yielded striking antitumor effects in many cancers, yet the proportion of benefited patients is still limited. As key mediators of tumor suppression, CD8+ T cells are crucial for cancer immunotherapy. It has been widely appreciated that the modulation of CD8+ T cell immunity could be an effective way to further improve the therapeutic benefit of immunotherapy. Recent Advances: Emerging evidence has underlined a close link between metabolism and immune functions, providing a metabolism-immune axis that is increasingly investigated for understanding CD8+ T cell regulation. On the other hand, growing findings have reported that tumors adopt multiple approaches to induce metabolic reprogramming of CD8+ T cells, leading to compromised immunotherapy. Critical Issues: CD8+ T cell metabolism in the tumor microenvironment (TME) is often adapted to diminish antitumor immune responses and thereby evade from immune surveillance. A better understanding of metabolic regulation of CD8+ T cells in the TME is believed to hold promise for opening a new therapeutic window to further improve the benefit of immunotherapy. We herein review the mechanistic understanding of how CD8+ T cell metabolism is reprogrammed in the TME, mainly focusing on the impact of nutrient availability and bioactive molecules secreted by surrounding cells. Future Directions: Future research should pay attention to tumor heterogeneity in the metabolic microenvironment and associated immune responses. It is also important to include the trending opinion of "precision medicine" in cancer immunotherapies to tailor metabolic interventions for individual patients in combination with immunotherapy treatments. Antioxid. Redox Signal. 37, 1234-1253.
Collapse
Affiliation(s)
- Ying Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaomin Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Min Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
20
|
Wißfeld J, Werner A, Yan X, ten Bosch N, Cui G. Metabolic regulation of immune responses to cancer. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0381. [PMID: 36269001 PMCID: PMC9724228 DOI: 10.20892/j.issn.2095-3941.2022.0381] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The tumor microenvironment is an ecosystem composed of multiple types of cells, such as tumor cells, immune cells, and cancer-associated fibroblasts. Cancer cells grow faster than non-cancerous cells and consume larger amounts of nutrients. The rapid growth characteristic of cancer cells fundamentally alters nutrient availability in the tumor microenvironment and results in reprogramming of immune cell metabolic pathways. Accumulating evidence suggests that cellular metabolism of nutrients, such as lipids and amino acids, beyond being essential to meet the bioenergetic and biosynthetic demands of immune cells, also regulates a broad spectrum of cellular signal transduction, and influences immune cell survival, differentiation, and anti-tumor effector function. The cancer immunometabolism research field is rapidly evolving, and exciting new discoveries are reported in high-profile journals nearly weekly. Therefore, all new findings in this field cannot be summarized within this short review. Instead, this review is intended to provide a brief introduction to this rapidly developing research field, with a focus on the metabolism of two classes of important nutrients-lipids and amino acids-in immune cells. We highlight recent research on the roles of lipids and amino acids in regulating the metabolic fitness and immunological functions of T cells, macrophages, and natural killer cells in the tumor microenvironment. Furthermore, we discuss the possibility of "editing" metabolic pathways in immune cells to act synergistically with currently available immunotherapies in enhancing anti-tumor immune responses.
Collapse
Affiliation(s)
- Jannis Wißfeld
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Anke Werner
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Xin Yan
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg 69120, Germany
| | - Nora ten Bosch
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Guoliang Cui
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz 55131, Germany,T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg 69120, Germany,Correspondence to: Guoliang Cui, E-mail:
| |
Collapse
|
21
|
Forcados C, Joaquina S, Casey NP, Caulier B, Wälchli S. How CAR T Cells Breathe. Cells 2022; 11:cells11091454. [PMID: 35563759 PMCID: PMC9102061 DOI: 10.3390/cells11091454] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
The manufacture of efficacious CAR T cells represents a major challenge in cellular therapy. An important aspect of their quality concerns energy production and consumption, known as metabolism. T cells tend to adopt diverse metabolic profiles depending on their differentiation state and their stimulation level. It is therefore expected that the introduction of a synthetic molecule such as CAR, activating endogenous signaling pathways, will affect metabolism. In addition, upon patient treatment, the tumor microenvironment might influence the CAR T cell metabolism by compromising the energy resources. The access to novel technology with higher throughput and reduced cost has led to an increased interest in studying metabolism. Indeed, methods to quantify glycolysis and mitochondrial respiration have been available for decades but were rarely applied in the context of CAR T cell therapy before the release of the Seahorse XF apparatus. The present review will focus on the use of this instrument in the context of studies describing the impact of CAR on T cell metabolism and the strategies to render of CAR T cells more metabolically fit.
Collapse
Affiliation(s)
- Christopher Forcados
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, 0379 Oslo, Norway; (C.F.); (S.J.); (N.P.C.); (B.C.)
| | - Sandy Joaquina
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, 0379 Oslo, Norway; (C.F.); (S.J.); (N.P.C.); (B.C.)
| | - Nicholas Paul Casey
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, 0379 Oslo, Norway; (C.F.); (S.J.); (N.P.C.); (B.C.)
| | - Benjamin Caulier
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, 0379 Oslo, Norway; (C.F.); (S.J.); (N.P.C.); (B.C.)
- Center for Cancer Cell Reprogramming (CanCell), Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Department of Cellular Therapy, Oslo University Hospital, 0379 Oslo, Norway; (C.F.); (S.J.); (N.P.C.); (B.C.)
- Correspondence:
| |
Collapse
|
22
|
Stirling ER, Bronson SM, Mackert JD, Cook KL, Triozzi PL, Soto-Pantoja DR. Metabolic Implications of Immune Checkpoint Proteins in Cancer. Cells 2022; 11:179. [PMID: 35011741 PMCID: PMC8750774 DOI: 10.3390/cells11010179] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 12/29/2022] Open
Abstract
Expression of immune checkpoint proteins restrict immunosurveillance in the tumor microenvironment; thus, FDA-approved checkpoint inhibitor drugs, specifically PD-1/PD-L1 and CTLA-4 inhibitors, promote a cytotoxic antitumor immune response. Aside from inflammatory signaling, immune checkpoint proteins invoke metabolic reprogramming that affects immune cell function, autonomous cancer cell bioenergetics, and patient response. Therefore, this review will focus on the metabolic alterations in immune and cancer cells regulated by currently approved immune checkpoint target proteins and the effect of costimulatory receptor signaling on immunometabolism. Additionally, we explore how diet and the microbiome impact immune checkpoint blockade therapy response. The metabolic reprogramming caused by targeting these proteins is essential in understanding immune-related adverse events and therapeutic resistance. This can provide valuable information for potential biomarkers or combination therapy strategies targeting metabolic pathways with immune checkpoint blockade to enhance patient response.
Collapse
Affiliation(s)
- Elizabeth R. Stirling
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (E.R.S.); (K.L.C.); (P.L.T.)
| | - Steven M. Bronson
- Department of Pathology, Section of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jessica D. Mackert
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
| | - Katherine L. Cook
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (E.R.S.); (K.L.C.); (P.L.T.)
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - Pierre L. Triozzi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (E.R.S.); (K.L.C.); (P.L.T.)
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
- Department of Hematology and Oncology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - David R. Soto-Pantoja
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (E.R.S.); (K.L.C.); (P.L.T.)
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
23
|
Ginefra P, Carrasco Hope H, Spagna M, Zecchillo A, Vannini N. Ionic Regulation of T-Cell Function and Anti-Tumour Immunity. Int J Mol Sci 2021; 22:ijms222413668. [PMID: 34948472 PMCID: PMC8705279 DOI: 10.3390/ijms222413668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/02/2022] Open
Abstract
The capacity of T cells to identify and kill cancer cells has become a central pillar of immune-based cancer therapies. However, T cells are characterized by a dysfunctional state in most tumours. A major obstacle for proper T-cell function is the metabolic constraints posed by the tumour microenvironment (TME). In the TME, T cells compete with cancer cells for macronutrients (sugar, proteins, and lipid) and micronutrients (vitamins and minerals/ions). While the role of macronutrients in T-cell activation and function is well characterized, the contribution of micronutrients and especially ions in anti-tumour T-cell activities is still under investigation. Notably, ions are important for most of the signalling pathways regulating T-cell anti-tumour function. In this review, we discuss the role of six biologically relevant ions in T-cell function and in anti-tumour immunity, elucidating potential strategies to adopt to improve immunotherapy via modulation of ion metabolism.
Collapse
|