1
|
Kathiresan DS, Balasubramani R, Marudhachalam K, Jaiswal P, Ramesh N, Sureshbabu SG, Puthamohan VM, Vijayan M. Role of Mitochondrial Dysfunctions in Neurodegenerative Disorders: Advances in Mitochondrial Biology. Mol Neurobiol 2025; 62:6827-6855. [PMID: 39269547 DOI: 10.1007/s12035-024-04469-x] [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: 04/04/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
Mitochondria, essential organelles responsible for cellular energy production, emerge as a key factor in the pathogenesis of neurodegenerative disorders. This review explores advancements in mitochondrial biology studies that highlight the pivotal connection between mitochondrial dysfunctions and neurological conditions such as Alzheimer's, Parkinson's, Huntington's, ischemic stroke, and vascular dementia. Mitochondrial DNA mutations, impaired dynamics, and disruptions in the ETC contribute to compromised energy production and heightened oxidative stress. These factors, in turn, lead to neuronal damage and cell death. Recent research has unveiled potential therapeutic strategies targeting mitochondrial dysfunction, including mitochondria targeted therapies and antioxidants. Furthermore, the identification of reliable biomarkers for assessing mitochondrial dysfunction opens new avenues for early diagnosis and monitoring of disease progression. By delving into these advancements, this review underscores the significance of understanding mitochondrial biology in unraveling the mechanisms underlying neurodegenerative disorders. It lays the groundwork for developing targeted treatments to combat these devastating neurological conditions.
Collapse
Affiliation(s)
- Divya Sri Kathiresan
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Rubadevi Balasubramani
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Kamalesh Marudhachalam
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Piyush Jaiswal
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Nivedha Ramesh
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Suruthi Gunna Sureshbabu
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Vinayaga Moorthi Puthamohan
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India.
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
2
|
Zhou M, Zang J, Qian Y, Zhang Q, Wang Y, Yao T, Yan H, Zhang K, Cai X, Jiang L, Zheng Y. Mitochondrial Transplantation via Magnetically Responsive Artificial Cells Promotes Intracerebral Hemorrhage Recovery by Supporting Microglia Immunological Homeostasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2500303. [PMID: 39961067 PMCID: PMC11962678 DOI: 10.1002/adma.202500303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/02/2025] [Indexed: 04/03/2025]
Abstract
The immune-inflammatory responses in the brain represent a key therapeutic target to ameliorate brain injury following intracerebral hemorrhage (ICH), where pro-inflammatory microglia and its mitochondrial dysfunction plays a pivotal role. Mitochondrial transplantation is a promising strategy to improve the cellular mitochondrial function and thus modulate their immune properties. However, the transplantation of naked mitochondria into the brain has been constrained by the peripheral clearance and the difficulty in achieving selective access to the brain. Here, a novel strategy for mitochondrial transplantation via intravenous injection of magnetically responsive artificial cells (ACs) are proposed. ACs can protect the loaded mitochondria and selectively accumulate around the lesion under an external magnetic field (EMF). In this study, mitochondria released from ACs can effectively improve microglial mitochondrial function, attenuate their pro-inflammatory attributes, and elevate the proportion of immunosuppressive microglia. In this way, microglia immune homeostasis in the brain is reestablished, and inflammation is attenuated, ultimately promoting functional recovery. This study presents an effective approach to transplant mitochondria into the brain, offering a promising alternative to modulate the immune-inflammatory cascade in the brain following ICH.
Collapse
Affiliation(s)
- Mi Zhou
- Shanghai Key Laboratory of Neuro‐Ultrasound for Diagnosis and TreatmentSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of Ultrasound in MedicineSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Jinhui Zang
- Shanghai Key Laboratory of Neuro‐Ultrasound for Diagnosis and TreatmentSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of Ultrasound in MedicineSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yuxuan Qian
- Shanghai Key Laboratory of Neuro‐Ultrasound for Diagnosis and TreatmentSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of Orthopedic SurgerySixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Qiang Zhang
- Institute of Diagnostic and Interventional RadiologySixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yifan Wang
- Department of EmergencySixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P.R. China
| | - Tingting Yao
- Institute of Diagnostic and Interventional RadiologySixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Hongyu Yan
- Shanghai Key Laboratory of Neuro‐Ultrasound for Diagnosis and TreatmentSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of Ultrasound in MedicineSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Kai Zhang
- Shanghai Key Laboratory of Neuro‐Ultrasound for Diagnosis and TreatmentSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of Ultrasound in MedicineSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Xiaojun Cai
- Shanghai Key Laboratory of Neuro‐Ultrasound for Diagnosis and TreatmentSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of Ultrasound in MedicineSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Lixian Jiang
- Shanghai Key Laboratory of Neuro‐Ultrasound for Diagnosis and TreatmentSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of Ultrasound in MedicineSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Yuanyi Zheng
- Shanghai Key Laboratory of Neuro‐Ultrasound for Diagnosis and TreatmentSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
- Department of Ultrasound in MedicineSixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| |
Collapse
|
3
|
Xie M, Li D, Zeng H, Huang Y, Xu R, Wang Z, Yu J, Sun Y. BAM8-22 targets spinal MrgC receptors to modulate UPR mt activity in the mechanism of bone cancer pain. Front Pharmacol 2025; 16:1575733. [PMID: 40230701 PMCID: PMC11994654 DOI: 10.3389/fphar.2025.1575733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Background Bone cancer pain (BCP) significantly impacts patients' overall quality of life. Cellular energy metabolism homeostasis is critically dependent on mitochondrial integrity, and emerging evidence suggests that mitochondrial dysfunction in chronic BCP exacerbates pain progression by disrupting nociceptive signaling pathways. Notably, G protein-coupled receptors (GPCRs), a major class of membrane receptors, modulate mitochondrial function through diverse molecular mechanisms. In this study, we investigated the role of Mas-related G protein-coupled receptor C (MrgC) in BCP pathogenesis and its regulatory effects on mitochondrial function. Methods Male C3H/HeN mice were utilized to establish a BCP model. Transmission electron microscopy and flow cytometry were employed to assess changes in mitochondrial ultrastructure, as well as levels of mtROS, ATP, and MMP in mice experiencing BCP. Following intrathecal injection of BAM8-22, we analyzed the effects of activated MrgC on mitochondrial unfolded protein response (UPRmt)-related molecules (ATF5, HSP60, LONP1, CLPP) and pain-related behaviors in BCP mice. The regulatory mechanism of MrgC on UPRmt was further explored in N2a and 293T cells. Results Mice with bone cancer pain showed improved mRNA and protein levels of UPRmt-related molecules, increased MMP and ATP, decreased mitochondrial ROS levels in the spinal cord after receiving an intrathecal injection of BAM8-22. Additionally, the paw withdrawal mechanical threshold in BCP mice increased, while the number of spontaneous foot lifts decreased. In complementary cellular studies, transfection-mediated overexpression of MrgC in N2a cells enhanced UPRmt biomarker expression, whereas RNA interference-mediated MrgC knockdown produced the opposite effect. Conclusion By activating spinal MrgC to mediate UPRmt activity and protect mitochondrial function, BAM8-22 contributes to the molecular development of BCP. This discovery suggests a new therapeutic target for BCP and offers a possible research avenue.
Collapse
Affiliation(s)
- Mingming Xie
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dan Li
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haohao Zeng
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rui Xu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhen Wang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jiacheng Yu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yu’e Sun
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
4
|
Liu Y, Shen C, Cao Y. Mediating Role of Blood Metabolites in the Relationship Between Immune Cell Traits and Heart Failure: A Mendelian Randomization and Mediation Analysis. J Am Heart Assoc 2025; 14:e037265. [PMID: 40079309 DOI: 10.1161/jaha.124.037265] [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] [Received: 06/23/2024] [Accepted: 01/30/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Observational studies have shown a significant association between immune cells and heart failure (HF). Nevertheless, the precise biological mechanisms underlying this association remain unclear. METHODS To investigate the causative relationships and underlying mechanisms between immune cell traits and adult HF, 3 main methods of Mendelian randomization were used: 2-sample Mendelian randomization, multivariable Mendelian randomization with controlling for several factors affecting HF, and mediation analysis. Results from the inverse variance-weighted model indicated that genetic predispositions for human leukocyte antigen-type DR (HLA DR) on CD33dim HLA DR+ CD11b+ (odds ratio, 0.967 [95% CI, 0.939-0.996]; P=0.028) may be associated with a reduced risk of HF. Although the association between HF and HLA DR on CD33 dim HLA DR+ CD11b+ did not withstand multiple-testing correction, the Mendelian randomization results (PIVW <0.05) decrease the likelihood that the observational results are due to chance. RESULTS Our 2-step mediation analysis demonstrated that genetic predispositions for HLA DR on CD33dim HLA DR+ CD11b+ (odds ratio,1.085 [95% CI, 1.020-1.155]; P=0.010) was associated with increased levels of the metabolite Octadecanedioate, while genetic predispositions for Octadecanedioate levels (odds ratio, 0.917 [95% CI, 0.849-0.991]; P=0.028) was associated with a reduced risk of HF. Moreover, our results also demonstrated that the association between HLA DR on CD33dim HLA DR+ CD11b+ and HF was possibly mediated by Octadecanedioate levels, with a mediation proportion of 21.4% [95% CI, 43.7 -0.998]. CONCLUSIONS These findings underscore the importance of HLA DR on CD33dim HLA DR+ CD11b+ in the development of HF, with Octadecanedioate levels acting as a possible mediator in this pathway.
Collapse
Affiliation(s)
- Yi Liu
- Department of Emergency Medicine, Laboratory of Emergency Medicine West China Hospital, West China School of Medicine, Sichuan University Chengdu China
| | - Chenfu Shen
- Department of Neurosurgery Xiangya Hospital, Central South University Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Yu Cao
- Department of Emergency Medicine, Laboratory of Emergency Medicine West China Hospital, West China School of Medicine, Sichuan University Chengdu China
| |
Collapse
|
5
|
Suzuki K, Watanabe N, Torii S, Arakawa S, Ochi K, Tsuchiya S, Yamada K, Kawamura Y, Ota S, Komatsu N, Shimizu S, Ando M, Takaku T. BCR::ABL1-induced mitochondrial morphological alterations as a potential clinical biomarker in chronic myeloid leukemia. Cancer Sci 2025; 116:673-689. [PMID: 39652455 PMCID: PMC11875769 DOI: 10.1111/cas.16424] [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: 08/03/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 03/05/2025] Open
Abstract
The BCR::ABL1 oncogene plays a crucial role in the development of chronic myeloid leukemia (CML). Previous studies have investigated the involvement of mitochondrial dynamics in various cancers, revealing potential therapeutic strategies. However, the impact of BCR::ABL1 on mitochondrial dynamics remains unclear. In this study, we demonstrated that BCR::ABL1 is sufficient to induce excessive mitochondrial fragmentation by activating dynamin-related protein (DRP)1 through the mitogen-activated protein kinase (MAPK) pathway. Leukocytes obtained from patients with CML and the BCR::ABL1-positive cell lines exhibited increased mitochondrial fragmentation compared to leukocytes obtained from healthy donors and BCR::ABL1-negative cells. Furthermore, the analysis of BCR::ABL1-transduced cells showed increased phosphorylation of DRP1 at serine 616 and extracellular signal-regulated kinase (ERK) 1/2. Moreover, the inhibition of DRP1 and upstream mitogen-activated extracellular signal-regulated kinase (MEK) 1/2 suppressed mitochondrial fragmentation. Strikingly, DRP1 inhibition effectively reduced the viability of BCR::ABL1-positive cells and induced necrotic cell death. Additionally, a label-free artificial intelligence-driven flow cytometry successfully identified not only the BCR::ABL1-transduced cells but also peripheral leukocytes from CML patients by assessing mitochondrial morphological alterations. These findings suggested the crucial role of BCR::ABL1-induced mitochondrial fragmentation in driving BCR::ABL1-positive cell proliferation, and the potential use of mitochondrial morphological alterations as a clinical biomarker for the label-free detection of CML cells.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Fusion Proteins, bcr-abl/metabolism
- Fusion Proteins, bcr-abl/genetics
- Mitochondria/metabolism
- Mitochondria/pathology
- Mitochondria/genetics
- Dynamins/metabolism
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Phosphorylation
- MAP Kinase Signaling System
- Mitochondrial Dynamics/genetics
Collapse
Affiliation(s)
- Kohjin Suzuki
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
- System Technologies Laboratory, Sysmex CorporationKobeJapan
| | - Naoki Watanabe
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Satoru Torii
- Department of Pathological Cell BiologyAdvanced Research Initiative, Institute of Science TokyoTokyoJapan
| | - Satoko Arakawa
- Department of Pathological Cell BiologyAdvanced Research Initiative, Institute of Science TokyoTokyoJapan
| | - Kiyosumi Ochi
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
- Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Shun Tsuchiya
- Department of HematologyJuntendo University Nerima HospitalTokyoJapan
| | | | | | - Sadao Ota
- ThinkCyteK.K.TokyoJapan
- Research Center for Advanced Science and TechnologyThe University of TokyoTokyoJapan
| | - Norio Komatsu
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Shigeomi Shimizu
- Department of Pathological Cell BiologyAdvanced Research Initiative, Institute of Science TokyoTokyoJapan
| | - Miki Ando
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Tomoiku Takaku
- Department of HematologyJuntendo University Graduate School of MedicineTokyoJapan
- Department of HematologySaitama Medical UniversitySaitamaJapan
| |
Collapse
|
6
|
Chen C, Yan Y, Wu J, Gan WB. GCTransNet: 3D mitochondrial instance segmentation based on Global Context Vision Transformers. J Struct Biol 2025; 217:108170. [PMID: 39842559 DOI: 10.1016/j.jsb.2025.108170] [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: 11/07/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
Mitochondria are double membrane-bound organelles essential for generating energy in eukaryotic cells. Mitochondria can be readily visualized in 3D using Volume Electron Microscopy (vEM), and accurate image segmentation is vital for quantitative analysis of mitochondrial morphology and function. To address the challenge of segmenting small mitochondrial compartments in vEM images, we propose an automated mitochondrial segmentation method called GCTransNet. This method employs grayscale migration technology to preprocess images, effectively reducing intensity distribution differences across EM images. By utilizing 3D Global Context Vision Transformers (GC-ViT) combined with global context self-attention modules and local self-attention modules, GCTransNet precisely models long-range and short-range spatial interactions. The long-range interactions enable the model to capture the global structural relationships within the mitochondrial segmentation network, while the short-range interactions refine local details and boundaries. In our approach, the encoder of the 3D U-Net network, a classical multi-scale learning architecture that retains high-resolution features through skip connections and combines multi-scale features for precise segmentation, is replaced by a 3D GC-ViT. The GC-ViT leverages shifted window-based self-attention, capturing long-range dependencies and offering improved segmentation accuracy compared to traditional U-Net encoders. In the MitoEM mitochondrial segmentation challenge, GCTransNet achieved state-of-the-art results, demonstrating its superiority in automated mitochondrial segmentation. The code and its documentation are publicly available at https://github.com/GanLab123/GCTransNet.
Collapse
Affiliation(s)
- Chaoyi Chen
- Collage of Biological Sciences, China Agricultural University, Beijing 100091, China; Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yidan Yan
- Collage of Biological Sciences, China Agricultural University, Beijing 100091, China
| | | | - Wen-Biao Gan
- Shenzhen Bay Laboratory, Shenzhen 518132, China; Lingang Laboratory, Shanghai 200032, China.
| |
Collapse
|
7
|
Li Y, Huang X, Qiao Q, Li Y, Han X, Chen C, Chen Y, Guo S, Zhang Y, Gao W, Liu H, Sun T. Suppression of Sepsis Cytokine Storm by Escherichia Coli Cell Wall-Derived Carbon Dots. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414237. [PMID: 39775885 DOI: 10.1002/adma.202414237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Sepsis is a life-threatening disease caused by a dysregulated immune response to infection, often involving the translocation of Gram-negative bacteria such as Escherichia coli (E. coli) into the bloodstream, triggering a cytokine storm. Despite its severity, no effective drugs currently exist for sepsis treatment. This study explores whether pathogen-derived carbon dots can mitigate their inherent toxicity while leveraging their structural similarity to pathogens to competitively bind pattern recognition receptors, thereby inhibiting sepsis. Based on this concept, E. coli wall-derived carbon dots (E-CDs) are synthesized and shown to reduce inflammatory cytokine production, protect organ function, and improve survival in septic mice. Mechanistic studies reveal that E-CDs competitively bind to lipopolysaccharide-binding protein with lipopolysaccharide, promoting toll-like receptor 4 degradation via the lysosomal pathway and inhibiting nuclear factor kappa-B (NF-κB) activation. Additionally, E-CDs exhibit antioxidant properties, reducing oxidative stress and mitochondrial DNA release, thereby suppressing overactivation of the stimulator of interferon genes pathway. In septic cynomolgus monkeys and patient-derived peripheral blood mononuclear cells, E-CDs alleviate inflammation and oxidative stress. Overall, this study demonstrates that E-CDs can suppress the cytokine storm in sepsis by co-silencing innate immune pathways, suggesting that converting pathogens into carbon dots offers a novel therapeutic strategy.
Collapse
Affiliation(s)
- Yinan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Xiu Huang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Qingqing Qiao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Yingying Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Xu Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Caihong Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Yang Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Shuang Guo
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Yang Zhang
- Department of Anesthesiology, Tianjin Fourth Central Hospital, Tianjin, 300142, China
| | - Wenqing Gao
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Huijuan Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| |
Collapse
|
8
|
Yang S, Su Z, Huo M, Zhong C, Wang F, Zhang Y, Song Y, Shi Y. Effect of Supplementation of Quercetagetin on the Antioxidant Function, Liver Mitochondrial Function and Gut Microbiota of Broilers at High Stocking Density. Animals (Basel) 2025; 15:398. [PMID: 39943168 PMCID: PMC11816227 DOI: 10.3390/ani15030398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
This study investigates the effects of quercetagetin (QG) supplementation on the antioxidant capacity, liver mitochondrial function, and cecal microbiota of broilers raised under high-density conditions. A 2 × 2 factorial design with 144 one-day-old WOD168 broilers, which were allocated to two stocking densities (LD: 4 birds per cage, equivalent to 11.1 birds per square meter; HD: eight birds per cage, equivalent to 22.2 birds per square meter) and two levels of dietary supplementation of QG (0 and 20 mg/kg). At the conclusion of day 21, broilers of similar body weights were randomly allocated into four groups (22 to 42 d): control (CON), QG treatment (QG), high stocking density (HSD), and high stocking density with QG supplementation (H_QG). The results demonstrated that HD groups significantly reduced broiler growth performance, including body weight (BW), average daily gain (ADG), and average daily feed intake (ADFI) (p < 0.05). Additionally, HD groups increased serum stress hormone levels (CORT and ACTH), pro-inflammatory cytokines (IL-1β and IL-6) (p < 0.05), while decreasing liver antioxidant enzyme activities (GSH-Px, T-SOD), serum CAT and T-SOD activities, and mitochondrial function (GSH, complex I-III, ATP contents) (p < 0.05). However, dietary supplementation with 20 mg/kg QG significantly alleviated the negative effects induced by HSD, restoring growth performance, stress hormone levels, immune parameters, and liver antioxidant and mitochondrial function. Moreover, QG supplementation markedly improved cecal microbiota composition, enhancing gut health. Correlation analysis revealed a strong association between microbial composition and overall broiler health, indicating that gut microbiota plays a critical role in mediating these beneficial effects. In conclusion, QG exhibits protective effects against oxidative stress, mitochondrial dysfunction, and gut microbiota imbalance induced by high-density rearing, suggesting its potential as a functional feed additive to improve broiler health under intensive farming conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yuxiang Shi
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan 056038, China; (S.Y.); (Z.S.); (M.H.); (C.Z.); (F.W.); (Y.Z.); (Y.S.)
| |
Collapse
|
9
|
Yadav S, Ganta VC, Varadarajan S, Ong V, Shi Y, Das A, Ash D, Nagarkoti S, McMenamin M, Kelley S, Fukai T, Ushio-Fukai M. Myeloid DRP1 deficiency limits revascularization in ischemic muscles via inflammatory macrophage polarization and metabolic reprogramming. JCI Insight 2025; 10:e177334. [PMID: 39589842 PMCID: PMC11721294 DOI: 10.1172/jci.insight.177334] [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: 11/10/2023] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Macrophages play a crucial role in promoting perfusion recovery and revascularization after ischemia through antiinflammatory polarization, a process essential for the treatment of peripheral artery disease (PAD). Mitochondrial dynamics, particularly regulated by the fission protein DRP1, are closely linked to macrophage metabolism and inflammation. However, the role of DRP1 in reparative neovascularization remains unexplored. Here, we show that DRP1 expression was increased in F4/80+ macrophages within ischemic muscle on day 3 after hind limb ischemia (HLI), an animal model of PAD. Mice lacking Drp1 in myeloid cells exhibited impaired limb perfusion recovery, angiogenesis, and muscle regeneration after HLI. These effects were associated with increased proinflammatory M1-like macrophages, p-NF-κB, and TNF-α, and reduced antiinflammatory M2-like macrophages and p-AMPK in ischemic muscle of myeloid Drp1-/- mice. In vitro, Drp1-deficient macrophages under hypoxia serum starvation (HSS), an in vitro PAD model, demonstrated enhanced glycolysis via reducing p-AMPK as well as mitochondrial dysfunction, and excessive mitochondrial ROS production, resulting in increased proinflammatory M1-gene and reduced antiinflammatory M2-gene expression. Conditioned media from HSS-treated Drp1-/- macrophages exhibited increased proinflammatory cytokine secretion, leading to suppressed angiogenesis in endothelial cells. Thus, macrophage DRP1 deficiency under ischemia drives proinflammatory metabolic reprogramming and macrophage polarization, limiting revascularization in experimental PAD.
Collapse
Affiliation(s)
| | - Vijay C. Ganta
- Vascular Biology Center
- Department of Medicine (Cardiology), and
| | - Sudhahar Varadarajan
- Vascular Biology Center
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Vy Ong
- Biostatistics and Bioinformatics Core, Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yang Shi
- Biostatistics and Bioinformatics Core, Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Archita Das
- Vascular Biology Center
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Dipankar Ash
- Vascular Biology Center
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | | | - Malgorzata McMenamin
- Vascular Biology Center
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | | | - Tohru Fukai
- Vascular Biology Center
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | | |
Collapse
|
10
|
Tabassum S, Wu S, Lee CH, Yang BSK, Gusdon AM, Choi HA, Ren XS. Mitochondrial-targeted therapies in traumatic brain injury: From bench to bedside. Neurotherapeutics 2025; 22:e00515. [PMID: 39721917 PMCID: PMC11840356 DOI: 10.1016/j.neurot.2024.e00515] [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: 07/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide, with limited effective therapeutic options currently available. Recent research has highlighted the pivotal role of mitochondrial dysfunction in the pathophysiology of TBI, making mitochondria an attractive target for therapeutic intervention. This review comprehensively examines advancements in mitochondrial-targeted therapies for TBI, bridging the gap from basic research to clinical applications. We discuss the underlying mechanisms of mitochondrial damage in TBI, including oxidative stress, impaired bioenergetics, mitochondrial dynamics, and apoptotic pathways. Furthermore, we highlight the complex interplay between mitochondrial dysfunction, inflammation, and blood-brain barrier (BBB) integrity, elucidating how these interactions exacerbate injury and impede recovery. We also evaluate various preclinical studies exploring pharmacological agents, gene therapy, and novel drug delivery systems designed to protect and restore mitochondrial function. Clinical trials and their outcomes are assessed to evaluate the translational potential of mitochondrial-targeted therapies in TBI. By integrating findings from bench to bedside, this review emphasizes promising therapeutic avenues and addresses remaining challenges. It also provides guidance for future research to pave the way for innovative treatments that improve patient outcomes in TBI.
Collapse
Affiliation(s)
- Sidra Tabassum
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Silin Wu
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Chang-Hun Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Bosco Seong Kyu Yang
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Aaron M Gusdon
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Huimahn A Choi
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Xuefang S Ren
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
11
|
Li P, Zhou M, Wang J, Tian J, Zhang L, Wei Y, Yang F, Xu Y, Wang G. Important Role of Mitochondrial Dysfunction in Immune Triggering and Inflammatory Response in Rheumatoid Arthritis. J Inflamm Res 2024; 17:11631-11657. [PMID: 39741752 PMCID: PMC11687318 DOI: 10.2147/jir.s499473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 12/15/2024] [Indexed: 01/03/2025] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease, primarily characterized by chronic symmetric synovial inflammation and erosive bone destruction.Mitochondria, the primary site of cellular energy production, play a crucial role in energy metabolism and possess homeostatic regulation capabilities. Mitochondrial function influences the differentiation, activation, and survival of both immune and non-immune cells involved in RA pathogenesis. If the organism experiences hypoxia, genetic predisposition, and oxidative stress, it leads to mitochondrial dysfunction, which further affects immune cell energy metabolism, synovial cell proliferation, apoptosis, and inflammatory signaling, causing the onset and progression of RA; and, mitochondrial regulation is becoming increasingly important in the treatment of RA.In this review, we examine the structure and function of mitochondria, analyze the potential causes of mitochondrial dysfunction in RA, and focus on the mechanisms by which mitochondrial dysfunction triggers chronic inflammation and immune disorders in RA. We also explore the effects of mitochondrial dysfunction on RA immune cells and osteoblasts, emphasizing its key role in the immune response and inflammatory processes in RA. Furthermore, we discuss potential biological processes that regulate mitochondrial homeostasis, which are of great importance for the prevention and treatment of RA.
Collapse
Affiliation(s)
- Pingshun Li
- College of Integrative Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Mengru Zhou
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Jia Wang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Jiexiang Tian
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Lihuan Zhang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Yong Wei
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Fang Yang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Yali Xu
- College of Integrative Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Gang Wang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| |
Collapse
|
12
|
Shen L, Dettmer U. Alpha-Synuclein Effects on Mitochondrial Quality Control in Parkinson's Disease. Biomolecules 2024; 14:1649. [PMID: 39766356 PMCID: PMC11674454 DOI: 10.3390/biom14121649] [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: 09/11/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The maintenance of healthy mitochondria is essential for neuronal survival and relies upon mitochondrial quality control pathways involved in mitochondrial biogenesis, mitochondrial dynamics, and mitochondrial autophagy (mitophagy). Mitochondrial dysfunction is critically implicated in Parkinson's disease (PD), a brain disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra. Consequently, impaired mitochondrial quality control may play a key role in PD pathology. This is affirmed by work indicating that genes such as PRKN and PINK1, which participate in multiple mitochondrial processes, harbor PD-associated mutations. Furthermore, mitochondrial complex-I-inhibiting toxins like MPTP and rotenone are known to cause Parkinson-like symptoms. At the heart of PD is alpha-synuclein (αS), a small synaptic protein that misfolds and aggregates to form the disease's hallmark Lewy bodies. The specific mechanisms through which aggregated αS exerts its neurotoxicity are still unknown; however, given the vital role of both αS and mitochondria to PD, an understanding of how αS influences mitochondrial maintenance may be essential to elucidating PD pathogenesis and discovering future therapeutic targets. Here, the current knowledge of the relationship between αS and mitochondrial quality control pathways in PD is reviewed, highlighting recent findings regarding αS effects on mitochondrial biogenesis, dynamics, and autophagy.
Collapse
Affiliation(s)
- Lydia Shen
- College of Arts & Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
13
|
Mosalmanzadeh N, Maurmann RM, Davis K, Schmitt BL, Makowski L, Pence BD. Modulatory Effects of Mdivi-1 on OxLDL-Induced Metabolic Alterations, Inflammatory Responses, and Foam Cell Formation in Human Monocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628145. [PMID: 39763794 PMCID: PMC11702575 DOI: 10.1101/2024.12.12.628145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Atherosclerosis, a major contributor to cardiovascular disease, involves lipid accumulation and inflammatory processes in arterial walls, with oxidized low-density lipoprotein (OxLDL) playing a central role. OxLDL is increased during aging and stimulates monocyte transformation into foam cells and induces metabolic reprogramming and pro-inflammatory responses, accelerating atherosclerosis progression and contributing to other age-related diseases. This study investigated the effects of Mdivi-1, a mitochondrial fission inhibitor, and S1QEL, a selective complex I-associated reactive oxygen species (ROS) inhibitor, on OxLDL-induced responses in monocytes. Healthy monocytes isolated from participants were treated with OxLDL, with or without Mdivi-1 or S1QEL, and assessed for metabolic shifts, inflammatory cytokine expression, foam cell formation, and ROS production. OxLDL treatment elevated glycolytic activity (ECAR) and expression of pro-inflammatory cytokines IL1B and CXCL8, promoting foam cell formation and mitochondrial ROS (mtROS) production. Mdivi-1 and S1QEL effectively reduced OxLDL-induced glycolytic reprogramming, inflammatory cytokine levels, and foam cell formation while limiting mtROS. These findings suggest that both Mdivi-1 and S1QEL modulate key monocyte responses to OxLDL, providing insights into potential therapeutic approaches for age-related diseases.
Collapse
Affiliation(s)
| | | | - Kierstin Davis
- College of Health Sciences, University of Memphis, Memphis, TN, USA
| | | | - Liza Makowski
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Brandt D Pence
- College of Health Sciences, University of Memphis, Memphis, TN, USA
- Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN, USA
| |
Collapse
|
14
|
Walker EC, Javati S, Todd EM, Matlam JP, Lin X, Bryant M, Krone E, Ramani R, Chandra P, Green TP, Anaya EP, Zhou JY, Alexander KA, Tong RS, Yuasi L, Boluarte S, Yang F, Greenberg L, Nerbonne JM, Greenberg MJ, Clemens RA, Philips JA, Wilson LD, Halabi CM, DeBosch BJ, Blyth CC, Druley TE, Kazura JW, Pomat WS, Morley SC. Novel coenzyme Q6 genetic variant increases susceptibility to pneumococcal disease. Nat Immunol 2024; 25:2247-2258. [PMID: 39496954 PMCID: PMC11908386 DOI: 10.1038/s41590-024-01998-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 09/30/2024] [Indexed: 11/06/2024]
Abstract
Acute lower respiratory tract infection (ALRI) remains a major worldwide cause of childhood mortality, compelling innovation in prevention and treatment. Children in Papua New Guinea (PNG) experience profound morbidity from ALRI caused by Streptococcus pneumoniae. As a result of evolutionary divergence, the human PNG population exhibits profound genetic variation and diversity. To address unmet health needs of children in PNG, we tested whether genetic variants increased ALRI morbidity. Whole-exome sequencing of a pilot child cohort identified homozygosity for a novel single-nucleotide variant (SNV) in coenzyme Q6 (COQ6) in cases with ALRI. COQ6 encodes a mitochondrial enzyme essential for biosynthesis of ubiquinone, an electron acceptor in the electron transport chain. A significant association of SNV homozygosity with ALRI was replicated in an independent ALRI cohort (P = 0.036). Mice homozygous for homologous mouse variant Coq6 exhibited increased mortality after pneumococcal lung infection, confirming causality. Bone marrow chimeric mice further revealed that expression of variant Coq6 in recipient (that is, nonhematopoietic) tissues conferred increased mortality. Variant Coq6 maintained ubiquinone biosynthesis, while accelerating metabolic remodeling after pneumococcal challenge. Identification of this COQ6 variant provides a genetic basis for increased pneumonia susceptibility in PNG and establishes a previously unrecognized role for the enzyme COQ6 in regulating inflammatory-mediated metabolic remodeling.
Collapse
Affiliation(s)
- Emma C Walker
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Program in Immunology, Division of Biological and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah Javati
- Infection and Immunity Unit, Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Elizabeth M Todd
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - John-Paul Matlam
- Infection and Immunity Unit, Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Xue Lin
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Michelle Bryant
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Emily Krone
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Rashmi Ramani
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Pallavi Chandra
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Taylor P Green
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Edgar P Anaya
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Julie Y Zhou
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine A Alexander
- Department of Pediatrics, Division of Hematology-Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - R Spencer Tong
- Department of Pediatrics, Division of Hematology-Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lapule Yuasi
- Infection and Immunity Unit, Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Sebastian Boluarte
- Department. of Pediatrics, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Fan Yang
- Department. of Pediatrics, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeanne M Nerbonne
- Departments of Developmental Biology and Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Regina A Clemens
- Department. of Pediatrics, Division of Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jennifer A Philips
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Leslie D Wilson
- Division of Comparative Medicine, Research Animal Diagnostic Laboratory, Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen M Halabi
- Department of Pediatrics, Division of Nephrology and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian J DeBosch
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, Division of Gastroenterology, Hepatology & Nutrition, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher C Blyth
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute and School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
- Department of Infectious Diseases, Perth Children's Hospital, Nedlands, Western Australia, Australia
- Department of Microbiology, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Todd E Druley
- Department of Pediatrics, Division of Hematology-Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - James W Kazura
- Center for Global Health & Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - William S Pomat
- Infection and Immunity Unit, Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute and School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Sharon Celeste Morley
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, USA.
- Program in Immunology, Division of Biological and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA.
- Dept. of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
15
|
Grima-Terrén M, Campanario S, Ramírez-Pardo I, Cisneros A, Hong X, Perdiguero E, Serrano AL, Isern J, Muñoz-Cánoves P. Muscle aging and sarcopenia: The pathology, etiology, and most promising therapeutic targets. Mol Aspects Med 2024; 100:101319. [PMID: 39312874 DOI: 10.1016/j.mam.2024.101319] [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: 02/27/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Sarcopenia is a progressive muscle wasting disorder that severely impacts the quality of life of elderly individuals. Although the natural aging process primarily causes sarcopenia, it can develop in response to other conditions. Because muscle function is influenced by numerous changes that occur with age, the etiology of sarcopenia remains unclear. However, recent characterizations of the aging muscle transcriptional landscape, signaling pathway disruptions, fiber and extracellular matrix compositions, systemic metabolomic and inflammatory responses, mitochondrial function, and neurological inputs offer insights and hope for future treatments. This review will discuss age-related changes in healthy muscle and our current understanding of how this can deteriorate into sarcopenia. As our elderly population continues to grow, we must understand sarcopenia and find treatments that allow individuals to maintain independence and dignity throughout an extended lifespan.
Collapse
Affiliation(s)
- Mercedes Grima-Terrén
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Silvia Campanario
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Ignacio Ramírez-Pardo
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Andrés Cisneros
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Xiaotong Hong
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | | | - Antonio L Serrano
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | - Joan Isern
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | - Pura Muñoz-Cánoves
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain.
| |
Collapse
|
16
|
Liu Y, Li Y, Zhu Y, Wang M, Luan Z. Construction of a novel mitochondrial oxidative stress-related genes prognostic system and molecular subtype characterization for breast cancer. Discov Oncol 2024; 15:631. [PMID: 39514138 PMCID: PMC11549074 DOI: 10.1007/s12672-024-01522-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE Breast cancer (BRCA) is the most common malignant tumor among women, characterized by high incidence rates and mortality rates. Oxidative stress and immunity, particularly in relation to mitochondria, have emerged as pivotal factors in breast carcinogenesis. Nonetheless, limited research has explored the specific contribution of mitochondrial oxidative stress to the prognosis of BRCA. METHOD In this study, we conducted univariate and multivariate Cox regression analyses to pinpoint independent prognostic genes associated with mitochondrial oxidative stress (MOSRGs) and their correlation with BRCA clinical outcomes. Subsequently, we developed a robust and accurate MOS scoring system for BRCA patients based on these identified independent prognostic MOSRGs. RESULT Our findings were further substantiated by immune infiltration and somatic mutation analyses, providing additional evidence that the MOS scoring system holds predictive value for clinical outcomes in patients and correlates directly with three subtypes of BRCA. In vitro experiments in the MCF7 cell and breast tissue further verified the mRNA and protein expression level of independent prognostic genes, validating the consistency of the MOS prognostic signatures in BRCA. CONCLUSION This research has unveiled a novel prognostic scoring system, providing valuable insights for improving patient prognosis assessment and developing individualized treatment strategies in BRCA patients.
Collapse
Affiliation(s)
- Ying Liu
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yang Li
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yanzheng Zhu
- Department of Internal Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Min Wang
- Department of Digestive Endoscopy, Dezhou Hospital, Qilu Hospital, Shandong University, Jinan, China
| | - Zheyao Luan
- Department of Physical Examination, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| |
Collapse
|
17
|
Johnston J, Jeon H, Choi YY, Kim G, Shi T, Khong C, Chang HC, Myung NV, Wang Y. Stimulative piezoelectric nanofibrous scaffolds for enhanced small extracellular vesicle production in 3D cultures. Biomater Sci 2024; 12:5728-5741. [PMID: 39403853 PMCID: PMC11474809 DOI: 10.1039/d4bm00504j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Small extracellular vesicles (sEVs) have great promise as effective carriers for drug delivery. However, the challenges associated with the efficient production of sEVs hinder their clinical applications. Herein, we report a stimulative 3D culture platform for enhanced sEV production. The proposed platform consists of a piezoelectric nanofibrous scaffold (PES) coupled with acoustic stimulation to enhance sEV production of cells in a 3D biomimetic microenvironment. Combining cell stimulation with a 3D culture platform in this stimulative PES enables a 15.7-fold increase in the production rate per cell with minimal deviations in particle size and protein composition compared with standard 2D cultures. We find that the enhanced sEV production is attributable to the activation and upregulation of crucial sEV production steps through the synergistic effect of stimulation and the 3D microenvironment. Moreover, changes in cell morphology lead to cytoskeleton redistribution through cell-matrix interactions in the 3D cultures. This in turn facilitates intracellular EV trafficking, which impacts the production rate. Overall, our work provides a promising 3D cell culture platform based on piezoelectric biomaterials for enhanced sEV production. This platform is expected to accelerate the potential use of sEVs for drug delivery and broad biomedical applications.
Collapse
Affiliation(s)
- James Johnston
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Hyunsu Jeon
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Yun Young Choi
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Gaeun Kim
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Tiger Shi
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Courtney Khong
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Nosang Vincent Myung
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Yichun Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
18
|
Raychaudhuri D, Singh P, Chakraborty B, Hennessey M, Tannir AJ, Byregowda S, Natarajan SM, Trujillo-Ocampo A, Im JS, Goswami S. Histone lactylation drives CD8 + T cell metabolism and function. Nat Immunol 2024; 25:2140-2151. [PMID: 39375549 DOI: 10.1038/s41590-024-01985-9] [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: 11/15/2023] [Accepted: 09/11/2024] [Indexed: 10/09/2024]
Abstract
The activation and functional differentiation of CD8+ T cells are linked to metabolic pathways that result in the production of lactate. Lactylation is a lactate-derived histone post-translational modification; however, the relevance of histone lactylation in the context of CD8+ T cell activation and function is not known. Here, we show the enrichment of H3K18 lactylation (H3K18la) and H3K9 lactylation (H3K9la) in human and mouse CD8+ T cells, which act as transcription initiators of key genes regulating CD8+ T cell function. Further, we note distinct patterns of H3K18la and H3K9la in CD8+ T cell subsets linked to their specific metabolic profiles. Additionally, we find that modulation of H3K18la and H3K9la by targeting metabolic and epigenetic pathways influence CD8+ T cell effector function, including antitumor immunity, in preclinical models. Overall, our study uncovers the potential roles of H3K18la and H3K9la in CD8+ T cells.
Collapse
Affiliation(s)
- Deblina Raychaudhuri
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pratishtha Singh
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bidisha Chakraborty
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mercedes Hennessey
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aminah J Tannir
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shrinidhi Byregowda
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Seanu Meena Natarajan
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abel Trujillo-Ocampo
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Jin Seon Im
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Sangeeta Goswami
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
19
|
Gaviraghi A, Barletta ABF, Silva TLAE, Oliveira MP, Sorgine MHF, Oliveira MF. Activation of innate immunity selectively compromises mitochondrial complex I, proline oxidation, and flight activity in the major arbovirus vector Aedes aegypti. Mol Microbiol 2024; 122:683-703. [PMID: 38720451 DOI: 10.1111/mmi.15269] [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: 10/20/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 11/26/2024]
Abstract
Aedes aegypti females are natural vectors of important arboviruses such as dengue, zika, and yellow fever. Mosquitoes activate innate immune response signaling pathways upon infection, as a resistance mechanism to fight pathogens and limit their propagation. Despite the beneficial effects of immune activation for insect vectors, phenotypic costs ultimately affect their fitness. However, the underlying mechanisms that mediate these fitness costs remain poorly understood. Given the high energy required to mount a proper immune response, we hypothesized that systemic activation of innate immunity would impair flight muscle mitochondrial function, compromising tissue energy demand and flight activity. Here, we investigated the dynamic effects of activation of innate immunity by intra-thoracic zymosan injection on A. aegypti flight muscle mitochondrial metabolism. Zymosan injection significantly increased defensin A expression in fat bodies in a time-dependent manner that compromised flight activity. Although oxidant levels in flight muscle were hardly altered, ATP-linked respiratory rates driven by mitochondrial pyruvate+proline oxidation were significantly reduced at 24 h upon zymosan injection. Oxidative phosphorylation coupling was preserved regardless of innate immune response activation along 24 h. Importantly, rotenone-sensitive respiration and complex I-III activity were specifically reduced 24 h upon zymosan injection. Also, loss of complex I activity compromised ATP-linked and maximal respiratory rates mediated by mitochondrial proline oxidation. Finally, the magnitude of innate immune response activation negatively correlated with respiratory rates, regardless of the metabolic states. Collectively, we demonstrate that activation of innate immunity is strongly associated with reduced flight muscle complex I activity with direct consequences to mitochondrial proline oxidation and flight activity. Remarkably, our results indicate a trade-off between dispersal and immunity exists in an insect vector, underscoring the potential consequences of disrupted flight muscle mitochondrial energy metabolism to arbovirus transmission.
Collapse
Affiliation(s)
- Alessandro Gaviraghi
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Ana Beatriz F Barletta
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thiago Luiz Alves E Silva
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus P Oliveira
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Marcos H F Sorgine
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcus F Oliveira
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Li W, Cai P, Xu Y, Tian W, Jing L, Lv Q, Zhao Y, Wang H, Shao Q. Mitochondrial Quality Control Orchestrates the Symphony of B Cells and Plays Critical Roles in B Cell-Related Diseases. J Immunol Res 2024; 2024:5577506. [PMID: 39449998 PMCID: PMC11502133 DOI: 10.1155/2024/5577506] [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: 04/01/2024] [Revised: 09/04/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
B cells are essential for humoral immune response due to their ability to secrete antibodies. The development of B cells from the bone marrow to the periphery is tightly regulated by a complex set of immune signals, and each subset of B cells has a unique metabolic profile. Mitochondria, which serve as cellular energy powerhouses, play an essential role in regulating cell survival and immune responses. To maintain metabolic homeostasis, mitochondria dynamically adjust their morphology, distribution, and mass via biogenesis, fusion and fission, translocation, and mitophagy. Despite its extreme importance, the role of mitochondrial quality control (MQC) in B cells has not been thoroughly summarized, unlike in T cells. This article aims to review the mechanism of MQC that shapes B cell fate and functions. In addition, we will discuss the physiological and pathological implications of MQC in B cells, providing new insights into potential therapeutic targets for diseases associated with B cell abnormalities.
Collapse
Affiliation(s)
- Wuhao Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Peiyang Cai
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Ye Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Weihong Tian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Licong Jing
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Qiaoyi Lv
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Yangjing Zhao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Hui Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Qixiang Shao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
- Institute of Medical Genetics and Reproductive Immunity, The Digestive and Reproductive System Cancers Precise Prevention Engineering Research Center of Jiangsu Province, Jiangsu College of Nursing, Huai'an 223002, Jiangsu, China
| |
Collapse
|
21
|
Lu M, Li W, Zhou J, Shang J, Lin L, Liu Y, Zhu X. Integrative bioinformatics analysis for identifying the mitochondrial-related gene signature associated with immune infiltration in premature ovarian insufficiency. BMC Med 2024; 22:444. [PMID: 39379953 PMCID: PMC11462806 DOI: 10.1186/s12916-024-03675-7] [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] [Received: 05/14/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a reproductive disorder characterized by the cessation of ovarian function before the age of 40. Although mitochondrial dysfunction and immune disorders are believed to contribute to ovarian damage in POI, the interplay between these factors remains understudied. METHODS In this research, transcriptomic data related to POI were obtained from the NCBI GEO database. Hub biomarkers were identified through the construction of a protein‒protein interaction (PPI) network and further validated using RT‒qPCR and Western blot. Moreover, their expression across various cell types was elucidated via single-cell RNA sequencing analysis. A comprehensive investigation of the mitochondrial and immune profiles of POI was carried out through correlation analysis. Furthermore, potential therapeutic agents were predicted utilizing the cMap database. RESULTS A total of 119 mitochondria-related differentially expressed genes (MitoDEGs) were identified and shown to be significantly enriched in metabolic pathways. Among these genes, Hadhb, Cpt1a, Mrpl12, and Mrps7 were confirmed both in a POI model and in human granulosa cells (GCs), where they were found to accumulate in GCs and theca cells. Immune analysis revealed variations in macrophages, monocytes, and 15 other immune cell types between the POI and control groups. Notably, strong correlations were observed between seven hub-MitoDEGs (Hadhb, Cpt1a, Cpt2, Mrpl12, Mrps7, Mrpl51, and Eci1) and various functions, such as mitochondrial respiratory complexes, dynamics, mitophagy, mitochondrial metabolism, immune-related genes, and immunocytes. Additionally, nine potential drugs (calyculin, amodiaquine, eudesmic acid, cefotaxime, BX-912, prostratin, SCH-79797, HU-211, and pizotifen) targeting key genes were identified. CONCLUSIONS Our results highlight the crosstalk between mitochondrial function and the immune response in the development of POI. The identification of MitoDEGs could lead to reliable biomarkers for the early diagnosis, monitoring, and personalized treatment of POI.
Collapse
Affiliation(s)
- Minjun Lu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Wenxin Li
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Jiamin Zhou
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Junyu Shang
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Li Lin
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
- Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Yueqin Liu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China
| | - Xiaolan Zhu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), No. 20 Zhengdong Road, Zhenjiang, 212001, China.
| |
Collapse
|
22
|
Pandey S, Anang V, Schumacher MM. Tumor microenvironment induced switch to mitochondrial metabolism promotes suppressive functions in immune cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 389:67-103. [PMID: 39396850 DOI: 10.1016/bs.ircmb.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Understanding the intricacies of the metabolic phenotype in immune cells and its plasticity within the tumor microenvironment is pivotal in understanding the pathology and prognosis of cancer. Unfavorable conditions and cellular stress in the tumor microenvironment (TME) exert a profound impact on cellular functions in immune cells, thereby influencing both tumor progression and immune responses. Elevated AMP:ATP ratio, a consequence of limited glucose levels, activate AMP-activated protein kinase (AMPK) while concurrently repressing the activity of mechanistic target of rapamycin (mTOR) and hypoxia-inducible factor 1-alpha (HIF-1α). The intricate balance between AMPK, mTOR, and HIF-1α activities defines the metabolic phenotype of immune cells in the TME. These Changes in metabolic phenotype are strongly associated with immune cell functions and play a crucial role in creating a milieu conducive to tumor progression. Insufficiency of nutrient and oxygen supply leads to a metabolic shift in immune cells characterized by a decrease in glycolysis and an increase in oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) rates. In most cases, this shift in metabolism is accompanied by a compromise in the effector functions of these immune cells. This metabolic adaptation prompts immune cells to turn down their effector functions, entering a quiescent or immunosuppressive state that may support tumor growth. This article discusses how tumor microenvironment alters the metabolism in immune cells leading to their tolerance and tumor progression, with emphasis on mitochondrial metabolism (OXPHOS and FAO).
Collapse
Affiliation(s)
- Sanjay Pandey
- Department of Radiation Oncology, Montefiorke Medical Center, Bronx, NY, United States.
| | - Vandana Anang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.
| | - Michelle M Schumacher
- Department of Radiation Oncology, Montefiorke Medical Center, Bronx, NY, United States; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
23
|
Johnston J, Jeon H, Choi YY, Kim G, Shi T, Khong C, Chang HC, Myung NV, Wang Y. Stimulative piezoelectric nanofibrous scaffolds for enhanced small extracellular vesicle production in 3D cultures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589114. [PMID: 38659930 PMCID: PMC11042190 DOI: 10.1101/2024.04.12.589114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Small extracellular vesicles (sEVs) have great promise as effective carriers for drug delivery. However, the challenges associated with the efficient production of sEVs hinder their clinical applications. Herein, we report a stimulative 3D culture platform for enhanced sEV production. The proposed platform consists of a piezoelectric nanofibrous scaffold (PES) coupled with acoustic stimulation to enhance sEV production of cells in a 3D biomimetic microenvironment. Combining cell stimulation with a 3D culture platform in this stimulative PES enables a 15.7-fold increase in the production rate per cell with minimal deviations in particle size and protein composition compared with standard 2D cultures. We find that the enhanced sEV production is attributable to the activation and upregulation of crucial sEV production steps through the synergistic effect of stimulation and the 3D microenvironment. Moreover, changes in cell morphology lead to cytoskeleton redistribution through cell-matrix interactions in the 3D cultures. This in turn facilitates intracellular EV trafficking, which impacts the production rate. Overall, our work provides a promising 3D cell culture platform based on piezoelectric biomaterials for enhanced sEV production. This platform is expected to accelerate the potential use of sEVs for drug delivery and broad biomedical applications.
Collapse
|
24
|
Zhang Y, Wang X, Gao Z, Li X, Meng R, Wu X, Ding J, Shen W, Zhu J. Hypoxia-inducible factor-1α promotes macrophage functional activities in protecting hypoxia-tolerant large yellow croaker ( Larimichthys crocea) against Aeromonas hydrophila infection. Front Immunol 2024; 15:1410082. [PMID: 39156889 PMCID: PMC11327042 DOI: 10.3389/fimmu.2024.1410082] [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: 03/31/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024] Open
Abstract
The immune system requires a high energy expenditure to resist pathogen invasion. Macrophages undergo metabolic reprogramming to meet these energy requirements and immunologic activity and polarize to M1-type macrophages. Understanding the metabolic pathway switching in large yellow croaker (Larimichthys crocea) macrophages in response to lipopolysaccharide (LPS) stimulation and whether this switching affects immunity is helpful in explaining the stronger immunity of hypoxia-tolerant L. crocea. In this study, transcript levels of glycolytic pathway genes (Glut1 and Pdk1), mRNA levels or enzyme activities of glycolytic enzymes [hexokinase (HK), phosphofructokinase (PFK), pyruvate kinase (PK), and lactate dehydrogenase A (LDHA)], aerobic respiratory enzymes [pyruvate dehydrogenase (PDH), isocitrate dehydrogenase (IDH), and succinate dehydrogenase (SDH)], metabolites [lactic acid (LA) and adenosine triphosphate (ATP)], levels of bactericidal products [reactive oxygen species (ROS) and nitric oxide (NO)], and transcripts and level changes of inflammatory factors [IL1β, TNFα, and interferon (IFN) γ] were detected in LPS-stimulated L. crocea head kidney macrophages. We showed that glycolysis was significantly induced, the tricarboxylic acid (TCA) cycle was inhibited, and metabolic reprogramming occurred, showing the Warburg effect when immune cells were activated. To determine the potential regulatory mechanism behind these changes, LcHIF-1α was detected and found to be significantly induced and transferred to the nucleus after LPS stimulation. LcHif-1α interference led to a significant reduction in glycolytic pathway gene transcript expression, enzyme activity, metabolites, bactericidal substances, and inflammatory factor levels; a significant increase in the aerobic respiration enzymes; and decreased migration, invasion, and phagocytosis. Further ultrastructural observation by electron microscopy showed that fewer microspheres contained phagocytes and that more cells were damaged after LcHif-1α interference. LcHif-1α overexpression L. crocea head kidney macrophages showed the opposite trend, and promoter activities of Ldha and Il1β were significantly enhanced after LcHif-1α overexpression in HEK293T cells. Our data showed that LcHIF-1α acted as a metabolic switch in L. crocea macrophages and was important in polarization. Hypoxia-tolerant L. crocea head kidney showed a stronger Warburg effect and inhibited the TCA cycle, higher metabolites, and bactericidal substance levels. These results collectively revealed that LcHif-1α may promote the functional activities of head kidney macrophages in protecting hypoxia-tolerant L. crocea from Aeromonas hydrophila infection.
Collapse
Affiliation(s)
- Yibo Zhang
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Ningbo Academy of Oceanology and Fishery, Ningbo, Zhejiang, China
| | - Xuelei Wang
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Ningbo Academy of Oceanology and Fishery, Ningbo, Zhejiang, China
| | - Zhenyu Gao
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Ningbo Academy of Oceanology and Fishery, Ningbo, Zhejiang, China
| | - XuJie Li
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Ningbo Academy of Oceanology and Fishery, Ningbo, Zhejiang, China
| | - Ran Meng
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Ningbo Academy of Oceanology and Fishery, Ningbo, Zhejiang, China
| | - Xiongfei Wu
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Ningbo Academy of Oceanology and Fishery, Ningbo, Zhejiang, China
| | - Jie Ding
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Ningbo Academy of Oceanology and Fishery, Ningbo, Zhejiang, China
| | - Weiliang Shen
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Ningbo Academy of Oceanology and Fishery, Ningbo, Zhejiang, China
| | - Junquan Zhu
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, and Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
25
|
Priyanka, Sharma S, Sharma M. Role of PE/PPE proteins of Mycobacterium tuberculosis in triad of host mitochondria, oxidative stress and cell death. Microb Pathog 2024; 193:106757. [PMID: 38908454 DOI: 10.1016/j.micpath.2024.106757] [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: 03/11/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The PE and PPE family proteins of Mycobacterium tuberculosis (Mtb) is exclusively found in pathogenic Mycobacterium species, comprising approximately 8-10 % of the Mtb genome. These emerging virulent factors have been observed to play pivotal roles in Mtb pathogenesis and immune evasion through various strategies. These immunogenic proteins are known to modulate the host immune response and cell-death pathways by targeting the powerhouse of the cell, the mitochondria to support Mtb survival. In this article, we are focused on how PE/PPE family proteins target host mitochondria to induce mitochondrial perturbations, modulate the levels of cellular ROS (Reactive oxygen species) and control cell death pathways. We observed that the time of expression of these proteins at different stages of infection is crucial for elucidating their impact on the cell death pathways and eventually on the outcome of infection. This article focuses on understanding the contributions of the PE/PPE proteins by unravelling the triad of host mitochondria, oxidative stress and cell death pathways that facilitate the Mtb persistence. Understanding the role of these proteins in host cellular pathways and the intricate mechanisms paves the way for the development of novel therapeutic strategies to combat TB infections.
Collapse
Affiliation(s)
- Priyanka
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Sadhna Sharma
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Monika Sharma
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
26
|
Zimmermann JA, Lucht K, Stecher M, Badhan C, Glaser KM, Epple MW, Koch LR, Deboutte W, Manke T, Ebnet K, Brinkmann F, Fehler O, Vogl T, Schuster EM, Bremser A, Buescher JM, Rambold AS. Functional multi-organelle units control inflammatory lipid metabolism of macrophages. Nat Cell Biol 2024; 26:1261-1273. [PMID: 38969763 PMCID: PMC11321999 DOI: 10.1038/s41556-024-01457-0] [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: 07/08/2023] [Accepted: 06/05/2024] [Indexed: 07/07/2024]
Abstract
Eukaryotic cells contain several membrane-separated organelles to compartmentalize distinct metabolic reactions. However, it has remained unclear how these organelle systems are coordinated when cells adapt metabolic pathways to support their development, survival or effector functions. Here we present OrgaPlexing, a multi-spectral organelle imaging approach for the comprehensive mapping of six key metabolic organelles and their interactions. We use this analysis on macrophages, immune cells that undergo rapid metabolic switches upon sensing bacterial and inflammatory stimuli. Our results identify lipid droplets (LDs) as primary inflammatory responder organelle, which forms three- and four-way interactions with other organelles. While clusters with endoplasmic reticulum (ER) and mitochondria (mitochondria-ER-LD unit) help supply fatty acids for LD growth, the additional recruitment of peroxisomes (mitochondria-ER-peroxisome-LD unit) supports fatty acid efflux from LDs. Interference with individual components of these units has direct functional consequences for inflammatory lipid mediator synthesis. Together, we show that macrophages form functional multi-organellar units to support metabolic adaptation and provide an experimental strategy to identify organelle-metabolic signalling hubs.
Collapse
Affiliation(s)
- Julia A Zimmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Center of Chronic Immunodeficiency, Medical Center University of Freiburg, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Kerstin Lucht
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Manuel Stecher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Chahat Badhan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katharina M Glaser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Maximilian W Epple
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Lena R Koch
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Ward Deboutte
- Bioinformatics Core Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Thomas Manke
- Bioinformatics Core Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Munster, Munster, Germany
| | - Frauke Brinkmann
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Munster, Munster, Germany
| | - Olesja Fehler
- Institute of Immunology, University of Munster, Munster, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Munster, Munster, Germany
| | - Ev-Marie Schuster
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Anna Bremser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Joerg M Buescher
- Metabolomics Core Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Angelika S Rambold
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
- Center of Chronic Immunodeficiency, Medical Center University of Freiburg, Freiburg, Germany.
| |
Collapse
|
27
|
Qi X, Liu C, Si J, Yin B, Huang J, Wang X, Huang J, Sun H, Zhu C, Zhang W. A bioenergetically-active ploy (glycerol sebacate)-based multiblock hydrogel improved diabetic wound healing through revitalizing mitochondrial metabolism. Cell Prolif 2024; 57:e13613. [PMID: 38351579 PMCID: PMC11216945 DOI: 10.1111/cpr.13613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 07/03/2024] Open
Abstract
Diabetic wounds impose significant burdens on patients' quality of life and healthcare resources due to impaired healing potential. Factors like hyperglycemia, oxidative stress, impaired angiogenesis and excessive inflammation contribute to the delayed healing trajectory. Mounting evidence indicates a close association between impaired mitochondrial function and diabetic complications, including chronic wounds. Mitochondria are critical for providing energy essential to wound healing processes. However, mitochondrial dysfunction exacerbates other pathological factors, creating detrimental cycles that hinder healing. This study conducted correlation analysis using clinical specimens, revealing a positive correlation between mitochondrial dysfunction and oxidative stress, inflammatory response and impaired angiogenesis in diabetic wounds. Restoring mitochondrial function becomes imperative for developing targeted therapies. Herein, we synthesized a biodegradable poly (glycerol sebacate)-based multiblock hydrogel, named poly (glycerol sebacate)-co-poly (ethylene glycol)-co-poly (propylene glycol) (PEPGS), which can be degraded in vivo to release glycerol, a crucial component in cellular metabolism, including mitochondrial respiration. We demonstrate the potential of PEPGS-based hydrogels to improve outcomes in diabetic wound healing by revitalizing mitochondrial metabolism. Furthermore, we investigate the underlying mechanism through proteomics analysis, unravelling the regulation of ATP and nicotinamide adenine dinucleotide metabolic processes, biosynthetic process and generation during mitochondrial metabolism. These findings highlight the therapeutic potential of PEPGS-based hydrogels as advanced wound dressings for diabetic wound healing.
Collapse
Affiliation(s)
- Xin Qi
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Orthopedic Surgery, Shanghai Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chenjun Liu
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jingyi Si
- Department of Gastroenterology and Hepatology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Bohao Yin
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jingjing Huang
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin Wang
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jinghuan Huang
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hui Sun
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Changfeng Zhu
- Department of Gastroenterology and Hepatology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Wei Zhang
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
28
|
Raychaudhuri D, Singh P, Hennessey M, Chakraborty B, Tannir AJ, Trujillo-Ocampo A, Im JS, Goswami S. Histone Lactylation Drives CD8 T Cell Metabolism and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.25.554830. [PMID: 38854142 PMCID: PMC11160580 DOI: 10.1101/2023.08.25.554830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The activation and functional differentiation of CD8 T cells are linked to metabolic pathways that result in the production of lactate. Lactylation is a lactate-derived histone post-translational modification (hPTM); however, the relevance of histone lactylation in the context of CD8 T cell activation and function is not known. Here, we show the enrichment of H3K18-lactylation (H3K18la) and H3K9-lactylation (H3K9la) in human and murine CD8 T cells which act as transcription initiators of key genes regulating CD8 T cell phenotype and function. Further, we note distinct impacts of H3K18la and H3K9la on CD8 T cell subsets linked to their specific metabolic profiles. Importantly, we demonstrate that modulation of H3K18la and H3K9la by targeting metabolic and epigenetic pathways regulates CD8 T cell effector function including anti-tumor immunity in preclinical models. Overall, our study uncovers the unique contributions of H3K18la and H3K9la in modulating CD8 T cell phenotype and function intricately associated with metabolic state.
Collapse
|
29
|
Bamidele AO, Mishra SK, Piovezani Ramos G, Hirsova P, Klatt EE, Abdelrahman LM, Sagstetter MR, Davidson HM, Fehrenbach PJ, Valenzuela-Pérez L, Kim Lee HS, Zhang S, Aguirre Lopez A, Kurdi AT, Westphal MS, Gonzalez MM, Gaballa JM, Kosinsky RL, Lee HE, Smyrk TC, Bantug G, Gades NM, Faubion WA. Interleukin 21 Drives a Hypermetabolic State and CD4 + T-Cell-Associated Pathogenicity in Chronic Intestinal Inflammation. Gastroenterology 2024; 166:826-841.e19. [PMID: 38266738 PMCID: PMC11034723 DOI: 10.1053/j.gastro.2024.01.026] [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: 04/09/2023] [Revised: 11/23/2023] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND & AIMS Incapacitated regulatory T cells (Tregs) contribute to immune-mediated diseases. Inflammatory Tregs are evident during human inflammatory bowel disease; however, mechanisms driving the development of these cells and their function are not well understood. Therefore, we investigated the role of cellular metabolism in Tregs relevant to gut homeostasis. METHODS Using human Tregs, we performed mitochondrial ultrastructural studies via electron microscopy and confocal imaging, biochemical and protein analyses using proximity ligation assay, immunoblotting, mass cytometry and fluorescence-activated cell sorting, metabolomics, gene expression analysis, and real-time metabolic profiling utilizing the Seahorse XF analyzer. We used a Crohn's disease single-cell RNA sequencing dataset to infer the therapeutic relevance of targeting metabolic pathways in inflammatory Tregs. We examined the superior functionality of genetically modified Tregs in CD4+ T-cell-induced murine colitis models. RESULTS Mitochondria-endoplasmic reticulum appositions, known to mediate pyruvate entry into mitochondria via voltage-dependent anion channel 1 (VDAC1), are abundant in Tregs. VDAC1 inhibition perturbed pyruvate metabolism, eliciting sensitization to other inflammatory signals reversible by membrane-permeable methyl pyruvate supplementation. Notably, interleukin (IL) 21 diminished mitochondria-endoplasmic reticulum appositions, resulting in enhanced enzymatic function of glycogen synthase kinase 3 β, a putative negative regulator of VDAC1, and a hypermetabolic state that amplified Treg inflammatory response. Methyl pyruvate and glycogen synthase kinase 3 β pharmacologic inhibitor (LY2090314) reversed IL21-induced metabolic rewiring and inflammatory state. Moreover, IL21-induced metabolic genes in Tregs in vitro were enriched in human Crohn's disease intestinal Tregs. Adoptively transferred Il21r-/- Tregs efficiently rescued murine colitis in contrast to wild-type Tregs. CONCLUSIONS IL21 triggers metabolic dysfunction associated with Treg inflammatory response. Inhibiting IL21-induced metabolism in Tregs may mitigate CD4+ T-cell-driven chronic intestinal inflammation.
Collapse
Affiliation(s)
- Adebowale O Bamidele
- Immunometabolism and Mucosal Immunity Laboratory, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Department of Immunology, Mayo Clinic, Rochester, Minnesota.
| | - Shravan K Mishra
- Immunometabolism and Mucosal Immunity Laboratory, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Emily E Klatt
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Leena M Abdelrahman
- Immunometabolism and Mucosal Immunity Laboratory, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Mary R Sagstetter
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Heidi M Davidson
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Patrick J Fehrenbach
- Immunometabolism and Mucosal Immunity Laboratory, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Hyun Se Kim Lee
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Song Zhang
- Mayo Clinic Metabolomics Core, Mayo Clinic, Rochester, Minnesota; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Abner Aguirre Lopez
- Immunometabolism and Mucosal Immunity Laboratory, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Ahmed T Kurdi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Maria S Westphal
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Michelle M Gonzalez
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Joseph M Gaballa
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Hee Eun Lee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Thomas C Smyrk
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Glenn Bantug
- Immunobiology Laboratory, Department of Biomedicine, University Hospital of Basel, Basel, Switzerland
| | - Naomi M Gades
- Department of Comparative Medicine, Mayo Clinic, Scottsdale, Arizona
| | - William A Faubion
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
30
|
Wang Y, Wang Y, Wang S, Wang C, Tang Y, Zhang C, Yu D, Hou S, Lin N. Comprehensive analysis of CYBB as a prognostic marker and therapeutic target in glioma: A bioinformatics approach. Heliyon 2024; 10:e29549. [PMID: 38655339 PMCID: PMC11036048 DOI: 10.1016/j.heliyon.2024.e29549] [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: 07/19/2023] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Background In the central nervous system, glioma is the most common malignant tumor, and patients have a poor prognosis. Identification of novel marker genes and establishment of prognostic models are important for early diagnosis and prognosis determination. Methods Download glioma data from the CGGA and TCG databases. Application of bioinformatics to analyze the impact of CYBB on the clinicopathological characteristics, immunological features and prognosis of gliomas. Using single-cell sequencing data from 7 glioblastoma patients in the CGGA database, the role of CYBB in the tumor microenvironment was analyzed. In addition, a prognostic model was constructed based on CYBB high and low differentially expressed genes and mitochondrial genes. Results The expression of CYBB is closely related to various clinical features, immune cell infiltration level, immune checkpoint and survival time of patients. A 10-gene prediction model was constructed based on the differentially expressed genes of low and high CYBB and mitochondria-related genes. Glioma patients with higher risk scores had significantly lower survival probabilities. Receiver operating characteristic curves and nomograms were plotted over time to show the predictive accuracy and predictive value of the 10-gene prognostic model. Conclusions Our study shows that CYBB is strongly correlated with clinical characteristics features and prognosis of glioma patients, and can be used as a potential therapeutic target. Prognostic models based on CYBB and mitochondrial genes have good performance in predicting prognosis of glioma patients.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| | - Yuhao Wang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| | - Shuai Wang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| | - Chengcheng Wang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| | - Yuhang Tang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| | - Chao Zhang
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| | - Dong Yu
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| | - Shiqiang Hou
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| | - Ning Lin
- Department of Neurosurgery, The Affliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, 239000, China
| |
Collapse
|
31
|
Fries-Craft K, Bobeck EA. Coccidiosis and necrotic enteritis model may have a greater impact than dietary anti-interleukin-10 on broiler chicken systemic immunometabolic responses. Poult Sci 2024; 103:103551. [PMID: 38417332 PMCID: PMC10909892 DOI: 10.1016/j.psj.2024.103551] [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: 12/07/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 03/01/2024] Open
Abstract
Dietary egg yolk-derived anti-interleukin (IL)-10 may preserve broiler chicken performance during coccidiosis due to Eimeria spp. infection while effects on secondary Clostridium perfringens (necrotic enteritis) are unknown. Some necrotic enteritis models implement Salmonella Typhimurium to improve repeatability; however, Salmonella upregulation of IL-10 may be a confounder when evaluating anti-IL-10. The study objective was to investigate anti-IL-10 effects on systemic cytokine concentrations and immunometabolism during E. maxima ± C. perfringens challenge in models ± S. Typhimurium. Three 25 d replicate studies using Ross 308 chicks were conducted in wire-floor cages (32 cages/ replicate) with chicks assigned to diets ± 0.03% anti-IL-10. 640 chicks (20/ cage; replicates 1 and 2) were inoculated with sterile saline ± 1×108 colony forming units (CFU) S. Typhimurium while 480 chicks (15/ cage) were placed in replicate 3. In all replicates, blood samples were collected on d 14 (6 chicks/treatment) before administering 15,000 sporulated E. maxima M6 oocysts to S. Typhimurium-inoculated (replicates 1 and 2) or challenge-designated chicks (replicate 3). Half the E. maxima-challenged chicks received 1×108 CFU C. perfringens on d 18 and 19. Blood samples were collected at 1, 3, 7, and 11 d post-inoculation (dpi) with E. maxima and 1, 3, and 7 dpi with secondary C. perfringens. Plasma cytokines were determined by ELISA while immunometabolic assays evaluated peripheral blood mononuclear cell ATP production and glycolytic rate responses. Data were analyzed with diet and challenge fixed effects plus associated interactions (SAS 9.4; P ≤ 0.05). Replicates 1 and 2 showed few immunometabolic responses within 3 dpi with E. maxima, but 25 to 31% increased ATP production and 32% increased compensatory glycolysis at 1 dpi with C. perfringens in challenged vs. unchallenged chicks (P ≤ 0.04). In replicate 3, total ATP production and compensatory glycolysis were increased 25 and 40%, respectively, by the E. maxima main effect at 1dpi (P ≤ 0.05) with unobserved responsiveness to C. perfringens. These outcomes indicate that model type had greater impacts on systemic immunity than anti-IL-10.
Collapse
Affiliation(s)
- K Fries-Craft
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - E A Bobeck
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
32
|
Tsubouchi A, An Y, Kawamura Y, Yanagihashi Y, Nakayama H, Murata Y, Teranishi K, Ishiguro S, Aburatani H, Yachie N, Ota S. Pooled CRISPR screening of high-content cellular phenotypes using ghost cytometry. CELL REPORTS METHODS 2024; 4:100737. [PMID: 38531306 PMCID: PMC10985231 DOI: 10.1016/j.crmeth.2024.100737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/30/2023] [Accepted: 02/27/2024] [Indexed: 03/28/2024]
Abstract
Recent advancements in image-based pooled CRISPR screening have facilitated the mapping of diverse genotype-phenotype associations within mammalian cells. However, the rapid enrichment of cells based on morphological information continues to pose a challenge, constraining the capacity for large-scale gene perturbation screening across diverse high-content cellular phenotypes. In this study, we demonstrate the applicability of multimodal ghost cytometry-based cell sorting, including both fluorescent and label-free high-content phenotypes, for rapid pooled CRISPR screening within vast cell populations. Using the high-content cell sorter operating in fluorescence mode, we successfully executed kinase-specific CRISPR screening targeting genes influencing the nuclear translocation of RelA. Furthermore, using the multiparametric, label-free mode, we performed large-scale screening to identify genes involved in macrophage polarization. Notably, the label-free platform can enrich target phenotypes without requiring invasive staining, preserving untouched cells for downstream assays and expanding the potential for screening cellular phenotypes even when suitable markers are absent.
Collapse
Affiliation(s)
| | - Yuri An
- ThinkCyte Inc., Tokyo 113-8654, Japan
| | | | | | | | | | | | - Soh Ishiguro
- School of Biomedical Engineering, Faculty of Medicine and Faculty of Applied Science, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hiroyuki Aburatani
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
| | - Nozomu Yachie
- School of Biomedical Engineering, Faculty of Medicine and Faculty of Applied Science, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
| | - Sadao Ota
- ThinkCyte Inc., Tokyo 113-8654, Japan; Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan.
| |
Collapse
|
33
|
Wang SF, Chang YL, Liu TY, Huang KH, Fang WL, Li AFY, Yeh TS, Hung GY, Lee HC. Mitochondrial dysfunction decreases cisplatin sensitivity in gastric cancer cells through upregulation of integrated stress response and mitokine GDF15. FEBS J 2024; 291:1131-1150. [PMID: 37935441 DOI: 10.1111/febs.16992] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/18/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Gastric neoplasm is a high-mortality cancer worldwide. Chemoresistance is the obstacle against gastric cancer treatment. Mitochondrial dysfunction has been observed to promote malignant progression. However, the underlying mechanism is still unclear. The mitokine growth differentiation factor 15 (GDF15) is a significant biomarker for mitochondrial disorder and is activated by the integrated stress response (ISR) pathway. The serum level of GDF15 was found to be correlated with the poor prognosis of gastric cancer patients. In this study, we found that high GDF15 protein expression might increase disease recurrence in adjuvant chemotherapy-treated gastric cancer patients. Moreover, treatment with mitochondrial inhibitors, especially oligomycin (a complex V inhibitor) and salubrinal (an ISR activator), respectively, was found to upregulate GDF15 and enhance cisplatin insensitivity of human gastric cancer cells. Mechanistically, it was found that the activating transcription factor 4-C/EBP homologous protein pathway has a crucial function in the heightened manifestation of GDF15. In addition, reactive oxygen species-activated general control nonderepressible 2 mediates the oligomycin-induced ISR, and upregulates GDF15. The GDF15-glial cell-derived neurotrophic factor family receptor a-like-ISR-cystine/glutamate transporter-enhanced glutathione production was found to be involved in cisplatin resistance. These results suggest that mitochondrial dysfunction might enhance cisplatin insensitivity through GDF15 upregulation, and targeting mitokine GDF15-ISR regulation might be a strategy against cisplatin resistance of gastric cancer.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, Taiwan
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taiwan
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuh-Lih Chang
- Department of Pharmacy, Taipei Veterans General Hospital, Taiwan
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Yu Liu
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kuo-Hung Huang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan
- Department of Surgery, Gastric Cancer Medical Center, Taipei Veterans General Hospital, Taiwan
| | - Wen-Liang Fang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan
- Department of Surgery, Gastric Cancer Medical Center, Taipei Veterans General Hospital, Taiwan
| | - Anna Fen-Yau Li
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Anatomical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Tien-Shun Yeh
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Giun-Yi Hung
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Taipei Veterans General Hospital, Taiwan
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
34
|
Trinchese G, Cimmino F, Catapano A, Cavaliere G, Mollica MP. Mitochondria: the gatekeepers between metabolism and immunity. Front Immunol 2024; 15:1334006. [PMID: 38464536 PMCID: PMC10920337 DOI: 10.3389/fimmu.2024.1334006] [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: 11/06/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
Metabolism and immunity are crucial monitors of the whole-body homeodynamics. All cells require energy to perform their basic functions. One of the most important metabolic skills of the cell is the ability to optimally adapt metabolism according to demand or availability, known as metabolic flexibility. The immune cells, first line of host defense that circulate in the body and migrate between tissues, need to function also in environments in which nutrients are not always available. The resilience of immune cells consists precisely in their high adaptive capacity, a challenge that arises especially in the framework of sustained immune responses. Pubmed and Scopus databases were consulted to construct the extensive background explored in this review, from the Kennedy and Lehninger studies on mitochondrial biochemistry of the 1950s to the most recent findings on immunometabolism. In detail, we first focus on how metabolic reconfiguration influences the action steps of the immune system and modulates immune cell fate and function. Then, we highlighted the evidence for considering mitochondria, besides conventional cellular energy suppliers, as the powerhouses of immunometabolism. Finally, we explored the main immunometabolic hubs in the organism emphasizing in them the reciprocal impact between metabolic and immune components in both physiological and pathological conditions.
Collapse
Affiliation(s)
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
35
|
Wang C, Chen Q, Chen S, Fan L, Gan Z, Zhao M, Shi L, Bin P, Yang G, Zhou X, Ren W. Serine synthesis sustains macrophage IL-1β production via NAD +-dependent protein acetylation. Mol Cell 2024; 84:744-759.e6. [PMID: 38266638 DOI: 10.1016/j.molcel.2024.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/10/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024]
Abstract
Serine metabolism is involved in the fate decisions of immune cells; however, whether and how de novo serine synthesis shapes innate immune cell function remain unknown. Here, we first demonstrated that inflammatory macrophages have high expression of phosphoglycerate dehydrogenase (PHGDH, the rate-limiting enzyme of de novo serine synthesis) via nuclear factor κB signaling. Notably, the pharmacological inhibition or genetic modulation of PHGDH limits macrophage interleukin (IL)-1β production through NAD+ accumulation and subsequent NAD+-dependent SIRT1 and SIRT3 expression and activity. Mechanistically, PHGDH not only sustains IL-1β expression through H3K9/27 acetylation-mediated transcriptional activation of Toll-like receptor 4 but also supports IL-1β maturation via NLRP3-K21/22/24/ASC-K21/22/24 acetylation-mediated activation of the NLRP3 inflammasome. Moreover, mice with myeloid-specific depletion of Phgdh show alleviated inflammatory responses in lipopolysaccharide-induced systemic inflammation. This study reveals a network by which a metabolic enzyme, involved in de novo serine synthesis, mediates post-translational modifications and epigenetic regulation to orchestrate IL-1β production, providing a potential inflammatory disease target.
Collapse
Affiliation(s)
- Chuanlong Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qingyi Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siyuan Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Lijuan Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhending Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Muyang Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Lexuan Shi
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Peng Bin
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Guan Yang
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Xihong Zhou
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
36
|
Nakano H, Nakano A. The role of metabolism in cardiac development. Curr Top Dev Biol 2024; 156:201-243. [PMID: 38556424 DOI: 10.1016/bs.ctdb.2024.01.005] [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: 04/02/2024]
Abstract
Metabolism is the fundamental process that sustains life. The heart, in particular, is an organ of high energy demand, and its energy substrates have been studied for more than a century. In recent years, there has been a growing interest in understanding the role of metabolism in the early differentiation of pluripotent stem cells and in cancer research. Studies have revealed that metabolic intermediates from glycolysis and the tricarboxylic acid cycle act as co-factors for intracellular signal transduction, playing crucial roles in regulating cell behaviors. Mitochondria, as the central hub of metabolism, are also under intensive investigation regarding the regulation of their dynamics. The metabolic environment of the fetus is intricately linked to the maternal metabolic status, and the impact of the mother's nutrition and metabolic health on fetal development is significant. For instance, it is well known that maternal diabetes increases the risk of cardiac and nervous system malformations in the fetus. Another notable example is the decrease in the risk of neural tube defects when pregnant women are supplemented with folic acid. These examples highlight the profound influence of the maternal metabolic environment on the fetal organ development program. Therefore, gaining insights into the metabolic environment within developing fetal organs is critical for deepening our understanding of normal organ development. This review aims to summarize recent findings that build upon the historical recognition of the environmental and metabolic factors involved in the developing embryo.
Collapse
Affiliation(s)
- Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States; Cardiology Division, Department of Medicine, UCLA, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, United States; Molecular Biology Institute, UCLA, Los Angeles, CA, United States; Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
37
|
Xiong S, Zhang Y, Zeng J, Zhou J, Liu S, Wei P, Liu H, Yi F, Wan Z, Xiong L, Zhang B, Li J. DLP fabrication of HA scaffold with customized porous structures to regulate immune microenvironment and macrophage polarization for enhancing bone regeneration. Mater Today Bio 2024; 24:100929. [PMID: 38229884 PMCID: PMC10789648 DOI: 10.1016/j.mtbio.2023.100929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/24/2023] [Accepted: 12/23/2023] [Indexed: 01/18/2024] Open
Abstract
The immune microenvironment plays a pivotal role in osteoanagenesis. Biomaterials can modulate osteogenic efficacy by inducing specific local immune reactions. As 3D-printing technology advances, digital light projection printing has emerged as a promising method for creating large scale, high-precision biomaterial scaffolds. By adjusting the solid content and the sintering conditions during printing, the pore size of biomaterials can be meticulously controlled. Yet, the systematic influence of pore size on the immune microenvironment remains uncharted. We fabricated 3D-printed hydroxyapatite bioceramic scaffolds with three distinct pore sizes: 400 μm, 600 μm, and 800 μm. Our study revealed that scaffolds with a pore size of 600 μm promote macrophage M2 polarization, which is achieved by upregulating interferon-beta and HIF-1α production. When these materials were implanted subcutaneously in rats and within rabbit skulls, we observed that the 600 μm scaffolds notably improved the long-term inflammatory response, fostered vascular proliferation, and augmented new bone growth. This research paves the way for innovative therapeutic strategies for treating large segmental bone defects in clinical settings.
Collapse
Affiliation(s)
- Shilang Xiong
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Yinuo Zhang
- Department of Orthopedics, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Jianhua Zeng
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jingyu Zhou
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Shiwei Liu
- Department of Orthopedics, Ganzhou People's Hospital No.16, Mei Guan Road, Zhang Gong District, Ganzhou, Jiangxi, 341000, China
| | - Peng Wei
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Hantian Liu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Feng Yi
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zongmiao Wan
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Long Xiong
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Bin Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Jingtang Li
- Department of Traumatology, Jiangxi Provincial People's Hospital the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
38
|
Glaser KM, Doon-Ralls J, Walters N, Rima XY, Rambold AS, Réategui E, Lämmermann T. Arp2/3 complex and the pentose phosphate pathway regulate late phases of neutrophil swarming. iScience 2024; 27:108656. [PMID: 38205244 PMCID: PMC10777075 DOI: 10.1016/j.isci.2023.108656] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/29/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024] Open
Abstract
Neutrophil swarming is an essential process of the neutrophil response to many pathological conditions. Resultant neutrophil accumulations are hallmarks of acute tissue inflammation and infection, but little is known about their dynamic regulation. Technical limitations to spatiotemporally resolve individual cells in dense neutrophil clusters and manipulate these clusters in situ have hampered recent progress. We here adapted an in vitro swarming-on-a-chip platform for the use with confocal laser-scanning microscopy to unravel the complexity of single-cell responses during neutrophil crowding. Confocal sectioning allowed the live visualization of subcellular components, including mitochondria, cell membranes, cortical actin, and phagocytic cups, inside neutrophil clusters. Based on this experimental setup, we identify that chemical inhibition of the Arp2/3 complex causes cell death in crowding neutrophils. By visualizing spatiotemporal patterns of reactive oxygen species (ROS) production in developing neutrophil swarms, we further demonstrate a regulatory role of the metabolic pentose phosphate pathway for ROS production and neutrophil cluster growth.
Collapse
Affiliation(s)
- Katharina M. Glaser
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics and Metabolism (IMPRS-IEM), 79108 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Jacob Doon-Ralls
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Nicole Walters
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Xilal Y. Rima
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Angelika S. Rambold
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Eduardo Réategui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, 48149 Münster, Germany
| |
Collapse
|
39
|
Elhage R, Kelly M, Goudin N, Megret J, Legrand A, Nemazanyy I, Patitucci C, Quellec V, Wai T, Hamaï A, Ezine S. Mitochondrial dynamics and metabolic regulation control T cell fate in the thymus. Front Immunol 2024; 14:1270268. [PMID: 38288115 PMCID: PMC10822881 DOI: 10.3389/fimmu.2023.1270268] [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: 07/31/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
Several studies demonstrated that mitochondrial dynamics and metabolic pathways control T cell fate in the periphery. However, little is known about their implication in thymocyte development. Our results showed that thymic progenitors (CD3-CD4-CD8- triple negative, TN), in active division, have essentially a fused mitochondrial morphology and rely on high glycolysis and mitochondrial oxidative phosphorylation (OXPHOS). As TN cells differentiate to double positive (DP, CD4+CD8+) and single positive (SP, CD4+ and CD8+) stages, they became more quiescent, their mitochondria fragment and they downregulate glycolysis and OXPHOS. Accordingly, in vitro inhibition of the mitochondrial fission during progenitor differentiation on OP9-DL4 stroma, affected the TN to DP thymocyte transition by enhancing the percentage of TN and reducing that of DP, leading to a decrease in the total number of thymic cells including SP T cells. We demonstrated that the stage 3 triple negative pre-T (TN3) and the stage 4 triple negative pre-T (TN4) have different metabolic and functional behaviors. While their mitochondrial morphologies are both essentially fused, the LC-MS based analysis of their metabolome showed that they are distinct: TN3 rely more on OXPHOS whereas TN4 are more glycolytic. In line with this, TN4 display an increased Hexokinase II expression in comparison to TN3, associated with high proliferation and glycolysis. The in vivo inhibition of glycolysis using 2-deoxyglucose (2-DG) and the absence of IL-7 signaling, led to a decline in glucose metabolism and mitochondrial membrane potential. In addition, the glucose/IL-7R connection affects the TN3 to TN4 transition (also called β-selection transition), by enhancing the percentage of TN3, leading to a decrease in the total number of thymocytes. Thus, we identified additional components, essential during β-selection transition and playing a major role in thymic development.
Collapse
Affiliation(s)
- Rima Elhage
- Institut Necker Enfant-Malades (INEM), INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Mairead Kelly
- Institut Necker Enfant-Malades (INEM), INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Nicolas Goudin
- Platform for Image Analysis Center, SFR Necker, INSERM US 24 - CNRS UMS 3633, Paris, France
| | - Jérôme Megret
- Platform for Cytometry, SFR Necker, INSERM US 24 - CNRS UMS 3633, Paris, France
| | - Agnès Legrand
- Institut Necker Enfant-Malades (INEM), INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, SFR Necker, INSERM US 24 - CNRS UMS 3633, Paris, France
| | - Cécilia Patitucci
- Mitochondrial Biology Group, Institut Pasteur, CNRS UMR 3691, Paris, France
| | - Véronique Quellec
- Institut Necker Enfant-Malades (INEM), INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Timothy Wai
- Mitochondrial Biology Group, Institut Pasteur, CNRS UMR 3691, Paris, France
| | - Ahmed Hamaï
- Institut Necker Enfant-Malades (INEM), INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| | - Sophie Ezine
- Institut Necker Enfant-Malades (INEM), INSERM U1151-CNRS UMR 8253, Université de Paris, Paris, France
| |
Collapse
|
40
|
Jia C, Liu M, Yao L, Zhao F, Liu S, Li Z, Han Y. Multi-omics analysis reveals cuproptosis and mitochondria-based signature for assessing prognosis and immune landscape in osteosarcoma. Front Immunol 2024; 14:1280945. [PMID: 38250070 PMCID: PMC10796547 DOI: 10.3389/fimmu.2023.1280945] [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: 08/21/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
Background Osteosarcoma (OSA), the most common primary mesenchymal bone tumor, is a health threat to children and adolescents with a dismal prognosis. While cuproptosis and mitochondria dysfunction have been demonstrated to exert a crucial role in tumor progression and development, the mechanisms by which they are regulated in OSA still await clarification. Methods Two independent OSA cohorts containing transcriptome data and clinical information were collected from public databases. The heterogeneity of OSA were evaluated by single cell RNA (scRNA) analysis. To identify a newly molecular subtype, unsupervised consensus clustering was conducted. Cox relevant regression methods were utilized to establish a prognostic gene signature. Wet lab experiments were performed to confirm the effect of model gene in OSA cells. Results We determined 30 distinct cell clusters and assessed OSA heterogeneity and stemness scRNA analysis. Then, univariate Cox analysis identified 24 candidate genes which were greatly associated with the prognosis of OSA. Based on these prognostic genes, we obtained two molecular subgroups. After conducting step Cox regression, three model genes were selected to construct a signature showing a favorable performance to forecast clinical outcome. Our proposed signature could also evaluate the response to chemotherapy and immunotherapy of OSA cases. Conclusion We generated a novel risk model based on cuproptosis and mitochondria-related genes in OSA with powerful predictive ability in prognosis and immune landscape.
Collapse
Affiliation(s)
- Chenguang Jia
- Department of Osteonecrosis and Hip Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Orthopedics, Hebei Chest Hospital, Shijiazhuang, China
| | - Mei Liu
- Molecular Biology Laboratory, Hebei Chest Hospital, Shijiazhuang, China
| | - Liming Yao
- Department of Orthopedics, Hebei Chest Hospital, Shijiazhuang, China
| | - Fangchao Zhao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuren Liu
- Department of Orthopedics, Hebei Chest Hospital, Shijiazhuang, China
| | - Zhuo Li
- Department of Orthopedics, Hebei Chest Hospital, Shijiazhuang, China
| | - Yongtai Han
- Department of Osteonecrosis and Hip Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
41
|
Liu H, Xu K, He Y, Huang F. Mitochondria in Multi-Directional Differentiation of Dental-Derived Mesenchymal Stem Cells. Biomolecules 2023; 14:12. [PMID: 38275753 PMCID: PMC10813276 DOI: 10.3390/biom14010012] [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: 10/18/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The pursuit of tissue regeneration has fueled decades of research in regenerative medicine. Among the numerous types of mesenchymal stem cells (MSCs), dental-derived mesenchymal stem cells (DMSCs) have recently emerged as a particularly promising candidate for tissue repair and regeneration. In recent years, evidence has highlighted the pivotal role of mitochondria in directing and orchestrating the differentiation processes of DMSCs. Beyond mitochondrial energy metabolism, the multifaceted functions of mitochondria are governed by the mitochondrial quality control (MQC) system, encompassing biogenesis, autophagy, and dynamics. Notably, mitochondrial energy metabolism not only governs the decision to differentiate but also exerts a substantial influence on the determination of differentiation directions. Furthermore, the MQC system exerts a nuanced impact on the differentiation of DMSCs by finely regulating the quality and mass of mitochondria. The review aims to provide a comprehensive overview of the regulatory mechanisms governing the multi-directional differentiation of DMSCs, mediated by both mitochondrial energy metabolism and the MQC system. We also focus on a new idea based on the analysis of data from many research groups never considered before, namely, DMSC-based regenerative medicine applications.
Collapse
Affiliation(s)
| | | | - Yifan He
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510000, China; (H.L.); (K.X.)
| | - Fang Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510000, China; (H.L.); (K.X.)
| |
Collapse
|
42
|
Zhang G, Jiang P, Tang W, Wang Y, Qiu F, An J, Zheng Y, Wu D, Zhou J, Neculai D, Shi Y, Sheng W. CPT1A induction following epigenetic perturbation promotes MAVS palmitoylation and activation to potentiate antitumor immunity. Mol Cell 2023; 83:4370-4385.e9. [PMID: 38016475 DOI: 10.1016/j.molcel.2023.10.043] [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: 03/14/2023] [Revised: 07/31/2023] [Accepted: 10/31/2023] [Indexed: 11/30/2023]
Abstract
Targeting epigenetic regulators to potentiate anti-PD-1 immunotherapy converges on the activation of type I interferon (IFN-I) response, mimicking cellular response to viral infection, but how its strength and duration are regulated to impact combination therapy efficacy remains largely unknown. Here, we show that mitochondrial CPT1A downregulation following viral infection restrains, while its induction by epigenetic perturbations sustains, a double-stranded RNA-activated IFN-I response. Mechanistically, CPT1A recruits the endoplasmic reticulum-localized ZDHHC4 to catalyze MAVS Cys79-palmitoylation, which promotes MAVS stabilization and activation by inhibiting K48- but facilitating K63-linked ubiquitination. Further elevation of CPT1A incrementally increases MAVS palmitoylation and amplifies the IFN-I response, which enhances control of viral infection and epigenetic perturbation-induced antitumor immunity. Moreover, CPT1A chemical inducers augment the therapeutic effect of combined epigenetic treatment with PD-1 blockade in refractory tumors. Our study identifies CPT1A as a stabilizer of MAVS activation, and its link to epigenetic perturbation can be exploited for cancer immunotherapy.
Collapse
Affiliation(s)
- Guiheng Zhang
- Institute of Immunology, and Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, Zhejiang, China
| | - Peishan Jiang
- Institute of Immunology, and Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, Zhejiang, China
| | - Wen Tang
- Department of Human Anatomy, Histology and Embryology, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yunyi Wang
- Ludwig Institute for Cancer Research, University of Oxford, Oxford OX3 7DQ, UK
| | - Fengqi Qiu
- Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Jie An
- Institute of Immunology, and Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, Zhejiang, China
| | - Yuping Zheng
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, Zhejiang, China
| | - Dandan Wu
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, Zhejiang, China
| | - Jianya Zhou
- Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Dante Neculai
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, Zhejiang, China
| | - Yang Shi
- Ludwig Institute for Cancer Research, University of Oxford, Oxford OX3 7DQ, UK.
| | - Wanqiang Sheng
- Institute of Immunology, and Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, Zhejiang, China.
| |
Collapse
|
43
|
Chen X, He Q, Zhai Q, Tang H, Li D, Zhu X, Zheng X, Jian G, Cannon RD, Mei L, Wang S, Ji P, Song J, Chen T. Adaptive Nanoparticle-Mediated Modulation of Mitochondrial Homeostasis and Inflammation to Enhance Infected Bone Defect Healing. ACS NANO 2023; 17:22960-22978. [PMID: 37930276 DOI: 10.1021/acsnano.3c08165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Infected bone defects (IBDs) exhibit impaired healing due to excessive inflammation triggered by pathogen-associated molecular patterns (PAMPs) from bacteria. As a vital factor in orchestrating immune responses, mitochondrial homeostasis maintenance is central to inflammation blockade. This research developed a chameleon-like nanoplatform by covering hydroxyapatite nanoparticles with a cerium ion coordinated tannic acid supramolecular network (HA@Ce-TA), which adaptively functions to regulate mitochondrial homeostasis based on intra- and extracellular environments. Extracellularly, acidic conditions activate HA@Ce-TA's peroxidase/oxidase-mimicking activity to produce reactive oxygen species (ROS), and external near-infrared (NIR) irradiation excites nanoscale Ce-TA to produce hyperthermia, which is found and explained by chemical computation. ROS production with photothermal therapy can eliminate bacteria effectively and reduce mitochondrial stress. Intracellularly, HA@Ce-TA remodels mitochondrial dynamics by upregulating mitochondrial fusion genes and eliminates excessive ROS by mimicking superoxidase/catalase. Consequently, this comprehensive modulation of mitochondrial homeostasis inhibits inflammasome overactivation. In vitro and in vivo studies showed HA@Ce-TA can modulate the mitochondria-centered inflammatory cascade to enhance IBD treatment, highlighting the potential of engineering nanotherapeutics to recalibrate mitochondrial homeostasis as an infected disease-modifying intervention.
Collapse
Affiliation(s)
- Xu Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Qingqing He
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Qiming Zhai
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Han Tang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Dize Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Xingyu Zhu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Xinhui Zheng
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Guangyu Jian
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Richard D Cannon
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9054, New Zealand
| | - Li Mei
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9054, New Zealand
| | - Shan Wang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Ping Ji
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| | - Tao Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, P. R. China
| |
Collapse
|
44
|
Guo J, Zhang H, Lin W, Lu L, Su J, Chen X. Signaling pathways and targeted therapies for psoriasis. Signal Transduct Target Ther 2023; 8:437. [PMID: 38008779 PMCID: PMC10679229 DOI: 10.1038/s41392-023-01655-6] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 11/28/2023] Open
Abstract
Psoriasis is a common, chronic, and inflammatory skin disease with a high burden on individuals, health systems, and society worldwide. With the immunological pathologies and pathogenesis of psoriasis becoming gradually revealed, the therapeutic approaches for this disease have gained revolutionary progress. Nevertheless, the mechanisms of less common forms of psoriasis remain elusive. Furthermore, severe adverse effects and the recurrence of disease upon treatment cessation should be noted and addressed during the treatment, which, however, has been rarely explored with the integration of preliminary findings. Therefore, it is crucial to have a comprehensive understanding of the mechanisms behind psoriasis pathogenesis, which might offer new insights for research and lead to more substantive progress in therapeutic approaches and expand clinical options for psoriasis treatment. In this review, we looked to briefly introduce the epidemiology, clinical subtypes, pathophysiology, and comorbidities of psoriasis and systematically discuss the signaling pathways involving extracellular cytokines and intracellular transmission, as well as the cross-talk between them. In the discussion, we also paid more attention to the potential metabolic and epigenetic mechanisms of psoriasis and the molecular mechanistic cascades related to its comorbidities. This review also outlined current treatment for psoriasis, especially targeted therapies and novel therapeutic strategies, as well as the potential mechanism of disease recurrence.
Collapse
Affiliation(s)
- Jia Guo
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Hanyi Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Wenrui Lin
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Lixia Lu
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China.
| |
Collapse
|
45
|
Ninfali C, Cortés M, Martínez-Campanario MC, Domínguez V, Han L, Tobías E, Esteve-Codina A, Enrich C, Pintado B, Garrabou G, Postigo A. The adaptive antioxidant response during fasting-induced muscle atrophy is oppositely regulated by ZEB1 and ZEB2. Proc Natl Acad Sci U S A 2023; 120:e2301120120. [PMID: 37948583 PMCID: PMC10655555 DOI: 10.1073/pnas.2301120120] [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: 01/20/2023] [Accepted: 09/26/2023] [Indexed: 11/12/2023] Open
Abstract
Reactive oxygen species (ROS) serve important homeostatic functions but must be constantly neutralized by an adaptive antioxidant response to prevent supraphysiological levels of ROS from causing oxidative damage to cellular components. Here, we report that the cellular plasticity transcription factors ZEB1 and ZEB2 modulate in opposing directions the adaptive antioxidant response to fasting in skeletal muscle. Using transgenic mice in which Zeb1 or Zeb2 were specifically deleted in skeletal myofibers, we show that in fasted mice, the deletion of Zeb1, but not Zeb2, increased ROS production and that the adaptive antioxidant response to fasting essentially requires ZEB1 and is inhibited by ZEB2. ZEB1 expression increased in fasted muscles and protected them from atrophy; conversely, ZEB2 expression in muscles decreased during fasting and exacerbated muscle atrophy. In fasted muscles, ZEB1 reduces mitochondrial damage and increases mitochondrial respiratory activity; meanwhile, ZEB2 did the opposite. Treatment of fasting mice with Zeb1-deficient myofibers with the antioxidant triterpenoid 1[2-cyano-3,12-dioxool-eana-1,9(11)-dien-28-oyl] trifluoro-ethylamide (CDDO-TFEA) completely reversed their altered phenotype to that observed in fasted control mice. These results set ZEB factors as potential therapeutic targets to modulate the adaptive antioxidant response in physiopathological conditions and diseases caused by redox imbalance.
Collapse
Affiliation(s)
- Chiara Ninfali
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Marlies Cortés
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - M. C. Martínez-Campanario
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Verónica Domínguez
- National Center of Biotechnology (CSIC-CNB) and Center for Molecular Biology Severo Ochoa (CSIC-CBMSO), Transgenesis Facility, High Research Council (CSIC) and Autonomous University of Madrid, Cantoblanco, Madrid28049, Spain
| | - Lu Han
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Ester Tobías
- Group of Muscle Research and Mitochondrial Function, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), University of Barcelona School of Medicine, Hospital Clínic of Barcelona, and Rare Diseases Networking Biomedical Research Center (CIBERer), Barcelona08036, Spain
| | | | - Carlos Enrich
- Department of Biomedicine, University of Barcelona School of Medicine, and Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Belén Pintado
- National Center of Biotechnology (CSIC-CNB) and Center for Molecular Biology Severo Ochoa (CSIC-CBMSO), Transgenesis Facility, High Research Council (CSIC) and Autonomous University of Madrid, Cantoblanco, Madrid28049, Spain
| | - Gloria Garrabou
- Group of Muscle Research and Mitochondrial Function, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), University of Barcelona School of Medicine, Hospital Clínic of Barcelona, and Rare Diseases Networking Biomedical Research Center (CIBERer), Barcelona08036, Spain
| | - Antonio Postigo
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
- Molecular Targets Program, Department of Medicine, James Graham Brown Cancer Center, Louisville, KY40202
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona08010, Spain
| |
Collapse
|
46
|
Yadav S, Ganta V, Sudhahar V, Ash D, Nagarkoti S, Das A, McMenamin M, Kelley S, Fukai T, Ushio-Fukai M. Myeloid Drp1 Deficiency Limits Revascularization in Ischemic Muscles via Inflammatory Macrophage Polarization and Metabolic Reprograming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.04.565656. [PMID: 37961122 PMCID: PMC10635146 DOI: 10.1101/2023.11.04.565656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In the preclinical model of peripheral arterial disease (PAD), M2-like anti-inflammatory macrophage polarization and angiogenesis are required for revascularization. The regulation of cell metabolism and inflammation in macrophages is tightly linked to mitochondrial dynamics. Drp1, a mitochondrial fission protein, has shown context-dependent macrophage phenotypes with both pro- and anti-inflammatory characteristics. However, the role of macrophage Drp1 in reparative neovascularization remains unexplored. Here we show that Drp1 expression was significantly increased in F4/80+ macrophages within ischemic muscle at day 3 following hindlimb ischemia (HLI), an animal model of PAD. Myeloid-specific Drp1 -/- mice exhibited reduced limb perfusion recovery, angiogenesis and muscle regeneration after HLI. These effects were concomitant with enhancement of pro-inflammatory M1-like macrophages, p-NFkB, and TNFα levels, while showing reduction in anti-inflammatory M2-like macrophages and p-AMPK in ischemic muscle of myeloid Drp1 -/- mice. In vitro, Drp1 -/- macrophages under hypoxia serum starvation (HSS), an in vitro PAD model, demonstrated enhanced glycolysis via reducing p-AMPK as well as mitochondrial dysfunction and excessive mitochondrial ROS, resulting in increased M1-gene and reduced M2-gene expression. Conditioned media from HSS-treated Drp1 -/- macrophages exhibited increased secretion of pro-inflammatory cytokines and suppressed angiogenic responses in cultured endothelial cells. Thus, Drp1 deficiency in macrophages under ischemia drives inflammatory metabolic reprogramming and macrophage polarization, thereby limiting revascularization in experimental PAD.
Collapse
|
47
|
Fries-Craft K, Graham D, Hargis BM, Bobeck EA. Evaluating a Salmonella Typhimurium, Eimeria maxima, and Clostridium perfringens coinfection necrotic enteritis model in broiler chickens: repeatability, dosing, and immune outcomes. Poult Sci 2023; 102:103018. [PMID: 37651774 PMCID: PMC10480656 DOI: 10.1016/j.psj.2023.103018] [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: 06/06/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/02/2023] Open
Abstract
Coccidiosis and necrotic enteritis negatively impact poultry production, making challenge model repeatability important for evaluating mitigation strategies. Study objectives were: 1) evaluate Salmonella Typhimurium-Eimeria maxima-Clostridium perfringens necrotic enteritis coinfection model repeatability and 2) investigate E. maxima dose and early S. Typhimurium challenge on immune responses. Three trials using Ross 308 chicks assigned to 12 cages/trial (7 chicks/cage) in wire-floor brooders were completed. Trials 1 and 2 determined E. maxima dose for necrotic enteritis challenge in trial 3. Chicks in trials 1 and 2 were inoculated with 0 (control), 5, 15, or 25,000 sporulated E. maxima M6 oocysts on d 14 and jejunal lesion scores evaluated on d 20. In trial 3, chicks were assigned to control or necrotic enteritis challenge (42 chicks/group). Necrotic enteritis challenge chicks were inoculated with 1 × 105 colony forming units (CFU) S. Typhimurium on d 1, 15,000 E. maxima oocysts on d 14, and 1 × 108 CFU C. perfringens on d 19 and 20 with lesion scoring on d 22. Bird and feeder weights were recorded throughout each trial. Peripheral blood mononuclear cells (PBMC) were isolated from 1 chick/cage at baseline (all trials), 4 chicks/dose (trials 1 and 2) or 8 chicks/challenge (trial 3) 24 h post-inoculation (pi) with E. maxima for immunometabolic assays and immune cell profiling. Data were analyzed by mixed procedure (SAS 9.4) with challenge and timepoint fixed effects (P ≤ 0.05, trends 0.05 ≤ P ≤ 0.01). Inoculating chicks with 15,000 E. maxima oocysts increased d 14 to 20 FCR 79 points (trials 1 and 2; P = 0.009) vs. unchallenged chicks and achieved a target lesion score of 2. While C. perfringens challenge reduced trial 3 performance, average lesion scores were <1. Salmonella inoculation on d 1 tended to increase PBMC ATP production 41.6% 24 hpi with E. maxima vs. chicks challenged with E. maxima only (P = 0.06). These results provide insight for future model optimization and considerations regarding S. Typhimurium's effect on E. maxima immune response timelines.
Collapse
Affiliation(s)
- K Fries-Craft
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - D Graham
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - B M Hargis
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - E A Bobeck
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
48
|
Yi X, Chang ML, Zhou ZD, Yi L, Yuan H, Qi J, Yi L, Huan JN, Huang XQ. LPS induces SGPP2 to participate metabolic reprogramming in endothelial cells. Free Radic Biol Med 2023; 208:780-793. [PMID: 37703934 DOI: 10.1016/j.freeradbiomed.2023.09.007] [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] [Received: 12/29/2022] [Revised: 08/12/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
Sepsis often causes organ dysfunction and is manifested in increased endothelial cell permeability in blood vessels. Early-stage inflammation is accompanied by metabolic changes, but it is unclear how the metabolic alterations in the endothelial cells following lipopolysaccharide (LPS) stimulation affect endothelial cell function. In this study, the effects of 1 μg/ml of LPS on the metabolism of human umbilical vein endothelial cells (HUVECs) were investigated, and the metabolic changes after LPS stimulation were explained from the perspective of mRNA expression, chromatin openness and metabolic flux. We found changes in the central metabolism of endothelial cells after LPS stimulation, such as enhanced glycolysis function, decreased mitochondrial membrane potential, and increased production of reactive oxygen species (ROS). Sphingolipid metabolic pathways change at the transcriptome level, and sphingosine-1-phosphatase 2 (SGPP2) was upregulated in LPS-stimulated endothelial cells and zebrafish models. Overexpression of SGPP2 improved cell barrier function, enhanced mitochondrial respiration capacity, but also produced oxidative respiration chain uncoupling. In addition, SGPP2 overexpression inhibited the degradation of HIF-1α protein. The molecular and biochemical processes identified in this study are not only beneficial for understanding the metabolic-related mechanisms of LPS-induced endothelial injury, but also for the discovery of general therapeutic targets for inflammation and inflammation-related diseases.
Collapse
Affiliation(s)
- Xin Yi
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng-Ling Chang
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeng-Ding Zhou
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Yi
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Yuan
- Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Qi
- Department of Orthopedics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Yi
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jing-Ning Huan
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiao-Qin Huang
- Department of Burn, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
49
|
Maes ME, Colombo G, Schoot Uiterkamp FE, Sternberg F, Venturino A, Pohl EE, Siegert S. Mitochondrial network adaptations of microglia reveal sex-specific stress response after injury and UCP2 knockout. iScience 2023; 26:107780. [PMID: 37731609 PMCID: PMC10507162 DOI: 10.1016/j.isci.2023.107780] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/10/2023] [Accepted: 08/28/2023] [Indexed: 09/22/2023] Open
Abstract
Mitochondrial networks remodel their connectivity, content, and subcellular localization to support optimized energy production in conditions of increased environmental or cellular stress. Microglia rely on mitochondria to respond to these stressors, however our knowledge about mitochondrial networks and their adaptations in microglia in vivo is limited. Here, we generate a mouse model that selectively labels mitochondria in microglia. We identify that mitochondrial networks are more fragmented with increased content and perinuclear localization in vitro vs. in vivo. Mitochondrial networks adapt similarly in microglia closest to the injury site after optic nerve crush. Preventing microglial UCP2 increase after injury by selective knockout induces cellular stress. This results in mitochondrial hyperfusion in male microglia, a phenotype absent in females due to circulating estrogens. Our results establish the foundation for mitochondrial network analysis of microglia in vivo, emphasizing the importance of mitochondrial-based sex effects of microglia in other pathologies.
Collapse
Affiliation(s)
- Margaret E. Maes
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Gloria Colombo
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | | | - Felix Sternberg
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Alessandro Venturino
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Elena E. Pohl
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Sandra Siegert
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| |
Collapse
|
50
|
Socodato R, Rodrigues-Santos A, Tedim-Moreira J, Almeida TO, Canedo T, Portugal CC, Relvas JB. RhoA balances microglial reactivity and survival during neuroinflammation. Cell Death Dis 2023; 14:690. [PMID: 37863874 PMCID: PMC10589285 DOI: 10.1038/s41419-023-06217-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 09/29/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Microglia are the largest myeloid cell population in the brain. During injury, disease, or inflammation, microglia adopt different functional states primarily involved in restoring brain homeostasis. However, sustained or exacerbated microglia inflammatory reactivity can lead to brain damage. Dynamic cytoskeleton reorganization correlates with alterations of microglial reactivity driven by external cues, and proteins controlling cytoskeletal reorganization, such as the Rho GTPase RhoA, are well positioned to refine or adjust the functional state of the microglia during injury, disease, or inflammation. Here, we use multi-biosensor-based live-cell imaging approaches and tissue-specific conditional gene ablation in mice to understand the role of RhoA in microglial response to inflammation. We found that a decrease in RhoA activity is an absolute requirement for microglial metabolic reprogramming and reactivity to inflammation. However, without RhoA, inflammation disrupts Ca2+ and pH homeostasis, dampening mitochondrial function, worsening microglial necrosis, and triggering microglial apoptosis. Our results suggest that a minimum level of RhoA activity is obligatory to concatenate microglia inflammatory reactivity and survival during neuroinflammation.
Collapse
Affiliation(s)
- Renato Socodato
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
| | - Artur Rodrigues-Santos
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Joana Tedim-Moreira
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Tiago O Almeida
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- ICBAS - School of Medicine and Biomedical Sciences, Porto, Portugal
| | - Teresa Canedo
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Camila C Portugal
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - João B Relvas
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.
| |
Collapse
|