1
|
Curran CS, Cui X, Li Y, Gamble T, Sun J, Minkove S, Livinski AA, Eichacker PQ, Torabi-Parizi P. Meta-analysis of niacin and NAD metabolite treatment in infectious disease animal studies suggests benefit but requires confirmation in clinically relevant models. Sci Rep 2025; 15:12621. [PMID: 40221506 PMCID: PMC11993703 DOI: 10.1038/s41598-025-95735-y] [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: 10/20/2024] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Disruption of nicotinamide adenine dinucleotide (NAD) biosynthesis and function during infection may impair host defenses and aggravate inflammatory and oxidative organ injury. Increasingly, studies are investigating whether niacin or NAD metabolite treatment is beneficial in infection and sepsis animal models. We examined whether this preclinical experience supports clinical trials. A systematic review of three data bases was conducted through 2/29/2024 and a meta-analysis was performed comparing niacin or NAD metabolite treatment to control in adult animal models employing microbial challenges. Fifty-six studies met inclusion criteria, with 24 published after 2019. Most studies employed mouse (n = 40 studies) or rat (n = 12) models and administered either a bacterial toxin (n = 28) or bacterial (n = 19) challenge. Four and three studies employed viral or fungal challenges respectively. Studies investigated an NAD metabolite alone (n = 44), niacin alone (n = 9), or both (n = 3), usually administered before or within 24h after challenge (n = 50). Only three and four studies included standard antimicrobial support or started treatment > 24h after challenge respectively. In similar patterns with differing animal types (p ≥ 0.06), compared to control across those studies investigating the parameter, niacin or NAD treatment decreased the odds ratio of mortality [95% confidence interval (CI)] [0.28 (0.17, 0.49)] and in blood or tissue increased antioxidant levels [standardized mean differences (95%CI)] (SMD) [3.61 (2.20,5.02)] and decreased levels of microbes [- 2.44 (- 3.34, - 1.55)], histologic and permeability organ injury scoring [- 1.62 (- 2.27, - 0.98) and - 1.31(- 1.77, - 0.86) respectively], levels of TNFα, IL-6 and IL-1β [- 2.47 (- 3.30, - 1.64), - 3.17 (- 4.74, - 1.60) and - 8.44 (- 12.4, - 4.5) respectively] and myeloperoxidase (MPO) [- 1.60 (- 2.06, - 1.14)], although with significant, primarily quantitative heterogeneity for each (I2 ≥ 53%, p < 0.01) except MPO. Treatment increased blood or tissue NAD+ levels and decreased chemical organ injury measures and oxidation markers but differently comparing species (p ≤ 0.05). Only 2 and 9 survival studies described power analyses or animal randomization respectively and no study described treatment or non-histologic outcome measure blinding. Among survival studies, Egger's analysis (p = 0.002) suggested publication bias. While suggestive, published animal studies do not yet support clinical trials testing niacin and NAD metabolite treatment for infection and sepsis. Animal studies simulating clinical conditions and with randomized, blinded designs are needed to investigate this potentially promising therapeutic approach.
Collapse
Affiliation(s)
- Colleen S Curran
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tom Gamble
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Samuel Minkove
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Alicia A Livinski
- National Institutes of Health Library, Office of Research Services, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Peter Q Eichacker
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Parizad Torabi-Parizi
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10, Room 2C138, Bethesda, USA.
| |
Collapse
|
2
|
Jelcic I, Naghavian R, Fanaswala I, Macnair W, Esposito C, Calini D, Han Y, Marti Z, Raposo C, Sarabia Del Castillo J, Oldrati P, Erny D, Kana V, Zheleznyakova G, Al Nimer F, Tackenberg B, Reichen I, Khademi M, Piehl F, Robinson MD, Jelcic I, Sospedra M, Pelkmans L, Malhotra D, Reynolds R, Jagodic M, Martin R. T-bet+ CXCR3+ B cells drive hyperreactive B-T cell interactions in multiple sclerosis. Cell Rep Med 2025; 6:102027. [PMID: 40107244 PMCID: PMC11970401 DOI: 10.1016/j.xcrm.2025.102027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 05/16/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS). Self-peptide-dependent autoproliferation (AP) of B and T cells is a key mechanism in MS. Here, we show that pro-inflammatory B-T cell-enriched cell clusters (BTECs) form during AP and mirror features of a germinal center reaction. T-bet+CXCR3+ B cells are the main cell subset amplifying and sustaining their counterpart Th1 cells via interferon (IFN)-γ and are present in highly inflamed meningeal tissue. The underlying B cell activation signature is reflected by epigenetic modifications and receptor-ligand interactions with self-reactive T cells. AP+ CXCR3+ B cells show marked clonal evolution from memory to somatically hypermutated plasmablasts and upregulation of IFN-γ-related genes. Our data underscore a key role of T-bet+CXCR3+ B cells in the pathogenesis of MS in both the peripheral immune system and the CNS compartment, and thus they appear to be involved in both early relapsing-remitting disease and the chronic stage.
Collapse
Affiliation(s)
- Ivan Jelcic
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Reza Naghavian
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Imran Fanaswala
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Will Macnair
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Cinzia Esposito
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Daniela Calini
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Yanan Han
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Zoe Marti
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Catarina Raposo
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Pietro Oldrati
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Daniel Erny
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Veronika Kana
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Galina Zheleznyakova
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Faiez Al Nimer
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Björn Tackenberg
- Product Development Medical Affairs, Neuroscience and Rare Disease, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Ina Reichen
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mohsen Khademi
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mark D Robinson
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Ilijas Jelcic
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Lucas Pelkmans
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Dheeraj Malhotra
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Roland Martin
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland; Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden; Cellerys AG, Schlieren, Switzerland.
| |
Collapse
|
3
|
Wu J, Ji K, Kang G, Zhang M, Wang J, Wang L, Gao M, Jia X, Lu X, Wang Y, Gao X, Guo Y, Zhu Z, Wang Q, Zhao Z, Liu Q, Huang H. Butyrate-engineered yeast activates Nppa and Sgcg genes and reduces radiation-induced heart damage via the gut-heart axis. Pharmacol Res 2025; 213:107642. [PMID: 39909125 DOI: 10.1016/j.phrs.2025.107642] [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: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Radiotherapy is a method of treating cancer through radiation aimed at killing cancer cells or inhibiting their growth. However, radiotherapy has numerous side effects because it kills tumors while causing damage to normal cells or tissues. The literature shows that radiation can cause damage to heart tissue. This study found that engineered yeast that produced butyrate can maintain small intestinal barrier function by recovering GPR109A to reduce intestinal damage caused by abdominal irradiation in mice. We unexpectedly found that engineered yeast could mitigate irradiation-induced heart damage via the gut-heart axis. Mechanistically, engineered yeast enhanced taurine and nicotinamide metabolism by increasing the relative abundance of Akkermansia and Lachnospiraceae_NK4A136; then, yeast modulated cardiac function by activating the Sgcg and Nppa genes to attenuate cardiac damage induced by abdominal irradiation. Finally, we confirmed that engineered yeast mitigated cardiac damage caused by total body irradiation, which protected other vital organs through the intestinal tract. This study has a profound impact on cancer treatment, the emergence of engineered yeast will alleviate radiotherapy side effects and benefit patients.
Collapse
Affiliation(s)
- Jiahao Wu
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Kaihua Ji
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Guangbo Kang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Manman Zhang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Jigang Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lina Wang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Mengxue Gao
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Xiaoxiao Jia
- Department of Anatomy, Shandong Second Medical University, Weifang 261053, China
| | - Xinran Lu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Yan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Xinran Gao
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Yufei Guo
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Zhixin Zhu
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Qinghua Wang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Zhenyu Zhao
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China; School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| | - He Huang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China.
| |
Collapse
|
4
|
Zhang L, Wang L, Xu Z, Zhang X, Guan S, Liu Z, Gu S, Zhao L, Bai W, Li T, Zhao X. eNAMPT/Ac-STAT3/DIRAS2 Axis Promotes Development and Cancer Stemness in Triple-Negative Breast Cancer by Enhancing Cytokine Crosstalk Between Tumor-Associated Macrophages and Cancer Cells. Int J Biol Sci 2025; 21:2027-2047. [PMID: 40083697 PMCID: PMC11900821 DOI: 10.7150/ijbs.103723] [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: 09/15/2024] [Accepted: 02/03/2025] [Indexed: 03/16/2025] Open
Abstract
The intricate relationship between tumor-associated macrophages (TAMs) and cancer cells is pivotal for carcinogenesis, with TAMs being integral to the tumor microenvironment (TME). This study explores the novel mechanisms by which TAMs regulate the progression of triple-negative breast cancer (TNBC) within the TME. Using a co-culture system and methodologies such as cytokine arrays, proteomics, and CRISPR-Cas9, we investigated the crosstalk between TAMs and TNBC cells. We found that high levels of CD163+ TAMs in TNBC tissues correlate with poor prognosis. TNBC cell-conditioned medium induces macrophage polarization towards the M2 phenotype, enhancing TNBC cell migration, invasion, and stemness through the secretion of extracellular nicotinamide phosphoribosyltransferase (eNAMPT). eNAMPT binding to CCR5 on TNBC cells activates STAT3, leading to the downregulation of the tumor suppressor DIRAS2 and an increase in CCL2, which promotes a macrophage recruitment loop. Intervention at the eNAMPT/CCR5 or CCL2 level disrupts this loop, mitigating TAM-induced effects. Our findings uncover a cytokine communication mechanism between immune and cancer cells, suggesting potential targets for TNBC detection and treatment.
Collapse
Affiliation(s)
- Lifen Zhang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Lu Wang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ziyao Xu
- Senior Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xingmei Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Shaoyu Guan
- Pharmaceutical Sciences Research Division, Department of Pharmacy, Medical Supplies Centre of PLA General Hospital, Beijing 100853, China
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Shanzhi Gu
- Department of Forensic Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Lin Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Weichao Bai
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Xinhan Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
5
|
Zubiaur M, Terrón-Camero LC, Gordillo-González F, Andrés-León E, Barroso-del Jesús A, Canet-Antequera LM, Pérez Sánchez-Cañete MM, Martínez-Blanco Á, Domínguez-Pantoja M, Botia-Sánchez M, Pérez-Cabrera S, Bello-Iglesias N, Alcina A, Abadía-Molina AC, Matesanz F, Zumaquero E, Merino R, Sancho J. CD38 deficiency leads to a defective short-lived transcriptomic response to chronic graft-versus-host disease induction, involving purinergic signaling-related genes and distinct transcriptomic signatures associated with lupus. Front Immunol 2025; 16:1441981. [PMID: 39995666 PMCID: PMC11847871 DOI: 10.3389/fimmu.2025.1441981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
This study aimed to elucidate the transcriptomic signatures and dysregulated pathways associated with the autoimmune response in Cd38-/- mice compared to wild-type (WT) mice within the bm12 chronic graft-versus-host disease (cGVHD) lupus model. We conducted bulk RNA sequencing on peritoneal exudate cells (PECs) and spleen cells (SPC) at two and four weeks following adoptive cell transfer. We also analyzed cells from healthy, untreated mice. These analyses revealed a sustained upregulation of a transcriptional profile of purinergic receptors and ectonucleotidases in cGVHD WT PECs, which displayed a coordinated expression with several type I interferon-stimulated genes (ISGs) and with key molecules involved in the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway, two hallmarks in the lupus pathology. A second purinergic receptor transcriptomic profile, which included P2rx7 and P2rx4, showed a coordinated gene expression of the components of the NLRP3 inflammasome with its potential activators. These processes were transcriptionally less active in cGVHD Cd38-/- PECs than in WT PECs. We have also shown evidence of a distinct enrichment in pathways signatures that define processes such as Ca2+ ion homeostasis, cell division, phagosome, autophagy, senescence, cytokine/cytokine receptor interactions, Th17 and Th1/Th2 cell differentiation in Cd38-/- versus WT samples, which reflected the milder inflammatory and autoimmune response elicited in Cd38-/- mice relative to WT counterparts in response to the allogeneic challenge. Last, we have shown an intense metabolic reprogramming toward oxidative phosphorylation in PECs and SPC from cGVHD WT mice, which may reflect an increased cellular demand for oxygen consumption, in contrast to PECs and SPC from cGVHD Cd38-/- mice, which showed a short-lived metabolic effect at the transcriptomic level. Overall, these findings support the pro-inflammatory and immunomodulatory role of CD38 during the development of the cGVHD-lupus disease.
Collapse
Affiliation(s)
- Mercedes Zubiaur
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | | | | | | | | | | | | | - África Martínez-Blanco
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Marilú Domínguez-Pantoja
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - María Botia-Sánchez
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Sonia Pérez-Cabrera
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Nerea Bello-Iglesias
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Antonio Alcina
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Ana-Clara Abadía-Molina
- Department of Biochemistry, Molecular Biology and Immunology III, School of Medicine, University of Granada (UGR), Granada, Spain
| | - Fuencisla Matesanz
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Esther Zumaquero
- Department of Microbiology, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| | - Ramón Merino
- Department of Cell and Molecular Signaling, Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), University of Cantabria (UC) and CSIC, Santander, Spain
| | - Jaime Sancho
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine “López-Neyra” (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| |
Collapse
|
6
|
Zhang L, Yan Y, Li S, Lin Y, Wang Y, Wang Y, Li N, Lu F, Sun X, Zhang L, Zhou J, Ding Y, Li Q. Nicotinamide Phosphoribosyltransferase Acetylation Mediating Muscle Dysfunction Contributes to Sleep Apnoea in Obesity. J Cachexia Sarcopenia Muscle 2025; 16:e13693. [PMID: 39901373 PMCID: PMC11790607 DOI: 10.1002/jcsm.13693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/24/2024] [Accepted: 01/02/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Obstructive sleep apnoea (OSA) occurs frequently among individuals with obesity, which is attributed to upper airway muscle dysfunction. Muscle function is regulated by the dynamic balance of the nicotinamide adenine dinucleotide (NAD+) and its reduced form (NADH), which is controlled by the enzyme nicotinamide phosphoribosyltransferase (NAMPT). Elevated NAMPT levels have been found in individuals with obesity. However, the role of NAMPT in obesity-induced muscle impairment has not been fully clarified. METHODS A total of 110 participants (70 moderate-to-severe OSA vs. 40 mild or no OSA) underwent electrical impedance mammography and polysomnography. C57BL/6J mice with high-fat diet-induced obesity (DIO) and control group were utilized for their characterizations, which included forced running wheel tests, glucose tolerance tests, haematoxylin and eosin staining, immunostaining, magnetic resonance imaging, whole-body plethysmography, electromyographic techniques, western blot, NAMPT enzymatic activity assays and NAD+/NADH ratio measurements. RESULTS Patients with moderate-severe OSA have a significant decrease in lean mass percentage of upper airway muscles compared with those in controls (p < 0.01). In vivo, a high-fat diet reduced the levels of NAD-dependent deacetylase sirtuin-1 (SIRT1) (p < 0.01), which plays a crucial role in the deacetylation of NAMPT. The reduction in SIRT1-mediated NAMPT deacetylation (p < 0.001) resulted in decreased NAMPT activity (p < 0.01), leading to a decrease in NAD+/NADH ratio (p < 0.05) and decreased the myosin heavy chain isoform (MyHC) I level (p < 0.05), thereby affecting the effectiveness of upper airway muscle and ultimately leading to upper airway collapse (101.0 vs. 81.7 pixels, p = 0.02). The introduction of estradiol mitigated high-fat diet-induced muscle dysfunction by enhancing expression of SIRT1 and inhibiting the acetylation of NAMPT, reducing upper airway collapse (81.7 vs. 96.7 pixels, p = 0.06). CONCLUSIONS These findings highlight the crucial role of SIRT1-mediated NAMPT deacetylation on obesity-induced muscle dysfunction, suggesting targeting NAMPT has the potential to reverse the obesity induced muscle dysfunction and provide effective treatment options for OSA.
Collapse
Affiliation(s)
- Liu Zhang
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ya Ru Yan
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shi Qi Li
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Ni Lin
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yu Qing Wang
- Department of Internal Medicine, Montefiore Medical CenterAlbeit Einstein College of MedicineNew YorkNew YorkUSA
| | - Ning Li
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang Ying Lu
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xian Wen Sun
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Li Yue Zhang
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jian Ping Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yong Jie Ding
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qing Yun Li
- Department of Respiratory and Critical Care Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Respiratory DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
7
|
Li Z, Chen W, Yao S, Peng Z, Liu H, Tang Y, Feng Y. Single-cell mitophagy patterns within the tumor microenvironment modulate intercellular communication, impacting the progression and prognosis of hepatocellular carcinoma. Front Immunol 2025; 15:1448878. [PMID: 39835122 PMCID: PMC11742944 DOI: 10.3389/fimmu.2024.1448878] [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: 06/18/2024] [Accepted: 09/30/2024] [Indexed: 01/22/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignant tumor of the digestive system with a high incidence that seriously threatens patients' lives and health. However, with the rise and application of new treatments, such as immunotherapy, there are still some restrictions in the treatment and diagnosis of HCC, and the therapeutic effects on patients are not ideal. Methods Two single-cell RNA sequencing (scRNA-seq) datasets from HCC patients, encompassing 25,189 cells, were analyzed in the study. We utilized non-negative matrix factorization (NMF) clustering to identify mitophagy patterns in HCC TME cells, including cancer-associated fibroblasts (CAFs), T cells, B cells, and tumor-associated macrophages (TAMs). Cell-to-cell communication was analyzed using the CellChat package, and pseudotime trajectory analysis was performed using the Monocle package. Gene regulatory networks were investigated with the SCENIC package, and survival analyses were conducted with mitophagy-related signatures. Results HCC samples analysis identified 22 clusters, including 7 principal cell types. Complex cell communications were observed among these cell types. Mitophagy-related CAFs, TAMs, CD8+ T cells, and B cells were identified. These subtypes had different biological states, cell-cell communications, and metabolic pathways. Mitophagy levels were elevated in tumor samples. Changes in mitophagy-related genes within specific cell subtypes were associated with different overall survival rates. However, mitophagy did not seem to affect the effectiveness of immunotherapy. Conclusion This study provides evidence that mitophagy within the HCC TME modulates intercellular communication, influencing tumor progression and patient prognosis. Targeting mitophagy may offer a promising approach to improve the long-term prognosis of HCC patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Yongliang Tang
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Feng
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
8
|
Cabulong RB, Kafle SR, Singh A, Sharma M, Kim BS. Biological production of nicotinamide mononucleotide: a review. Crit Rev Biotechnol 2024:1-18. [PMID: 39675885 DOI: 10.1080/07388551.2024.2433993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/30/2024] [Accepted: 10/27/2024] [Indexed: 12/17/2024]
Abstract
Nicotinamide mononucleotide (NMN) presents significant therapeutic potential against aging-related conditions, such as Alzheimer's disease, due to its consistent and strong pharmacological effects. Aside from its anti-aging effect, NMN is also an emerging noncanonical cofactor for orthogonal metabolic pathways in the field of biomanufacturing. This has significant advantages in the field of metabolic engineering, allowing cells to produce unnatural chemicals without disrupting the natural cellular processes. NMN is produced through both the chemical and biological methods, with the latter being more environmentally sustainable. The primary biological production pathway centers on the enzyme nicotinamide phosphoribosyltransferase, which transforms nicotinamide and phosphoribosyl pyrophosphate to NMN. Efforts to increase NMN production have been explored in microorganisms, such as: Escherichia coli, Bacillus subtilis, and yeast, serving as biocatalysts, by rewiring their metabolic processes. Although most researchers are focusing on genetically and metabolically manipulating microorganisms to act as biocatalysts, a growing number of studies on cell-free synthesis are emerging as a promising strategy for producing NMN. This review explores the different biological production techniques of NMN employing microorganisms. This article, in particular, is essential to those who are working on NMN production using microbial strain engineering and cell-free systems.
Collapse
Affiliation(s)
- Rhudith B Cabulong
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Saroj Raj Kafle
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Anju Singh
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Mukesh Sharma
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Beom Soo Kim
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
9
|
Kim MI, Lee C. Identification of virus-rich intermediate cells as crucial players in SARS-CoV-2 infection and differentiation dynamics of human airway epithelium. Front Microbiol 2024; 15:1507852. [PMID: 39735182 PMCID: PMC11681626 DOI: 10.3389/fmicb.2024.1507852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/19/2024] [Indexed: 12/31/2024] Open
Abstract
Understanding the early interactions between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human airway epithelial cells is essential for unraveling viral replication and spread mechanisms. In this study, we investigated the early dynamics of airway epithelial cells during SARS-CoV-2 infection using well-differentiated human nasal and tracheal epithelial cell cultures by incorporating three publicly available single-cell RNA sequencing datasets. We identified a previously uncharacterized cell population, termed virus-rich intermediate (VRI) cells, representing an intermediate differentiation stage between basal and ciliated cells. These VRI cells exhibited high viral loads at all infection time points, strong interferon and inflammatory responses, increased mRNA expression of microvilli-related genes (PAK1, PAK4, VIL1), and suppression of apoptosis markers (BAX, CASP3) alongside increased anti-apoptotic gene expression (BCL2). Cell-cell interaction analysis revealed that VRI cells send signals to basal cells via receptor-ligand pathways such as EPHA and VEGF, likely promoting basal cell differentiation and proliferation through MAPK signaling. These findings suggest that SARS-CoV-2 utilizes VRI cells as a primary site for replication and spread, leveraging these cells' unique differentiation state to evade host cell death and facilitate viral propagation. This study provides insights into the early cellular responses to SARS-CoV-2 infection and highlights potential therapeutic targets to limit viral spread.
Collapse
Affiliation(s)
| | - Choongho Lee
- College of Pharmacy, Dongguk University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Marín-Blázquez M, Rovira J, Ramírez-Bajo MJ, Zapata-Pérez R, Rabadán-Ros R. NAD + enhancers as therapeutic agents in the cardiorenal axis. Cell Commun Signal 2024; 22:537. [PMID: 39516787 PMCID: PMC11546376 DOI: 10.1186/s12964-024-01903-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Cardiorenal diseases represent a complex interplay between heart failure and renal dysfunction, being clinically classified as cardiorenal syndromes (CRS). Recently, the contributions of altered nicotinamide adenine dinucleotide (NAD+) metabolism, through deficient NAD+ synthesis and/or elevated consumption, have proved to be decisive in the onset and progress of cardiorenal disease. NAD+ is a pivotal coenzyme in cellular metabolism, being significant in various signaling pathways, such as energy metabolism, DNA damage repair, gene expression, and stress response. Convincing evidence suggests that strategies designed to boost cellular NAD+ levels are a promising therapeutic option to address cardiovascular and renal disorders. Here, we review and discuss the implications of NAD+ metabolism in cardiorenal diseases, focusing on the propitious NAD+ boosting therapeutic strategies, based on the use of NAD+ precursors, poly(ADP-ribose) polymerase inhibitors, sirtuin activators, and other alternative approaches, such as CD38 blockade, nicotinamide phosphoribosyltransferase activation and combined interventions.
Collapse
Affiliation(s)
- Mariano Marín-Blázquez
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Casanova 143 CRB CELLEX sector 2B, Barcelona, 08036, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), Madrid, Spain
| | - María José Ramírez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Casanova 143 CRB CELLEX sector 2B, Barcelona, 08036, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), Madrid, Spain
| | - Rubén Zapata-Pérez
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain.
| | - Rubén Rabadán-Ros
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain.
| |
Collapse
|
11
|
Sekiya M, Sakakibara Y, Hirota Y, Ito N, Chikamatsu S, Takei K, Nishijima R, Iijima KM. Decreased plasma nicotinamide and altered NAD + metabolism in glial cells surrounding Aβ plaques in a mouse model of Alzheimer's disease. Neurobiol Dis 2024; 202:106694. [PMID: 39374707 DOI: 10.1016/j.nbd.2024.106694] [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: 02/20/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and a leading cause of senile dementia. Amyloid-β (Aβ) accumulation triggers chronic neuroinflammation, initiating AD pathogenesis. Recent clinical trials for anti-Aβ immunotherapy underscore that blood-based biomarkers have significant advantages and applicability over conventional diagnostics and are an unmet clinical need. To further advance ongoing clinical trials and identify novel therapeutic targets for AD, developing additional plasma biomarkers closely associated with pathogenic mechanisms downstream of Aβ accumulation is critically important. To identify plasma metabolites reflective of neuroinflammation caused by Aβ pathology, we performed untargeted metabolomic analyses of the plasma by capillary electrophoresis time-of-flight mass spectrometry (CE-TOFMS) and analyzed the potential roles of the identified metabolic changes in the brain neuroinflammatory response using the female App knock-in (AppNLGF) mouse model of Aβ amyloidosis. The CE-TOFMS analysis of plasma samples from female wild-type (WT) and AppNLGF mice revealed that plasma levels of nicotinamide, a nicotinamide adenine dinucleotide (NAD+) precursor, were decreased in AppNLGF mice, and altered metabolite profiles were enriched for nicotinate/nicotinamide metabolism. In AppNLGF mouse brains, NAD+ levels were unaltered, but mRNA levels of NAD+-synthesizing nicotinate phosphoribosyltransferase (Naprt) and NAD+-degrading Cd38 genes were increased. These enzymes were induced in reactive astrocytes and microglia surrounding Aβ plaques in the cortex and hippocampus of female AppNLGF mouse brains, suggesting neuroinflammation increases NAD+ metabolism. This study suggests plasma nicotinamide could be indicative of the neuroinflammatory response and that nicotinate and nicotinamide metabolism are potential therapeutic targets for AD, by targeting both neuroinflammation and neuroprotection.
Collapse
Affiliation(s)
- Michiko Sekiya
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Yasufumi Sakakibara
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yu Hirota
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Reseach Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Naoki Ito
- Brain-Skeletal Muscle Connection in Aging Project Team, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Sachie Chikamatsu
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kimi Takei
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Risa Nishijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Koichi M Iijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| |
Collapse
|
12
|
Peng A, Li J, Xing J, Yao Y, Niu X, Zhang K. The function of nicotinamide phosphoribosyl transferase (NAMPT) and its role in diseases. Front Mol Biosci 2024; 11:1480617. [PMID: 39513038 PMCID: PMC11540786 DOI: 10.3389/fmolb.2024.1480617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024] Open
Abstract
Nicotinamide phosphoribosyl transferase (NAMPT) is a rate-limiting enzyme in the mammalian nicotinamide adenine dinucleotide (NAD) salvage pathway, and plays a vital role in the regulation of cell metabolic activity, reprogramming, aging and apoptosis. NAMPT synthesizes nicotinamide mononucleotide (NMN) through enzymatic action, which is a key protein involved in host defense mechanism and plays an important role in metabolic homeostasis and cell survival. NAMPT is involved in NAD metabolism and maintains intracellular NAD levels. Sirtuins (SIRTs) are a family of nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylases (HDACs), the members are capable of sensing cellular NAD+ levels. NAMPT-NAD and SIRT constitute a powerful anti-stress defense system. In this paper, the structure, biological function and correlation with diseases of NAMPT are introduced, aiming to provide new ideas for the targeted therapy of related diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Center Hospital, Taiyuan, China
| |
Collapse
|
13
|
Li Y, Pang S, Guo H, Yang S. The renal apical sodium-dependent bile acid transporter expression rescue attenuates renal damage in diabetic nephropathy via farnesoid X receptor activation. Nephrology (Carlton) 2024; 29:627-635. [PMID: 38925891 DOI: 10.1111/nep.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/28/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
AIM Bile acids (BA) function as signalling molecules regulating glucose-lipid homeostasis and energy expenditure. However, the expression of the apical sodium-dependent bile acid transporter (ASBT) in the kidney, responsible for renal BA reabsorption, is downregulated in patients with diabetic kidney disease (DKD). Using the db/db mouse model of DKD, this study aimed to investigate the effects of rescuing ASBT expression via adeno-associated virus-mediated delivery of ASBT (AAVASBT) on kidney protection. METHODS Six-week-old male db/db mice received an intraparenchymal injection of AAVASBT at a dose of 1 × 1011 viral genomes (vg)/animal and were subsequently fed a chow diet for 2 weeks. Male db/m mice served as controls. For drug treatment, daily intraperitoneal (i.p.) injections of the farnesoid X receptor (FXR) antagonist guggulsterone (GS, 10 mg/kg) were administered one day after initiating the experiment. RESULTS AAVASBT treatment rescued renal ASBT expression and reduced the urinary BA output in db/db mice. AAVASBT treatment activated kidney mitochondrial biogenesis and ameliorated renal impairment associated with diabetes by activating FXR. In addition, the injection of FXR antagonist GS in DKD mice would reverse these beneficial effects by AAVASBT treatment. CONCLUSION Our work indicated that restoring renal ASBT expression slowed the course of DKD via activating FXR. FXR activation stimulates mitochondrial biogenesis while reducing renal oxidative stress and lipid build up, indicating FXR activation's crucial role in preventing DKD. These findings further suggest that the maintenance of renal BA reabsorption could be a viable treatment for DKD.
Collapse
Affiliation(s)
- Youmei Li
- Department of Endocrinology, Jinan Central Hospital, Shandong University, Jinan, China
- Department of Endocrinology, Changqing District People's Hospital, Jinan, China
| | - Shuguang Pang
- Department of Endocrinology, Jinan Central Hospital, Shandong University, Jinan, China
- Department of Endocrinology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Clinical Medicine, Weifang Medical College, Weifang, China
| | - Honggang Guo
- Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Shuo Yang
- Department of Clinical Medicine, Weifang Medical College, Weifang, China
| |
Collapse
|
14
|
Hernando-Redondo J, Malcampo M, Pérez-Vega KA, Paz-Graniel I, Martínez-González MÁ, Corella D, Estruch R, Salas-Salvadó J, Pintó X, Arós F, Bautista-Castaño I, Romaguera D, Lapetra J, Ros E, Cueto-Galán R, Fitó M, Castañer O. Mediterranean Diet Modulation of Neuroinflammation-Related Genes in Elderly Adults at High Cardiovascular Risk. Nutrients 2024; 16:3147. [PMID: 39339745 PMCID: PMC11434799 DOI: 10.3390/nu16183147] [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: 09/04/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Individuals with dementia and neurodegenerative diseases (NDDs) often suffer from cardiovascular diseases (CVDs). Neuroinflammation driven by conditions involved in CVDs is linked to disruptions in the central nervous system triggering immune reactions, perpetuating an "inflammatory-like" environment. The Mediterranean diet (MedDiet), known for its anti-inflammatory and antioxidant properties, has been proposed as a key factor to attenuate these risks. Blood nuclear cell samples were collected from 134 participants of the PREDIMED trial, which randomized participants to three diets: one supplemented with extra-virgin olive oil (MedDiet-EVOO), another with nuts (MedDiet-Nuts), and a low-fat control diet. These samples were analyzed at baseline and 12-month follow-up to assess the impact of these dietary interventions on gene expression markers. We first selected target genes by analyzing intersections between NDD and CVD associations. Significant gene expression changes from baseline to 12 months were observed in the participants allocated to the MedDiet-EVOO, particularly in CDKN2A, IFNG, NLRP3, PIK3CB, and TGFB2. Additionally, TGFB2 expression changed over time in the MedDiet-Nuts group. Comparative analyses showed significant differences in TGFB2 between MedDiet-EVOO and control, and in NAMPT between MedDiet-Nuts and control. Longitudinal models adjusted for different covariates also revealed significant effects for TGFB2 and NAMPT. In conclusion, our results suggest that one year of traditional MedDiet, especially MedDiet-EVOO, modulates gene expression associated with CVD risk and NDDs in older adults at high CV risk.
Collapse
Affiliation(s)
- Javier Hernando-Redondo
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Unit of Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute, 08024 Barcelona, Spain (O.C.)
- Ph.D. Program in Food Science and Nutrition, University of Barcelona, 08028 Barcelona, Spain
| | - Mireia Malcampo
- Unit of Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute, 08024 Barcelona, Spain (O.C.)
| | - Karla Alejandra Pérez-Vega
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Unit of Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute, 08024 Barcelona, Spain (O.C.)
| | - Indira Paz-Graniel
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Departament de Bioquímica i Biotecnologia, Alimentació, Nutrició, Desenvolupament i Salut Mental ANUT-DSM, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Miguel Ángel Martínez-González
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Department of Preventive Medicine and Public Health, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Universidad de Navarra, 31009 Pamplona, Spain
| | - Dolores Corella
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Departament of Preventive Medicine, University of Valencia, 46010 Valencia, Spain
| | - Ramón Estruch
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Departament of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, 46010 Barcelona, Spain
| | - Jordi Salas-Salvadó
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Departament de Bioquímica i Biotecnologia, Alimentació, Nutrició, Desenvolupament i Salut Mental ANUT-DSM, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Xavier Pintó
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Lipids and Vascular Risk Unit, Internal Medicine, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Universitario de Bellvitge, University of Barcelona, 08028 Barcelona, Spain
| | - Fernando Arós
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Cardiology Department, Organización Sanitaria Integrada Araba (OSI ARABA), University Hospital of Araba, 01009 Gasteiz, Spain
- University of País Vasco/Euskal Herria Unibersitatea (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
| | - Inmaculada Bautista-Castaño
- Institute for Biomedical Research, University of Las Palmas de Gran Canaria, 35001 Las Palmas de Gran Canaria, Spain;
| | - Dora Romaguera
- Research Group in Nutritional Epidemiology and Cardiovascular Pathophysiology, Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - José Lapetra
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Department of Family Medicine, Research Unity, Distrito Sanitario Atención Primaria Sevilla, 41013 Seville, Spain
| | - Emilio Ros
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 46010 Barcelona, Spain
| | - Raquel Cueto-Galán
- Preventive Medicine and Public Health Department, School of Medicine, University of Malaga, Spain, Biomedical Research Institute of Malaga (IBIMA), 29071 Malaga, Spain;
| | - Montserrat Fitó
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.H.-R.); (K.A.P.-V.); (I.P.-G.); (M.Á.M.-G.); (J.S.-S.); (F.A.); (E.R.)
- Unit of Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute, 08024 Barcelona, Spain (O.C.)
| | - Olga Castañer
- Unit of Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute, 08024 Barcelona, Spain (O.C.)
- CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
15
|
Gao L, Ramirez FJ, Cabrera JTO, Varghese MV, Watanabe M, Tsuji-Hosokawa A, Zheng Q, Yang M, Razan MR, Kempf CL, Camp SM, Wang J, Garcia JGN, Makino A. eNAMPT is a novel therapeutic target for mitigation of coronary microvascular disease in type 2 diabetes. Diabetologia 2024; 67:1998-2011. [PMID: 38898303 PMCID: PMC11410976 DOI: 10.1007/s00125-024-06201-9] [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: 01/09/2024] [Accepted: 04/25/2024] [Indexed: 06/21/2024]
Abstract
AIMS/HYPOTHESIS Individuals with diabetes are at high risk of cardiovascular complications, which significantly increase morbidity/mortality. Coronary microvascular disease (CMD) is recognised as a critical contributor to the increased cardiac mortality observed in people with diabetes. Therefore, there is an urgent need for treatments that are specific to CMD. eNAMPT (extracellular nicotinamide phosphoribosyltransferase) is a damage-associated molecular pattern and TLR4 ligand, whose plasma levels are elevated in people with diabetes. This study was thus designed to investigate the pathogenic role of intracellular nicotinamide phosphoribosyltransferase (iNAMPT) and eNAMPT in promoting the development of CMD in a preclinical murine model of type 2 diabetes. METHODS An inducible type 2 diabetic mouse model was generated by a single injection of low-dose streptozocin (75 mg/kg, i.p.) combined with a high-fat diet for 16 weeks. The in vivo effects of i/eNAMPT inhibition on cardiac endothelial cell (CEC) function were evaluated by using Nampt+/- heterozygous mice, chronic administration of eNAMPT-neutralising monoclonal antibody (mAb) or use of an NAMPT enzymatic inhibitor (FK866). RESULTS As expected, diabetic wild-type mice exhibited significantly lower coronary flow velocity reserve (CFVR), a determinant of coronary microvascular function, compared with control wild-type mice. eNAMPT plasma levels or expression in CECs were significantly greater in diabetic mice than in control mice. Furthermore, in comparison with diabetic wild-type mice, diabetic Nampt+/- heterozygous mice showed markedly improved CFVR, accompanied by increased left ventricular capillary density and augmented endothelium-dependent relaxation (EDR) in the coronary artery. NAMPT inhibition by FK866 or an eNAMPT-neutralising mAb significantly increased CFVR in diabetic mice. Furthermore, administration of the eNAMPT mAb upregulated expression of angiogenesis- and EDR-related genes in CECs from diabetic mice. Treatment with either eNAMPT or NAD+ significantly decreased CEC migration and reduced EDR in coronary arteries, partly linked to increased production of mitochondrial reactive oxygen species. CONCLUSIONS/INTERPRETATION These data indicate that increased i/eNAMPT expression contributes to the development of diabetic coronary microvascular dysfunction, and provide compelling support for eNAMPT inhibition as a novel and effective therapeutic strategy for CMD in diabetes.
Collapse
Affiliation(s)
- Lei Gao
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Francisco J Ramirez
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Jody Tori O Cabrera
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Makiko Watanabe
- Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | | | - Qiuyu Zheng
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingya Yang
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Md Rahatullah Razan
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Carrie L Kempf
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Sara M Camp
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Joe G N Garcia
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA.
- Department of Physiology, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
16
|
Chen L, Xing X, Zhang P, Chen L, Pei H. Homeostatic regulation of NAD(H) and NADP(H) in cells. Genes Dis 2024; 11:101146. [PMID: 38988322 PMCID: PMC11233901 DOI: 10.1016/j.gendis.2023.101146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 07/12/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+)/reduced NAD+ (NADH) and nicotinamide adenine dinucleotide phosphate (NADP+)/reduced NADP+ (NADPH) are essential metabolites involved in multiple metabolic pathways and cellular processes. NAD+ and NADH redox couple plays a vital role in catabolic redox reactions, while NADPH is crucial for cellular anabolism and antioxidant responses. Maintaining NAD(H) and NADP(H) homeostasis is crucial for normal physiological activity and is tightly regulated through various mechanisms, such as biosynthesis, consumption, recycling, and conversion between NAD(H) and NADP(H). The conversions between NAD(H) and NADP(H) are controlled by NAD kinases (NADKs) and NADP(H) phosphatases [specifically, metazoan SpoT homolog-1 (MESH1) and nocturnin (NOCT)]. NADKs facilitate the synthesis of NADP+ from NAD+, while MESH1 and NOCT convert NADP(H) into NAD(H). In this review, we summarize the physiological roles of NAD(H) and NADP(H) and discuss the regulatory mechanisms governing NAD(H) and NADP(H) homeostasis in three key aspects: the transcriptional and posttranslational regulation of NADKs, the role of MESH1 and NOCT in maintaining NAD(H) and NADP(H) homeostasis, and the influence of the circadian clock on NAD(H) and NADP(H) homeostasis. In conclusion, NADKs, MESH1, and NOCT are integral to various cellular processes, regulating NAD(H) and NADP(H) homeostasis. Dysregulation of these enzymes results in various human diseases, such as cancers and metabolic disorders. Hence, strategies aiming to restore NAD(H) and NADP(H) homeostasis hold promise as novel therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Luojun Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
| | - Xiaoke Xing
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430062, China
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
17
|
Cvekl A, Vijg J. Aging of the eye: Lessons from cataracts and age-related macular degeneration. Ageing Res Rev 2024; 99:102407. [PMID: 38977082 DOI: 10.1016/j.arr.2024.102407] [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: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
Aging is the greatest risk factor for chronic human diseases, including many eye diseases. Geroscience aims to understand the effects of the aging process on these diseases, including the genetic, molecular, and cellular mechanisms that underlie the increased risk of disease over the lifetime. Understanding of the aging eye increases general knowledge of the cellular physiology impacted by aging processes at various biological extremes. Two major diseases, age-related cataract and age-related macular degeneration (AMD) are caused by dysfunction of the lens and retina, respectively. Lens transparency and light refraction are mediated by lens fiber cells lacking nuclei and other organelles, which provides a unique opportunity to study a single aging hallmark, i.e., loss of proteostasis, within an environment of limited metabolism. In AMD, local dysfunction of the photoreceptors/retinal pigmented epithelium/Bruch's membrane/choriocapillaris complex in the macula leads to the loss of photoreceptors and eventually loss of central vision, and is driven by nearly all the hallmarks of aging and shares features with Alzheimer's disease, Parkinson's disease, cardiovascular disease, and diabetes. The aging eye can function as a model for studying basic mechanisms of aging and, vice versa, well-defined hallmarks of aging can be used as tools to understand age-related eye disease.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Jan Vijg
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
18
|
Marini S, Huber A, Cash MN, Salemi M, Cook RL, Borsa P, Mavian CN. Oral Cannabidiol Treatment Is Associated with an Anti-Inflammatory Gene Expression Signature in Myeloid Cells of People Living with HIV. Cannabis Cannabinoid Res 2024; 9:1028-1037. [PMID: 38252549 DOI: 10.1089/can.2023.0139] [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: 01/24/2024] Open
Abstract
Introduction: HIV-related comorbidities appear to be related to chronic inflammation, a condition characterizing people living with HIV (PLWH). Prior work indicates that cannabidiol (CBD) might reduce inflammation; however, the genetics underpinning of this effect are not well investigated. Our main objective is to detect gene expression alterations in human peripheral blood mononuclear cells (PBMCs) from PLWH after at least 1 month of CBD treatment. Materials and Methods: We analyzed ∼41,000 PBMCs from three PLWH at baseline and after CBD treatment (27-60 days) through single-cell RNA sequencing. Results: We obtained a coherent signature, characterized by an anti-inflammatory activity, of differentially expressed genes in myeloid cells. Conclusions: Our study shows how CBD is associated with alterations of gene expression in myeloid cells after CBD treatment. Clinical Trial Registration: NCT05209867.
Collapse
Affiliation(s)
- Simone Marini
- Department of Epidemiology, University of Florida, Gainesville, Florida, USA
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | - Amanda Huber
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Melanie N Cash
- Department of Pathology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Marco Salemi
- Department of Pathology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Robert L Cook
- Department of Epidemiology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Paul Borsa
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Carla N Mavian
- Department of Pathology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
19
|
Gasparrini M, Giovannuzzi S, Nocentini A, Raffaelli N, Supuran CT. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) in cancer: a patent review. Expert Opin Ther Pat 2024; 34:565-582. [PMID: 38861278 DOI: 10.1080/13543776.2024.2367006] [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: 03/13/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
INTRODUCTION Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme in the biosynthesis of nicotinamide adenine dinucleotide (NAD) from nicotinamide. In addition to its role as essential redox cofactor, NAD also functions as a substrate for NAD-consuming enzymes, regulating multiple cellular processes such as DNA repair and gene expression, fundamental to sustain energetic needs for tumor growth. In this sense, NAMPT over-expression represents a common strategy that several tumor types adopt to sustain NAD production. In addition to its enzymatic role, NAMPT behaves as cytokine-like protein with pro-inflammatory function. Increasing evidence demonstrated that NAMPT inhibition represents a promising anti-cancer strategy to deplete NAD and impair cellular metabolism in cancer conditions. AREAS COVERED By using Espacenet, we collected the patents which identified new molecules, compounds, formulations and methods able to inhibit NAMPT from 2007 to date. EXPERT OPINION Most of the collected patents focused the attention on the ability of different compounds to inhibit the enzymatic activity of NAMPT, lacking other important aspects related to the extracellular role of NAMPT and the ability of alternative enzymes to counteract NAMPT-mediated NAD depletion. It is necessary to consider also these aspects to promote novel strategies and create novel inhibitors and molecules useful as anti-cancer compounds.
Collapse
Affiliation(s)
- Massimiliano Gasparrini
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Simone Giovannuzzi
- NEUROFARBA Department, Pharmaceutical Chemistry Section, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Alessio Nocentini
- NEUROFARBA Department, Pharmaceutical Chemistry Section, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Nadia Raffaelli
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Pharmaceutical Chemistry Section, University of Florence, Sesto Fiorentino, Florence, Italy
| |
Collapse
|
20
|
Hong D, Kim HK, Yang W, Yoon C, Kim M, Yang CS, Yoon S. Integrative analysis of single-cell RNA-seq and gut microbiome metabarcoding data elucidates macrophage dysfunction in mice with DSS-induced ulcerative colitis. Commun Biol 2024; 7:731. [PMID: 38879692 PMCID: PMC11180211 DOI: 10.1038/s42003-024-06409-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/03/2024] [Indexed: 06/19/2024] Open
Abstract
Ulcerative colitis (UC) is a significant inflammatory bowel disease caused by an abnormal immune response to gut microbes. However, there are still gaps in our understanding of how immune and metabolic changes specifically contribute to this disease. Our research aims to address this gap by examining mouse colons after inducing ulcerative colitis-like symptoms. Employing single-cell RNA-seq and 16 s rRNA amplicon sequencing to analyze distinct cell clusters and microbiomes in the mouse colon at different time points after induction with dextran sodium sulfate. We observe a significant reduction in epithelial populations during acute colitis, indicating tissue damage, with a partial recovery observed in chronic inflammation. Analyses of cell-cell interactions demonstrate shifts in networking patterns among different cell types during disease progression. Notably, macrophage phenotypes exhibit diversity, with a pronounced polarization towards the pro-inflammatory M1 phenotype in chronic conditions, suggesting the role of macrophage heterogeneity in disease severity. Increased expression of Nampt and NOX2 complex subunits in chronic UC macrophages contributes to the inflammatory processes. The chronic UC microbiome exhibits reduced taxonomic diversity compared to healthy conditions and acute UC. The study also highlights the role of T cell differentiation in the context of dysbiosis and its implications in colitis progression, emphasizing the need for targeted interventions to modulate the inflammatory response and immune balance in colitis.
Collapse
Affiliation(s)
- Dawon Hong
- RNA Cell Biology Laboratory, Graduate Department of Bioconvergence Engineering, Dankook University, Yongin, Republic of Korea
| | - Hyo Keun Kim
- Dept of Molecular and Life Science and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan-si, Korea
| | - Wonhee Yang
- Department of AI-based Convergence, Dankook University, Yongin, Republic of Korea
| | - Chanjin Yoon
- Dept of Molecular and Life Science and Institute of Natural Science and Technology, Hanyang University, Ansan-si, Korea
| | - Minsoo Kim
- Department of Computer Science, College of SW Convergence, Dankook University, Yongin, Republic of Korea
| | - Chul-Su Yang
- Dept of Medicinal and Life Science and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan-si, Korea.
| | - Seokhyun Yoon
- Department of Electronics & Electrical Engineering, College of Engineering, Dankook University, Yongin, Republic of Korea.
| |
Collapse
|
21
|
Nunes PR, Pereira DA, Passeti LFP, Coura LLF, Gomes KB, Sandrim VC, Luizon MR. The interplay between extracellular NAMPT and inflammatory cytokines in preeclampsia. J Reprod Immunol 2024; 163:104248. [PMID: 38703439 DOI: 10.1016/j.jri.2024.104248] [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: 12/11/2023] [Revised: 03/21/2024] [Accepted: 04/09/2024] [Indexed: 05/06/2024]
Abstract
Preeclampsia (PE) is the major cause of maternal-fetal mortality and morbidity. Its pathophysiology is not elucidated, but there is evidence for the role of visfatin/nicotinamide phosphoribosyl transferase (NAMPT), mainly due to its relation to endothelial dysfunction, a hallmark of PE. However, there is heterogeneous data regarding visfatin/NAMPT in healthy pregnancy (HP) and PE. Therefore, we performed a search on MEDLINE/PubMed using the terms "visfatin and preeclampsia" and "NAMPT and preeclampsia, and we selected 23 original articles: 12 articles reported increased levels in PE compared to HP, only four articles showed lower levels and eight articles did not find differences regarding visfatin/NAMPT in the groups studied. It is widely acknowledged that levels detected in plasma, serum, or placenta can be influenced by the size of the population and sample analyzed, as well as genetic factors. We further discussed the correlations of visfatin/NAMPT with clinical biomarkers in PE and inflammatory pathways. Considering the common inflammatory mechanisms between PE and visfatin/NAMPT, few studies have recently performed serum or plasma dosages. In conclusion, further studies are needed to highlight the potential role of visfatin/NAMPT in the pathophysiology of PE. This will provide comparative evidence to establish it as a biomarker for disease outcomes and treatment.
Collapse
Affiliation(s)
- Priscila Rezeck Nunes
- Department of Pharmacology and Biophysics, Institute of Biosciences, Sao Paulo State University (Unesp), Sao Paulo, Brazil.
| | - Daniela Alves Pereira
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Luis Fernando Pereira Passeti
- Department of Pharmacology and Biophysics, Institute of Biosciences, Sao Paulo State University (Unesp), Sao Paulo, Brazil
| | - Lídia Lana Ferreira Coura
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Karina Braga Gomes
- Department of Clinical and Toxicological Analysis, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Valeria Cristina Sandrim
- Department of Pharmacology and Biophysics, Institute of Biosciences, Sao Paulo State University (Unesp), Sao Paulo, Brazil
| | - Marcelo Rizzatti Luizon
- Department of Pharmacology and Biophysics, Institute of Biosciences, Sao Paulo State University (Unesp), Sao Paulo, Brazil; Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
22
|
Huang Y, Niu Y, Wang X, Li X, He Y, Liu X. Identification of novel biomarkers related to neutrophilic inflammation in COPD. Front Immunol 2024; 15:1410158. [PMID: 38873611 PMCID: PMC11169582 DOI: 10.3389/fimmu.2024.1410158] [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: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is one of the most prevalent chronic respiratory diseases and the fourth cause of mortality globally. Neutrophilic inflammation has a vital role in the occurrence and progression of COPD. This study aimed to identify the novel hub genes involved in neutrophilic inflammation in COPD through bioinformatic prediction and experimental validation. Methods Both the single-cell RNA sequencing (scRNA-seq) dataset (GSE173896) and the RNA sequencing (RNA-seq) dataset (GSE57148) were downloaded from the Gene Expression Omnibus (GEO) database. The Seurat package was used for quality control, dimensions reduction, and cell identification of scRNA-seq. The irGSEA package was used for scoring individual cells. The Monocle2 package was used for the trajectory analysis of neutrophils. The CIBERSORT algorithm was used for analysis of immune cell infiltration in the lungs of COPD patients and controls in RNA-seq dataset, and weighted gene co-expression network analysis (WGCNA) correlated gene modules with neutrophil infiltration. The Mendelian randomization (MR) analysis explored the causal relationship between feature DEGs and COPD. The protein-protein interaction (PPI) network of novel hub genes was constructed, and real-time quantitative polymerase chain reaction (qRT-PCR) was used to validate novel hub genes in clinical specimens. Results In scRNA-seq, the gene sets upregulated in COPD samples were related to the neutrophilic inflammatory response and TNF-α activation of the NF-κB signaling pathway. In RNA-seq, immune infiltration analysis showed neutrophils were upregulated in COPD lung tissue. We combined data from differential and modular genes and identified 51 differential genes associated with neutrophilic inflammation. Using MR analysis, 6 genes were explored to be causally associated with COPD. Meanwhile, 11 hub genes were identified by PPI network analysis, and all of them were upregulated. qRT-PCR experiments validated 9 out of 11 genes in peripheral blood leukocytes of COPD patients. Furthermore, 5 genes negatively correlated with lung function in COPD patients. Finally, a network of transcription factors for NAMPT and PTGS2 was constructed. Conclusion This study identified nine novel hub genes related to the neutrophilic inflammation in COPD, and two genes were risk factors of COPD, which may serve as potential biomarkers for the clinical severity of COPD.
Collapse
Affiliation(s)
- Yuchen Huang
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yang Niu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xuezhao Wang
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xiaochen Li
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yuanzhou He
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xiansheng Liu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Pulmonary Diseases of National Health Commission, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| |
Collapse
|
23
|
Zhou W, Su P, Wang Y, Li Z, Liu L. Exploration of the molecular linkage between endometriosis and Crohn disease by bioinformatics methods. Medicine (Baltimore) 2024; 103:e38097. [PMID: 38758892 PMCID: PMC11098239 DOI: 10.1097/md.0000000000038097] [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: 12/26/2023] [Accepted: 04/11/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Endometriosis (EMT) is a common disease in reproductive-age woman and Crohn disease (CD) is a chronic inflammatory disorder in gastrointestinal tract. Previous studies reported that patients with EMT had an increased risk of CD. However, the linkage between EMT and CD remains unclear. In this study, we aimed to investigate the potential molecular mechanism of EMT and CD. METHODS The microarray data of EMT and CD were downloaded from Gene Expression Omnibus. Common genes of EMT and CD were obtained to perform the Gene Ontology and Kyoto Encyclopedia of Gene Genomes enrichments. The protein-protein interaction network was constructed by Cytoscape software and the hub genes were identified by CytoHubba plug-in. Finally we predicted the transcription factors (TFs) of hub genes and constructed a TFs-hub genes regulation network. RESULTS A total of 50 common genes were identified. Kyoto Encyclopedia of Gene Genomes enrichment showed that the common genes mainly enriched in MAPK pathway, VEGF pathway, Wnt pathway, TGF-beta pathway, and Ras pathway. Fifteen hub genes were collected from the protein-protein interaction network, including FMOD, FRZB, CPE, SST, ISG15, EFEMP1, KDR, ADRA2A, FZD7, AQP1, IGFBP5, NAMPT, PLUA, FGF9, and FHL2. Among them, FGF9, FZD7, IGFBP5, KDR, and NAMPT were both validated in the other 2 datasets. Finally TFs-hub genes regulation network were constructed. CONCLUSION Our findings firstly revealed the linkage between EMT and CD, including inflammation, angiogenesis, immune regulation, and cell behaviors, which may lead to the risk of CD in EMT. FGF9, FZD7, IGFBP5, KDR, and NAMPT may closely relate to the linkage.
Collapse
Affiliation(s)
- Weijie Zhou
- Department of Gastroenterology, The Six Affiliated Hospital of South China University of Technology, Foshan City, Guangdong Province, China
| | - Peizhu Su
- Department of Gastroenterology, The First People’s Hospital of Foshan, Foshan City, Guangdong Province, China
| | - Yilin Wang
- Department of Gastroenterology, The First People’s Hospital of Foshan, Foshan City, Guangdong Province, China
| | - Zhaotao Li
- Department of Gastroenterology, The First People’s Hospital of Foshan, Foshan City, Guangdong Province, China
| | - Liu Liu
- Department of Gastroenterology, The Six Affiliated Hospital of South China University of Technology, Foshan City, Guangdong Province, China
| |
Collapse
|
24
|
Jia S, Zhao F. Single-cell transcriptomic profiling of the neonatal oviduct and uterus reveals new insights into upper Müllerian duct regionalization. FASEB J 2024; 38:e23632. [PMID: 38686936 PMCID: PMC11095678 DOI: 10.1096/fj.202400303r] [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: 02/07/2024] [Revised: 03/20/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024]
Abstract
The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper MD development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.
Collapse
Affiliation(s)
- Shuai Jia
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Fei Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
25
|
Halpern MD, Gupta A, Zaghloul N, Thulasingam S, Calton CM, Camp SM, Garcia JGN, Ahmed M. Extracellular Nicotinamide Phosphoribosyltransferase Is a Therapeutic Target in Experimental Necrotizing Enterocolitis. Biomedicines 2024; 12:970. [PMID: 38790933 PMCID: PMC11118767 DOI: 10.3390/biomedicines12050970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency of prematurity. Postulated mechanisms leading to inflammatory necrosis of the ileum and colon include activation of the pathogen recognition receptor Toll-like receptor 4 (TLR4) and decreased levels of transforming growth factor beta (TGFβ). Extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a novel damage-associated molecular pattern (DAMP), is a TLR4 ligand and plays a role in a number of inflammatory disease processes. To test the hypothesis that eNAMPT is involved in NEC, an eNAMPT-neutralizing monoclonal antibody, ALT-100, was used in a well-established animal model of NEC. Preterm Sprague-Dawley pups delivered prematurely from timed-pregnant dams were exposed to hypoxia/hypothermia and randomized to control-foster mother dam-fed rats, injected IP with saline (vehicle) 48 h after delivery; control + mAB-foster dam-fed rats, injected IP with 10 µg of ALT-100 at 48 h post-delivery; NEC-orally gavaged, formula-fed rats injected with saline; and NEC + mAb-formula-fed rats, injected IP with 10 µg of ALT-100 at 48 h. The distal ileum was processed 96 h after C-section delivery for histological, biochemical, molecular, and RNA sequencing studies. Saline-treated NEC pups exhibited markedly increased fecal blood and histologic ileal damage compared to controls (q < 0.0001), and findings significantly reduced in ALT-100 mAb-treated NEC pups (q < 0.01). Real-time PCR in ileal tissues revealed increased NAMPT in NEC pups compared to pups that received the ALT-100 mAb (p < 0.01). Elevated serum levels of tumor necrosis factor alpha (TNFα), interleukin 6 (IL-6), interleukin-8 (IL-8), and NAMPT were observed in NEC pups compared to NEC + mAb pups (p < 0.01). Finally, RNA-Seq confirmed dysregulated TGFβ and TLR4 signaling pathways in NEC pups that were attenuated by ALT-100 mAb treatment. These data strongly support the involvement of eNAMPT in NEC pathobiology and eNAMPT neutralization as a strategy to address the unmet need for NEC therapeutics.
Collapse
Affiliation(s)
- Melissa D. Halpern
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Akash Gupta
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Nahla Zaghloul
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Senthilkumar Thulasingam
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Christine M. Calton
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Sara M. Camp
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Joe G. N. Garcia
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Mohamed Ahmed
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
26
|
Velma G, Krider IS, Alves ETM, Courey JM, Laham MS, Thatcher GRJ. Channeling Nicotinamide Phosphoribosyltransferase (NAMPT) to Address Life and Death. J Med Chem 2024; 67:5999-6026. [PMID: 38580317 PMCID: PMC11056997 DOI: 10.1021/acs.jmedchem.3c02112] [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: 11/11/2023] [Revised: 02/22/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step in NAD+ biosynthesis via salvage of NAM formed from catabolism of NAD+ by proteins with NADase activity (e.g., PARPs, SIRTs, CD38). Depletion of NAD+ in aging, neurodegeneration, and metabolic disorders is addressed by NAD+ supplementation. Conversely, NAMPT inhibitors have been developed for cancer therapy: many discovered by phenotypic screening for cancer cell death have low nanomolar potency in cellular models. No NAMPT inhibitor is yet FDA-approved. The ability of inhibitors to act as NAMPT substrates may be associated with efficacy and toxicity. Some 3-pyridyl inhibitors become 4-pyridyl activators or "NAD+ boosters". NAMPT positive allosteric modulators (N-PAMs) and boosters may increase enzyme activity by relieving substrate/product inhibition. Binding to a "rear channel" extending from the NAMPT active site is key for inhibitors, boosters, and N-PAMs. A deeper understanding may fulfill the potential of NAMPT ligands to regulate cellular life and death.
Collapse
Affiliation(s)
- Ganga
Reddy Velma
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Isabella S. Krider
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Erick T. M. Alves
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Jenna M. Courey
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Megan S. Laham
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Gregory R. J. Thatcher
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
27
|
Messana VG, Fascì A, Vitale N, Micillo M, Rovere M, Pesce NA, Martines C, Efremov DG, Vaisitti T, Deaglio S. A molecular circuit linking the BCR to the NAD biosynthetic enzyme NAMPT is an actionable target in Richter syndrome. Blood Adv 2024; 8:1920-1933. [PMID: 38359376 PMCID: PMC11021907 DOI: 10.1182/bloodadvances.2023011690] [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: 09/14/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/17/2024] Open
Abstract
ABSTRACT This works defines, to the best of our knowledge, for the first time a molecular circuit connecting nicotinamide mononucleoside phosphoribosyl transferase (NAMPT) activity to the B-cell receptor (BCR) pathway. Using 4 distinct xenograft models derived from patients with Richter syndrome (RS-PDX), we show that BCR cross-linking results in transcriptional activation of the nicotinamide adenine dinucleotide (NAD) biosynthetic enzyme NAMPT, with increased protein expression, in turn, positively affecting global cellular NAD levels and sirtuins activity. NAMPT blockade, by using the novel OT-82 inhibitor in combination with either BTK or PI3K inhibitors (BTKi or PI3Ki), induces rapid and potent apoptotic responses in all 4 models, independently of their mutational profile and the expression of the other NAD biosynthetic enzymes, including nicotinate phosphoribosyltransferase. The connecting link in the circuit is represented by AKT that is both tyrosine- and serine-phosphorylated by PI3K and deacetylated by sirtuin 1 and 2 to obtain full kinase activation. Acetylation (ie, inhibition) of AKT after OT-82 administration was shown by 2-dimensional gel electrophoresis and immunoprecipitation. Consistently, pharmacological inhibition or silencing of sirtuin 1 and 2 impairs AKT activation and induces apoptosis of RS cells in combination with PI3Ki or BTKi. Lastly, treatment of RS-PDX mice with the combination of PI3Ki and OT-82 results in significant inhibition of tumor growth, with evidence of in vivo activation of apoptosis. Collectively, these data highlight a novel application for NAMPT inhibitors in combination with BTKi or PI3Ki in aggressive lymphomas.
Collapse
Affiliation(s)
- Vincenzo G. Messana
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Amelia Fascì
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Nicoletta Vitale
- Department of Molecular Biotechnologies and Health Science, University of Turin, Turin, Italy
| | - Matilde Micillo
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Matteo Rovere
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Noemi A. Pesce
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Claudio Martines
- Molecular Hematology Unit, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Dimitar G. Efremov
- Molecular Hematology Unit, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Tiziana Vaisitti
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Laboratory of Functional Genomics, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
28
|
Zhang K, Mishra A, Jagannath C. New insight into arginine and tryptophan metabolism in macrophage activation during tuberculosis. Front Immunol 2024; 15:1363938. [PMID: 38605962 PMCID: PMC11008464 DOI: 10.3389/fimmu.2024.1363938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Arginine and tryptophan are pivotal in orchestrating cytokine-driven macrophage polarization and immune activation. Specifically, interferon-gamma (IFN-γ) stimulates inducible nitric oxide synthase (iNOS) expression), leading to the conversion of arginine into citrulline and nitric oxide (NO), while Interleukin-4 (IL4) promotes arginase activation, shifting arginine metabolism toward ornithine. Concomitantly, IFN-γ triggers indoleamine 2,3-dioxygenase 1 (IDO1) and Interleukin-4 induced 1 (IL4i1), resulting in the conversion of tryptophan into kynurenine and indole-3-pyruvic acid. These metabolic pathways are tightly regulated by NAD+-dependent sirtuin proteins, with Sirt2 and Sirt5 playing integral roles. In this review, we present novel insights that augment our understanding of the metabolic pathways of arginine and tryptophan following Mycobacterium tuberculosis infection, particularly their relevance in macrophage responses. Additionally, we discuss arginine methylation and demethylation and the role of Sirt2 and Sirt5 in regulating tryptophan metabolism and arginine metabolism, potentially driving macrophage polarization.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| |
Collapse
|
29
|
Chen KC, Dhar T, Chen CR, Chen ECY, Peng CC. Nicotinamide phosphoribosyltransferase modulates PD-L1 in bladder cancer and enhances immunotherapeutic sensitivity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167106. [PMID: 38428685 DOI: 10.1016/j.bbadis.2024.167106] [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: 10/31/2023] [Revised: 02/11/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Bladder cancer (BLCA) is one of the most prevalent malignancies worldwide with a high mortality rate and poor response to immunotherapy in patients expressing lower programmed death ligand 1 (PD-L1) levels. Nicotinamide phosphoribosyltransferase (NAMPT), a rate-limiting enzyme responsible for the biosynthesis of nicotinamide adenine dinucleotide (NAD+) from nicotinamide was reported to be overexpressed in various cancers; however, the role of NAMPT in BLCA is obscure. Immunohistochemistry of tissue microarrays, a real-time polymerase chain reaction, Western blotting, proliferation assay, NAD+ quantification, transwell-migration assay, and colony-formation assay were performed to measure NAMPT and PD-L1 expression levels in patients and the effect of NAMPT inhibition on T24 cells. Our study revealed that NAMPT expression was upregulated in BLCA patients with different grades and associated with poor T-cell infiltration. Notably, FK866-mediated NAMPT inhibition decreased cell viability by depleting NAD+, and reducing the migration ability and colony-formation ability of T24 cells. Interestingly, NAMPT negatively regulated PD-L1 under an interferon (IFN)-γ-mediated microenvironment. However, exogenous NAMPT activator has no effect on PD-L1. NAD+ supplementation also only increased PD-L1 in the absence of IFN-γ. Conclusively, NAMPT is crucial for BLCA tumorigenic properties, and it regulates expression of the PD-L1 immune checkpoint protein. NAMPT could be considered a target for modulating sensitivity to immunotherapy.
Collapse
Affiliation(s)
- Kuan-Chou Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Urology, Taipei Medical University Shuang-Ho Hospital, Zhong-He District, New Taipei City 23561, Taiwan; Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; TMU-Research Center of Urology and Kidney, Taipei Medical University, Taipei, 11031, Taiwan
| | - Trayee Dhar
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chang-Rong Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Eugene Chang-Yu Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chiung-Chi Peng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
30
|
Ibragimova M, Kussainova A, Aripova A, Bersimbaev R, Bulgakova O. The Molecular Mechanisms in Senescent Cells Induced by Natural Aging and Ionizing Radiation. Cells 2024; 13:550. [PMID: 38534394 PMCID: PMC10969416 DOI: 10.3390/cells13060550] [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: 02/20/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
This review discusses the relationship between cellular senescence and radiation exposure. Given the wide range of ionizing radiation sources encountered by people in professional and medical spheres, as well as the influence of natural background radiation, the question of the effect of radiation on biological processes, particularly on aging processes, remains highly relevant. The parallel relationship between natural and radiation-induced cellular senescence reveals the common aspects underlying these processes. Based on recent scientific data, the key points of the effects of ionizing radiation on cellular processes associated with aging, such as genome instability, mitochondrial dysfunction, altered expression of miRNAs, epigenetic profile, and manifestation of the senescence-associated secretory phenotype (SASP), are discussed. Unraveling the molecular mechanisms of cellular senescence can make a valuable contribution to the understanding of the molecular genetic basis of age-associated diseases in the context of environmental exposure.
Collapse
Affiliation(s)
- Milana Ibragimova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Assiya Kussainova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
- Department of Health Sciences, University of Genova, Via Pastore 1, 16132 Genoa, Italy
| | - Akmaral Aripova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Rakhmetkazhi Bersimbaev
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Olga Bulgakova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| |
Collapse
|
31
|
Park JW, Park SE, Koh W, Jang WH, Choi JH, Roh E, Kang GM, Kim SJ, Lim HS, Park CB, Jeong SY, Moon SY, Lee CH, Kim SY, Choi HJ, Min SH, Lee CJ, Kim MS. Hypothalamic astrocyte NAD + salvage pathway mediates the coupling of dietary fat overconsumption in a mouse model of obesity. Nat Commun 2024; 15:2102. [PMID: 38453901 PMCID: PMC10920699 DOI: 10.1038/s41467-024-46009-0] [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: 10/01/2023] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD)+ serves as a crucial coenzyme in numerous essential biological reactions, and its cellular availability relies on the activity of the nicotinamide phosphoribosyltransferase (NAMPT)-catalyzed salvage pathway. Here we show that treatment with saturated fatty acids activates the NAD+ salvage pathway in hypothalamic astrocytes. Furthermore, inhibition of this pathway mitigates hypothalamic inflammation and attenuates the development of obesity in male mice fed a high-fat diet (HFD). Mechanistically, CD38 functions downstream of the NAD+ salvage pathway in hypothalamic astrocytes burdened with excess fat. The activation of the astrocytic NAMPT-NAD+-CD38 axis in response to fat overload induces proinflammatory responses in the hypothalamus. It also leads to aberrantly activated basal Ca2+ signals and compromised Ca2+ responses to metabolic hormones such as insulin, leptin, and glucagon-like peptide 1, ultimately resulting in dysfunctional hypothalamic astrocytes. Our findings highlight the significant contribution of the hypothalamic astrocytic NAD+ salvage pathway, along with its downstream CD38, to HFD-induced obesity.
Collapse
Affiliation(s)
- Jae Woo Park
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Se Eun Park
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science, Daejeon, 34126, Korea
| | - Won Hee Jang
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Jong Han Choi
- Division of Endocrinology and Metabolism, Konkuk University Medical Center, Seoul, 05030, Korea
| | - Eun Roh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, 14068, Korea
| | - Gil Myoung Kang
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea
| | - Seong Jun Kim
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Hyo Sun Lim
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Chae Beom Park
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - So Yeon Jeong
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Sang Yun Moon
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Chan Hee Lee
- Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, 24252, Korea
| | - Sang Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Korea
| | - Hyung Jin Choi
- Department of Biomedical Sciences, Wide River Institute of Immunology, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Se Hee Min
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea
- Division of Endocrinology and Metabolism, Asan Diabetes Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science, Daejeon, 34126, Korea
| | - Min-Seon Kim
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea.
- Division of Endocrinology and Metabolism, Asan Diabetes Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.
| |
Collapse
|
32
|
Ghanem MS, Caffa I, Monacelli F, Nencioni A. Inhibitors of NAD + Production in Cancer Treatment: State of the Art and Perspectives. Int J Mol Sci 2024; 25:2092. [PMID: 38396769 PMCID: PMC10889166 DOI: 10.3390/ijms25042092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
The addiction of tumors to elevated nicotinamide adenine dinucleotide (NAD+) levels is a hallmark of cancer metabolism. Obstructing NAD+ biosynthesis in tumors is a new and promising antineoplastic strategy. Inhibitors developed against nicotinamide phosphoribosyltransferase (NAMPT), the main enzyme in NAD+ production from nicotinamide, elicited robust anticancer activity in preclinical models but not in patients, implying that other NAD+-biosynthetic pathways are also active in tumors and provide sufficient NAD+ amounts despite NAMPT obstruction. Recent studies show that NAD+ biosynthesis through the so-called "Preiss-Handler (PH) pathway", which utilizes nicotinate as a precursor, actively operates in many tumors and accounts for tumor resistance to NAMPT inhibitors. The PH pathway consists of three sequential enzymatic steps that are catalyzed by nicotinate phosphoribosyltransferase (NAPRT), nicotinamide mononucleotide adenylyltransferases (NMNATs), and NAD+ synthetase (NADSYN1). Here, we focus on these enzymes as emerging targets in cancer drug discovery, summarizing their reported inhibitors and describing their current or potential exploitation as anticancer agents. Finally, we also focus on additional NAD+-producing enzymes acting in alternative NAD+-producing routes that could also be relevant in tumors and thus become viable targets for drug discovery.
Collapse
Affiliation(s)
- Moustafa S. Ghanem
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (I.C.); (F.M.)
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (I.C.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (I.C.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy; (I.C.); (F.M.)
- Ospedale Policlinico San Martino IRCCS, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
33
|
Zhang S, Zhang B, Liu Y, Li L. Adipokines in atopic dermatitis: the link between obesity and atopic dermatitis. Lipids Health Dis 2024; 23:26. [PMID: 38263019 PMCID: PMC10804547 DOI: 10.1186/s12944-024-02009-z] [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: 10/12/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic skin condition with intense pruritus, eczema, and dry skin. The recurrent intense pruritus and numerous complications in patients with AD can profoundly affect their quality of life. Obesity is one of its comorbidities that has been confirmed to be the hazard factor of AD and also worsen its severity. Nevertheless, the specific mechanisms that explain the connection between obesity and AD remain incompletely recognized. Recent studies have built hopes on various adipokines to explain this connection. Adipokines, which are disturbed by an obese state, may lead to immune system imbalances in people with AD and promote the development of the disease. This review focuses on the abnormal expression patterns of adipokines in patients with AD and their potential regulatory molecular mechanisms associated with AD. The connection between AD and obesity is elucidated through the involvement of adipokines. This conduces to the in-depth exploration of AD pathogenesis and provides a new perspective to develop therapeutic targets.
Collapse
Affiliation(s)
- Shiyun Zhang
- Eight-year Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China, No. 1 Shuaifuyuan, 100730
| | - Bingjie Zhang
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China, No. 1 Shuaifuyuan, 100730
| | - Yuehua Liu
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China, No. 1 Shuaifuyuan, 100730
| | - Li Li
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China, No. 1 Shuaifuyuan, 100730.
| |
Collapse
|
34
|
Cicchinelli S, Pignataro G, Gemma S, Piccioni A, Picozzi D, Ojetti V, Franceschi F, Candelli M. PAMPs and DAMPs in Sepsis: A Review of Their Molecular Features and Potential Clinical Implications. Int J Mol Sci 2024; 25:962. [PMID: 38256033 PMCID: PMC10815927 DOI: 10.3390/ijms25020962] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Sepsis is a serious organ dysfunction caused by a dysregulated immune host reaction to a pathogen. The innate immunity is programmed to react immediately to conserved molecules, released by the pathogens (PAMPs), and the host (DAMPs). We aimed to review the molecular mechanisms of the early phases of sepsis, focusing on PAMPs, DAMPs, and their related pathways, to identify potential biomarkers. We included studies published in English and searched on PubMed® and Cochrane®. After a detailed discussion on the actual knowledge of PAMPs/DAMPs, we analyzed their role in the different organs affected by sepsis, trying to elucidate the molecular basis of some of the most-used prognostic scores for sepsis. Furthermore, we described a chronological trend for the release of PAMPs/DAMPs that may be useful to identify different subsets of septic patients, who may benefit from targeted therapies. These findings are preliminary since these pathways seem to be strongly influenced by the peculiar characteristics of different pathogens and host features. Due to these reasons, while initial findings are promising, additional studies are necessary to clarify the potential involvement of these molecular patterns in the natural evolution of sepsis and to facilitate their transition into the clinical setting.
Collapse
Affiliation(s)
- Sara Cicchinelli
- Department of Emergency, S.S. Filippo e Nicola Hospital, 67051 Avezzano, Italy;
| | - Giulia Pignataro
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Stefania Gemma
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Andrea Piccioni
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Domitilla Picozzi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Veronica Ojetti
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| |
Collapse
|
35
|
Chini CCS, Cordeiro HS, Tran NLK, Chini EN. NAD metabolism: Role in senescence regulation and aging. Aging Cell 2024; 23:e13920. [PMID: 37424179 PMCID: PMC10776128 DOI: 10.1111/acel.13920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023] Open
Abstract
The geroscience hypothesis proposes that addressing the biology of aging could directly prevent the onset or mitigate the severity of multiple chronic diseases. Understanding the interplay between key aspects of the biological hallmarks of aging is essential in delivering the promises of the geroscience hypothesis. Notably, the nucleotide nicotinamide adenine dinucleotide (NAD) interfaces with several biological hallmarks of aging, including cellular senescence, and changes in NAD metabolism have been shown to be involved in the aging process. The relationship between NAD metabolism and cellular senescence appears to be complex. On the one hand, the accumulation of DNA damage and mitochondrial dysfunction induced by low NAD+ can promote the development of senescence. On the other hand, the low NAD+ state that occurs during aging may inhibit SASP development as this secretory phenotype and the development of cellular senescence are both highly metabolically demanding. However, to date, the impact of NAD+ metabolism on the progression of the cellular senescence phenotype has not been fully characterized. Therefore, to explore the implications of NAD metabolism and NAD replacement therapies, it is essential to consider their interactions with other hallmarks of aging, including cellular senescence. We propose that a comprehensive understanding of the interplay between NAD boosting strategies and senolytic agents is necessary to advance the field.
Collapse
Affiliation(s)
- Claudia Christiano Silva Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Heidi Soares Cordeiro
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Ngan Le Kim Tran
- Center for Clinical and Translational Science and Mayo Clinic Graduate School of Biomedical SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Eduardo Nunes Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| |
Collapse
|
36
|
Tilahun L, Asrat A, Wessel GM, Simachew A. Ancestors in the Extreme: A Genomics View of Microbial Diversity in Hypersaline Aquatic Environments. Results Probl Cell Differ 2024; 71:185-212. [PMID: 37996679 DOI: 10.1007/978-3-031-37936-9_10] [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: 11/25/2023]
Abstract
The origin of eukaryotic cells, and especially naturally occurring syncytial cells, remains debatable. While a majority of our biomedical research focuses on the eukaryotic result of evolution, our data remain limiting on the prokaryotic precursors of these cells. This is particularly evident when considering extremophile biology, especially in how the genomes of organisms in extreme environments must have evolved and adapted to unique habitats. Might these rapidly diversifying organisms have created new genetic tools eventually used to enhance the evolution of the eukaryotic single nuclear or syncytial cells? Many organisms are capable of surviving, or even thriving, in conditions of extreme temperature, acidity, organic composition, and then rapidly adapt to yet new conditions. This study identified organisms found in extremes of salinity. A lake and a nearby pond in the Ethiopian Rift Valley were interrogated for life by sequencing the DNA of populations of organism collected from the water in these sites. Remarkably, a vast diversity of microbes were identified, and even though the two sites were nearby each other, the populations of organisms were distinctly different. Since these microbes are capable of living in what for humans would be inhospitable conditions, the DNA sequences identified should inform the next step in these investigations; what new gene families, or modifications to common genes, do these organisms employ to survive in these extreme conditions. The relationship between organisms and their environment can be revealed by decoding genomes of organisms living in extreme environments. These genomes disclose new biological mechanisms that enable life outside moderate environmental conditions, new gene functions for application in biotechnology, and may even result in identification of new species. In this study, we have collected samples from two hypersaline sites in the Danakil depression, the shorelines of Lake As'ale and an actively mixing salt pond called Muda'ara (MUP), to identify the microbial community by metagenomics. Shotgun sequencing was applied to high density sampling, and the relative abundance of Operational Taxonomic Units (OTUs) was calculated. Despite the broad taxonomic similarities among the salt-saturated metagenomes analyzed, MUP stood out from Lake As'ale samples. In each sample site, Archaea accounted for 95% of the total OTUs, largely to the class Halobacteria. The remaining 5% of organisms were eubacteria, with an unclassified strain of Salinibacter ruber as the dominant OTU in both the Lake and the Pond. More than 40 different genes coding for stress proteins were identified in the three sample sites of Lake As'ale, and more than 50% of the predicted stress-related genes were associated with oxidative stress response proteins. Chaperone proteins (DnaK, DnaJ, GrpE, and ClpB) were predicted, with percentage of query coverage and similarities ranging between 9.5% and 99.2%. Long reads for ClpB homologous protein from Lake As'ale metagenome datasets were modeled, and compact 3D structures were generated. Considering the extreme environmental conditions of the Danakil depression, this metagenomics dataset can add and complement other studies on unique gene functions on stress response mechanisms of thriving bio-communities that could have contributed to cellular changes leading to single and/or multinucleated eukaryotic cells.
Collapse
Affiliation(s)
- Lulit Tilahun
- Institute of Biotechnology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Asfawossen Asrat
- Department of Mining and Geological Engineering, Botswana International University of Science and Technology, Palapye, Botswana
- School of Earth Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Gary M Wessel
- Department of Molecular and Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| | - Addis Simachew
- Institute of Biotechnology, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
37
|
Iwatani Y, Hayashi H, Yamamoto H, Minamikawa H, Ichikawa M, Orikawa H, Masuda A, Tada N, Moriyama Y, Takagi N. Pathogenic role of NAMPT in the perivascular regions after ischemic stroke in mice with type 2 diabetes mellitus. Exp Neurol 2024; 371:114584. [PMID: 37884188 DOI: 10.1016/j.expneurol.2023.114584] [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: 07/13/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Ischemic stroke in patients with abnormal glucose tolerance results in poor outcomes. Nicotinamide phosphoribosyltransferase (NAMPT), an adipocytokine, exerts neuroprotective effects. However, the pathophysiological role of NAMPT after ischemic stroke with diabetes and the relationship of NAMPT with cerebrovascular lesions are unclear. The purpose of this study was to clarify the pathophysiological role of NAMPT in cerebral ischemia with diabetes, using db/db mice as a type 2 diabetes animal model. The number of degenerating neurons increased after middle cerebral artery occlusion and reperfusion (MCAO/R) in db/db mice compared with the degenerating neurons in db/+ mice. Extracellular NAMPT (eNAMPT) levels, especially monomeric eNAMPT, increased significantly in db/db MCAO/R mice but not db/+ mice in isolated brain microvessels. The increased eNAMPT levels were associated with increased expression of inflammatory cytokine mRNA. Immunohistochemical analysis demonstrated that NAMPT colocalized with GFAP-positive cells after MCAO/R. In addition, both dimeric and monomeric eNAMPT levels increased in the conditioned medium of primary cortical astrocytes under high glucose conditions subsequent oxygen/glucose deprivation. Our findings are the first to demonstrate the ability of increased monomeric eNAMPT to induce inflammatory responses in brain microvessels, which may be located near astrocyte foot processes.
Collapse
Affiliation(s)
- Yui Iwatani
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hideki Hayashi
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Haruna Yamamoto
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hayato Minamikawa
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Mitsuki Ichikawa
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hayato Orikawa
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Aya Masuda
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Natsumi Tada
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yoshiyuki Moriyama
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Norio Takagi
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
38
|
Liu Z, Sammani S, Barber CJ, Kempf CL, Li F, Yang Z, Bermudez RT, Camp SM, Herndon VR, Furenlid LR, Martin DR, Garcia JGN. An eNAMPT-neutralizing mAb reduces post-infarct myocardial fibrosis and left ventricular dysfunction. Biomed Pharmacother 2024; 170:116103. [PMID: 38160623 PMCID: PMC10872269 DOI: 10.1016/j.biopha.2023.116103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
Myocardial infarction (MI) triggers adverse ventricular remodeling (VR), cardiac fibrosis, and subsequent heart failure. Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) is postulated to play a significant role in VR processing via activation of the TLR4 inflammatory pathway. We hypothesized that an eNAMPT specific monoclonal antibody (mAb) could target and neutralize overexpressed eNAMPT post-MI and attenuate chronic cardiac inflammation and fibrosis. We investigated humanized ALT-100 and ALT-300 mAb with high eNAMPT-neutralizing capacity in an infarct rat model to test our hypothesis. ALT-300 was 99mTc-labeled to generate 99mTc-ALT-300 for imaging myocardial eNAMPT expression at 2 hours, 1 week, and 4 weeks post-IRI. The eNAMPT-neutralizing ALT-100 mAb (0.4 mg/kg) or saline was administered intraperitoneally at 1 hour and 24 hours post-reperfusion and twice a week for 4 weeks. Cardiac function changes were determined by echocardiography at 3 days and 4 weeks post-IRI. 99mTc-ALT-300 uptake was initially localized to the ischemic area at risk (IAR) of the left ventricle (LV) and subsequently extended to adjacent non-ischemic areas 2 hours to 4 weeks post-IRI. Radioactive uptake (%ID/g) of 99mTc-ALT-300 in the IAR increased from 1 week to 4 weeks (0.54 ± 0.16 vs. 0.78 ± 0.13, P < 0.01). Rats receiving ALT-100 mAb exhibited significantly improved myocardial histopathology and cardiac function at 4 weeks, with a significant reduction in the collagen volume fraction (%LV) compared to controls (21.5 ± 6.1% vs. 29.5 ± 9.9%, P < 0.05). Neutralization of the eNAMPT/TLR4 inflammatory cascade is a promising therapeutic strategy for MI by reducing chronic inflammation, fibrosis, and preserving cardiac function.
Collapse
Affiliation(s)
- Zhonglin Liu
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States; Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States.
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Christy J Barber
- Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Carrie L Kempf
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| | - Feng Li
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Zhen Yang
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Rosendo T Bermudez
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Sara M Camp
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| | - Vivian Reyes Herndon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Lars R Furenlid
- Department of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Diego R Martin
- Translational Imaging Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States.
| | - Joe G N Garcia
- University of Florida UF Scripps Research Institute, Jupiter, FL, United States
| |
Collapse
|
39
|
Jia S, Zhao F. Single-cell transcriptomic profiling of the neonatal oviduct and uterus reveals new insights into upper Müllerian duct regionalization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572607. [PMID: 38187777 PMCID: PMC10769252 DOI: 10.1101/2023.12.20.572607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper Müllerian duct development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.
Collapse
|
40
|
Zou A, Chen Y, Liu T, Yang T, Zhou B. Identification and verification of three autophagy-related genes as potential biomarkers for the diagnosis of psoriasis. Sci Rep 2023; 13:22918. [PMID: 38129460 PMCID: PMC10739819 DOI: 10.1038/s41598-023-49764-0] [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: 11/24/2022] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Psoriasis vulgaris is the most common form of the four clinical types. However, early diagnosis of psoriasis vulgaris is difficult due to the lack of effective biomarkers. The aim of this study was to screen potential biomarkers for the diagnosis of psoriasis. In our study, we downloaded the original data from GSE30999 and GSE41664, and the autophagy-related genes list from human autophagy database to identify differentially expressed autophagy-related genes (DERAGs) by R software. Then Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed for DERAGs. DERAGs were validated by the other four databases (GSE13355, GSE14905, GSE6710, and GSE55201) to screen biomarkers with high diagnostic value for the early diagnosis of psoriasis vulgaris. Finally, DERAGs were verified in our clinical blood samples by ELISA. A total of 12 DERAGs were identified between 123 paired non-lesional and lesional skin samples from patients with psoriasis vulgaris. GO and KEGG enrichment analysis indicated the TORC2 complex was more enriched and the NOD-like receptor signaling pathway was mostly enriched. Three autophagy-related genes (BIRC5, NAMPT and BCL2) were identified through bioinformatics analysis and verified by ELISA in clinical blood samples. And these genes showed high diagnostic value for the early diagnosis of psoriasis vulgaris. We identified three autophagy-related genes (BIRC5, NAMPT and BCL2) with high diagnostic value for the early diagnosis of psoriasis vulgaris through bioinformatics analysis and clinical samples. Therefore, we proposed that BIRC5, NAMPT and BCL2 may be as potential biomarkers for the early diagnosis of psoriasis vulgaris. In addition, BIRC5, NAMPT and BCL2 may affect the development of psoriasis by regulating autophagy.
Collapse
Affiliation(s)
- Ailing Zou
- Department of Dermatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, China
| | - Yongjun Chen
- Department of Dermatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, China
| | - Tangsheng Liu
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, China
- Department of Stomatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Ting Yang
- Department of Dermatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, China
| | - Bei Zhou
- Department of Dermatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China.
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, China.
| |
Collapse
|
41
|
Yong J, Cai S, Zeng Z. Targeting NAD + metabolism: dual roles in cancer treatment. Front Immunol 2023; 14:1269896. [PMID: 38116009 PMCID: PMC10728650 DOI: 10.3389/fimmu.2023.1269896] [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: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is indispensable for various oxidation-reduction reactions in mammalian cells, particularly during energy production. Malignant cells increase the expression levels of NAD+ biosynthesis enzymes for rapid proliferation and biomass production. Furthermore, mounting proof has indicated that NAD-degrading enzymes (NADases) play a role in creating the immunosuppressive tumor microenvironment (TME). Interestingly, both inhibiting NAD+ synthesis and targeting NADase have positive implications for cancer treatment. Here we summarize the detrimental outcomes of increased NAD+ production, the functions of NAD+ metabolic enzymes in creating an immunosuppressive TME, and discuss the progress and clinical translational potential of inhibitors for NAD+ synthesis and therapies targeting NADase.
Collapse
Affiliation(s)
- Jiaxin Yong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Songqing Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhaolei Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
42
|
Meng Y, Ye F, Nie P, Zhao Q, An L, Wang W, Qu S, Shen Z, Cao Z, Zhang X, Jiao S, Wu D, Zhou Z, Wei L. Immunosuppressive CD10 +ALPL + neutrophils promote resistance to anti-PD-1 therapy in HCC by mediating irreversible exhaustion of T cells. J Hepatol 2023; 79:1435-1449. [PMID: 37689322 DOI: 10.1016/j.jhep.2023.08.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 07/20/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND & AIMS Remodeling the tumor microenvironment is a critical strategy for treating advanced hepatocellular carcinoma (HCC). Yet, how distinct cell populations in the microenvironment mediate tumor resistance to immunotherapies, such as anti-PD-1, remains poorly understood. METHODS We analyzed the transcriptomic profile, at a single-cell resolution, of tumor tissues from patients with HCC scheduled to receive anti-PD-1-based immunotherapy. Our comparative analysis and experimental validation using flow cytometry and histopathological analysis uncovered a discrete subpopulation of cells associated with resistance to anti-PD-1 treatment in patients and a rat model. A TurboID-based proximity labeling approach was deployed to gain mechanistic insights into the reprogramming of the HCC microenvironment. RESULTS We identified CD10+ALPL+ neutrophils as being associated with resistance to anti-PD-1 treatment. These neutrophils exhibited a strong immunosuppressive activity by inducing an apparent "irreversible" exhaustion of T cells in terms of cell number, frequency, and gene profile. Mechanistically, CD10+ALPL+ neutrophils were induced by tumor cells, i.e., tumor-secreted NAMPT reprogrammed CD10+ALPL+ neutrophils through NTRK1, maintaining them in an immature state and inhibiting their maturation and activation. CONCLUSIONS Collectively, our results reveal a fundamental mechanism by which CD10+ALPL+ neutrophils contribute to tumor immune escape from durable anti-PD-1 treatment. These data also provide further insights into novel immunotherapy targets and possible synergistic treatment regimens. IMPACT AND IMPLICATIONS Herein, we discovered that tumor cells reprogrammed CD10+ALPL+ neutrophils to induce the "irreversible" exhaustion of T cells and hence allow tumors to escape from the intended effects of anti-PD-1 treatment. Our data provided a new theoretical basis for the elucidation of special cell populations and revealed a molecular mechanism underpinning resistance to immunotherapy. Targeting these cells alongside existing immunotherapy could be looked at as a potentially more effective therapeutic approach.
Collapse
Affiliation(s)
- Yan Meng
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China; Department of Stomatology, Shanghai Tenth People's Hospital, Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai 200072, China
| | - Fei Ye
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China; School of Medicine, Shanghai University, Shanghai 200444, China
| | - Pingping Nie
- Department of Stomatology, Shanghai Tenth People's Hospital, Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai 200072, China
| | - Qiudong Zhao
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Liwei An
- Department of Stomatology, Shanghai Tenth People's Hospital, Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai 200072, China
| | - Wenjia Wang
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Shuping Qu
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Zhemin Shen
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Zhifa Cao
- Department of Stomatology, Shanghai Tenth People's Hospital, Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaobing Zhang
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Shi Jiao
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Dong Wu
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China.
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China.
| |
Collapse
|
43
|
Shastry A, Dunham-Snary K. Metabolomics and mitochondrial dysfunction in cardiometabolic disease. Life Sci 2023; 333:122137. [PMID: 37788764 DOI: 10.1016/j.lfs.2023.122137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
Circulating metabolites are indicators of systemic metabolic dysfunction and can be detected through contemporary techniques in metabolomics. These metabolites are involved in numerous mitochondrial metabolic processes including glycolysis, fatty acid β-oxidation, and amino acid catabolism, and changes in the abundance of these metabolites is implicated in the pathogenesis of cardiometabolic diseases (CMDs). Epigenetic regulation and direct metabolite-protein interactions modulate metabolism, both within cells and in the circulation. Dysfunction of multiple mitochondrial components stemming from mitochondrial DNA mutations are implicated in disease pathogenesis. This review will summarize the current state of knowledge regarding: i) the interactions between metabolites found within the mitochondrial environment during CMDs, ii) various metabolites' effects on cellular and systemic function, iii) how harnessing the power of metabolomic analyses represents the next frontier of precision medicine, and iv) how these concepts integrate to expand the clinical potential for translational cardiometabolic medicine.
Collapse
Affiliation(s)
- Abhishek Shastry
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Kimberly Dunham-Snary
- Department of Medicine, Queen's University, Kingston, ON, Canada; Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
44
|
Akrida I, Papadaki H. Adipokines and epithelial-mesenchymal transition (EMT) in cancer. Mol Cell Biochem 2023; 478:2419-2433. [PMID: 36715963 DOI: 10.1007/s11010-023-04670-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 01/17/2023] [Indexed: 01/31/2023]
Abstract
Obesity is a significant risk factor for cancer development. Within the tumor microenvironment, adipocytes interact with cancer cells, immune cells, fibroblasts and endothelial cells, and orchestrate several signaling pathways by secreting bioactive molecules, including adipokines. Adipokines or adipocytokines are produced predominantly by adipocytes and function as autocrine, paracrine and endocrine mediators. Adipokines can exert pro- and anti-inflammatory functions, and they play a pivotal role in the state of chronic low-grade inflammation that characterizes obesity. Epithelial-mesenchymal transition (EMT), a complex biological process whereby epithelial cells acquire the invasive, migratory mesenchymal phenotype is well-known to be implicated in cancer progression and metastasis. Emerging evidence suggests that there is a link between adipokines and EMT. This may contribute to the correlation that has been documented between obesity and cancer progression. This review summarizes the existing body of evidence supporting an association between the process of EMT in cancer and the adipokines leptin, adiponectin, resistin, visfatin/NAMPT, lipocalin-2/NGAL, as well as other newly discovered adipokines including chemerin, nesfatin-1/nucleobindin-2, AZGP1, SFRP5 and FABP4.
Collapse
Affiliation(s)
- Ioanna Akrida
- Department of General Surgery, University General Hospital of Patras, Rion, Greece.
- Department of Anatomy-Histology-Embryology, University of Patras Medical School, Rion, Greece.
- Department of Surgery, Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, 26504, Rion, Greece.
| | - Helen Papadaki
- Department of Anatomy-Histology-Embryology, University of Patras Medical School, Rion, Greece
| |
Collapse
|
45
|
Curran CS, Kopp JB. The complexity of nicotinamide adenine dinucleotide (NAD), hypoxic, and aryl hydrocarbon receptor cell signaling in chronic kidney disease. J Transl Med 2023; 21:706. [PMID: 37814337 PMCID: PMC10563221 DOI: 10.1186/s12967-023-04584-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023] Open
Abstract
Early-stage detection of chronic kidney diseases (CKD) is important to treatment that may slow and occasionally halt CKD progression. CKD of diverse etiologies share similar histologic patterns of glomerulosclerosis, tubular atrophy, and interstitial fibrosis. Macro-vascular disease and micro-vascular disease promote tissue ischemia, contributing to injury. Tissue ischemia promotes hypoxia, and this in turn activates the hypoxia-inducible transcription factors (HIFs). HIF-1α and HIF-2α, share a dimer partner, HIF-1β, with the aryl hydrocarbon receptor (AHR) and are each activated in CKD and associated with kidney cellular nicotinamide adenine dinucleotide (NAD) depletion. The Preiss-Handler, salvage, and de novo pathways regulate NAD biosynthesis and gap-junctions regulate NAD cellular retention. In the Preiss-Handler pathway, niacin forms NAD. Niacin also exhibits crosstalk with HIF and AHR cell signals in the regulation of insulin sensitivity, which is a complication in CKD. Dysregulated enzyme activity in the NAD de novo pathway increases the levels of circulating tryptophan metabolites that activate AHR, resulting in poly-ADP ribose polymerase activation, thrombosis, endothelial dysfunction, and immunosuppression. Therapeutically, metabolites from the NAD salvage pathway increase NAD production and subsequent sirtuin deacetylase activity, resulting in reduced activation of retinoic acid-inducible gene I, p53, NF-κB and SMAD2 but increased activation of FOXO1, PGC-1α, and DNA methyltransferase-1. These post-translational responses may also be initiated through non-coding RNAs (ncRNAs), which are additionally altered in CKD. Nanoparticles traverse biological systems and can penetrate almost all tissues as disease biomarkers and drug delivery carriers. Targeted delivery of non-coding RNAs or NAD metabolites with nanoparticles may enable the development of more effective diagnostics and therapies to treat CKD.
Collapse
Affiliation(s)
- Colleen S Curran
- National Heart Lung and Blood Institute, NIH, BG 10 RM 2C135, 10 Center Drive, Bethesda, MD, 20814, USA.
| | | |
Collapse
|
46
|
Fang J, Hou P, Liu S, Zuo M, Liu Z, Chen W, Han Y, Li Y, Wang T, Feng C, Li P, Shao C, Shi Y. NAD + salvage governs the immunosuppressive capacity of mesenchymal stem cells. Cell Mol Immunol 2023; 20:1171-1185. [PMID: 37580400 PMCID: PMC10541442 DOI: 10.1038/s41423-023-01073-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/24/2023] [Indexed: 08/16/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) possess robust immunoregulatory functions and are promising therapeutics for inflammatory disorders. This capacity is not innate but is activated or 'licensed' by inflammatory cytokines. The licensing mechanism remains unclear. Here, we examined whether inflammatory cytokines metabolically reprogrammed MSCs to confer this immunoregulatory capacity. In response to stimulation by inflammatory cytokines, MSCs exhibited a dramatic increase in the consumption of glucose, which was accompanied by an enhanced use of nicotinamide adenine dinucleotide (NAD+) and increased expression of nicotinamide phosphoribosyltransferase (NAMPT), a central enzyme in the salvage pathway for NAD+ production. When NAD+ synthesis was blocked by inhibiting or depleting NAMPT, the immunosuppressive function of MSCs induced by inflammatory cytokines was greatly attenuated. Consequently, when NAD+ metabolism in MSCs was perturbed, their therapeutic benefit was decreased in mice suffering from inflammatory bowel disease and acute liver injury. Further analysis revealed that NAMPT-driven production of NAD+ was critical for the inflammatory cytokine-induced increase in glycolysis in MSCs. Furthermore, the increase in glycolysis led to succinate accumulation in the tricarboxylic acid cycle, which led to hypoxia-inducible factor 1α (HIF-1α) stabilization and subsequently increased the transcription of key glycolytic genes, thereby persistently maintaining glycolytic flux. This study demonstrated that unlike its proinflammatory role in immune cells, NAD+ metabolism governs the anti-inflammatory function of MSCs during inflammation.
Collapse
Affiliation(s)
- Jiankai Fang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Pengbo Hou
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Shisong Liu
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Muqiu Zuo
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhanhong Liu
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Wangwang Chen
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yuyi Han
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Yanan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Tingting Wang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chao Feng
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Peishan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
47
|
Shah M, Anwar A, Qasim A, Jaan S, Sarfraz A, Ullah R, Ali EA, Nishan U, Shehroz M, Zaman A, Ojha SC. Proteome level analysis of drug-resistant Prevotella melaninogenica for the identification of novel therapeutic candidates. Front Microbiol 2023; 14:1271798. [PMID: 37808310 PMCID: PMC10556700 DOI: 10.3389/fmicb.2023.1271798] [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: 08/02/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
The management of infectious diseases has become more critical due to the development of novel pathogenic strains with enhanced resistance. Prevotella melaninogenica, a gram-negative bacterium, was found to be involved in various infections of the respiratory tract, aerodigestive tract, and gastrointestinal tract. The need to explore novel drug and vaccine targets against this pathogen was triggered by the emergence of antimicrobial resistance against reported antibiotics to combat P. melaninogenica infections. The study involves core genes acquired from 14 complete P. melaninogenica strain genome sequences, where promiscuous drug and vaccine candidates were explored by state-of-the-art subtractive proteomics and reverse vaccinology approaches. A stringent bioinformatics analysis enlisted 18 targets as novel, essential, and non-homologous to humans and having druggability potential. Moreover, the extracellular and outer membrane proteins were subjected to antigenicity, allergenicity, and physicochemical analysis for the identification of the candidate proteins to design multi-epitope vaccines. Two candidate proteins (ADK95685.1 and ADK97014.1) were selected as the best target for the designing of a vaccine construct. Lead B- and T-cell overlapped epitopes were joined to generate potential chimeric vaccine constructs in combination with adjuvants and linkers. Finally, a prioritized vaccine construct was found to have stable interactions with the human immune cell receptors as confirmed by molecular docking and MD simulation studies. The vaccine construct was found to have cloning and expression ability in the bacterial cloning system. Immune simulation ensured the elicitation of significant immune responses against the designed vaccine. In conclusion, our study reported novel drug and vaccine targets and designed a multi-epitope vaccine against the P. melaninogenica infection. Further experimental validation will help open new avenues in the treatment of this multi-drug-resistant pathogen.
Collapse
Affiliation(s)
- Mohibullah Shah
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Amna Anwar
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Aqsa Qasim
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Samavia Jaan
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Asifa Sarfraz
- Department of Biochemistry, Bahauddin Zakariya University, Multan, Pakistan
| | - Riaz Ullah
- Medicinal Aromatic and Poisonous Plants Research Center, College of Pharmacy King Saud University, Riyadh, Saudi Arabia
| | - Essam A. Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Umar Nishan
- Department of Chemistry, Kohat University of Science and Technology, Kohat, Pakistan
| | - Muhammad Shehroz
- Department of Bioinformatics, Kohsar University Murree, Murree, Pakistan
| | - Aqal Zaman
- Department of Microbiology and Molecular Genetics, Bahauddin Zakariya University, Multan, Pakistan
| | - Suvash Chandra Ojha
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
48
|
Zhu X, Li J, Wang H, Gasior FM, Lee C, Lin S, Justice CN, O’Donnell JM, Vanden Hoek TL. Nicotinamide restores tissue NAD+ and improves survival in rodent models of cardiac arrest. PLoS One 2023; 18:e0291598. [PMID: 37713442 PMCID: PMC10503771 DOI: 10.1371/journal.pone.0291598] [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: 04/19/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023] Open
Abstract
Metabolic suppression in the ischemic heart is characterized by reduced levels of NAD+ and ATP. Since NAD+ is required for most metabolic processes that generate ATP, we hypothesized that nicotinamide restores ischemic tissue NAD+ and improves cardiac function in cardiomyocytes and isolated hearts, and enhances survival in a mouse model of cardiac arrest. Mouse cardiomyocytes were exposed to 30 min simulated ischemia and 90 min reperfusion. NAD+ content dropped 40% by the end of ischemia compared to pre-ischemia. Treatment with 100 μM nicotinamide (NAM) at the start of reperfusion completely restored the cellular level of NAD+ at 15 min of reperfusion. This rescue of NAD+ depletion was associated with improved contractile recovery as early as 10 min post-reperfusion. In a mouse model of cardiac arrest, 100 mg/kg NAM administered IV immediately after cardiopulmonary resuscitation resulted in 100% survival at 4 h as compared to 50% in the saline group. In an isolated rat heart model, the effect of NAM on cardiac function was measured for 20 min following 18 min global ischemia. Rate pressure product was reduced by 26% in the control group following arrest. Cardiac contractile function was completely recovered with NAM treatment given at the start of reperfusion. NAM restored tissue NAD+ and enhanced production of lactate and ATP, while reducing glucose diversion to sorbitol in the heart. We conclude that NAM can rapidly restore cardiac NAD+ following ischemia and enhance glycolysis and contractile recovery, with improved survival in a mouse model of cardiac arrest.
Collapse
Affiliation(s)
- Xiangdong Zhu
- Center for Advanced Resuscitation Medicine and Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| | - Jing Li
- Center for Advanced Resuscitation Medicine and Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| | - Huashan Wang
- Center for Advanced Resuscitation Medicine and Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| | - Filip M. Gasior
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| | - Chunpei Lee
- Center for Advanced Resuscitation Medicine and Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| | - Shaoxia Lin
- Center for Advanced Resuscitation Medicine and Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| | - Cody N. Justice
- Center for Advanced Resuscitation Medicine and Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| | - J. Michael O’Donnell
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| | - Terry L. Vanden Hoek
- Center for Advanced Resuscitation Medicine and Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, Chicago, Illinois, United States of America
| |
Collapse
|
49
|
Novak EA, Crawford EC, Mentrup HL, Griffith BD, Fletcher DM, Flanagan MR, Schneider C, Firek B, Rogers MB, Morowitz MJ, Piganelli JD, Wang Q, Mollen KP. Epithelial NAD + depletion drives mitochondrial dysfunction and contributes to intestinal inflammation. Front Immunol 2023; 14:1231700. [PMID: 37744380 PMCID: PMC10512956 DOI: 10.3389/fimmu.2023.1231700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction We have previously demonstrated that a pathologic downregulation of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1α) within the intestinal epithelium contributes to the pathogenesis of inflammatory bowel disease (IBD). However, the mechanism underlying downregulation of PGC1α expression and activity during IBD is not yet clear. Methods Mice (male; C57Bl/6, Villincre/+;Pgc1afl/fl mice, and Pgc1afl/fl) were subjected to experimental colitis and treated with nicotinamide riboside. Western blot, high-resolution respirometry, nicotinamide adenine dinucleotide (NAD+) quantification, and immunoprecipitation were used to in this study. Results We demonstrate a significant depletion in the NAD+ levels within the intestinal epithelium of mice undergoing experimental colitis, as well as humans with ulcerative colitis. While we found no decrease in the levels of NAD+-synthesizing enzymes within the intestinal epithelium of mice undergoing experimental colitis, we did find an increase in the mRNA level, as well as the enzymatic activity, of the NAD+-consuming enzyme poly(ADP-ribose) polymerase-1 (PARP1). Treatment of mice undergoing experimental colitis with an NAD+ precursor reduced the severity of colitis, restored mitochondrial function, and increased active PGC1α levels; however, NAD+ repletion did not benefit transgenic mice that lack PGC1α within the intestinal epithelium, suggesting that the therapeutic effects require an intact PGC1α axis. Discussion Our results emphasize the importance of PGC1α expression to both mitochondrial health and homeostasis within the intestinal epithelium and suggest a novel therapeutic approach for disease management. These findings also provide a mechanistic basis for clinical trials of nicotinamide riboside in IBD patients.
Collapse
Affiliation(s)
- Elizabeth A. Novak
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Erin C. Crawford
- Division of Gastroenterology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Heather L. Mentrup
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Brian D. Griffith
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - David M. Fletcher
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | | | - Corinne Schneider
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Brian Firek
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Matthew B. Rogers
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Michael J. Morowitz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Jon D. Piganelli
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Qian Wang
- Department of Pathology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Kevin P. Mollen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
50
|
Lesnak JB, Mazhar K, Price TJ. Neuroimmune Mechanisms Underlying Post-acute Sequelae of SARS-CoV-2 (PASC) Pain, Predictions from a Ligand-Receptor Interactome. Curr Rheumatol Rep 2023; 25:169-181. [PMID: 37300737 PMCID: PMC10256978 DOI: 10.1007/s11926-023-01107-8] [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] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
PURPOSE OF REVIEW Individuals with post-acute sequelae of SARS-CoV-2 (PASC) complain of persistent musculoskeletal pain. Determining how COVID-19 infection produces persistent pain would be valuable for the development of therapeutics aimed at alleviating these symptoms. RECENT FINDINGS To generate hypotheses regarding neuroimmune interactions in PASC, we used a ligand-receptor interactome to make predictions about how ligands from PBMCs in individuals with COVID-19 communicate with dorsal root ganglia (DRG) neurons to induce persistent pain. In a structured literature review of -omics COVID-19 studies, we identified ligands capable of binding to receptors on DRG neurons, which stimulate signaling pathways including immune cell activation and chemotaxis, the complement system, and type I interferon signaling. The most consistent finding across immune cell types was an upregulation of genes encoding the alarmins S100A8/9 and MHC-I. This ligand-receptor interactome, from our hypothesis-generating literature review, can be used to guide future research surrounding mechanisms of PASC-induced pain.
Collapse
Affiliation(s)
- Joseph B Lesnak
- School for Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, BSB 14.102G, Richardson, TX, 75080, USA
| | - Khadijah Mazhar
- School for Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, BSB 14.102G, Richardson, TX, 75080, USA
| | - Theodore J Price
- School for Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, BSB 14.102G, Richardson, TX, 75080, USA.
| |
Collapse
|