1
|
Shi S, Ou X, Liu C, Wen H, Jiang K. Immunoproteasome acted as immunotherapy 'coffee companion' in advanced carcinoma therapy. Front Immunol 2024; 15:1464267. [PMID: 39281672 PMCID: PMC11392738 DOI: 10.3389/fimmu.2024.1464267] [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: 07/13/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Immunoproteasome is a specialized form of proteasome which plays a crucial role in antigen processing and presentation, and enhances immune responses against malignant cells. This review explores the role of immunoproteasome in the anti-tumor immune responses, including immune surveillance and modulation of the tumor microenvironment, as well as its potential as a target for cancer immunotherapy. Furthermore, we have also discussed the therapeutic potential of immunoproteasome inhibitors, strategies to enhance antigen presentation and combination therapies. The ongoing trials and case studies in urology, melanoma, lung, colorectal, and breast cancers have also been summarized. Finally, the challenges facing clinical translation of immunoproteasome-targeted therapies, such as toxicity and resistance mechanisms, and the future research directions have been addressed. This review underscores the significance of targeting the immunoproteasome in combination with other immunotherapies for solid tumors and its potential broader applications in other diseases.
Collapse
Affiliation(s)
- Shaoyan Shi
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xuehai Ou
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chao Liu
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hao Wen
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ke Jiang
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
2
|
Chen H, Xiao J, Huang B, Que J, Liu M. Geraniol (GER) attenuated chronic sleep restriction (CSR)-induced neuroinflammation in adolescent mice. J Neuroimmunol 2024; 393:578400. [PMID: 38991453 DOI: 10.1016/j.jneuroim.2024.578400] [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: 03/27/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
Sleep insufficiency is a significant health problem worldwide, and adolescent sleep restriction (SR) could induce multiple neurodevelopmental disorders in the central nervous system (CNS). Microglial-mediated neuroinflammation plays a vital role in multiple neurological diseases, and recent research showed the regulation effect of immunoproteasome on microglia functions. Geraniol (GER), an important ingredient in many essential oils, possesses diverse pharmacological properties like anti-inflammatory and antioxidant. The present study was designed to evaluate the neuroprotective effect of GER on SR in adolescent mice and further investigate the underlying mechanisms. Our results displayed that 14 days of chronic sleep restriction (CSR) induced cognitive decline, and anxiety-like and attention-deficit behaviors, which were mitigated by GER pretreatment. GER administration also reversed microglial pro-inflammatory response under CSR stimulation in the anterior cingulate cortex (ACC) regions by reducing the expression and secretion of cytokines like IL-1β and TNF-α. Mechanism research showed that LMP7 mRNA was selectively up-regulated under CSR treatment but down-regulated by GER administration. Proteasome activity and protein expression of LMP7 were consistent with mRNA data. ONX-0914 was applied to inhibit LMP7 selectively, and data validated that GER might alleviate CSR-induced neuroinflammation by regulating LMP7. Our study provides evidence that LMP7 is a critical regulator of CSR-induced proinflammation, and geraniol might be a promising therapy against CSR-induced neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hengdao Chen
- Department of Anesthesiology and Perioperative Medicine, The 900 Hospital of the Joint Logistic Support Force of the People's Liberation Army of China, Fuzhou 350025, China
| | - Jinrong Xiao
- Department of Anesthesiology and Perioperative Medicine, The 900 Hospital of the Joint Logistic Support Force of the People's Liberation Army of China, Fuzhou 350025, China
| | - Bin Huang
- Department of Anesthesiology and Perioperative Medicine, The 900 Hospital of the Joint Logistic Support Force of the People's Liberation Army of China, Fuzhou 350025, China
| | - Jun Que
- Department of Anesthesiology and Perioperative Medicine, The 900 Hospital of the Joint Logistic Support Force of the People's Liberation Army of China, Fuzhou 350025, China
| | - Minsheng Liu
- Department of Anesthesiology and Perioperative Medicine, The 900 Hospital of the Joint Logistic Support Force of the People's Liberation Army of China, Fuzhou 350025, China.
| |
Collapse
|
3
|
Amin N, Abbasi IN, Wu F, Shi Z, Sundus J, Badry A, Yuan X, Zhao BX, Pan J, Mi XD, Luo Y, Geng Y, Fang M. The Janus face of HIF-1α in ischemic stroke and the possible associated pathways. Neurochem Int 2024; 177:105747. [PMID: 38657682 DOI: 10.1016/j.neuint.2024.105747] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Stroke is the most devastating disease, causing paralysis and eventually death. Many clinical and experimental trials have been done in search of a new safe and efficient medicine; nevertheless, scientists have yet to discover successful remedies that are also free of adverse effects. This is owing to the variability in intensity, localization, medication routes, and each patient's immune system reaction. HIF-1α represents the modern tool employed to treat stroke diseases due to its functions: downstream genes such as glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Its role can be achieved via two downstream EPO and VEGF strongly related to apoptosis and antioxidant processes. Recently, scientists paid more attention to drugs dealing with the HIF-1 pathway. This review focuses on medicines used for ischemia treatment and their potential HIF-1α pathways. Furthermore, we discussed the interaction between HIF-1α and other biological pathways such as oxidative stress; however, a spotlight has been focused on certain potential signalling contributed to the HIF-1α pathway. HIF-1α is an essential regulator of oxygen balance within cells which affects and controls the expression of thousands of genes related to sustaining homeostasis as oxygen levels fluctuate. HIF-1α's role in ischemic stroke strongly depends on the duration and severity of brain damage after onset. HIF-1α remains difficult to investigate, particularly in ischemic stroke, due to alterations in the acute and chronic phases of the disease, as well as discrepancies between the penumbra and ischemic core. This review emphasizes these contrasts and analyzes the future of this intriguing and demanding field.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Zoology, Faculty of Science, Aswan University, Egypt; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Irum Naz Abbasi
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Javaria Sundus
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Yuan
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jie Pan
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiao-Dan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Marong Fang
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
4
|
Besteher B, Rocktäschel T, Garza AP, Machnik M, Ballez J, Helbing DL, Finke K, Reuken P, Güllmar D, Gaser C, Walter M, Opel N, Rita Dunay I. Cortical thickness alterations and systemic inflammation define long-COVID patients with cognitive impairment. Brain Behav Immun 2024; 116:175-184. [PMID: 38036270 DOI: 10.1016/j.bbi.2023.11.028] [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: 07/20/2023] [Revised: 10/26/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
As the heterogeneity of symptoms is increasingly recognized among long-COVID patients, it appears highly relevant to study potential pathophysiological differences along the different subtypes. Preliminary evidence suggests distinct alterations in brain structure and systemic inflammatory patterns in specific groups of long-COVID patients. To this end, we analyzed differences in cortical thickness and peripheral immune signature between clinical subgroups based on 3 T-MRI scans and signature inflammatory markers in n = 120 participants comprising healthy never-infected controls (n = 30), healthy COVID-19 survivors (n = 29), and subgroups of long-COVID patients with (n = 26) and without (n = 35) cognitive impairment according to screening with Montreal Cognitive Assessment. Whole-brain comparison of cortical thickness between the 4 groups was conducted by surface-based morphometry. We identified distinct cortical areas showing a progressive increase in cortical thickness across different groups, starting from healthy individuals who had never been infected with COVID-19, followed by healthy COVID-19 survivors, long-COVID patients without cognitive deficits (MoCA ≥ 26), and finally, long-COVID patients exhibiting significant cognitive deficits (MoCA < 26). These findings highlight the continuum of cortical thickness alterations associated with COVID-19, with more pronounced changes observed in individuals experiencing cognitive impairment (p < 0.05, FWE-corrected). Affected cortical regions covered prefrontal and temporal gyri, insula, posterior cingulate, parahippocampal gyrus, and parietal areas. Additionally, we discovered a distinct immunophenotype, with elevated levels of IL-10, IFNγ, and sTREM2 in long-COVID patients, especially in the group suffering from cognitive impairment. We demonstrate lingering cortical and immunological alterations in healthy and impaired subgroups of COVID-19 survivors. This implies a complex underlying pathomechanism in long-COVID and emphasizes the necessity to investigate the whole spectrum of post-COVID biology to determine targeted treatment strategies targeting specific sub-groups.
Collapse
Affiliation(s)
- Bianca Besteher
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany.
| | - Tonia Rocktäschel
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Alejandra P Garza
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University, Magdeburg, Germany
| | - Marlene Machnik
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany
| | - Johanna Ballez
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany
| | - Dario-Lucas Helbing
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Kathrin Finke
- Department of Neurology, Jena University Hospital, Germany
| | - Philipp Reuken
- Department of Internal Medicine IV, Gastroenterology, Hepatology and Infectious Diseases, Jena University Hospital, Germany
| | - Daniel Güllmar
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Germany
| | - Christian Gaser
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany; Department of Neurology, Jena University Hospital, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Nils Opel
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Germany; German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Ildiko Rita Dunay
- German Center for Mental Health (DZPG), Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany; Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
5
|
Wagner T, Bangoura B, Wiedmer S, Daugschies A, Dunay IR. Phytohormones regulate asexual Toxoplasma gondii replication. Parasitol Res 2023; 122:2835-2846. [PMID: 37725257 DOI: 10.1007/s00436-023-07968-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
The protozoan Toxoplasma gondii (T. gondii) is a zoonotic disease agent causing systemic infection in warm-blooded intermediate hosts including humans. During the acute infection, the parasite infects host cells and multiplies intracellularly in the asexual tachyzoite stage. In this stage of the life cycle, invasion, multiplication, and egress are the most critical events in parasite replication. T. gondii features diverse cell organelles to support these processes, including the apicoplast, an endosymbiont-derived vestigial plastid originating from an alga ancestor. Previous studies have highlighted that phytohormones can modify the calcium-mediated secretion, e.g., of adhesins involved in parasite movement and cell invasion processes. The present study aimed to elucidate the influence of different plant hormones on the replication of asexual tachyzoites in a human foreskin fibroblast (HFF) host cell culture. T. gondii replication was measured by the determination of T. gondii DNA copies via qPCR. Three selected phytohormones, namely abscisic acid (ABA), gibberellic acid (GIBB), and kinetin (KIN) as representatives of different plant hormone groups were tested. Moreover, the influence of typical cell culture media components on the phytohormone effects was assessed. Our results indicate that ABA is able to induce a significant increase of T. gondii DNA copies in a typical supplemented cell culture medium when applied in concentrations of 20 ng/μl or 2 ng/μl, respectively. In contrast, depending on the culture medium composition, GIBB may potentially serve as T. gondii growth inhibitor and may be further investigated as a potential treatment for toxoplasmosis.
Collapse
Affiliation(s)
- Tina Wagner
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Berit Bangoura
- Department of Veterinary Sciences, Wyoming State Veterinary Laboratory, University of Wyoming, Laramie, WY, 82070, USA.
| | - Stefanie Wiedmer
- Faculty of Biology, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Arwid Daugschies
- Institute of Parasitology, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
6
|
Mendoza-Larios LA, García-Dolores F, Sánchez-Anguiano LF, Antuna-Salcido EI, Hernández-Tinoco J, Rocha-Salais A, Segoviano-Mendoza MA, Sifuentes-Álvarez A, Alvarado-Esquivel C. Toxoplasma gondii infection of the central nervous system and suicide: A case-control study of decedents. Eur J Microbiol Immunol (Bp) 2023; 13:77-82. [PMID: 37982867 PMCID: PMC10668923 DOI: 10.1556/1886.2023.00033] [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: 09/07/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
We sought to determine the association between Toxoplasma gondii (T. gondii) infection of the central nervous system and suicide in a sample of decedents in Mexico City. One hundred and forty-seven decedents (87 who committed suicide and 60 who did not commit suicide) were studied. Brain tissues (amygdala and prefrontal cortex) of decedents were examined for the detection of T. gondii using immunohistochemistry. Detection of T. gondii was positive in 7 (8.0%) of the 87 cases (6 found in prefrontal cortex and one in amygdala), and in one (1.7%) of the 60 controls (found in prefrontal cortex) (OR: 5.16; 95% CI: 0.61-43.10; P = 0.14). Results suggest that T. gondii infection in brain is not associated with suicide. Further studies to confirm this finding are needed.
Collapse
Affiliation(s)
- Laura Alejandra Mendoza-Larios
- Instituto de Ciencias Forenses de la Ciudad de México, Avenida Niños Héroes 130, Colonia Doctores, Delegación Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Fernando García-Dolores
- Instituto de Ciencias Forenses de la Ciudad de México, Avenida Niños Héroes 130, Colonia Doctores, Delegación Cuauhtémoc, 06720 Ciudad de México, Mexico
| | - Luis Francisco Sánchez-Anguiano
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juárez University of Durango State, Avenida Universidad S/N. 34000 Durango, Mexico
| | - Elizabeth Irasema Antuna-Salcido
- Institute for Scientific Research “Dr. Roberto Rivera Damm”, Juárez University of Durango State, Avenida Universidad S/N, 34000 Durango, Mexico
| | - Jesús Hernández-Tinoco
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juárez University of Durango State, Avenida Universidad S/N. 34000 Durango, Mexico
| | - Adriana Rocha-Salais
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juárez University of Durango State, Avenida Universidad S/N. 34000 Durango, Mexico
| | - Marcela Araceli Segoviano-Mendoza
- Department of Microbiology, Faculty of Medicine and Nutrition, Juárez University of Durango State, Avenida Universidad S/N. 34000 Durango, Mexico
| | - Antonio Sifuentes-Álvarez
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juárez University of Durango State, Avenida Universidad S/N. 34000 Durango, Mexico
| | - Cosme Alvarado-Esquivel
- Biomedical Research Laboratory, Faculty of Medicine and Nutrition, Juárez University of Durango State, Avenida Universidad S/N. 34000 Durango, Mexico
| |
Collapse
|
7
|
Figueiredo CA, Düsedau HP, Steffen J, Ehrentraut S, Dunay MP, Toth G, Reglödi D, Heimesaat MM, Dunay IR. The neuropeptide PACAP alleviates T. gondii infection-induced neuroinflammation and neuronal impairment. J Neuroinflammation 2022; 19:274. [PMCID: PMC9675261 DOI: 10.1186/s12974-022-02639-z] [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: 04/13/2022] [Accepted: 10/23/2022] [Indexed: 11/21/2022] Open
Abstract
Background Cerebral infection with the protozoan Toxoplasma gondii (T. gondii) is responsible for inflammation of the central nervous system (CNS) contributing to subtle neuronal alterations. Albeit essential for brain parasite control, continuous microglia activation and recruitment of peripheral immune cells entail distinct neuronal impairment upon infection-induced neuroinflammation. PACAP is an endogenous neuropeptide known to inhibit inflammation and promote neuronal survival. Since PACAP is actively transported into the CNS, we aimed to assess the impact of PACAP on the T. gondii-induced neuroinflammation and subsequent effects on neuronal homeostasis. Methods Exogenous PACAP was administered intraperitoneally in the chronic stage of T. gondii infection, and brains were isolated for histopathological analysis and determination of pathogen levels. Immune cells from the brain, blood, and spleen were analyzed by flow cytometry, and the further production of inflammatory mediators was investigated by intracellular protein staining as well as expression levels by RT-qPCR. Neuronal and synaptic alterations were assessed on the transcriptional and protein level, focusing on neurotrophins, neurotrophin-receptors and signature synaptic markers. Results Here, we reveal that PACAP administration reduced the inflammatory foci and the number of apoptotic cells in the brain parenchyma and restrained the activation of microglia and recruitment of monocytes. The neuropeptide reduced the expression of inflammatory mediators such as IFN-γ, IL-6, iNOS, and IL-1β. Moreover, PACAP diminished IFN-γ production by recruited CD4+ T cells in the CNS. Importantly, PACAP promoted neuronal health via increased expression of the neurotrophin BDNF and reduction of p75NTR, a receptor related to neuronal cell death. In addition, PACAP administration was associated with increased expression of transporters involved in glutamatergic and GABAergic signaling that are particularly affected during cerebral toxoplasmosis. Conclusions Together, our findings unravel the beneficial effects of exogenous PACAP treatment upon infection-induced neuroinflammation, highlighting the potential implication of neuropeptides to promote neuronal survival and minimize synaptic prejudice. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02639-z.
Collapse
Affiliation(s)
- Caio Andreeta Figueiredo
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany
| | - Henning Peter Düsedau
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany
| | - Johannes Steffen
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany
| | - Stefanie Ehrentraut
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany
| | - Miklos P. Dunay
- grid.483037.b0000 0001 2226 5083Department and Clinic of Surgery and Ophthalmology, University of Veterinary Medicine, Budapest, Hungary
| | - Gabor Toth
- grid.9008.10000 0001 1016 9625Department of Medical Chemistry, University of Szeged, Budapest, Hungary
| | - Dora Reglödi
- grid.9679.10000 0001 0663 9479Department of Anatomy, MTA-PTE PACAP Research Team and Szentagothai Research Center, University of Pecs Medical School, Pecs, Hungary
| | - Markus M. Heimesaat
- grid.6363.00000 0001 2218 4662Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Ildiko Rita Dunay
- grid.5807.a0000 0001 1018 4307Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology and Inflammation (GC-I3), Otto-Von-Guericke University, Magdeburg, Germany ,grid.418723.b0000 0001 2109 6265Center for Behavioral Brain Sciences – CBBS, Magdeburg, Germany
| |
Collapse
|
8
|
Experimental infection of Toxoplasma gondii in specific pathogen-free and commercial broiler chicks. Comp Immunol Microbiol Infect Dis 2022; 90-91:101890. [DOI: 10.1016/j.cimid.2022.101890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/18/2022] [Accepted: 09/29/2022] [Indexed: 11/20/2022]
|
9
|
Dimasuay KG, Schaunaman N, Berg B, Cervantes D, Kruger E, Heppner FL, Ferrington DA, Chu HW. Airway epithelial immunoproteasome subunit LMP7 protects against rhinovirus infection. Sci Rep 2022; 12:14507. [PMID: 36008456 PMCID: PMC9403975 DOI: 10.1038/s41598-022-18807-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/19/2022] [Indexed: 11/20/2022] Open
Abstract
Immunoproteasomes (IP) serve as an important modulator of immune responses to pathogens and other pathological factors. LMP7/β5i, one of the IP subunits, plays a critical role in autoimmune diseases by downregulating inflammation. Rhinovirus (RV) infection is a major risk factor in the exacerbations of respiratory inflammatory diseases, but whether LMP7 regulates RV-mediated inflammation in the lung particularly in the airway epithelium, the first line of defense against RV infection, remains unclear. In this study, we determined whether airway epithelial LMP7 promotes the resolution of RV-mediated lung inflammation. Inducible airway epithelial-specific LMP7-deficient (conditional knockout, CKO) mice were generated to reveal the in vivo anti-inflammatory and antiviral functions of LMP7. By using LMP7-deficient primary human airway epithelial cells generated by CRISPR-Cas9, we confirmed that airway epithelial LMP7 decreased pro-inflammatory cytokines and viral load during RV infection. Additionally, airway epithelial LMP7 enhanced the expression of a negative immune regulator A20/TNFAIP3 during viral infection that may contribute to the anti-inflammatory function of LMP7. We also discovered that induction of LMP7 by a low dose of polyinosinic:polycytidylic acid (PI:C) reduced RV-mediated inflammation in our CKO mice infected with RV. Our findings suggest that airway epithelial LMP7 has anti-inflammatory and antiviral functions that is critical to the resolution of RV-mediated lung inflammation. Induction of airway epithelial LMP7 may open a novel avenue for therapeutic intervention against RV infection.
Collapse
Affiliation(s)
| | - Niccolette Schaunaman
- grid.240341.00000 0004 0396 0728Department of Medicine, National Jewish Health, Denver, CO USA
| | - Bruce Berg
- grid.240341.00000 0004 0396 0728Department of Medicine, National Jewish Health, Denver, CO USA
| | - Diana Cervantes
- grid.240341.00000 0004 0396 0728Department of Medicine, National Jewish Health, Denver, CO USA
| | - Elke Kruger
- grid.412469.c0000 0000 9116 8976Institute for Medicine Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Frank L. Heppner
- grid.6363.00000 0001 2218 4662Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Deborah A. Ferrington
- grid.19006.3e0000 0000 9632 6718Doheny Eye Institute, University of California Los Angeles, Pasadena, CA USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO, USA.
| |
Collapse
|
10
|
Oliveri F, Basler M, Rao TN, Fehling HJ, Groettrup M. Immunoproteasome Inhibition Reduces the T Helper 2 Response in Mouse Models of Allergic Airway Inflammation. Front Immunol 2022; 13:870720. [PMID: 35711460 PMCID: PMC9197384 DOI: 10.3389/fimmu.2022.870720] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background Allergic asthma is a chronic disease and medical treatment often fails to fully control the disease in the long term, leading to a great need for new therapeutic approaches. Immunoproteasome inhibition impairs T helper cell function and is effective in many (auto-) inflammatory settings but its effect on allergic airway inflammation is unknown. Methods Immunoproteasome expression was analyzed in in vitro polarized T helper cell subsets. To study Th2 cells in vivo acute allergic airway inflammation was induced in GATIR (GATA-3-vYFP reporter) mice using ovalbumin and house dust mite extract. Mice were treated with the immunoproteasome inhibitor ONX 0914 or vehicle during the challenge phase and the induction of airway inflammation was analyzed. Results In vitro polarized T helper cell subsets (Th1, Th2, Th17, and Treg) express high levels of immunoproteasome subunits. GATIR mice proved to be a useful tool for identification of Th2 cells. Immunoproteasome inhibition reduced the Th2 response in both airway inflammation models. Furthermore, T cell activation and antigen-specific cytokine secretion was impaired and a reduced infiltration of eosinophils and professional antigen-presenting cells into the lung and the bronchoalveolar space was observed in the ovalbumin model. Conclusion These results show the importance of the immunoproteasome in Th2 cells and airway inflammation. Our data provides first insight into the potential of using immunoproteasome inhibition to target the aberrant Th2 response, e.g. in allergic airway inflammation.
Collapse
Affiliation(s)
- Franziska Oliveri
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Michael Basler
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| | | | | | - Marcus Groettrup
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
- *Correspondence: Marcus Groettrup,
| |
Collapse
|
11
|
Steffen J, Ehrentraut S, Bank U, Biswas A, Figueiredo CA, Hölsken O, Düsedau HP, Dovhan V, Knop L, Thode J, Romero-Suárez S, Duarte CI, Gigley J, Romagnani C, Diefenbach A, Klose CSN, Schüler T, Dunay IR. Type 1 innate lymphoid cells regulate the onset of Toxoplasma gondii-induced neuroinflammation. Cell Rep 2022; 38:110564. [PMID: 35354032 DOI: 10.1016/j.celrep.2022.110564] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 12/21/2021] [Accepted: 03/03/2022] [Indexed: 12/31/2022] Open
Abstract
Cerebral infections are restrained by a complex interplay of tissue-resident and recruited peripheral immune cells. Whether innate lymphoid cells (ILCs) are involved in the orchestration of the neuroinflammatory dynamics is not fully understood. Here, we demonstrate that ILCs accumulate in the cerebral parenchyma, the choroid plexus, and the meninges in the onset of cerebral Toxoplasma gondii infection. Antibody-mediated depletion of conventional natural killer (cNK) cells and ILC1s in the early stage of infection results in diminished cytokine and chemokine expression and increased cerebral parasite burden. Using cNK- and ILC1-deficient murine models, we demonstrate that exclusively the lack of ILC1s affects cerebral immune responses. In summary, our results provide evidence that ILC1s are an early source of IFN-γ and TNF in response to cerebral T. gondii infection, thereby inducing host defense factors and initiating the development of a neuroinflammatory response.
Collapse
Affiliation(s)
- Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Stefanie Ehrentraut
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Ute Bank
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto-von-Guericke University, Magdeburg, Germany
| | - Aindrila Biswas
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Caio Andreeta Figueiredo
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Oliver Hölsken
- Mucosal and Developmental Immunology, German Rheuma Research Center Berlin (DRFZ), Berlin, Germany; Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases, and Immunology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Vladyslava Dovhan
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto-von-Guericke University, Magdeburg, Germany
| | - Laura Knop
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto-von-Guericke University, Magdeburg, Germany
| | - Jacqueline Thode
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Silvina Romero-Suárez
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Infante Duarte
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jason Gigley
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | - Chiara Romagnani
- Innate Immunity, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany; Medical Department I, Charité - Universitätsmedizin, Berlin, Germany
| | - Andreas Diefenbach
- Mucosal and Developmental Immunology, German Rheuma Research Center Berlin (DRFZ), Berlin, Germany; Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases, and Immunology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Christoph S N Klose
- Neuro-immune Interactions, Institute of Microbiology, Infectious Diseases, and Immunology, Charité - Universitätsmedizin, Berlin, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto-von-Guericke University, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Health Campus Immunology, Infectiology, and Inflammation (GC-I(3)), Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
12
|
Targeted inhibition of the immunoproteasome blocks endothelial MHC class II antigen presentation to CD4 + T cells in chronic liver injury. Int Immunopharmacol 2022; 107:108639. [PMID: 35219165 DOI: 10.1016/j.intimp.2022.108639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2023]
Abstract
Chronic or overwhelming liver injury is frequently associated with fibrosis, which is the main histological characteristic of non-alcoholic steatohepatitis (NASH). Currently, there is no effective treatment for liver fibrosis. Adaptive immunity is one of the perpetrators of liver inflammation and involves the antigen-specific activation of lymphocytes. Targeting adaptive immunity has been proposed as a novel therapeutic approach for NASH. In this study, we demonstrated that liver endothelial cells contribute to MHC class II (MHC-II) antigen presentation to CD4+ T cells after chronic liver injury. In human cirrhotic liver samples, we observed an increased expression of endothelial MHC-II and of the antigen presentation-associated protein LMP7, which is one of the proteolytically active subunits of the immunoproteasome. In a CCl4-induced chronic injury model or a diet- and chemical-induced NASH model, endothelial MHC-II and LMP7 expression was induced to increase. PR-957, a selective inhibitor of the immunoproteasome, inhibited MHC-II expression in endothelial cells and CD4+ T cell response after chronic liver injury. In vitro experiment demonstrated PR-957 also reversed IFN-γ-induced upregulation of MHC-II in endothelial cells. Furthermore, PR-957 treatment or CD4+ T cell depletion in chronic liver injury alleviated liver fibrosis and reduced inflammation, as indicated by the downregulation of inflammatory response markers (F4/80, IL-1, IL-6 and IL-18). In conclusion, targeted inhibition of the immunoproteasome blocks endothelial MHC-II antigen presentation to CD4+ T cells in chronic liver injury. In this regard, the PR-957 inhibitor is a promising candidate for the development of future therapies against NASH.
Collapse
|
13
|
Ross EC, Olivera GC, Barragan A. Early passage of Toxoplasma gondii across the blood–brain barrier. Trends Parasitol 2022; 38:450-461. [DOI: 10.1016/j.pt.2022.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/29/2022]
|
14
|
On the Role of the Immunoproteasome in Protein Homeostasis. Cells 2021; 10:cells10113216. [PMID: 34831438 PMCID: PMC8621243 DOI: 10.3390/cells10113216] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/28/2022] Open
Abstract
Numerous cellular processes are controlled by the proteasome, a multicatalytic protease in the cytosol and nucleus of all eukaryotic cells, through regulated protein degradation. The immunoproteasome is a special type of proteasome which is inducible under inflammatory conditions and constitutively expressed in hematopoietic cells. MECL-1 (β2i), LMP2 (β1i), and LMP7 (β5i) are the proteolytically active subunits of the immunoproteasome (IP), which is known to shape the antigenic repertoire presented on major histocompatibility complex (MHC) class I molecules. Furthermore, the immunoproteasome is involved in T cell expansion and inflammatory diseases. In recent years, targeting the immunoproteasome in cancer, autoimmune diseases, and transplantation proved to be therapeutically effective in preclinical animal models. However, the prime function of standard proteasomes and immunoproteasomes is the control of protein homeostasis in cells. To maintain protein homeostasis in cells, proteasomes remove proteins which are not properly folded, which are damaged by stress conditions such as reactive oxygen species formation, or which have to be degraded on the basis of regular protein turnover. In this review we summarize the latest insights on how the immunoproteasome influences protein homeostasis.
Collapse
|