1
|
Brady S, Poulton J, Muller S. Inclusion body myositis: Correcting impaired mitochondrial and lysosomal autophagy as a potential therapeutic strategy. Autoimmun Rev 2024; 23:103644. [PMID: 39306221 DOI: 10.1016/j.autrev.2024.103644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Inclusion body myositis (IBM) is a late onset sporadic myopathy with a characteristic clinical presentation, but as yet unknown aetiology or effective treatment. Typical clinical features are early predominant asymmetric weakness of finger flexor and knee extensor muscles. Muscle biopsy shows endomysial inflammatory infiltrate, mitochondrial changes, and protein aggregation. Proteostasis (protein turnover) appears to be impaired, linked to potentially dysregulated chaperone-mediated autophagy and mitophagy (a type of mitochondrial quality control). In this review, we bring together the most recent clinical and biological data describing IBM. We then address the question of diagnosing this pathology and the relevance of the current biological markers that characterize IBM. In these descriptions, we put a particular emphasis on data related to the deregulation of autophagic processes and to the mitochondrial-lysosomal crosstalk. Finally, after a short description of current treatments, an overview is provided pointing towards novel therapeutic targets and emerging regulatory molecules that are being explored for treating IBM. Special attention is paid to autophagy inhibitors that may offer innovative breakthrough therapies for patients with IBM.
Collapse
Affiliation(s)
- Stefen Brady
- Oxford Adult Muscle Service, John Radcliffe Hospital, Oxford University Hospital Trust, Oxford, UK
| | - Joanna Poulton
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Sylviane Muller
- CNRS and Strasbourg University Unit Biotechnology and Cell signalling/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France; University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.
| |
Collapse
|
2
|
Dong Y, Wang T, Wu H. The role of cytokines from salivary gland epithelial cells in the immunopathology of Sjögren's syndrome. Front Immunol 2024; 15:1443455. [PMID: 39346911 PMCID: PMC11427401 DOI: 10.3389/fimmu.2024.1443455] [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: 06/04/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
In the pathogenesis and progression of Sjögren's syndrome (SS), hematopoietic cells in the peripheral circulation, tissue-resident immune cells, and parenchymal cells of salivary gland tissues (such as epithelial cells, endothelial cells, fibroblasts, etc.) all play crucial roles. These diverse cells form intricate networks and interact with each other, leading to tissue destruction and persistent chronic inflammation, ultimately causing irreversible damage in glandular function. Among these, salivary gland epithelial cells (SGECs) consistently hold a key position, characterized by their functions in expressing co-stimulatory and antigen-presenting molecules and secreting pro-inflammatory cytokines and chemokines. Moreover, SGECs actively engage in and facilitate the development of specific pathological structures within the salivary gland, such as lymphoepithelial lesions (LELs) and tertiary lymphoid structures (TLSs), thereby substantially elevating the risk of mucosa-associated lymphoid tissue (MALT) lymphoma. Overall, SGECs are recognized for their essential and irreplaceable contributions to the pathogenesis of SS. This review article initially delves into the anatomical composition of salivary gland epithelial cells, subsequently focusing on elucidating the different cytokines derived from SGECs, encompassing chemokines, pro-inflammatory cytokines, anti-inflammatory cytokines, pro-survival cytokines, and damage-associated molecular patterns (DAMPs), to explore their key roles in the pathogenesis of SS.
Collapse
Affiliation(s)
- Yuanji Dong
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ting Wang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huaxiang Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Zhou J, Pathak JL, Liu Q, Hu S, Cao T, Watanabe N, Huo Y, Li J. Modes and Mechanisms of Salivary Gland Epithelial Cell Death in Sjogren's Syndrome. Adv Biol (Weinh) 2023; 7:e2300173. [PMID: 37409392 DOI: 10.1002/adbi.202300173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/16/2023] [Indexed: 07/07/2023]
Abstract
Sjogren's syndrome is an autoimmune disease in middle and old-aged women with a dry mucosal surface, which is caused by the dysfunction of secretory glands, such as the oral cavity, eyeballs, and pharynx. Pathologically, Sjogren's syndrome are characterized by lymphocyte infiltration into the exocrine glands and epithelial cell destruction caused by autoantibodies Ro/SSA and La/SSB. At present, the exact pathogenesis of Sjogren's syndrome is unclear. Evidence suggests epithelial cell death and the subsequent dysfunction of salivary glands as the main causes of xerostomia. This review summarizes the modes of salivary gland epithelial cell death and their role in Sjogren's syndrome progression. The molecular mechanisms involved in salivary gland epithelial cell death during Sjogren's syndrome as potential leads to treating the disease are also discussed.
Collapse
Affiliation(s)
- Jiannan Zhou
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Janak Lal Pathak
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Qianwen Liu
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Shilin Hu
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Tingting Cao
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Nobumoto Watanabe
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama, 351-0198, Japan
| | - Yongliang Huo
- Experimental Animal Center, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Jiang Li
- Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| |
Collapse
|
4
|
Nakamura H, Tanaka T, Zheng C, Afione SA, Warner BM, Noguchi M, Atsumi T, Chiorini JA. Lysosome-Associated Membrane Protein 3 Induces Lysosome-Dependent Cell Death by Impairing Autophagic Caspase 8 Degradation in the Salivary Glands of Individuals With Sjögren's Disease. Arthritis Rheumatol 2023; 75:1586-1598. [PMID: 37096570 PMCID: PMC11132095 DOI: 10.1002/art.42540] [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: 03/03/2023] [Revised: 03/03/2023] [Accepted: 04/10/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE Lysosome-associated membrane protein 3 (LAMP3) overexpression is implicated in the development and progression of Sjögren's disease (SjD) by inducing lysosomal membrane permeabilization (LMP) and apoptotic cell death in salivary gland epithelium. The aim of this study was to clarify the molecular details of LAMP3-induced lysosome-dependent cell death and to test lysosomal biogenesis as a therapeutic intervention. METHODS Human labial minor salivary gland biopsies were analyzed using immunofluorescence staining for LAMP3 expression levels and galectin-3 puncta formation, a marker of LMP. Expression level of caspase 8, an initiator of LMP, was determined by Western blotting in cell culture. Galectin-3 puncta formation and apoptosis were evaluated in cell cultures and a mouse model treated with glucagon-like peptide 1 receptor (GLP-1R) agonists, a known promoter of lysosomal biogenesis. RESULTS Galectin-3 puncta formation was more frequent in the salivary glands of SjD patients compared to control glands. The proportion of galectin-3 puncta-positive cells was positively correlated with LAMP3 expression levels in the glands. LAMP3 overexpression increased caspase 8 expression, and knockdown of caspase 8 decreased galectin-3 puncta formation and apoptosis in LAMP3-overexpressing cells. Inhibition of autophagy increased caspase 8 expression, while restoration of lysosomal function using GLP-1R agonists decreased caspase 8 expression, which reduced galectin-3 puncta formation and apoptosis in both LAMP3-overexpressing cells and mice. CONCLUSION LAMP3 overexpression induced lysosomal dysfunction, resulting in lysosome-dependent cell death via impaired autophagic caspase 8 degradation, and restoring lysosomal function using GLP-1R agonists could prevent this. These findings suggested that LAMP3-induced lysosomal dysfunction is central to disease development and is a target for therapeutic intervention in SjD.
Collapse
Affiliation(s)
- Hiroyuki Nakamura
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Tsutomu Tanaka
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Changyu Zheng
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Sandra A Afione
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Blake M. Warner
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Masayuki Noguchi
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - John A. Chiorini
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Muller S. The abscopal effect: Implications for drug discovery in autoimmunity. Autoimmun Rev 2023; 22:103315. [PMID: 36924921 DOI: 10.1016/j.autrev.2023.103315] [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: 02/24/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
The emergence of novel targeted therapies and the tools that increase the stability and delivery of drugs have greatly improved treatment outcomes in autoimmune diseases (ADs). Recently-developed strategies deplete specific deleterious T- and B-cell subsets, interrupt receptor-ligand interactions, and/or inhibit the secretion or activity of inflammatory mediators linked to tissue damage. Although generally efficient, these lines of intervention have limitations, with documented cases of drug-resistance and undesired side effects. They are also difficult to apply to non-organ-specific ADs, where the trigger and effector antigens are unknown and in which autoimmune activity is widely spread throughout the body. The potential of cellular modulators that act at a distance from the affected site, by abscopal effect, as described in the case of cancer radio- and immuno-therapy might be especially efficient in the context of ADs. Future research to discover small molecule- and peptide-based treatments will need to explore potential drugs with abscopal effects that could elicit potent immune tolerance and clinical quiescence to restore quality of life of affected patients.
Collapse
Affiliation(s)
- Sylviane Muller
- CNRS and Strasbourg University Unit Biotechnology and Cell signalling/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France; Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France; University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.
| |
Collapse
|
6
|
The role of lysosomes in metabolic and autoimmune diseases. Nat Rev Nephrol 2023; 19:366-383. [PMID: 36894628 DOI: 10.1038/s41581-023-00692-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2023] [Indexed: 03/11/2023]
Abstract
Lysosomes are catabolic organelles that contribute to the degradation of intracellular constituents through autophagy and of extracellular components through endocytosis, phagocytosis and macropinocytosis. They also have roles in secretory mechanisms, the generation of extracellular vesicles and certain cell death pathways. These functions make lysosomes central organelles in cell homeostasis, metabolic regulation and responses to environment changes including nutrient stresses, endoplasmic reticulum stress and defects in proteostasis. Lysosomes also have important roles in inflammation, antigen presentation and the maintenance of long-lived immune cells. Their functions are tightly regulated by transcriptional modulation via TFEB and TFE3, as well as by major signalling pathways that lead to activation of mTORC1 and mTORC2, lysosome motility and fusion with other compartments. Lysosome dysfunction and alterations in autophagy processes have been identified in a wide variety of diseases, including autoimmune, metabolic and kidney diseases. Deregulation of autophagy can contribute to inflammation, and lysosomal defects in immune cells and/or kidney cells have been reported in inflammatory and autoimmune pathologies with kidney involvement. Defects in lysosomal activity have also been identified in several pathologies with disturbances in proteostasis, including autoimmune and metabolic diseases such as Parkinson disease, diabetes mellitus and lysosomal storage diseases. Targeting lysosomes is therefore a potential therapeutic strategy to regulate inflammation and metabolism in a variety of pathologies.
Collapse
|
7
|
Liu Y, Tan YQ, Zhou G. Melatonin: a potential therapeutic approach for the management of primary Sjögren's syndrome. Immunol Res 2023; 71:373-387. [PMID: 36715831 DOI: 10.1007/s12026-023-09360-w] [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: 07/19/2022] [Accepted: 01/03/2023] [Indexed: 01/31/2023]
Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune disease that primarily affects the exocrine glands and is mainly characterized by sicca symptoms of the eyes and mouth. Approximately 30-50% of pSS patients develop systemic multi-organ disorders including malignant lymphoma. The etiology of pSS is not well understood; growing evidence suggests that uncontrolled immune/inflammatory responses, excessive oxidative stress, defected apoptosis, dysregulated autophagy, exosomes, and exogenous virus infections may participate in the pathogenesis of pSS. There is no ideal therapeutic method for pSS; the management of pSS is mainly palliative, which aims to alleviate sicca symptoms. Melatonin, as the main secretory product of the pineal gland, has been evidenced to show various physiological functions, including effects of immunoregulation, capability of antioxidation, moderation of autophagy, suppressive activities of apoptosis, regulative capacity of exosomes, properties of anti-infection, and improvement of sleep. The beneficial effects of melatonin have been already validated in some autoimmune diseases such as multiple sclerosis (MS), type 1 diabetes mellitus (T1DM), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD). However, our previous research firstly revealed that melatonin might inhibit pathogenic responses of peripheral Th17 and double-negative (DN) T cells in pSS. More importantly, melatonin administration alleviated the development of pSS in animal models with reduced infiltrating lymphocytes, improved functional activity of salivary gland, and decreased production of inflammatory factors as well as autoantibodies. Owing to the important biological properties reported in melatonin are characteristics closely related to the treatment of pSS; the potential role and underlying mechanisms of melatonin in the administration of pSS are certainly worth further investigations. Consequently, the aim of this review is to give a deep insight to the therapeutic potency of melatonin for pSS.
Collapse
Affiliation(s)
- Yi Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ya-Qin Tan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, China
| | - Gang Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China. .,Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, China.
| |
Collapse
|
8
|
Renaudineau Y, Muller S, Hedrich CM, Chauveau D, Bellière J, De Almeida S, Damoiseaux J, Scherlinger M, Guery JC, Sailler L, Bost C. Immunological and translational key challenges in systemic lupus erythematosus: A symposium update. J Transl Autoimmun 2023; 6:100199. [PMID: 37065621 PMCID: PMC10090709 DOI: 10.1016/j.jtauto.2023.100199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
The first LBMR-Tim (Toulouse Referral Medical Laboratory of Immunology) symposium convened on December 16, 2022 in Toulouse, France to address challenging questions in systemic lupus erythematosus (SLE). Special focus was put on (i) the role played by genes, sex, TLR7, and platelets on SLE pathophysiology; (ii) autoantibodies, urinary proteins, and thrombocytopenia contribution at the time of diagnosis and during follow-up; (iii) neuropsychiatric involvement, vaccine response in the COVID-19 era, and lupus nephritis management at the clinical frontline; and (iv) therapeutic perspectives in patients with lupus nephritis and the unexpected adventure of the Lupuzor/P140 peptide. The multidisciplinary panel of experts further supports the concept that a global approach including basic sciences, translational research, clinical expertise, and therapeutic development have to be prioritized in order to better understand and then improve the management of this complex syndrome.
Collapse
|
9
|
The Therapeutic Effect of Phosphopeptide P140 Attenuates Inflammation Induced by Uric Acid Crystals in Gout Arthritis Mouse Model. Cells 2022; 11:cells11233709. [PMID: 36496970 PMCID: PMC9740613 DOI: 10.3390/cells11233709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Gout is a painful form of inflammatory arthritis characterized by the deposition of monosodium urate (MSU) crystals in the joints. The aim of this study was to investigate the effect of peptide P140 on the inflammatory responses in crystal-induced mouse models of gout and cell models including MSU-treated human cells. Injection of MSU crystals into the knee joint of mice induced neutrophil influx and inflammatory hypernociception. Injection of MSU crystals subcutaneously into the hind paw induced edema and increased pro-inflammatory cytokines levels. Treatment with P140 effectively reduced hypernociception, the neutrophil influx, and pro-inflammatory cytokine levels in these experimental models. Furthermore, P140 modulated neutrophils chemotaxis in vitro and increased apoptosis pathways through augmented caspase 3 activity and reduced NFκB phosphorylation. Moreover, P140 increased the production of the pro-resolving mediator annexin A1 and decreased the expression of the autophagy-related ATG5-ATG12 complex and HSPA8 chaperone protein. Overall, these findings suggest that P140 exerts a significant beneficial effect in a neutrophilic inflammation observed in the model of gout that can be of special interest in the design of new therapeutic strategies.
Collapse
|
10
|
Ling Y, Liang H, Tang Q. Protective Role of Bone Marrow Mesenchymal Stem Cells (BMSCs) in Repairing Epithelial Cells of Diabetic Retinopathy. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Diabetic retinopathy (DR) is one of the main causes of blindness. By directly employing mesenchymal stem cells to repair damaged retinal tissues, we aim to study the underlying repair mechanisms. 30 DR patients were included, along with 30 healthy control cases. Western-blot and qRT-PCR
were conducted to measure PI3K/Akt pathway-related genes. The PI3K/Akt antagonist (Rigosertib) was utilized in the induction process of cell differentiation to analyze the effects of PI3K/Akt pathwayspecific proteins and mRNAs. DR patients showed significantly elevated expression of PI3K/Akt
compared to control. With prolongation of induction, the expression of normal epithelial cell-related genes (SpC, SpB, SpA, CK18, KGF and Occludin) was elevated along with upregulated Occludin and KGF, two specific proteins of healthy epithelial cells. Meanwhile, the quantities of Occludin
and KGF in cell culture medium showed a gradual downward trend. In the differentiation of BMSCs towards epithelial cells, addition of PI3K/Akt antagonist Rigosertib was negatively correlated with the expression of several genes (IGF-1, shh, EGF, mTOR, AKT and PI3K) and decreased the quantities
of PI3K/Akt pathway-specific proteins (mTOR, PI3K and AKT). In conclusion, BMSCs can effectively reduce the release of cytokines in DR and promote the repair of damaged diabetic retina, possibly through regulation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yu Ling
- Department of Ophthalmology, The Fifth Affiliated Hospital of Guangxi Medical University, The First People’s Hospital of Nanning, Nanning, Guangxi, 530022, China
| | - Haiming Liang
- Department of Ophthalmology, The Fifth Affiliated Hospital of Guangxi Medical University, The First People’s Hospital of Nanning, Nanning, Guangxi, 530022, China
| | - Qi Tang
- Department of Ophthalmology, The Fifth Affiliated Hospital of Guangxi Medical University, The First People’s Hospital of Nanning, Nanning, Guangxi, 530022, China
| |
Collapse
|
11
|
Akiyama K, Aung KT, Talamini L, Huck O, Kuboki T, Muller S. Therapeutic effects of peptide P140 in a mouse periodontitis model. Cell Mol Life Sci 2022; 79:518. [DOI: 10.1007/s00018-022-04537-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/25/2022]
|
12
|
Wang R, Yuan J. Regulation of Autophagy and Inflammation Improves the Corneal Injury in the Model of Rats with Xerophthalmia. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study assessed the mechanism of regulation of autophagy and inflammation on corneal injury in the model of rats with xerophthalmia. The level of inducer and inhibitor of autophagy in the model of rats with xerophthalmia was detected and cell proliferation was evaluated by MTT assay
together with analysis of colony formation, cell apoptosis and cycle by FCM. The effect of inducer on the corneal injury and inflammation was assessed. The level of autophagy marker LC3 was elevated significantly after treatment with autophagy inducer along with increased cell proliferation
and migration and strengthened sensibility of corneal epithelial cells on corneal injury and inflammation and autophagy rate. In addition, cells in the established model was blocked at G2/M phase. Moreover, autophagy inducer significantly upregulated MMP-10 expression. Furthermore, there was
a target relationship between LC3 and P62. In conclusion, the cell migration, growth and autophagy is induced with autophagy inducer in the model of rats with xerophthalmia, indicating that autophagy inducer might be a brand-new therapeutic target spot for the treatment of xerophthalmia.
Collapse
Affiliation(s)
- Rui Wang
- Department of Ophthalmology, Jianghan University Affiliated Hospital, Wuhan, Hubei, 430015, China
| | - Jing Yuan
- Department of Ophthalmology, Jianghan University Affiliated Hospital, Wuhan, Hubei, 430015, China
| |
Collapse
|
13
|
Schall N, Talamini L, Wilhelm M, Jouvin-Marche E, Muller S. P140 Peptide Leads to Clearance of Autoreactive Lymphocytes and Normalizes Immune Response in Lupus-Prone Mice. Front Immunol 2022; 13:904669. [PMID: 35720371 PMCID: PMC9199391 DOI: 10.3389/fimmu.2022.904669] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
In systemic lupus erythematosus, T cells display multiple abnormalities. They are abnormally activated, secrete pro-inflammatory cytokines, help B cells to generate pathogenic autoantibodies, and provoke the accumulation of autoreactive memory T cells. P140, a synthetic peptide evaluated in phase-III clinical trials for lupus, binds HSPA8/HSC70 chaperone protein. In vitro and in vivo, it interferes with hyperactivated chaperone-mediated autophagy, modifying overexpression of major histocompatibility complex class II molecules and antigen presentation to autoreactive T cells. Here, we show that in P140-treated lupus mice, abnormalities affecting T and B cells are no longer detectable in secondary lymphoid tissue and peripheral blood. Data indicate that P140 acts by depleting hyper-activated autoreactive T and B cells and restores normal immune homeostasis. Our findings suggest that P140 belongs to a new family of non-immunosuppressive immunoregulators that do not correct T and B cell abnormalities but rather contribute to the clearance of deleterious T and B cells.
Collapse
Affiliation(s)
- Nicolas Schall
- CNRS and Strasbourg University, Unit Biotechnology and Cell signaling, UMR7242/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Laura Talamini
- CNRS and Strasbourg University, Unit Biotechnology and Cell signaling, UMR7242/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Maud Wilhelm
- CNRS and Strasbourg University, Unit Biotechnology and Cell signaling, UMR7242/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Evelyne Jouvin-Marche
- Institute for Advanced Biosciences, Research Centre Université Grenoble Alpes (UGA)-Inserm U1209-CNRS UMR 5309, La Tronche, France
| | - Sylviane Muller
- CNRS and Strasbourg University, Unit Biotechnology and Cell signaling, UMR7242/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France.,University of Strasbourg Institute for Advanced Study, Strasbourg, France
| |
Collapse
|
14
|
Suzuki A, Iwaya C, Ogata K, Yoshioka H, Shim J, Tanida I, Komatsu M, Tada N, Iwata J. Impaired GATE16-mediated exocytosis in exocrine tissues causes Sjögren's syndrome-like exocrinopathy. Cell Mol Life Sci 2022; 79:307. [PMID: 35593968 PMCID: PMC11071900 DOI: 10.1007/s00018-022-04334-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/03/2022]
Abstract
Sjögren's syndrome (SjS) is a chronic autoimmune disease characterized by immune cell infiltration of the exocrine glands, mainly the salivary and lacrimal glands. Despite recent advances in the clinical and mechanistic characterization of the disease, its etiology remains largely unknown. Here, we report that mice with a deficiency for either Atg7 or Atg3, which are enzymes involved in the ubiquitin modification pathway, in the salivary glands exhibit a SjS-like phenotype, characterized by immune cell infiltration with autoantibody detection, acinar cell death, and dry mouth. Prior to the onset of the SjS-like phenotype in these null mice, we detected an accumulation of secretory vesicles in the acinar cells of the salivary glands and found that GATE16, an uncharacterized autophagy-related molecule activated by ATG7 (E1-like enzyme) and ATG3 (E2-like enzyme), was highly expressed in these cells. Notably, GATE16 was activated by isoproterenol, an exocytosis inducer, and localized on the secretory vesicles in the acinar cells of the salivary glands. Failure to activate GATE16 was correlated with exocytosis defects in the acinar cells of the salivary glands in Atg7 and Atg3 cKO mice. Taken together, our results show that GATE16 activation regulated by the autophagic machinery is crucial for exocytosis and that defects in this pathway cause SjS.
Collapse
Affiliation(s)
- Akiko Suzuki
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Center for Craniofacial Research, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Chihiro Iwaya
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Center for Craniofacial Research, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Kenichi Ogata
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Center for Craniofacial Research, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Hiroki Yoshioka
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Center for Craniofacial Research, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Junbo Shim
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Center for Craniofacial Research, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Isei Tanida
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, 113-8431, Japan
| | - Masaaki Komatsu
- Department of Organ and Cell Physiology, Juntendo University Graduate School of Medicine, Tokyo, 113-8431, Japan
| | - Norihiro Tada
- Division of Genome Research, Research Institute for Diseases of Old Ages, Juntendo University School of Medicine, Tokyo, 113-8431, Japan
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA.
- Center for Craniofacial Research, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA.
- Pediatric Research Center, School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Celia AI, Colafrancesco S, Barbati C, Alessandri C, Conti F. Autophagy in Rheumatic Diseases: Role in the Pathogenesis and Therapeutic Approaches. Cells 2022; 11:cells11081359. [PMID: 35456038 PMCID: PMC9025357 DOI: 10.3390/cells11081359] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/23/2022] Open
Abstract
Autophagy is a lysosomal pathway for the degradation of damaged proteins and intracellular components that promotes cell survival under specific conditions. Apoptosis is, in contrast, a critical programmed cell death mechanism, and the relationship between these two processes influences cell fate. Recent evidence suggests that autophagy and apoptosis are involved in the self-tolerance promotion and in the regulatory mechanisms contributing to disease susceptibility and immune regulation in rheumatic diseases. The aim of this review is to discuss how the balance between autophagy and apoptosis may be dysregulated in multiple rheumatic diseases and to dissect the role of autophagy in the pathogenesis of rheumatoid arthritis, systemic lupus erythematosus, and Sjögren’s syndrome. Furthermore, to discuss the potential capacity of currently used disease-modifying antirheumatic drugs (DMARDs) to target and modulate autophagic processes.
Collapse
|
16
|
Targeting the endo-lysosomal autophagy pathway to treat inflammatory bowel diseases. J Autoimmun 2022; 128:102814. [PMID: 35298976 DOI: 10.1016/j.jaut.2022.102814] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 01/18/2023]
Abstract
Inflammatory bowel disease (IBD) is a serious public health problem in Western society with a continuing increase in incidence worldwide. Safe, targeted medicines for IBD are not yet available. Autophagy, a vital process implicated in normal cell homeostasis, provides a potential point of entry for the treatment of IBDs, as several autophagy-related genes are associated with IBD risk. We conducted a series of experiments in three distinct mouse models of colitis to test the effectiveness of therapeutic P140, a phosphopeptide that corrects autophagy dysfunctions in other autoimmune and inflammatory diseases. Colitis was experimentally induced in mice by administering dextran sodium sulfate and 2,4,6 trinitrobenzene sulfonic acid. Transgenic mice lacking both il-10 and iRhom2 - involved in tumor necrosis factor α secretion - were also used. In the three models investigated, P140 treatment attenuated the clinical and histological severity of colitis. Post-treatment, altered expression of several macroautophagy and chaperone-mediated autophagy markers, and of pro-inflammatory mediators was corrected. Our results demonstrate that therapeutic intervention with an autophagy modulator improves colitis in animal models. These findings highlight the potential of therapeutic peptide P140 for use in the treatment of IBD.
Collapse
|
17
|
Qu C, Zhao Y, Zhang H, Xu W, Zhang X. Bone Marrow Mesenchymal Stem Cells (BMSCs)-Derived Exosomes Promotes Proliferation and Differentiation of Retinal Neuron-Like Cells to Repair Corneal Epithelial Damage. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dry eye disease (DED) is a common ocular surface disease. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into various cells, and BMSC-derived exosomes (BMSC-exo) is essential to maintaining BMSCs stemness. This study aimed to elucidate the mechanism underlying BMSCexo
in DED. Sixty rats with corneal epithelial injury were treated with BMSCs or BMSC-exo and untreated (each group, n = 20) followed by analysis of the effect of BMSCs and BMSC-exo by evaluating the corneal epithelium damage via measuring the Basso-Beattie-Bresnahan (BBB) score on 1st,
3rd, 7th, 14th, 28th day after treatments. TUNEL staining assessed cell apoptosis, NF200 expression and the number of BrdU-positive cells. There was no significant difference in BBB scores among three groups on the 1st and 3rd day after treatment (p > 0.05) with significant difference
on the 7th, 14th, and 28th day (p <0.05); compared with control group, BMSCs group and combination group had significantly higher BBB score (p < 0.05). The amount of apoptotic cells rose on 3rd and then gradually decreased since 7th day. Moreover, BMSCs and BMSC-exo decreased
the apoptotic index and increased absorbance of NF200 and BrdU-positive rate (p < 0.05). BMSC-exo alleviates corneal epithelial damage in DED and facilitates wound healing possibly through reducing cell apoptosis and increasing retinal neuron-like cell proliferation protein.
Collapse
Affiliation(s)
- Cuiping Qu
- Department of Medical Cosmetology, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, 161000, China
| | - Yue Zhao
- Department of Ophthalmology, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, 161000, China
| | - Huijuan Zhang
- International Medical Department, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, 116001, China
| | - Wenshuang Xu
- Department of Ophthalmology, Qiqihar Wuguan Hospital, Qiqihar, Heilongjiang, 161006, China
| | - Xiaofeng Zhang
- Department of Ophthalmology, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, 161000, China
| |
Collapse
|
18
|
CIDP: Current Treatments and Identification of Targets for Future Specific Therapeutic Intervention. IMMUNO 2022. [DOI: 10.3390/immuno2010009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is an acquired immune-mediated inflammatory disorder of the peripheral nervous system. This clinically heterogeneous neurological disorder is closely related to Guillain–Barré syndrome and is considered the chronic counterpart of that acute disease. Currently available treatments are mostly empirical; they include corticosteroids, intravenous immunoglobulins, plasma exchange and chronic immunosuppressive agents, either alone or in combination. Recent advances in the understanding of the underlying pathogenic mechanisms in CIDP have brought a number of novel ways of possible intervention for use in CIDP. This review summarizes selected pre-clinical and clinical findings, highlights the importance of using adapted animal models to evaluate the efficacy of novel treatments, and proposes the outlines of future directions to ameliorate the conditions of patients with CIDP.
Collapse
|
19
|
Bonam SR, Tranchant C, Muller S. Autophagy-Lysosomal Pathway as Potential Therapeutic Target in Parkinson's Disease. Cells 2021; 10:3547. [PMID: 34944054 PMCID: PMC8700067 DOI: 10.3390/cells10123547] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
Cellular quality control systems have gained much attention in recent decades. Among these, autophagy is a natural self-preservation mechanism that continuously eliminates toxic cellular components and acts as an anti-ageing process. It is vital for cell survival and to preserve homeostasis. Several cell-type-dependent canonical or non-canonical autophagy pathways have been reported showing varying degrees of selectivity with regard to the substrates targeted. Here, we provide an updated review of the autophagy machinery and discuss the role of various forms of autophagy in neurodegenerative diseases, with a particular focus on Parkinson's disease. We describe recent findings that have led to the proposal of therapeutic strategies targeting autophagy to alter the course of Parkinson's disease progression.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Christine Tranchant
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France;
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM-U964/CNRS-UMR7104/Université de Strasbourg, 67400 Illkirch, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Sylviane Muller
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
- CNRS and Strasbourg University, Unit Biotechnology and Cell Signaling/Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), 67000 Strasbourg, France
| |
Collapse
|
20
|
Targeting lysosomes in human disease: from basic research to clinical applications. Signal Transduct Target Ther 2021; 6:379. [PMID: 34744168 PMCID: PMC8572923 DOI: 10.1038/s41392-021-00778-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/26/2021] [Indexed: 01/18/2023] Open
Abstract
In recent years, accumulating evidence has elucidated the role of lysosomes in dynamically regulating cellular and organismal homeostasis. Lysosomal changes and dysfunction have been correlated with the development of numerous diseases. In this review, we interpreted the key biological functions of lysosomes in four areas: cellular metabolism, cell proliferation and differentiation, immunity, and cell death. More importantly, we actively sought to determine the characteristic changes and dysfunction of lysosomes in cells affected by these diseases, the causes of these changes and dysfunction, and their significance to the development and treatment of human disease. Furthermore, we outlined currently available targeting strategies: (1) targeting lysosomal acidification; (2) targeting lysosomal cathepsins; (3) targeting lysosomal membrane permeability and integrity; (4) targeting lysosomal calcium signaling; (5) targeting mTOR signaling; and (6) emerging potential targeting strategies. Moreover, we systematically summarized the corresponding drugs and their application in clinical trials. By integrating basic research with clinical findings, we discussed the current opportunities and challenges of targeting lysosomes in human disease.
Collapse
|
21
|
Daubeuf F, Schall N, Petit-Demoulière N, Frossard N, Muller S. An Autophagy Modulator Peptide Prevents Lung Function Decrease and Corrects Established Inflammation in Murine Models of Airway Allergy. Cells 2021; 10:cells10092468. [PMID: 34572117 PMCID: PMC8472429 DOI: 10.3390/cells10092468] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 12/18/2022] Open
Abstract
The involvement of autophagy and its dysfunction in asthma is still poorly documented. By using a murine model of chronic house dust mite (HDM)-induced airway inflammation, we tested the expression of several autophagy markers in the lung and spleen of asthma-like animals. Compared to control mice, in HDM-sensitized and challenged mice, the expression of sequestosome-1/p62, a multifunctional adaptor protein that plays an important role in the autophagy machinery, was raised in the splenocytes. In contrast, its expression was decreased in the neutrophils recovered from the bronchoalveolar fluid, indicating that autophagy was independently regulated in these two compartments. In a strategy of drug repositioning, we treated allergen-sensitized mice with the therapeutic peptide P140 known to target chaperone-mediated autophagy. A single intravenous administration of P140 in these mice resulted in a significant reduction in airway resistance and elastance, and a reduction in the number of neutrophils and eosinophils present in the bronchoalveolar fluid. It corrected the autophagic alteration without showing any suppressive effect in the production of IgG1 and IgE. Collectively, these findings show that autophagy processes are altered in allergic airway inflammation. This cellular pathway may represent a potential therapeutic target for treating selected patients with asthma.
Collapse
Affiliation(s)
- François Daubeuf
- CNRS-Strasbourg University Laboratoire d’Innovation Thérapeutique/Strasbourg Drug Discovery and Development Institute (IMS), Faculté de Pharmacie, 67400 Illkirch, France; (F.D.); (N.P.-D.); (N.F.)
- CNRS UMS3286, Plate-Forme de Chimie Biologique Intégrative de Strasbourg/Strasbourg Drug Discovery and Development Institute (IMS), 67400 Illkirch, France
| | - Nicolas Schall
- CNRS-Strasbourg University Unit Biotechnology and Cell Signaling/Strasbourg Drug Discovery and Development Institute (IMS), Ecole Supérieure de Biotechnologie de Strasbourg, 67400 Illkirch, France;
| | - Nathalie Petit-Demoulière
- CNRS-Strasbourg University Laboratoire d’Innovation Thérapeutique/Strasbourg Drug Discovery and Development Institute (IMS), Faculté de Pharmacie, 67400 Illkirch, France; (F.D.); (N.P.-D.); (N.F.)
- CNRS-Strasbourg University Unit Biotechnology and Cell Signaling/Strasbourg Drug Discovery and Development Institute (IMS), Ecole Supérieure de Biotechnologie de Strasbourg, 67400 Illkirch, France;
| | - Nelly Frossard
- CNRS-Strasbourg University Laboratoire d’Innovation Thérapeutique/Strasbourg Drug Discovery and Development Institute (IMS), Faculté de Pharmacie, 67400 Illkirch, France; (F.D.); (N.P.-D.); (N.F.)
| | - Sylviane Muller
- CNRS-Strasbourg University Unit Biotechnology and Cell Signaling/Strasbourg Drug Discovery and Development Institute (IMS), Ecole Supérieure de Biotechnologie de Strasbourg, 67400 Illkirch, France;
- Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, 67000 Strasbourg, France
- University of Strasbourg Institute for Advanced Study, 67000 Strasbourg, France
- Correspondence:
| |
Collapse
|
22
|
Wilhelm M, Bonam SR, Schall N, Bendorius M, Korganow AS, Lumbroso C, Muller S. Implication of a lysosomal antigen in the pathogenesis of lupus erythematosus. J Autoimmun 2021; 120:102633. [PMID: 33932829 DOI: 10.1016/j.jaut.2021.102633] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/21/2022]
Abstract
Naturally-occurring autoantibodies to certain components of autophagy processes have been described in a few autoimmune diseases, but their fine specificity, their relationships with clinical phenotypes, and their potential pathogenic functions remain elusive. Here, we explored IgG autoantibodies reacting with a panel of cytoplasmic endosomal/lysosomal antigens and individual heat-shock proteins, all of which share links to autophagy. Sera from autoimmune patients and from MRL/lpr and NZB/W lupus-prone mice reacted with the C-terminal residues of lysosome-associated membrane glycoprotein (LAMP)2A. No cross-reaction was observed with LAMP2B or LAMP2C variants, with dsDNA or mononucleosomes, or with heat-shock protein A8. Moreover, administering chromatography-purified LAMP2A autoantibodies to MRL/lpr mice accelerated mortality. Furthermore, flow cytometry revealed elevated cell-surface expression of LAMP2A on MRL/lpr B cells. These findings reveal the involvement of a new class of autoantibodies targeting the C-terminus of LAMP2A, a receptor for cytosolic proteins targeted for degradation via chaperone-mediated autophagy. These autoantibodies could affect the autophagy process, which is abnormally upregulated in lupus. The data presented support a novel connection between autophagy dysregulation, autoimmune processes and pathophysiology in lupus.
Collapse
Affiliation(s)
- Maud Wilhelm
- CNRS, Strasbourg University Unit Biotechnology and Cell Signaling / Strasbourg Drug Discovery and Development Institute (IMS); Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Srinivasa Reddy Bonam
- CNRS, Strasbourg University Unit Biotechnology and Cell Signaling / Strasbourg Drug Discovery and Development Institute (IMS); Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Nicolas Schall
- CNRS, Strasbourg University Unit Biotechnology and Cell Signaling / Strasbourg Drug Discovery and Development Institute (IMS); Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Mykolas Bendorius
- CNRS, Strasbourg University Unit Biotechnology and Cell Signaling / Strasbourg Drug Discovery and Development Institute (IMS); Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Anne-Sophie Korganow
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Hôpitaux Universitaires de Strasbourg, France; Strasbourg University, INSERM Unit Molecular ImmunoRheumatology, Strasbourg, France; Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, Strasbourg, France
| | | | - Sylviane Muller
- CNRS, Strasbourg University Unit Biotechnology and Cell Signaling / Strasbourg Drug Discovery and Development Institute (IMS); Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France; Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, Strasbourg, France; University of Strasbourg Institute for Advanced Study, Strasbourg, France.
| |
Collapse
|
23
|
Abstract
Lysosomes offer a unique arrangement of degradative, exocytic, and signaling capabilities that make their continued function critical to cellular homeostasis. Lysosomes owe their function to the activity of lysosomal ion channels and transporters, which maintain concentration gradients of H+, K+, Ca2+, Na+, and Cl- across the lysosomal membrane. This review examines the contributions of lysosomal ion channels to lysosome function, showing how ion channel function is integral to degradation and autophagy, maintaining lysosomal membrane potential, controlling Ca2+ signaling, and facilitating exocytosis. Evidence of lysosome dysfunction in a variety of disease pathologies creates a need to understand how lysosomal ion channels contribute to lysosome dysfunction. For example, the loss of function of the TRPML1 Ca2+ lysosome channel in multiple lysosome storage diseases leads to lysosome dysfunction and disease pathogenesis while neurodegenerative diseases are marked by lysosome dysfunction caused by changes in ion channel activity through the TRPML1, TPC, and TMEM175 ion channels. Autoimmune disease is marked by dysregulated autophagy, which is dependent on the function of multiple lysosomal ion channels. Understanding the role of lysosomal ion channel activity in lysosome membrane permeability and NLRP3 inflammasome activation could provide valuable mechanistic insight into NLRP3 inflammasome-mediated diseases. Finally, this review seeks to show that understanding the role of lysosomal ion channels in lysosome dysfunction could give mechanistic insight into the efficacy of certain drug classes, specifically those that target the lysosome, such as cationic amphiphilic drugs.
Collapse
Affiliation(s)
- Rebekah L Kendall
- Department of Biomedical and Pharmaceutical Sciences, Center for Environmental Health Sciences, University of Montana, Missoula, MT, USA
| | - Andrij Holian
- Department of Biomedical and Pharmaceutical Sciences, Center for Environmental Health Sciences, University of Montana, Missoula, MT, USA
| |
Collapse
|
24
|
Huijser E, Versnel MA. Making Sense of Intracellular Nucleic Acid Sensing in Type I Interferon Activation in Sjögren's Syndrome. J Clin Med 2021; 10:532. [PMID: 33540529 PMCID: PMC7867173 DOI: 10.3390/jcm10030532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is a systemic autoimmune rheumatic disease characterized by dryness of the eyes and mucous membranes, which can be accompanied by various extraglandular autoimmune manifestations. The majority of patients exhibit persistent systemic activation of the type I interferon (IFN) system, a feature that is shared with other systemic autoimmune diseases. Type I IFNs are integral to anti-viral immunity and are produced in response to stimulation of pattern recognition receptors, among which nucleic acid (NA) receptors. Dysregulated detection of endogenous NAs has been widely implicated in the pathogenesis of systemic autoimmune diseases. Stimulation of endosomal Toll-like receptors by NA-containing immune complexes are considered to contribute to the systemic type I IFN activation. Accumulating evidence suggest additional roles for cytosolic NA-sensing pathways in the pathogenesis of systemic autoimmune rheumatic diseases. In this review, we will provide an overview of the functions and signaling of intracellular RNA- and DNA-sensing receptors and summarize the evidence for a potential role of these receptors in the pathogenesis of pSS and the sustained systemic type I IFN activation.
Collapse
Affiliation(s)
| | - Marjan A. Versnel
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands;
| |
Collapse
|
25
|
In Vivo Remodeling of Altered Autophagy-Lysosomal Pathway by a Phosphopeptide in Lupus. Cells 2020; 9:cells9102328. [PMID: 33092174 PMCID: PMC7589999 DOI: 10.3390/cells9102328] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/30/2020] [Accepted: 10/16/2020] [Indexed: 12/22/2022] Open
Abstract
The phosphopeptide P140/Lupuzor, which improves the course of lupus disease in mice and patients, targets chaperone-mediated autophagy (CMA), a selective form of autophagy that is abnormally upregulated in lupus-prone MRL/lpr mice. Administered intravenously to diseased mice, P140 reduces the expression level of two major protein players of CMA, LAMP2A and HSPA8, and inhibits CMA in vitro in a cell line that stably expresses a CMA reporter. Here, we aimed to demonstrate that P140 also affects CMA in vivo and to unravel the precise cellular mechanism of how P140 interacts with the CMA process. MRL/lpr mice and CBA/J mice used as control received P140 or control peptides intravenously. Lysosome-enriched fractions of spleen or liver were prepared to examine lysosomal function. Highly purified lysosomes were further isolated and left to incubate with the CMA substrate to study at which cellular step P140 interacts with the CMA process. The data show that P140 effectively regulates CMA in vivo in MRL/lpr mice at the step of substrate lysosomal uptake and restores some alterations of defective lysosomes. For the first time, it is demonstrated that by occluding the intralysosome uptake of CMA substrates, a therapeutic molecule can attenuate excessive CMA activity in a pathological pro-inflammatory context and protect against hyperinflammation. This recovery effect of P140 on hyperactivated CMA is not only important for lupus therapy but potentially also for treating other (auto)inflammatory diseases, including neurologic and metabolic disorders, where CMA modulation would be highly beneficial.
Collapse
|
26
|
Colafrancesco S, Vomero M, Iannizzotto V, Minniti A, Barbati C, Arienzo F, Mastromanno L, Colasanti T, Izzo R, Nayar S, Pipi E, Cerbelli B, Giordano C, Ciccia F, Conti F, Valesini G, Barone F, Priori R, Alessandri C. Autophagy occurs in lymphocytes infiltrating Sjögren's syndrome minor salivary glands and correlates with histological severity of salivary gland lesions. Arthritis Res Ther 2020; 22:238. [PMID: 33050949 PMCID: PMC7557086 DOI: 10.1186/s13075-020-02317-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/11/2020] [Indexed: 12/09/2022] Open
Abstract
Backgrounds The organization of minor salivary glands (MSG) infiltrates, in patients with Sjögren’s syndrome (SS), associates with disease severity and progression. Aberrant regulation of lymphocyte autophagy is involved in autoimmunity, and in previous work, we provided the first evidence of upregulated autophagy in CD4+ T cells infiltrating SS MSG. The aim of this study was to further explore autophagy in SS infiltrating and circulating lymphocytes and to investigate its role in disease histopathological progression. Methods After collection of 20 SS MSG, the presence of lymphocyte aggregates (foci) and the formation of germinal center (GC)-like structures were observed by H&E and confirmed by immunohistochemistry. The expression of autophagy-related genes, Atg5 and MAP1LC3A, was detected by RT-PCR on microdissected salivary gland tissue and control tonsils. In MSG and tonsils, autophagic lymphocytes were identified by the detection of the autophagosome protein LC3B visualized as LC3 puncta staining by immunofluorescence. Peripheral blood autophagy was assessed by flow cytometry in SS and healthy controls (HC). Results Real-time PCR demonstrated higher expression in the autophagy genes Atg5 and MAP1LC3A in MSG GCs as compared to both small foci (p = 0.0075, p = 0.0002) and GCs from tonsils (p = 0.0001, p = 0.0037). In MSG, LC3 puncta staining was detectable on both CD3+ and CD20+ lymphocytes; in tonsils, LC3 puncta was almost undetectable on all lymphocytes. Compared to HC (n = 20), flow cytometry did not reveal any increase of autophagy in SS circulating lymphocytes (n = 30). Conclusions In SS MSG, lymphocytes’ autophagy is a feature of infiltrating T and B cells and is associated with histological severity. Interestingly, in MSG aberrant regulation of autophagy is detectable in GC-like structures possibly indicating its involvement in the development and persistence of the autoimmune process within the lesions.
Collapse
Affiliation(s)
- Serena Colafrancesco
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Marta Vomero
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Valentina Iannizzotto
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Antonina Minniti
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Cristiana Barbati
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Francesca Arienzo
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Linda Mastromanno
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Tania Colasanti
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Raffaella Izzo
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Elena Pipi
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Bruna Cerbelli
- Dipartimento di Radiologia, Oncologia e Scienze Patologiche, Sapienza University of Rome, Rome, Italy
| | - Carla Giordano
- Dipartimento di Radiologia, Oncologia e Scienze Patologiche, Sapienza University of Rome, Rome, Italy
| | - Francesco Ciccia
- Dipartimento di Medicina di Precisione, Rheumatology Unit, University of Campania "L. Vanvitelli", Naples, Italy
| | - Fabrizio Conti
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Guido Valesini
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, UK and Sandwell and West Birmingham Trust, Birmingham, UK
| | - Roberta Priori
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
27
|
Noguchi M, Hirata N, Tanaka T, Suizu F, Nakajima H, Chiorini JA. Autophagy as a modulator of cell death machinery. Cell Death Dis 2020; 11:517. [PMID: 32641772 PMCID: PMC7343815 DOI: 10.1038/s41419-020-2724-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 01/07/2023]
Abstract
The balance between cell death and survival is a critical parameter in the regulation of cells and the maintenance of homeostasis in vivo. Three major mechanisms for cell death have been identified in mammalian cells: apoptosis (type I), autophagic cell death (type II), and necrosis (type III). These three mechanisms have been suggested to engage in cross talk with each other. Among them, autophagy was originally characterized as a cell survival mechanism for amino acid recycling during starvation. Whether autophagy functions primarily in cell survival or cell death is a critical question yet to be answered. Here, we present a comprehensive review of the cell death-related events that take place during autophagy and their underlying mechanisms in cancer and autoimmune disease development.
Collapse
Affiliation(s)
- Masayuki Noguchi
- grid.39158.360000 0001 2173 7691Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Noriyuki Hirata
- grid.39158.360000 0001 2173 7691Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Tsutomu Tanaka
- grid.94365.3d0000 0001 2297 5165National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD USA
| | - Futoshi Suizu
- grid.39158.360000 0001 2173 7691Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Nakajima
- grid.136304.30000 0004 0370 1101Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - John A. Chiorini
- grid.94365.3d0000 0001 2297 5165National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
28
|
Bonam SR, Bayry J, Tschan MP, Muller S. Progress and Challenges in The Use of MAP1LC3 as a Legitimate Marker for Measuring Dynamic Autophagy In Vivo. Cells 2020; 9:E1321. [PMID: 32466347 PMCID: PMC7291013 DOI: 10.3390/cells9051321] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 01/02/2023] Open
Abstract
Tremendous efforts have been made these last decades to increase our knowledge of intracellular degradative systems, especially in the field of autophagy. The role of autophagy in the maintenance of cell homeostasis is well documented and the existence of defects in the autophagic machinery has been largely described in diseases and aging. Determining the alterations occurring in the many forms of autophagy that coexist in cells and tissues remains complicated, as this cellular process is highly dynamic in nature and can vary from organ to organ in the same individual. Although autophagy is extensively studied, its functioning in different tissues and its links with other biological processes is still poorly understood. Several assays have been developed to monitor autophagy activity in vitro, ex vivo, and in vivo, based on different markers, the use of various inhibitors and activators, and distinct techniques. This review emphasizes the methods applied to measure (macro-)autophagy in tissue samples and in vivo via a protein, which centrally intervenes in the autophagy pathway, the microtubule-associated protein 1A/1B-light chain 3 (MAP1LC3), which is the most widely used marker and the first identified to associate with autophagosomal structures. These approaches are presented and discussed in terms of pros and cons. Some recommendations are provided to improve the reliability of the interpretation of results.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- CNRS, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, 67412 Strasbourg University/Laboratory of Excellence Medalis, 67000 Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France;
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France;
| | - Mario P. Tschan
- Institute of Pathology, Division of Experimental Pathology, University of Bern, 3008 Bern, Switzerland;
| | - Sylviane Muller
- CNRS, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, 67412 Strasbourg University/Laboratory of Excellence Medalis, 67000 Strasbourg, France
- Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, 67000 Strasbourg, France
- University of Strasbourg Institute for Advanced Study, 67000 Strasbourg, France
| |
Collapse
|
29
|
Peptide-Based Vaccination Therapy for Rheumatic Diseases. J Immunol Res 2020; 2020:8060375. [PMID: 32258176 PMCID: PMC7104265 DOI: 10.1155/2020/8060375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
Rheumatic diseases are extremely heterogeneous diseases with substantial risks of morbidity and mortality, and there is a pressing need in developing more safe and cost-effective treatment strategies. Peptide-based vaccination is a highly desirable strategy in treating noninfection diseases, such as cancer and autoimmune diseases, and has gained increasing attentions. This review is aimed at providing a brief overview of the recent advances in peptide-based vaccination therapy for rheumatic diseases. Tremendous efforts have been made to develop effective peptide-based vaccinations against rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), while studies in other rheumatic diseases are still limited. Peptide-based active vaccination against pathogenic cytokines such as TNF-α and interferon-α (IFN-α) is shown to be promising in treating RA or SLE. Moreover, peptide-based tolerogenic vaccinations also have encouraging results in treating RA or SLE. However, most studies available now have been mainly based on animal models, while evidence from clinical studies is still lacking. The translation of these advances from experimental studies into clinical therapy remains impeded by some obstacles such as species difference in immunity, disease heterogeneity, and lack of safe delivery carriers or adjuvants. Nevertheless, advances in high-throughput technology, bioinformatics, and nanotechnology may help overcome these impediments and facilitate the successful development of peptide-based vaccination therapy for rheumatic diseases.
Collapse
|
30
|
Correction of autophagy impairment inhibits pathology in the NOD.H-2h4 mouse model of primary Sjögren's syndrome. J Autoimmun 2020; 108:102418. [PMID: 32029330 DOI: 10.1016/j.jaut.2020.102418] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/20/2020] [Accepted: 01/20/2020] [Indexed: 01/04/2023]
Abstract
Dysregulation of autophagy has been implicated in the development of various disease indications including autoimmune diseases. Here we identified hitherto unsuspected molecular alterations of autophagy occurring at an early stage of the macroautophagy pathway in the salivary glands and spleen of NOD.H-2h4 mice that develop a primary Sjögren's-like syndrome. In this study we investigated the capacity of phosphopeptide P140 to correct immune alteration in NOD.H-2h4 mice and the effect on neogenesis of tertiary lymphoid structures in salivary glands, which is hallmark characteristic of SS. Phosphopeptide P140 known to lower excessive autophagy processes, rescued sick NOD.H-2h4 mice from some autophagy defects and significantly reduced formation of tertiary lymphoid structures in salivary glands. Mechanistically, the frequency of activated CD44high/CD62Llow CD4+ T cell populations was significantly decreased and this reduction was correlated with an increased number of CD44low/CD62Lhigh resting T cells. The CD8 T cell compartment was not affected. P140 down-regulated the maturation and differentiation of B cells into plasma cells, and decreased IgG and autoantibody secretion. It had no effect on germinal centers B cells (B220+ FAS+GL-7+) that are an important compound of the B cell humoral immune response. Together with previous data generated in MRL/lpr mice that develop some features of Sjögren's syndrome associated to other inflammatory and autoimmune defects, our present findings strongly reinforce the potential of autophagy modulators, such as P140, for treating patients with Sjögren's syndrome.
Collapse
|
31
|
Abstract
Lysosomes are membrane-bound organelles with roles in processes involved in degrading and recycling cellular waste, cellular signalling and energy metabolism. Defects in genes encoding lysosomal proteins cause lysosomal storage disorders, in which enzyme replacement therapy has proved successful. Growing evidence also implicates roles for lysosomal dysfunction in more common diseases including inflammatory and autoimmune disorders, neurodegenerative diseases, cancer and metabolic disorders. With a focus on lysosomal dysfunction in autoimmune disorders and neurodegenerative diseases - including lupus, rheumatoid arthritis, multiple sclerosis, Alzheimer disease and Parkinson disease - this Review critically analyses progress and opportunities for therapeutically targeting lysosomal proteins and processes, particularly with small molecules and peptide drugs.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France
| | - Fengjuan Wang
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France.
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France.
- University of Strasbourg Institute for Advanced Study, Strasbourg, France.
- Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, Strasbourg, France.
| |
Collapse
|
32
|
Hong X, Min SN, Zhang YY, Lin YT, Wang F, Huang Y, Yu GY, Wu LL, Yang HY. TNF-α Suppresses Autophagic Flux in Acinar Cells in IgG4-Related Sialadenitis. J Dent Res 2019; 98:1386-1396. [PMID: 31461632 DOI: 10.1177/0022034519871890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
IgG4-related sialadenitis (IgG4-RS) is a newly recognized immune-mediated systemic fibroinflammatory disease that affects salivary glands and leads to hyposalivation. Tumor necrosis factor-α (TNF-α) is a critical proinflammatory cytokine involved in several salivary gland disorders, but its role and mechanism regarding acinar cell injury in IgG4-RS are unknown. Here, we found that TNF-α level was significantly increased in serum and submandibular gland (SMG) of patients and that serum TNF-α level was negatively correlated with saliva flow rate. Ultrastructural observations of IgG4-RS SMGs revealed accumulation of large autophagic vacuoles, as well as dense fibrous bundles, decreased secretory granules, widened intercellular spaces, swollen mitochondria, and expanded endoplasmic reticulum. Expression levels of LC3 and p62 were both increased in patients' SMGs. TNF-α treatment led to elevated levels of LC3II and p62 in both SMG-C6 cells and cultured human SMG tissues but did not further increase their levels when combined with bafilomycin A1 treatment. Moreover, transfection of Ad-mCherry-GFP-LC3B in SMG-C6 cells confirmed the suppression of autophagic flux after TNF-α treatment. Immunofluorescence imaging revealed that costaining of LC3 and the lysosomal marker LAMP2 was significantly decreased in patients, TNF-α-treated SMG-C6 cells, and cultured human SMGs, indicating a reduction in autophagosome-lysosome fusion. Furthermore, the ratio of pro/mature cathepsin D was elevated in vivo, ex vivo, and in vitro. TNF-α also appeared to induce abnormal acidification of lysosomes in acinar cells, as assessed by lysosomal pH and LysoTracker DND-26 fluorescence intensity. In addition, TNF-α treatment induced transcription factor EB (TFEB) redistribution in SMG-C6 cells, which was consistent with the changes observed in IgG4-RS patients. TNF-α increased the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, and inhibition of ERK1/2 by U0126 reversed TNF-α-induced TFEB redistribution, lysosomal dysfunction, and autophagic flux suppression. These findings suggest that TNF-α is a key cytokine related to acinar cell injury in IgG4-RS through ERK1/2-mediated autophagic flux suppression.
Collapse
Affiliation(s)
- X Hong
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University, the Hong Kong University of Science and Technology Medical Center, Shenzhen, P.R. China
| | - S N Min
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, P.R. China
| | - Y Y Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, P.R. China
| | - Y T Lin
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University, the Hong Kong University of Science and Technology Medical Center, Shenzhen, P.R. China
| | - F Wang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University, the Hong Kong University of Science and Technology Medical Center, Shenzhen, P.R. China
| | - Y Huang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, P.R. China
| | - G Y Yu
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University, the Hong Kong University of Science and Technology Medical Center, Shenzhen, P.R. China.,Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, P.R. China
| | - L L Wu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, P.R. China
| | - H Y Yang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University, the Hong Kong University of Science and Technology Medical Center, Shenzhen, P.R. China
| |
Collapse
|
33
|
Bonam SR, Ruff M, Muller S. HSPA8/HSC70 in Immune Disorders: A Molecular Rheostat that Adjusts Chaperone-Mediated Autophagy Substrates. Cells 2019; 8:E849. [PMID: 31394830 PMCID: PMC6721745 DOI: 10.3390/cells8080849] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022] Open
Abstract
HSPA8/HSC70 is a molecular chaperone involved in a wide variety of cellular processes. It plays a crucial role in protein quality control, ensuring the correct folding and re-folding of selected proteins, and controlling the elimination of abnormally-folded conformers and of proteins daily produced in excess in our cells. HSPA8 is a crucial molecular regulator of chaperone-mediated autophagy, as a detector of substrates that will be processed by this specialized autophagy pathway. In this review, we shortly summarize its structure and overall functions, dissect its implication in immune disorders, and list the known pharmacological tools that modulate its functions. We also exemplify the interest of targeting HSPA8 to regulate pathological immune dysfunctions.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Neuroimmunology & peptide therapy, Biotechnology and cell signaling, CNRS-University of Strasbourg, Illkirch 67412, France/Laboratory of excellence Medalis, 67000 Strasbourg, France
| | - Marc Ruff
- Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, 67404 Strasbourg, France
| | - Sylviane Muller
- Neuroimmunology & peptide therapy, Biotechnology and cell signaling, CNRS-University of Strasbourg, Illkirch 67412, France/Laboratory of excellence Medalis, 67000 Strasbourg, France.
- University of Strasbourg Institute for Advanced Study (USIAS), 67000 Strasbourg, France.
- Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, 67000 Strasbourg, France.
| |
Collapse
|
34
|
|
35
|
Retnakumar SV, Muller S. Pharmacological Autophagy Regulators as Therapeutic Agents for Inflammatory Bowel Diseases. Trends Mol Med 2019; 25:516-537. [PMID: 30952481 DOI: 10.1016/j.molmed.2019.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/03/2019] [Accepted: 03/06/2019] [Indexed: 12/12/2022]
Abstract
The arsenal of effective molecules to treat patients with chronic inflammatory bowel diseases (IBDs) remains limited. These remitting-relapsing diseases have become a global health issue and new therapeutic strategies are eagerly awaited to regulate the course of these disorders. Since the association between autophagy-related gene polymorphism and an increased risk of Crohn's disease (CD) has been discovered, a new domain of investigation has emerged, focused on the intracellular degradation system, with the objective of generating new medicines that are safer and more targeted. This review summarizes the drugs administered to IBD patients and describes recently emerged therapeutic agents. We compile evidence on the contribution of autophagy to IBD pathogenesis, give an overview of pharmacological autophagy regulators in animal models of colitis, and propose novel therapeutic avenues based on autophagy components.
Collapse
Affiliation(s)
- Sruthi Vijaya Retnakumar
- CNRS-University of Strasbourg, Biotechnology and Cell signaling, Institut de Science et d'ingénierie Supramoléculaire, 67000 Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell signaling, Institut de Science et d'ingénierie Supramoléculaire, 67000 Strasbourg, France; University of Strasbourg Institute for Advanced Study, 67000 Strasbourg, France.
| |
Collapse
|
36
|
Bendorius M, Po C, Muller S, Jeltsch-David H. From Systemic Inflammation to Neuroinflammation: The Case of Neurolupus. Int J Mol Sci 2018; 19:E3588. [PMID: 30428632 PMCID: PMC6274746 DOI: 10.3390/ijms19113588] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022] Open
Abstract
It took decades to arrive at the general consensus dismissing the notion that the immune system is independent of the central nervous system. In the case of uncontrolled systemic inflammation, the relationship between the two systems is thrown off balance and results in cognitive and emotional impairment. It is specifically true for autoimmune pathologies where the central nervous system is affected as a result of systemic inflammation. Along with boosting circulating cytokine levels, systemic inflammation can lead to aberrant brain-resident immune cell activation, leakage of the blood⁻brain barrier, and the production of circulating antibodies that cross-react with brain antigens. One of the most disabling autoimmune pathologies known to have an effect on the central nervous system secondary to the systemic disease is systemic lupus erythematosus. Its neuropsychiatric expression has been extensively studied in lupus-like disease murine models that develop an autoimmunity-associated behavioral syndrome. These models are very useful for studying how the peripheral immune system and systemic inflammation can influence brain functions. In this review, we summarize the experimental data reported on murine models developing autoimmune diseases and systemic inflammation, and we explore the underlying mechanisms explaining how systemic inflammation can result in behavioral deficits, with a special focus on in vivo neuroimaging techniques.
Collapse
Affiliation(s)
- Mykolas Bendorius
- UMR 7242 Biotechnologie et Signalisation Cellulaire, École Supérieure de Biotechnologie de Strasbourg (ESBS), Laboratoire d'Excellence Médalis, Université de Strasbourg/CNRS, 67412 Illkirch, France.
| | - Chrystelle Po
- ICube UMR 7357, Université de Strasbourg/CNRS, Fédération de Médecine Translationnelle de Strasbourg, 67000 Strasbourg, France.
| | - Sylviane Muller
- UMR 7242 Biotechnologie et Signalisation Cellulaire, École Supérieure de Biotechnologie de Strasbourg (ESBS), Laboratoire d'Excellence Médalis, Université de Strasbourg/CNRS, 67412 Illkirch, France.
- University of Strasbourg Institute for Advanced Study (USIAS), 67000 Strasbourg, France.
| | - Hélène Jeltsch-David
- UMR 7242 Biotechnologie et Signalisation Cellulaire, École Supérieure de Biotechnologie de Strasbourg (ESBS), Laboratoire d'Excellence Médalis, Université de Strasbourg/CNRS, 67412 Illkirch, France.
| |
Collapse
|
37
|
Bendorius M, Neeli I, Wang F, Bonam SR, Dombi E, Buron N, Borgne-Sanchez A, Poulton J, Radic M, Muller S. The Mitochondrion-lysosome Axis in Adaptive and Innate Immunity: Effect of Lupus Regulator Peptide P140 on Mitochondria Autophagy and NETosis. Front Immunol 2018; 9:2158. [PMID: 30319621 PMCID: PMC6168670 DOI: 10.3389/fimmu.2018.02158] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/31/2018] [Indexed: 12/13/2022] Open
Abstract
Mitochondria deserve special attention as sensors of cellular energy homeostasis and metabolic state. Moreover, mitochondria integrate intra- and extra-cellular signals to determine appropriate cellular responses that range from proliferation to cell death. In autoimmunity, as in other inflammatory chronic disorders, the metabolism of immune cells may be extensively remodeled, perturbing sensitive tolerogenic mechanisms. Here, we examine the distribution and effects of the therapeutic 21-mer peptide called P140, which shows remarkable efficacy in modulating immune responses in inflammatory settings. We measured P140 and control peptide effects on isolated mitochondria, the distribution of peptides in live cells, and their influence on the levels of key autophagy regulators. Our data indicate that while P140 targets macro- and chaperone-mediated autophagy processes, it has little effect, if any, on mitochondrial autophagy. Remarkably, however, it suppresses NET release from neutrophils exposed to immobilized NET-anti-DNA IgG complexes. Together, our results suggest that in the mitochondrion-lysosome axis, a likely driver of NETosis and inflammation, the P140 peptide does not operate by affecting mitochondria directly.
Collapse
Affiliation(s)
- Mykolas Bendorius
- Unit Biotechnology and Cell Signaling, Laboratory of Excellence Medalis, CNRS, Strasbourg University, Illkirch, France
| | - Indira Neeli
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Fengjuan Wang
- Unit Biotechnology and Cell Signaling, Laboratory of Excellence Medalis, CNRS, Strasbourg University, Illkirch, France
| | - Srinivasa Reddy Bonam
- Unit Biotechnology and Cell Signaling, Laboratory of Excellence Medalis, CNRS, Strasbourg University, Illkirch, France
| | - Eszter Dombi
- Nuffield Department of Women's and Reproductive Health, Women's Centre, Oxford, United Kingdom
| | | | | | - Joanna Poulton
- Nuffield Department of Women's and Reproductive Health, Women's Centre, Oxford, United Kingdom
| | - Marko Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sylviane Muller
- Unit Biotechnology and Cell Signaling, Laboratory of Excellence Medalis, CNRS, Strasbourg University, Illkirch, France.,Institute for Advanced Study, University of Strasbourg, Strasbourg, France
| |
Collapse
|
38
|
Bonam SR, Wang F, Muller S. Autophagy: A new concept in autoimmunity regulation and a novel therapeutic option. J Autoimmun 2018; 94:16-32. [PMID: 30219390 DOI: 10.1016/j.jaut.2018.08.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023]
Abstract
Nowadays, pharmacologic treatments of autoinflammatory diseases are largely palliative rather than curative. Most of them result in non-specific immunosuppression, which can be associated with broad disruption of natural and induced immunity with significant and sometimes serious unwanted injuries. Among the novel strategies that are under development, tools that modulate the immune system to restore normal tolerance mechanisms are central. In these approaches, peptide therapeutics constitute a class of agents that display many physicochemical advantages. Within this class of potent drugs, the phosphopeptide P140 is very promising for treating patients with lupus, and likely also patients with other chronic inflammatory diseases. We discovered that P140 targets autophagy, a finely orchestrated catabolic process, involved in the regulation of inflammation and in the biology of immune cells. In vitro, P140 acts directly on a particular form of autophagy called chaperone-mediated autophagy, which seems to be hyperactivated in certain subsets of lymphocytes in lupus and in other autoinflammatory settings. In lupus, the "correcting" effect of P140 on autophagy results in a weaker signaling of autoreactive T cells, leading to a significant improvement of pathophysiological status of treated mice. These findings also demonstrated ex vivo in human cells, open novel avenues of therapeutic intervention in pathological conditions, in which specific and not general targeting is highly pursued in the context of the new action plans for personalized medicines.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; CNRS-University of Strasbourg, Laboratory of Excellence Medalis, France
| | - Fengjuan Wang
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; CNRS-University of Strasbourg, Laboratory of Excellence Medalis, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; CNRS-University of Strasbourg, Laboratory of Excellence Medalis, France; University of Strasbourg Institute for Advanced Study, Strasbourg, France.
| |
Collapse
|