1
|
Liu C, Zheng J, Hao J, Kang W, Mao J, Hu C, Ouyang Y, Shen H. Lactylation-related genes serve as potential markers for the diagnosis and immune infiltration in rheumatoid arthritis. Autoimmunity 2025; 58:2474217. [PMID: 40048636 DOI: 10.1080/08916934.2025.2474217] [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: 06/25/2024] [Revised: 11/22/2024] [Accepted: 02/25/2025] [Indexed: 05/13/2025]
Abstract
Lactylation is widely involved in cellular processes and is pivotal in inflammation and immune regulation. However, the expression and clinical significance of lactylation in rheumatoid arthritis (RA) remain unclear. This study aimed to determine the role of lactylation in RA and its association with immune cell infiltration. We initially detected the levels of lactate in the plasma of RA patients and the levels of panlysine lactylation (Pan-Kla) in peripheral blood mononuclear cells (PBMCs). Next, we used differential expression analysis and weighted gene coexpression network analysis (WGCNA) to intersect with lactylation-related genes. We obtained lactylation-related differentially expressed genes (LADEGs) in RA and analyzed their functional enrichment. We subsequently used the CIBERSORT algorithm to analyze immune cell infiltration in RA synovial tissues and its correlation with LADEGs. Finally, key genes of LADEGs were validated in the Pathobiology of Early Arthritis Cohort (PEAC) study and our samples. Our study revealed elevated levels of lactate and lactylation in the peripheral blood of RA patients. IKAROS family zinc finger 1 (IKZF1), lymphocyte cytosolic protein 1 (LCP1), and WASP actin-nucleation promoting factor (WAS) may be potential biomarkers for early diagnosis and assessment of disease activity in RA.
Collapse
Affiliation(s)
- Chunhua Liu
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jianxiong Zheng
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiayao Hao
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wenjiao Kang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jing Mao
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Caiyun Hu
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuhong Ouyang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Haili Shen
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
2
|
Zou AE, Kongthong S, Mueller AA, Brenner MB. Fibroblasts in immune responses, inflammatory diseases and therapeutic implications. Nat Rev Rheumatol 2025:10.1038/s41584-025-01259-0. [PMID: 40369134 DOI: 10.1038/s41584-025-01259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/16/2025]
Abstract
Once regarded as passive bystander cells of the tissue stroma, fibroblasts have emerged as active orchestrators of tissue homeostasis and disease. From regulating immunity and controlling tissue remodelling to governing cell growth and differentiation, fibroblasts assume myriad roles in guiding normal tissue development, maintenance and repair. By comparison, in chronic inflammatory diseases such as rheumatoid arthritis, fibroblasts recruit and sustain inflammatory leukocytes, become dominant producers of pro-inflammatory factors and catalyse tissue destruction. In other disease contexts, fibroblasts promote fibrosis and impair host control of cancer. Single-cell studies have uncovered striking transcriptional and functional heterogeneity exhibited by fibroblasts in both normal tissues and diseased tissues. In particular, advances in the understanding of fibroblast pathology in rheumatoid arthritis have shed light on pathogenic fibroblast states in other chronic diseases. The differentiation and activation of these fibroblast states is driven by diverse physical and chemical cues within the tissue microenvironment and by cell-intrinsic signalling and epigenetic mechanisms. These insights into fibroblast behaviour and regulation have illuminated therapeutic opportunities for the targeted deletion or modulation of pathogenic fibroblasts across many diseases.
Collapse
Affiliation(s)
- Angela E Zou
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suppawat Kongthong
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alisa A Mueller
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA and Palo Alto Veterans Affairs Health Care System, Palo Alto, CA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Marshall L, Raychaudhuri S, Viatte S. Understanding rheumatic disease through continuous cell state analysis. Nat Rev Rheumatol 2025:10.1038/s41584-025-01253-6. [PMID: 40335652 DOI: 10.1038/s41584-025-01253-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2025] [Indexed: 05/09/2025]
Abstract
Autoimmune rheumatic diseases are a heterogeneous group of conditions, including rheumatoid arthritis (RA) and systemic lupus erythematosus. With the increasing availability of large single-cell datasets, novel disease-associated cell types continue to be identified and characterized at multiple omics layers, for example, 'T peripheral helper' (TPH) (CXCR5- PD-1hi) cells in RA and systemic lupus erythematosus and MerTK+ myeloid cells in RA. Despite efforts to define disease-relevant cell atlases, the very definition of a 'cell type' or 'lineage' has proven controversial as higher resolution assays emerge. This Review explores the cell types and states involved in disease pathogenesis, with a focus on the shifting perspectives on immune and stromal cell taxonomy. These understandings of cell identity are closely related to the computational methods adopted for analysis, with implications for the interpretation of single-cell data. Understanding the underlying cellular architecture of disease is also crucial for therapeutic research as ambiguity hinders translation to the clinical setting. We discuss the implications of different frameworks for cell identity for disease treatment and the discovery of predictive biomarkers for stratified medicine - an unmet clinical need for autoimmune rheumatic diseases.
Collapse
Affiliation(s)
- Lysette Marshall
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Soumya Raychaudhuri
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Divisions of Rheumatology, Inflammation and Immunity and Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Sebastien Viatte
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK.
- NIHR Manchester Musculoskeletal Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| |
Collapse
|
4
|
Andersen BB, Greisen S, Stengaard-Pedersen K, Junker P, Hørslev-Petersen K, Hetland ML, Østergaard M, Ørnbjerg LM, Hvid M, Deleuran B, Nielsen MA. Plasma haem oxygenase-1 may represent a first-in-class biomarker of oxidative stress in rheumatoid arthritis. Scand J Rheumatol 2025; 54:153-157. [PMID: 39287051 DOI: 10.1080/03009742.2024.2392364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVES This study explores the early identification of rheumatoid arthritis (RA) patients at elevated risk of progression. Haem-oxygenase-1 (HO-1) is a marker of oxidative stress in inflammation. Here, we investigate HO-1 as a biomarker of oxidative stress and its association with clinical disease activity and radiographic progression in RA. METHOD Baseline HO-1 was measured sequentially in plasma samples from patients with early rheumatoid arthritis (eRA) (n = 80). Disease Activity Score based on 28-joint count-C-reactive protein, Clinical Disease Activity Index, and total Sharp score were used to evaluate the disease course serially over 2 years. Paired plasma and synovial fluid samples were examined for HO-1 in active established rheumatoid arthritis (esRA) (n = 20). Plasma from healthy control subjects was also included (n = 35). RESULTS Plasma HO-1 levels were increased in eRA {1373 pg/mL [interquartile range (IQR) 1110-2050]} and esRA [2034 pg/mL (IQR 1630-2923)] compared with controls [1064 pg/mL (IQR 869.5-1378)]. HO-1 plasma levels decreased with treatment. Baseline HO-1 correlated with disease activity and radiographic progression. A strong, linear correlation was found between synovial and plasma HO-1 levels (r = 0.75, p < 0.001). CONCLUSION In eRA, plasma levels of HO-1 were increased and correlated with disease and radiographic progression. A baseline measurement of plasma HO-1 levels demonstrated superior performance to currently used clinical and serological disease markers in the prediction of radiographic progression. Plasma HO-1 may function as a first-in-class biomarker of synovial oxidative stress in RA.
Collapse
Affiliation(s)
- B B Andersen
- Department of Biomedicine, Health, Aarhus University, Aarhus, Denmark
| | - S Greisen
- Department of Biomedicine, Health, Aarhus University, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | | | - P Junker
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - K Hørslev-Petersen
- Danish Hospital for the Rheumatic Diseases, University of Southern Denmark, Odense, Denmark
| | - M Lund Hetland
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M Østergaard
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - L Midtbøll Ørnbjerg
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M Hvid
- Department of Biomedicine, Health, Aarhus University, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - B Deleuran
- Department of Biomedicine, Health, Aarhus University, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - M A Nielsen
- Department of Biomedicine, Health, Aarhus University, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- Department of Internal Medicine, Horsens Regional Hospital, Horsens, Denmark
| |
Collapse
|
5
|
Barbuti PA, Guardia-Laguarta C, Yun T, Chatila ZK, Flowers X, Wong C, Santos BFR, Larsen SB, Lotti JS, Hattori N, Bradshaw E, Dettmer U, Fanning S, Menon V, Reddy H, Teich AF, Krüger R, Area-Gomez E, Przedborski S. The role of alpha-synuclein in synucleinopathy: Impact on lipid regulation at mitochondria-ER membranes. NPJ Parkinsons Dis 2025; 11:103. [PMID: 40307230 PMCID: PMC12043847 DOI: 10.1038/s41531-025-00960-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
The protein alpha-synuclein (αSyn) plays a pivotal role in the pathogenesis of synucleinopathies, including Parkinson's disease and multiple system atrophy, with growing evidence indicating that lipid dyshomeostasis is a key phenotype in these neurodegenerative disorders. Previously, we identified that αSyn localizes, at least in part, to mitochondria-associated endoplasmic reticulum membranes (MAMs), which are transient functional domains containing proteins that regulate lipid metabolism, including the de novo synthesis of phosphatidylserine. In the present study, we analyzed the lipid composition of postmortem human samples, focusing on the substantia nigra pars compacta of Parkinson's disease and controls, as well as three less affected brain regions of Parkinson's donors. To further assess synucleinopathy-related lipidome alterations, similar analyses were performed on the striatum of multiple system atrophy cases. Our data reveal region- and disease-specific changes in the levels of lipid species. Specifically, our data revealed alterations in the levels of specific phosphatidylserine species in brain areas most affected in Parkinson's disease. Some of these alterations, albeit to a lesser degree, are also observed in multiple system atrophy. Using induced pluripotent stem cell-derived neurons, we show that αSyn regulates phosphatidylserine metabolism at MAM domains, and that αSyn dosage parallels the perturbation in phosphatidylserine levels. These findings support the notion that αSyn pathophysiology is linked to the dysregulation of lipid homeostasis, which may contribute to the vulnerability of specific brain regions in synucleinopathy. These findings have significant therapeutic implications.
Collapse
Affiliation(s)
- Peter A Barbuti
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Cristina Guardia-Laguarta
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Taekyung Yun
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, Spain
| | - Zena K Chatila
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Xena Flowers
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY, USA
| | - Chantel Wong
- Department of Neuroscience, Barnard College of Columbia University, New York, NY, USA
| | - Bruno F R Santos
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
- Disease Modelling and Screening Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - Simone B Larsen
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - James S Lotti
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Elizabeth Bradshaw
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Hasini Reddy
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew F Teich
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, Spain
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Serge Przedborski
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Neuroscience, Columbia University, New York, NY, USA.
| |
Collapse
|
6
|
Winthrop KL, Bathon J, Kerschbaumer A, Isaacs JD, Mease P, Gottenberg JE, Crow MK, Kay J, Crofford L, Baraliakos X, Bykerk V, Siebert S, Kloppenburg M, Aletaha D, McInnes IB, Huizinga T, Voll R, Gravallese EM, Breedveld FC, van Vollenhoven R, Smolen JS. Chasing the target: reports from the Advances in Targeted Therapies meeting, 2024. Ann Rheum Dis 2025:S0003-4967(25)00237-7. [PMID: 40240265 DOI: 10.1016/j.ard.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 04/18/2025]
Abstract
OBJECTIVES The Advances in Targeted Therapies annual meeting brings together experts within the field of rheumatology and immunology to highlight and discuss the latest scientific developments and needs in the field. The objective is to highlight unmet scientific needs in the field of rheumatology. METHODS The 24th annual Advances in Targeted Therapies meeting convened with more than 100 international clinicians and scientific researchers in rheumatology, immunology, and other specialities relating to all aspects of immune-mediated inflammatory diseases. During the meeting, we held 5 rheumatologic disease-specific discussion sections consisting of experts in each field. These groups included rheumatoid arthritis (RA), psoriatic arthritis (PsA), axial spondyloarthritis (axSpA), osteoarthritis (OA), and systemic lupus erythematosus (SLE). In each group, experts were asked to identify the top 2 to 3 most important overarching and disease-specific scientific unmet needs to be addressed in the next 5 years. RESULTS The overarching themes across disciplines included the need for precision medicine, improved classification of disease states, and the further identification of targets and associated therapies, including the potential role of chimeric antigen receptor (CAR) T cell therapies. Within RA, the group highlighted the lack of precision medicine and the need for better biomarkers. Further, the lack of targeted therapies against fibroblasts in RA was discussed, with the potential impact of targeting fibroblasts early in the disease as an unmet need. For PsA, there is a continued need for a better definition of disease endotypes and for the categorisation of those with complex and difficult-to-treat (D2T) diseases. The development of bispecific molecules and combination therapeutic approaches remain a high priority. For axSpA, the disease-modifying characteristics of nonsteroid anti-inflammatory drugs need further evaluation, as does the treatment of residual pain and fatigue frequently in the disease. In OA, new therapeutic targets remain an unmet need, and the discussion group prioritised potential experimental strategies that could lead to innovative therapeutic targets. Elucidating the specific signalling and target cells responsible for, or inhibiting, repair will be essential for developing targeted therapies. SLE experts emphasised the need to identify the most predictive biological contributions to disease progression in patients with early clinical precursors of SLE. The role of CAR T cell therapy must be further investigated, along with ancillary biologic studies (eg, immune system profiling) that provide critical insights into disease pathogenesis. Further, there is a need to determine the relationship of patient-relevant symptoms to the pathophysiology of SLE and identify new therapeutic targets for these symptoms. CONCLUSIONS There remain many unmet needs on the road to precision medicine with regard to identifying disease endotypes and biomarkers for disease progression or therapeutic response. For most diseases discussed, a strong unmet need remains with regard to identifying new targets and therapies for those with refractory or D2T disease. The ability to prevent or cure rheumatic disease remains the ultimate unmet need in rheumatology.
Collapse
Affiliation(s)
| | - Joan Bathon
- Division of Rheumatology, Columbia University, New York, NY, USA
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University and Musculoskeletal Unit, Newcastle Hospitals, Newcastle upon Tyne, UK
| | - Philip Mease
- Swedish Medical Center, University of Washington, Seattle, WA, USA
| | - Jaque-Eric Gottenberg
- Centre National de Référence des Maladies AutoImmunes Systémiques Rares, CHU Strasbourg-Hautepierre, Strasbourg, France
| | - Mary K Crow
- Division of Rheumatology, Hospital for Special Surgery, New York, NY, USA
| | - Jonathan Kay
- Division of Rheumatology, Department of Medicine, UMass Chan Medical School and UMass Memorial Medical Center, Worcester, MA, USA
| | - Leslie Crofford
- Division of Rheumatology and Immunology, Vanderbilt University, Nashville, TN, USA
| | | | - Vivian Bykerk
- Hospital for Special Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Stefan Siebert
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | | | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Iain B McInnes
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Thomas Huizinga
- Department of Rheumatology, University of Leiden, Leiden, The Netherlands
| | - Reinhard Voll
- Department of Rheumatology and Clinical Immunology, University of Freiburg, Freiburg, Germany
| | | | | | - Ronald van Vollenhoven
- Clinical Immunology and Rheumatology Department, VU Medical University, Amsterdam, The Netherlands
| | - Josef S Smolen
- Division of Rheumatology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Park S, Yoon K, Hong E, Kim MW, Kang MG, Mizuno S, Kim HJ, Lee MJ, Choi HJ, Heo JS, Bae JB, An H, Park N, Park H, Kim P, Son M, Pang K, Park JY, Takahashi S, Kwon YJ, Kang DW, Kim SJ. Tm4sf19 inhibition ameliorates inflammation and bone destruction in collagen-induced arthritis by suppressing TLR4-mediated inflammatory signaling and abnormal osteoclast activation. Bone Res 2025; 13:40. [PMID: 40128226 PMCID: PMC11933450 DOI: 10.1038/s41413-025-00419-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/02/2025] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by inflammation and abnormal osteoclast activation, leading to bone destruction. We previously demonstrated that the large extracellular loop (LEL) of Tm4sf19 is important for its function in osteoclast differentiation, and LEL-Fc, a competitive inhibitor of Tm4sf19, effectively suppresses osteoclast multinucleation and prevent bone loss associated with osteoporosis. This study aimed to investigate the role of Tm4sf19 in RA, an inflammatory and abnormal osteoclast disease, using a mouse model of collagen-induced arthritis (CIA). Tm4sf19 expression was observed in macrophages and osteoclasts within the inflamed synovium, and Tm4sf19 expression was increased together with inflammatory genes in the joint bones of CIA-induced mice compared with the sham control group. Inhibition of Tm4sf19 by LEL-Fc demonstrated both preventive and therapeutic effects in a CIA mouse model, reducing the CIA score, swelling, inflammation, cartilage damage, and bone damage. Knockout of Tm4sf19 gene or inhibition of Tm4sf19 activity by LEL-Fc suppressed LPS/IFN-γ-induced TLR4-mediated inflammatory signaling in macrophages. LEL-Fc disrupted not only the interaction between Tm4sf19 and TLR4/MD2, but also the interaction between TLR4 and MD2. μCT analysis showed that LEL-Fc treatment significantly reduced joint bone destruction and bone loss caused by hyperactivated osteoclasts in CIA mice. Taken together, these findings suggest that LEL-Fc may be a potential treatment for RA and RA-induced osteoporosis by simultaneously targeting joint inflammation and bone destruction caused by abnormal osteoclast activation.
Collapse
Affiliation(s)
- Sujin Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Eunji Hong
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Min Gi Kang
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | | | | | | | - Jin Sun Heo
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Haein An
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Naim Park
- Medpacto Inc., Seoul, Republic of Korea
| | - Hyeyeon Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Pyunggang Kim
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Minjung Son
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyoungwha Pang
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Je Yeun Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yong Jung Kwon
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Seong-Jin Kim
- GILO Institute, GILO Foundation, Seoul, Republic of Korea.
- Medpacto Inc., Seoul, Republic of Korea.
| |
Collapse
|
8
|
Bhamidipati K, McIntyre ABR, Kazerounian S, Ce G, Tran M, Prell SA, Lau R, Khedgikar V, Altmann C, Small A, Wong V, Madhu R, Presti S, Anufrieva KS, Blazar PE, Lange JK, Seifert J, Moreland LW, Croft AP, Lewis MJ, Thomas R, Jonsson AH, Pitzalis C, Gravallese EM, Brenner MB, Korsunsky I, Wechalekar MD, Wei K. Spatial patterning of fibroblast TGFβ signaling underlies treatment resistance in rheumatoid arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.642821. [PMID: 40166167 PMCID: PMC11956964 DOI: 10.1101/2025.03.14.642821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Treatment-refractory rheumatoid arthritis (RA) is a major unmet need, and the mechanisms driving treatment resistance are poorly understood. To identify molecular determinants of RA non-remission, we performed spatial transcriptomic profiling on pre- and post-treatment synovial tissue biopsies from treatment naïve patients who received conventional DMARDs or adalimumab for 6 months. In the baseline biopsies of non-remission patients, we identified significant expansion of fibrogenic fibroblasts marked by high expression of COMP, a fibrosis-associated extracellular matrix protein. COMPhi fibroblasts localized to perivascular niches that, unexpectedly, served as transcriptional hubs for TGFβ activity. We identified endothelial-derived Notch signaling as an upstream regulator of fibroblast TGFβ signaling via its dual role in driving TGFβ isoform expression and suppressing TGFβ receptors, generating a proximal-distal gradient of TGFβ activity. Further, disruption of steady-state Notch signaling in vitro enabled fibrogenic fibroblast activation. Analysis of post-treatment biopsies revealed marked expansion of COMPhi fibroblasts in non-remission RA patients, despite evidence of successful immune cell depletion, suggesting a spatiotemporal process of fibrogenic remodeling linked to treatment resistance. Collectively, our data implicates targeting of TGFβ signaling to prevent exuberant synovial tissue fibrosis as a potential therapeutic strategy for refractory RA.
Collapse
Affiliation(s)
- Kartik Bhamidipati
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Alexa B R McIntyre
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Shideh Kazerounian
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Gao Ce
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Miles Tran
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Sean A Prell
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Rachel Lau
- Centre for Experimental Medicine and Rheumatology, EULAR Centre of Excellence, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust, Barts Biomedical Research Centre (BRC), National Institute for Health and Care Research (NIHR), London, UK
| | - Vikram Khedgikar
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Christopher Altmann
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Annabelle Small
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Vincent Wong
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Roopa Madhu
- Division of Genetics, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Sonia Presti
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Ksenia S Anufrieva
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Philip E Blazar
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeffrey K Lange
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jennifer Seifert
- Division of Rheumatology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Larry W Moreland
- Division of Rheumatology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Adam P Croft
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, EULAR Centre of Excellence, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust, Barts Biomedical Research Centre (BRC), National Institute for Health and Care Research (NIHR), London, UK
| | - Ranjeny Thomas
- Frazer Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Anna H Jonsson
- Division of Rheumatology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, EULAR Centre of Excellence, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust, Barts Biomedical Research Centre (BRC), National Institute for Health and Care Research (NIHR), London, UK
| | - Ellen M Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| | - Ilya Korsunsky
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Mihir D Wechalekar
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital at Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Karsdal M, Cox TR, Parker AL, Willumsen N, Sand JMB, Jenkins G, Hansen HH, Oldenburger A, Geillinger-Kaestle KE, Larsen AT, Black D, Genovese F, Eckersley A, Heinz A, Nyström A, Holm Nielsen S, Bennink L, Johannsson L, Bay-Jensen AC, Orange DE, Friedman S, Røpke M, Fiore V, Schuppan D, Rieder F, Simona B, Borthwick L, Skarsfeldt M, Wennbo H, Thakker P, Stoffel R, Clarke GW, Kalluri R, Ruane D, Zannad F, Mortensen JH, Sinkeviciute D, Sundberg F, Coseno M, Thudium C, Croft AP, Khanna D, Cooreman M, Broermann A, Leeming DJ, Mobasheri A, Ricard-Blum S. Advances in Extracellular Matrix-Associated Diagnostics and Therapeutics. J Clin Med 2025; 14:1856. [PMID: 40142664 PMCID: PMC11943371 DOI: 10.3390/jcm14061856] [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: 12/20/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 03/28/2025] Open
Abstract
The extracellular matrix (ECM) is the common denominator of more than 50 chronic diseases. Some of these chronic pathologies lead to enhanced tissue formation and deposition, whereas others are associated with increased tissue degradation, and some exhibit a combination of both, leading to severe tissue alterations. To develop effective therapies for diseases affecting the lung, liver, kidney, skin, intestine, musculoskeletal system, heart, and solid tumors, we need to modulate the ECM's composition to restore its organization and function. Across diverse organ diseases, there are common denominators and distinguishing factors in this fibroinflammatory axis, which may be used to foster new insights into drug development across disease indications. The 2nd Extracellular Matrix Pharmacology Congress took place in Copenhagen, Denmark, from 17 to 19 June 2024 and was hosted by the International Society of Extracellular Matrix Pharmacology. The event was attended by 450 participants from 35 countries, among whom were prominent scientists who brought together state-of-the-art research on organ diseases and asked important questions to facilitate drug development. We highlight key aspects of the ECM in the liver, kidney, skin, intestine, musculoskeletal system, lungs, and solid tumors to advance our understanding of the ECM and its central targets in drug development. We also highlight key advances in the tools and technology that enable this drug development, thereby supporting the ECM.
Collapse
Affiliation(s)
- Morten Karsdal
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Thomas R. Cox
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Amelia L. Parker
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Nicholas Willumsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Jannie Marie Bülow Sand
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, NIHR Imperial Biomedical Research Centre, Imperial College London, London SW7 2AZ, UK;
| | | | | | - Kerstin E. Geillinger-Kaestle
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Anna Thorsø Larsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | - Federica Genovese
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Alexander Eckersley
- Wellcome Centre for Cell Matrix Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK;
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center—University of Freiburg, 79106 Breisgau, Germany;
| | - Signe Holm Nielsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | | | - Anne-Christine Bay-Jensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dana E. Orange
- Hospital for Special Surgery, The Rockefeller University, New York, NY 10065, USA;
| | - Scott Friedman
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA;
| | | | - Vincent Fiore
- Boehringer Ingelheim, 55218 Ingelheim am Rhein, Germany;
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Florian Rieder
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | | | - Lee Borthwick
- FibroFind Ltd., FibroFind Laboratories, Medical School, Newcastle upon Tyne NE2 4HH, UK;
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Mark Skarsfeldt
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Haakan Wennbo
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Paresh Thakker
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Ruedi Stoffel
- Roche Diagnostics International Ltd., 6343 Rotkreuz, Switzerland;
| | - Graham W. Clarke
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden;
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College, London E1 9RT, UK
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Darren Ruane
- Janssen Immunology, Translational Sciences and Medicine, La Jolla, CA 92037, USA;
| | - Faiez Zannad
- Division of Heart Failure and Hypertension, and of the Inserm CIC, University of Lorraine, 54000 Metz, France;
| | - Joachim Høg Mortensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dovile Sinkeviciute
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Fred Sundberg
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Molly Coseno
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Christian Thudium
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Adam P. Croft
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, University of Birmingham, Birmingham B15 2TT, UK;
- Institute of Inflammation and Ageing, Queen Elizabeth Hospital, University of Birmingham, Birmingham B15 2TT, UK
| | - Dinesh Khanna
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | | | - Andre Broermann
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Diana Julie Leeming
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Ali Mobasheri
- Faculty of Medicine, University of Oulu, 90570 Oulu, Finland;
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
- Faculté de Médecine, Université de Liège, 4000 Liège, Belgium
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Sylvie Ricard-Blum
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, ICBMS, University Lyon 1, 69622 Villeurbanne Cedex, France;
| |
Collapse
|
10
|
Jiang P, Zhao Y, Jia Y, Ma H, Guo Y, Yan W, Xi X. Multi-omics study on autophagic dysfunction molecular network in the pathogenesis of rheumatoid arthritis. J Transl Med 2025; 23:274. [PMID: 40045304 PMCID: PMC11881334 DOI: 10.1186/s12967-025-06288-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/23/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Autophagy is associated with the development of rheumatoid arthritis (RA), but its genetic pathological mechanisms remain incompletely understood. In this study, we employed summary-data-based Mendelian randomization (SMR) and co-localization analysis to systematically investigate the relationship between autophagy-related genes and RA. METHODS We obtained summary data on blood methylation (mQTL), gene expression (eQTL), and protein abundance (pQTL) from respective quantitative trait locus (QTL) studies. Genetic association data for RA were primarily derived from the FinnGen database, with validation performed using the UK Biobank (UKB) and GWAS Catalog databases. SMR analysis was conducted to evaluate the association between molecular characteristics of autophagy-related genes and RA. Subsequently, co-localization analysis was performed to determine whether the identified signals share the same causal genetic variants. RESULTS After integrating mQTL-eQTL multi-omics data, we identified two key autophagy genes, BCL2L1 and RAF1, which may have a causal relationship with RA. Significant associations were found for BCL2L1 (cg12873919, cg13989999) and RAF1 (cg26432171) in the SMR analysis of autophagy-related mQTL, eQTL, and GWAS data (p SMR < 0.05). In the integrated mQTL-eQTL SMR analysis, cg12873919 (p SMR = 1.40E-07, OR = 0.82, 95% CI [0.76-0.88]), cg13989999 (p SMR = 1.43E-06, OR = 0.78, 95% CI [0.71-0.87]), and cg26432171 (p SMR = 9.18E-09, OR = 1.83, 95% CI [1.49-2.25]) were all significantly validated. Methylation of cg12873919 and cg13989999 in BCL2L1 was associated with increased BCL2L1 expression, consistent with their negative impact on RA risk. Conversely, the cg26432171 site in RAF1 showed a positive correlation between gene methylation and expression. In the eQTL-GWAS SMR analysis, MAPK3 expression (p SMR = 7.24E-05, OR = 0.91, 95% CI [0.87-0.95]) was negatively correlated with RA risk, a finding supported by co-localization analysis (PPH4 > 0.5), suggesting that this gene may inhibit RA pathogenesis by regulating the autophagy process. Furthermore, protein level analysis also supported the protective role of MAPK3 (p SMR = 7.53E-05, OR = 0.89, 95% CI [0.84-0.94]). CONCLUSION We identified that autophagy-related genes BCL2L1 and RAF1 may be associated with RA risk, providing strong evidence from multi-omics data. This study identifies autophagy genes related to RA, potentially offering new insights into the pathogenesis of RA.
Collapse
Affiliation(s)
- Ping Jiang
- Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Ruijin Rehabilitation Hospital, Shanghai, 200023, China
| | - Yichen Zhao
- Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Youji Jia
- Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Honghong Ma
- Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yajuan Guo
- Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Yan
- Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiaobing Xi
- Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
11
|
Taylor PC. Towards better personalized care in rheumatoid arthritis. Rheumatology (Oxford) 2025; 64:ii1-ii2. [PMID: 39883572 PMCID: PMC11981329 DOI: 10.1093/rheumatology/keaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 02/01/2025] Open
Affiliation(s)
- Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Shim JV, Rehberg M, Wagenhuber B, van der Graaf PH, Chung DW. Combining mechanistic modeling with machine learning as a strategy to predict inflammatory bowel disease clinical scores. Front Pharmacol 2025; 16:1479666. [PMID: 40070575 PMCID: PMC11893853 DOI: 10.3389/fphar.2025.1479666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/27/2025] [Indexed: 03/14/2025] Open
Abstract
Disease activity scores are efficacy endpoints in clinical trials of inflammatory bowel disease (IBD) therapies. Crohn's disease activity index (CDAI), Mayo endoscopic score (MES) and Mayo score are frequently used in clinical trials. They rely on either the physician's observation of the inflammatory state of the patient's gastrointestinal tissue alone or combined with the patient's subjective evaluation of general wellbeing. Given the importance of these scores in evaluating the efficacy of drug treatment and disease severity, there has been interest in developing a computational approach to reliably predict these scores. A promising approach is using mechanistic models such as quantitative systems pharmacology (QSP) which simulate the mechanisms of the disease and its modulation by the drug pharmacology. However, extending QSP model simulations to clinical score predictions has been challenging due to the limited availability of gut biopsy measurements and the subjective nature of some of the evaluation criteria for these scores that cannot be described using mechanistic relationships. In this perspective, we examine details of IBD disease activity scores and current progress in building predictive models for these scores (such as biomarkers for disease activity). Then, we propose a method to leverage simulated markers of inflammation from a QSP model to predict IBD clinical scores using a machine learning algorithm. We will demonstrate how this combined approach can be used to (1) explore mechanistic insights underlying clinical observations; and (2) simulate novel therapeutic strategies that could potentially improve clinical outcomes.
Collapse
Affiliation(s)
- Jaehee V. Shim
- Certara Applied BioSimulation, Sheffield, United Kingdom
| | - Markus Rehberg
- Sanofi R&D, Translational Disease Modeling, Frankfurt amMain, Germany
| | - Britta Wagenhuber
- Sanofi R&D, Translational Disease Modeling, Frankfurt amMain, Germany
| | - Piet H. van der Graaf
- Certara Applied BioSimulation, Sheffield, United Kingdom
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | | |
Collapse
|
13
|
Gu F, Meyer AJ, Ježek F, Zhang S, Catalan T, Miller A, Schenk NA, Sturgess VE, Uceda D, Li R, Wittrup E, Hua X, Carlson BE, Tang YD, Raza F, Najarian K, Hummel SL, Beard DA. Identification of digital twins to guide interpretable AI for diagnosis and prognosis in heart failure. NPJ Digit Med 2025; 8:110. [PMID: 39966509 PMCID: PMC11836287 DOI: 10.1038/s41746-025-01501-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Heart failure (HF) is a highly heterogeneous condition, and current methods struggle to synthesize extensive clinical data for personalized care. Using data from 343 HF patients, we developed mechanistic computational models of the cardiovascular system to create digital twins. These twins, consisting of optimized measurable and unmeasurable parameters alongside simulations of cardiovascular function, provided comprehensive representations of individual disease states. Unsupervised machine learning applied to digital twin-derived features identified interpretable phenogroups and mechanistic drivers of cardiovascular death risk. Incorporating these features into prognostic AI models improved performance, transferability, and interpretability compared to models using only clinical variables. This framework demonstrates potential to enhance prognosis and guide therapy, paving the way for more precise, individualized HF management.
Collapse
Affiliation(s)
- Feng Gu
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48105, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Andrew J Meyer
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48105, USA
- Department of Mathematics, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Filip Ježek
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Shuangdi Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Tonimarie Catalan
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Alexandria Miller
- Department of Cardiology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Noah A Schenk
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Victoria E Sturgess
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Domingo Uceda
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Rui Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Emily Wittrup
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Xinwei Hua
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Brian E Carlson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Yi-Da Tang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Farhan Raza
- Department of Medicine-Cardiovascular Disease, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Kayvan Najarian
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48105, USA
- Max Harry Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48105, USA
- Michigan Institute for Data and AI in Society (MIDAS), University of Michigan, Ann Arbor, MI, 48105, USA
- Center for Data-Driven Drug Development and Treatment Assessment (DATA), Ann Arbor, MI, 48105, USA
| | - Scott L Hummel
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48105, USA
- Section of Cardiology, Department of Medicine, VA Ann Arbor Health System, Ann Arbor, MI, 48109, USA
| | - Daniel A Beard
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
14
|
Marks KE, Horisberger A, Solomon DH, Rao DA. Defining immune cell phenotypes that distinguish treatment responders and non-responders in RA. Semin Arthritis Rheum 2025; 70S:152581. [PMID: 39562275 DOI: 10.1016/j.semarthrit.2024.152581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/21/2024]
Affiliation(s)
- Kathryne E Marks
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Daniel H Solomon
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Zheng L, Gu M, Li X, Hu X, Chen C, Kang Y, Pan B, Chen W, Xian G, Wu X, Li C, Wang C, Li Z, Guan M, Zhou G, Mobasheri A, Song W, Peng S, Sheng P, Zhang Z. ITGA5 + synovial fibroblasts orchestrate proinflammatory niche formation by remodelling the local immune microenvironment in rheumatoid arthritis. Ann Rheum Dis 2025; 84:232-252. [PMID: 39919897 DOI: 10.1136/ard-2024-225778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVES To investigate the phenotypic heterogeneity of tissue-resident synovial fibroblasts and their role in inflammatory response in rheumatoid arthritis (RA). METHODS We used single-cell and spatial transcriptomics to profile synovial cells and spatial gene expressions of synovial tissues to identify phenotypic changes in patients with osteoarthritis, RA in sustained remission and active state. Immunohistology, multiplex immunofluorescence and flow cytometry were used to identify synovial fibroblasts subsets. Deconvolution methods further validated our findings in two cohorts (PEAC and R4RA) with treatment response. Cell coculture was used to access the potential cell-cell interactions. Adoptive transfer of synovial cells in collagen-induced arthritis (CIA) mice and bulk RNA sequencing of synovial joints further validate the cellular functions. RESULTS We identified a novel tissue-remodelling CD45-CD31-PDPN+ITGA5+ synovial fibroblast population with unique transcriptome of POSTN, COL3A1, CCL5 and TGFB1, and enriched in immunoregulatory pathways. This subset was upregulated in active and lympho-myeloid type of RA, associated with an increased risk of multidrug resistance. Transforming growth factor (TGF)-β1 might participate in the differentiation of this subset. Moreover, ITGA5+ synovial fibroblasts might occur in early stage of inflammation and induce the differentiation of CXCL13hiPD-1hi peripheral helper T cells (TPHs) from naïve CD4+ T cells, by secreting TGF-β1. Intra-articular injection of ITGA5+ synovial fibroblasts exacerbates RA development and upregulates TPHs in CIA mice. CONCLUSIONS We demonstrate that ITGA5+ synovial fibroblasts might regulate the RA progression by inducing the differentiation of CXCL13hiPD-1hi TPHs and remodelling the proinflammatory microenvironments. Therapeutic modulation of this subpopulation could therefore be a potential treatment strategy for RA.
Collapse
Affiliation(s)
- Linli Zheng
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Minghui Gu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xuantao Hu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Spine Surgery, Sun Yat-sen University Third Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chen Chen
- Trauma Orthopedics, Foot and Ankle Surgery, Sun Yat-sen Memorial Hostpial, Guangzhou, Guangdong, China; Institute of Precision Medicine, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Yunze Kang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Baiqi Pan
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Weishen Chen
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | | | - Xiaoyu Wu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chengxin Li
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chao Wang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Zhiwen Li
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Mingqiang Guan
- Department of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China
| | - Guanming Zhou
- Department of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Public Health Aspects of Musculoskeletal Health and Aging, World Health Organization Collaborating Centre, Liege, Belgium
| | - Weidong Song
- Trauma Orthopedics, Foot and Ankle Surgery, Sun Yat-sen Memorial Hostpial, Guangzhou, Guangdong, China
| | - Sui Peng
- Institute of Precision Medicine, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Clinical Trials Unit, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Gastroenterology and Hepatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| | - Puyi Sheng
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| | - Ziji Zhang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| |
Collapse
|
16
|
Smith MH, Bai Z, Lakhanpal A, Ramirez D, DiCarlo E, Donlin L, Orange D, Goodman SM. Characterizing molecular targets in difficult-to-treat rheumatoid arthritis. Semin Arthritis Rheum 2025; 70S:152588. [PMID: 39603842 PMCID: PMC11761353 DOI: 10.1016/j.semarthrit.2024.152588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Affiliation(s)
- Melanie H Smith
- Hospital for Special Surgery, New York, NY 10021, USA; Weill Cornell Medicine, New York, NY 10065, USA.
| | - Zilong Bai
- Weill Cornell Medicine, New York, NY 10065, USA
| | | | | | | | - Laura Donlin
- Hospital for Special Surgery, New York, NY 10021, USA; Weill Cornell Medicine, New York, NY 10065, USA
| | - Dana Orange
- Hospital for Special Surgery, New York, NY 10021, USA; Rockefeller University, New York, NY 10065, USA
| | - Susan M Goodman
- Hospital for Special Surgery, New York, NY 10021, USA; Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
17
|
Fantecelle CH, Polaco Covre L, Lopes PO, Sarmento IV, Decote-Ricardo D, Geraldo Freire-de-Lima C, de Matos Guedes HL, Pimentel MIF, Conceição-Silva F, Maretti-Mira AC, Borges VM, Pedreira de Carvalho L, de Carvalho EM, Mosser D, Falqueto A, Akbar AN, Gomes DCO. Senescence-related genes are associated with the immunopathology signature of American tegumentary leishmaniasis lesions and may predict progression to mucosal leishmaniasis. Clin Exp Immunol 2025; 219:uxae088. [PMID: 39428748 PMCID: PMC11771187 DOI: 10.1093/cei/uxae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/27/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024] Open
Abstract
The American tegumentary leishmaniasis (ATL) is caused by protozoans of the genus Leishmania and varies from mild localized cutaneous leishmaniasis (LCL) form to more severe manifestations such as the diffuse cutaneous leishmaniasis (DCL) form and the mucosal leishmaniasis (ML) form. Previously, we demonstrated the accumulation of senescent cells in skin lesions of patients with LCL. Moreover, lesional transcriptomic analyses revealed a robust co-induction of senescence and pro-inflammatory gene signatures, highlighting the critical role of senescent T cells in orchestrating pathology. In this work we hypothesized that senescent cells might operate differently among the ATL spectrum, potentially influencing immunopathological mechanisms and clinical outcome. We analysed previously published RNA-Seq datasets of skin biopsies of healthy subjects and lesional skin from DCL patients, LCL patients, and LCL patients that, after treatment, progressed to mucosal leishmaniasis (MLP). Our findings demonstrate a robust presence of a CD8 T-cell signature associated with both LCL and MLP lesions. Moreover, both inflammatory and cytotoxic signatures were significantly upregulated, showing a strong increase in MLP and LCL groups, but not DCL. The senescence signature was elevated between LCL and MLP groups, representing the only distinguishable signature of immunopathology between them. Interestingly, our analyses further revealed the senescence signature's capacity to predict progression from LCL to mucosal forms, which was not observed with other signatures. Both the senescence-signature score and specific senescence-associated genes demonstrated an increased capacity to predict mucosal progression, with correct predictions exceeding 97% of cases. Collectively, our findings contribute to a comprehensive understanding of immunosenescence in ATL and suggest that senescence may represent the latest and most important signature of the immunopathogenisis. This highlights its potential value in predicting disease severity.
Collapse
Affiliation(s)
| | - Luciana Polaco Covre
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, Brazil
- Division of Medicine, University College London, London, UK
| | - Paola Oliveira Lopes
- Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitória, Brazil
| | | | - Debora Decote-Ricardo
- Departamento de Veterinária, Universidade Federal Rural do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Herbert Leonel de Matos Guedes
- Instituto de Microbiologia Professor Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | | | - Ana C Maretti-Mira
- USC Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Valéria M Borges
- Instituto Gonçalo Muniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | | | | | - David Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Aloisio Falqueto
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitoria, Brazil
| | - Arne N Akbar
- Division of Medicine, University College London, London, UK
| | - Daniel Claudio Oliveira Gomes
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, Brazil
- Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitória, Brazil
| |
Collapse
|
18
|
Neofotistou-Themeli E, Goutakoli P, Chanis T, Semitekolou M, Sevdali E, Sidiropoulos P. Fibroblasts in rheumatoid arthritis: novel roles in joint inflammation and beyond. Front Med (Lausanne) 2025; 11:1376925. [PMID: 39906351 PMCID: PMC11790453 DOI: 10.3389/fmed.2024.1376925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025] Open
Abstract
High-throughput technologies in human and animal studies have revealed novel molecular and cellular pathways involved in tissue inflammation of rheumatoid arthritis (RA). Fibroblasts have been in the forefront of research for several decades. Subpopulations with specific phenotypic and functional properties have been characterized both in mouse models and human disease. Data supporting the active involvement of fibroblasts in immune responses and tissue remodeling processes, as well as their central role in promoting clinical relapses and contributing to treatment resistance, have clearly reshaped their role in disease evolution. The lung is an important non-synovial component of RA both from a clinical and an immunopathogenic aspect. Interstitial lung disease (ILD) is a significant contributor to disease burden affecting morbidity and mortality. Although our knowledge of ILD has progressed, significant gaps in both basic and clinical science remain, posing hurdles to efficient diagnosis, prediction of disease course and its effective treatment. The specific role and contribution of fibroblasts to this process has not been clearly defined. The focus of this review is on fibroblasts and their contribution to RA and RA-ILD, presenting data on genetics and immune responses associated with RA-ILD in humans and animal models.
Collapse
Affiliation(s)
- Elpida Neofotistou-Themeli
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece
| | - Panagiota Goutakoli
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece
| | - Theodoros Chanis
- Division of Immunology and Allergy, Department of Medicine, Karolinska Institute, Solna, Sweden
- Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Maria Semitekolou
- Dendritic Cells and Adaptive Immunity Unit, Immunology Department, Pasteur Institute, Paris, France
- Developmental Biology and Stem Cells, UMR3738 – National Center for Scientific Research (CNRS), Pasteur Institute, Paris, France
| | - Eirini Sevdali
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece
| | - Prodromos Sidiropoulos
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece
| |
Collapse
|
19
|
Yang Y, Zhao D, Luo J, Lin L, Lin Y, Shan B, Chen H, Qiao L. Quantitative Site-Specific Glycoproteomics Reveals Glyco-Signatures for Breast Cancer Diagnosis. Anal Chem 2025; 97:114-121. [PMID: 39810347 DOI: 10.1021/acs.analchem.4c03069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Intact glycopeptide characterization by mass spectrometry has proven to be a versatile tool for site-specific glycoproteomics analysis and biomarker screening. Here, we present a method using a new model of a Q-TOF instrument equipped with a Zeno trap for intact glycopeptide identification and demonstrate its ability to analyze large-cohort glycoproteomes. From 124 clinical serum samples of breast cancer, noncancerous diseases, and nondisease controls, a total of 6901 unique site-specific glycans on 807 glycosites of proteins were detected. Much more differences of glycoproteome were observed in breast diseases than the proteome. By employing machine learning, 15 site-specific glycans were determined as potential glyco-signatures in detecting breast cancer. The results demonstrate that our method provides a powerful tool in glycoproteomic studies.
Collapse
Affiliation(s)
- Yi Yang
- Department of Chemistry, and Minhang Hospital, Fudan University, Shanghai 200000, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Dan Zhao
- Department of Chemistry, and Minhang Hospital, Fudan University, Shanghai 200000, China
| | - Ji Luo
- SCIEX, Beijing 100015, China
| | - Ling Lin
- Department of Chemistry, and Minhang Hospital, Fudan University, Shanghai 200000, China
| | - Yuxiang Lin
- Department of Breast Surgery, Affiliated Union Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Baozhen Shan
- Bioinformatics Solutions Inc., Waterloo, Ontario N2L3K8, Canada
| | | | - Liang Qiao
- Department of Chemistry, and Minhang Hospital, Fudan University, Shanghai 200000, China
| |
Collapse
|
20
|
Periyakoil PK, Smith MH, Kshirsagar M, Ramirez D, DiCarlo EF, Goodman SM, Rudensky AY, Donlin LT, Leslie CS. Deep topic modeling of spatial transcriptomics in the rheumatoid arthritis synovium identifies distinct classes of ectopic lymphoid structures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631928. [PMID: 39829741 PMCID: PMC11741433 DOI: 10.1101/2025.01.08.631928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Single-cell RNA sequencing studies have revealed the heterogeneity of cell states present in the rheumatoid arthritis (RA) synovium. However, it remains unclear how these cell types interact with one another in situ and how synovial microenvironments shape observed cell states. Here, we use spatial transcriptomics (ST) to define stable microenvironments across eight synovial tissue samples from six RA patients and characterize the cellular composition of ectopic lymphoid structures (ELS). To identify disease-relevant cellular communities, we developed DeepTopics, a scalable reference-free deconvolution method based on a Dirichlet variational autoencoder architecture. DeepTopics identified 22 topics across tissue samples that were defined by specific cell types, activation states, and/or biological processes. Some topics were defined by multiple colocalizing cell types, such as CD34+ fibroblasts and LYVE1+ macrophages, suggesting functional interactions. Within ELS, we discovered two divergent cellular patterns that were stable across ELS in each patient and typified by the presence or absence of a "germinal-center-like" topic. DeepTopics is a versatile and computationally efficient method for identifying disease-relevant microenvironments from ST data, and our results highlight divergent cellular architectures in histologically similar RA synovial samples that have implications for disease pathogenesis.
Collapse
Affiliation(s)
- Preethi K Periyakoil
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Weill Cornell Medical College, New York, NY 10021, USA
| | - Melanie H Smith
- Weill Cornell Medical College, New York, NY 10021, USA
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY 10021, USA
| | | | - Daniel Ramirez
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Edward F DiCarlo
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Susan M Goodman
- Weill Cornell Medical College, New York, NY 10021, USA
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY 10021, USA
| | - Alexander Y Rudensky
- Howard Hughes Medical Institute and Immunology Program at Sloan Kettering Institute, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY,10065, USA
| | - Laura T Donlin
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY 10021, USA
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Christina S Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
21
|
Verma A, Patel P, Almalki WH, Sahebkar A, Kurmi BD, Kesharwani P. Recent Advances in Drug Delivery Approaches for Rheumatoid Arthritis. Curr Med Chem 2025; 32:396-415. [PMID: 37581524 DOI: 10.2174/0929867331666230815112818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/25/2023] [Accepted: 05/23/2023] [Indexed: 08/16/2023]
Abstract
Morbidity, disability, and healthcare expenses associated with rheumatoid arthritis (RA) impose a considerable health and economical burden on both patients and healthcare systems. This review aimed to examine the pathophysiological aspects of RA that may help design different types of drugs and drug delivery systems. These include monoclonal antibodies, immunoglobulins, tiny chemicals, and transgenes for gene therapy. These novel nanocarrier-based therapies target the underlying biological processes involved in RA while minimizing the systemic adverse effects of drugs.
Collapse
Affiliation(s)
- Abhishek Verma
- Department of Quality Assurance, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 24381, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Chennai, India
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
22
|
Kartnig F, Bonelli M, Goldmann U, Mészáros N, Krall N, Aletaha D, Heinz LX, Superti-Furga G. Ex vivo imaging-based high content phenotyping of patients with rheumatoid arthritis. EBioMedicine 2025; 111:105522. [PMID: 39729885 PMCID: PMC11732195 DOI: 10.1016/j.ebiom.2024.105522] [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: 05/23/2024] [Revised: 11/18/2024] [Accepted: 12/10/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND High content imaging-based functional precision medicine approaches have been developed and successfully applied in the field of haemato-oncology. For rheumatoid arthritis (RA), treatment selection is still based on a trial-and-error principle, and biomarkers for patient stratification and drug response prediction are needed. METHODS A high content, high throughput microscopy-based phenotyping pipeline for peripheral blood mononuclear cells (PBMCs) was developed, allowing for the quantification of cell type frequencies, cell type specific morphology and intercellular interactions from patients with RA (n = 65) and healthy controls (HC, n = 33). Samples were exposed to a curated set of RA-specific small molecules, biologicals and reference stimuli for 24 h to assess ex vivo drug effects. Data on ex vivo PBMC phenotypes were integrated with information on patients' in vivo medication and disease activity. FINDINGS The unbiased data from in total 6.9e8 individual cells were collected and allowed for the identification of PBMC phenotypes specific to disease activity as well as in vivo and ex vivo treatment. The arrayed ex vivo drug perturbation enabled the systematic characterization of drug effects, clustering by mode of action and uncovered morphologic alterations associated with biologic disease-modifying anti-rheumatic drug (DMARD) treatment. Individual in vivo treatment regimens translated into altered immune cell abundances in patients with a comedication of conventional synthetic DMARDs when compared to HCs. Global integration of PBMC characteristics led to clustering of patients according to disease activity and correlation with clinical data. INTERPRETATION The application of the developed screening tool demonstrates a technical proof-of-concept for feasibility of a functional precision medicine approach to the ex vivo immunophenotypic characterisation of patients with RA. FUNDING This work was supported by the Austrian Academy of Sciences, the Medical University of Vienna and a grant (RMG2235 to L.X.H.) from the European Alliance of Associations for Rheumatology (EULAR).
Collapse
Affiliation(s)
- Felix Kartnig
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna; Vienna, Austria
| | - Michael Bonelli
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna; Vienna, Austria
| | - Ulrich Goldmann
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Noemi Mészáros
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Nikolaus Krall
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Daniel Aletaha
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna; Vienna, Austria
| | - Leonhard X Heinz
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna; Vienna, Austria.
| | - Giulio Superti-Furga
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Centre for Physiology and Pharmacology, Medical University of Vienna; Vienna, Austria.
| |
Collapse
|
23
|
Mustonen AM, Malinen M, Paakinaho V, Lehenkari P, Palosaari S, Kärjä V, Nieminen P. RNA sequencing analysis reveals distinct gene expression patterns in infrapatellar fat pads of patients with end-stage osteoarthritis or rheumatoid arthritis. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159576. [PMID: 39489461 DOI: 10.1016/j.bbalip.2024.159576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Osteoarthritis (OA) and autoimmune-driven rheumatoid arthritis (RA) are inflammatory joint diseases that share partly similar symptoms but have different, inadequately understood pathogeneses. Adipose tissues, including intra-articular infrapatellar fat pad (IFP), may contribute to their development. Analysis of differentially expressed genes (DEGs) in IFPs could improve the diagnostics of these conditions and help to develop novel treatment strategies. The aim was to identify potentially crucial genes and pathways discriminating OA and RA IFPs using RNA sequencing analysis. We aimed to distinguish genetically distinct patient groups as a starting point for further translational studies with the eventual goal of personalized medicine. Samples were collected from arthritic knees during total knee arthroplasty of sex- and age-matched OA and seropositive RA patients (n = 5-6/group). Metabolic pathways of interest were investigated by whole transcriptome sequencing, and DEGs were analyzed with univariate tests, hierarchical clustering (HC), and pathway analyses. There was significant interindividual variation in mRNA expression patterns, but distinct subgroups of OA and RA patients emerged that reacted similarly to their disease states based on HC. Compared to OA, RA samples showed 703 genes to be upregulated and 691 genes to be downregulated. Signaling pathway analyses indicated that these DEGs had common pathways in lipid metabolism, fatty acid biosynthesis and degradation, adipocytokine and insulin signaling, inflammatory response, and extracellular matrix organization. The divergent mRNA expression profiles in RA and OA suggest contribution of IFP to the regulation of synovial inflammatory processes and articular cartilage degradation and could provide novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Anne-Mari Mustonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Department of Environmental and Biological Sciences, Faculty of Science, Forestry and Technology, University of Eastern Finland, P.O. Box 111, FI-80101 Joensuu, Finland.
| | - Marjo Malinen
- Department of Forestry and Environmental Engineering, South-Eastern Finland University of Applied Sciences, Paraatikenttä 7, FI-45100 Kouvola, Finland.
| | - Ville Paakinaho
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Petri Lehenkari
- Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland; Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland; Department of Surgery, Oulu University Hospital, P.O. Box 21, FI-90029 OYS, Finland.
| | - Sanna Palosaari
- Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland; Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland.
| | - Vesa Kärjä
- Department of Clinical Pathology, Kuopio University Hospital, Puijonlaaksontie 2, FI-70210 Kuopio, Finland.
| | - Petteri Nieminen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
24
|
Ruscitti P, Nunziato M, Caso F, Scarpa R, Di Maggio F, Giacomelli R, Salvatore F. Prevention of rheumatoid arthritis using a familial predictive medicine approach. Autoimmun Rev 2024; 23:103653. [PMID: 39370029 DOI: 10.1016/j.autrev.2024.103653] [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: 05/30/2024] [Revised: 09/03/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
Most of the chronic-degenerative diseases deserve a very early recognition of symptoms and signs for the earliest secondary prevention, which could be also very useful in many cases for the most precocious clinical approach. The periodic monitoring of a subject at risk of a specific disease, because of genomic predisposition by predictive medicine approach, may help to earlier detection of onset and/or the progression of the pathology itself, through intra-individual monitoring. This is particularly the case of rheumatoid arthritis (RA) for which an early diagnosis is undoubtedly the first step to ensure the most proper therapy for the patient. Thus, the earlier identification of individuals at high risk of RA could lead to ultra-preventive strategies to start for the best lifestyle performances and/or for any other effective therapeutic interventions to contrast the onset, and/or the evolution of the putative RA. This will also optimize both costs and medical resources, according to the health care policies of many countries.
Collapse
Affiliation(s)
- Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marcella Nunziato
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Via Sergio Pansini, 5, 80131 Napoli, NA, Italy; CEINGE - Biotecnologie Avanzate - Franco Salvatore, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy
| | - Raffaele Scarpa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, via S. Pansini 5, 80131 Naples, Italy
| | - Federica Di Maggio
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Via Sergio Pansini, 5, 80131 Napoli, NA, Italy; CEINGE - Biotecnologie Avanzate - Franco Salvatore, Via Gaetano Salvatore, 486, 80145 Napoli, Italy
| | - Roberto Giacomelli
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128 Rome, Italy; Rheumatology, Immunology and Clinical Medicine Unit, Department of Medicine, University of Rome "Campus Biomedico" School of Medicine, Rome, Italy.
| | - Francesco Salvatore
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Via Sergio Pansini, 5, 80131 Napoli, NA, Italy; CEINGE - Biotecnologie Avanzate - Franco Salvatore, Via Gaetano Salvatore, 486, 80145 Napoli, Italy.
| |
Collapse
|
25
|
Vandormael P, Fadlallah S, Ruytinx P, Pues A, Sleurs E, Liesenborgs J, Joly J, Agten A, Vandenabeele F, Fraussen J, Verschueren P, Somers V. Fibroblast-like synoviocyte targeting antibodies are associated with failure to reach early and sustained remission or low disease activity after first-line therapy in rheumatoid arthritis. RMD Open 2024; 10:e004743. [PMID: 39551578 PMCID: PMC11574395 DOI: 10.1136/rmdopen-2024-004743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/01/2024] [Indexed: 11/19/2024] Open
Abstract
OBJECTIVE To discover antibody biomarkers that can predict a lack of response to first-line therapy in rheumatoid arthritis (RA) patients. METHODS Two RA cDNA phage display libraries were screened for novel antibodies in baseline RA sera from the Care in early RA (CareRA) trial, differentiating between patients who did or did not reach remission after first-line therapy (n=20 each). Antibody reactivity to identified University Hasselt (UH)-RA antigens was validated in baseline samples from 136 additional CareRA participants. The novel antibodies' potential to predict failure to reach remission or low disease activity (LDA), according to the Disease Activity Score 28-joint C-reactive protein/erythrocyte sedimentation rate (DAS28CRP/ESR) and Clinical/Simplified Disease Activity Index (CDAI/SDAI), was studied by multivariate analyses. The presence of the antibody targets in RA synovial tissue and the fibroblast-like synoviocyte (FLS) cell line SW982 was determined by immunofluorescence. RESULTS We identified antibodies to 41 novel antigens. Antibodies against any of three antigens, UH-RA.305/318/329, discriminated between RA patients not reaching week (w)8 DAS28CRP remission and those that did (36% vs 13%,p=0.0031). In all patients, anti-UH-RA.305/318/329 antibody reactivity was associated with failure to reach week 8 DAS28CRP and DAS28ESR remission (OR 3.63,p=0.0031; OR 2.92,p=0.016; respectively), SDAI/CDAI sustained remission (OR 5.59,p=0.039 for both) and DAS28CRP and DAS28ESR sustained LDA (OR 3.7,p=0.009; OR 2.76,p=0.042; respectively). In rheumatoid factor/anti-citrullinated protein antibody (RF/ACPA) seronegative patients, these antibodies were strongly associated with failure to achieve week 8 DAS28CRP remission (OR 17.3,p=0.0029). Anti-UH-RA.305/329 antibodies were shown to target FLS in RA synovial tissue and SW982 cells. CONCLUSION We identified three antibody biomarkers that are associated with failure to achieve remission/LDA after first-line RA therapy.
Collapse
Affiliation(s)
- Patrick Vandormael
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Hasselt, Belgium
| | - Sukayna Fadlallah
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Hasselt, Belgium
| | - Pieter Ruytinx
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Hasselt, Belgium
| | - Astrid Pues
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Hasselt, Belgium
| | - Ellen Sleurs
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Hasselt, Belgium
| | - Jori Liesenborgs
- Expertise Centre for Digital Media Transnational University Limburg, UHasselt, Hasselt, Belgium
| | - Johan Joly
- Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Anouk Agten
- Faculty of Rehabilitation Sciences, REVAL-Rehabilitation Research Center, UHasselt, Hasselt, Belgium
| | - Frank Vandenabeele
- Faculty of Rehabilitation Sciences, REVAL-Rehabilitation Research Center, UHasselt, Hasselt, Belgium
| | - Judith Fraussen
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Hasselt, Belgium
| | - Patrick Verschueren
- Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, KU Leuven, Leuven, Belgium
| | - Veerle Somers
- Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Hasselt, Belgium
| |
Collapse
|
26
|
Figueiredo ML. Applications of single-cell RNA sequencing in rheumatoid arthritis. Front Immunol 2024; 15:1491318. [PMID: 39600707 PMCID: PMC11588722 DOI: 10.3389/fimmu.2024.1491318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Single cell RNA sequencing (scRNA-seq) is a relatively new technology that provides an unprecedented, detailed view of cellular heterogeneity and function by delineating the transcriptomic difference among individual cells. This will allow for mapping of cell-type-specific signaling during physiological and pathological processes, to build highly specific models of cellular signaling networks between the many discrete clusters that are present. This technology therefore provides a powerful approach to dissecting the cellular and molecular mechanisms that contribute to autoimmune diseases, including rheumatoid arthritis (RA). scRNA-seq can offer valuable insights into RA unique cellular states and transitions, potentially enabling development of novel drug targets. However, some challenges that still limit its mainstream utilization and include higher costs, a lower sensitivity for low-abundance transcripts, and a relatively complex data analysis workflow relative to bulk or traditional RNA-seq. This minireview explores the emerging application of scRNA-seq in RA research, highlighting its role in producing important insights that can help pave the way for innovative and more effective therapeutic strategies.
Collapse
Affiliation(s)
- Marxa L. Figueiredo
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue
University, West Lafayette, IN, United States
| |
Collapse
|
27
|
Gaigeard N, Cardon A, Le Goff B, Guicheux J, Boutet MA. Unveiling the macrophage dynamics in osteoarthritic joints: From inflammation to therapeutic strategies. Drug Discov Today 2024; 29:104187. [PMID: 39306233 DOI: 10.1016/j.drudis.2024.104187] [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: 06/19/2024] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024]
Abstract
Osteoarthritis (OA) is an incurable, painful, and debilitating joint disease affecting over 500 million people worldwide. The OA joint tissues are infiltrated by various immune cells, particularly macrophages, which are able to induce or perpetuate inflammation. Notably, synovitis and its macrophage component represent a target of interest for developing treatments. In this review, we describe the latest advances in understanding the heterogeneity of macrophage origins, phenotypes, and functions in the OA joint and the effect of current symptomatic therapies on these cells. We then highlight the therapeutic potential of anticytokines/chemokines, nano- and microdrug delivery, and future strategies to modulate macrophage functions in OA.
Collapse
Affiliation(s)
- Nicolas Gaigeard
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Anaïs Cardon
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Benoit Le Goff
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Marie-Astrid Boutet
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France; Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, EC1M6BQ London, UK.
| |
Collapse
|
28
|
Li JB, Liu PC, Chen L, Wu R. Infiltrations of plasma cells in synovium predict inadequate response to Adalimumab in Rheumatoid Arthritis patients. Arthritis Res Ther 2024; 26:186. [PMID: 39482773 PMCID: PMC11526639 DOI: 10.1186/s13075-024-03426-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a clinically heterogeneous and complex autoimmune disease, making the prediction of therapeutic responses a significant challenge. This study aims to assess the role of clinical and synovial biomarkers in predicting poor response to adalimumab treatment in RA patients. METHODS This single-center prospective study included 56 RA patients who had an inadequate response to methotrexate (MTX). At baseline, comprehensive assessments including complete blood count, liver and kidney function tests, blood glucose levels, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), rheumatoid factor (RF), anti-citrullinated protein antibody (ACPA), as well as counts of swollen and tender joints, Health Assessment Questionnaire (HAQ) score, pain visual analogue scale (VAS) scores, and DAS28-CRP scores were conducted. Synovial biopsies were performed, followed by an efficacy evaluation at 12 weeks of adalimumab treatment. Patients not meeting the ACR20 criteria were classified into the non-responder group, with the remainder categorized as the responder group. RESULTS Out of the participants, 24 (42.9%) failed to achieve ACR20 with adalimumab treatment. Non-responders exhibited higher infiltration of plasma cells in the synovium. Multivariate logistic regression analysis identified the presence of plasma cells as an independent risk factor for inadequate response to adalimumab. CONCLUSION Inadequate responses to adalimumab in RA patients were associated with increased plasma cell infiltrations in the synovium. These findings suggest a promising target for tailored therapies in rheumatoid arthritis.
Collapse
Affiliation(s)
- Jian Bin Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, 330006, China
| | - Peng Cheng Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, 330006, China
| | - Liming Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, 330006, China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, 330006, China.
| |
Collapse
|
29
|
Xiao J, Zhao Z, Zhou F, Xiong J, Yang Z, Gong B, Xiang L, Liu M, Cao F, Xiao H, Chen H, Zhang A, Wang K. TM9SF1 expression correlates with autoimmune disease activity and regulates antibody production through mTOR-dependent autophagy. BMC Med 2024; 22:502. [PMID: 39482663 PMCID: PMC11526568 DOI: 10.1186/s12916-024-03729-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/25/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Transmembrane 9 superfamily member 1 (TM9SF1) is involved in inflammation. Since both inflammatory and autoimmune diseases are linked to immune cells regulation, this study investigated the association between TM9SF1 expression and autoimmune disease activity. As B cell differentiation and autoantibody production exacerbate autoimmune disease, the signaling pathways involved in these processes were explored. METHODS Tm9sf1-/- mouse rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) models were used to verify the relationship between gene expression and disease severity. Peripheral blood mononuclear cells (PBMCs) from 156 RA and 145 SLE patients were used to explore the relationship between TM9SF1 expression and disease activity. The effectiveness of TM9SF1 as a predictor of disease activity was assessed using multiple logistic regression and receiver operating characteristic (ROC) curves. The signaling pathways regulated by TM9SF1 in B cell maturation and antibody production were conducted by plasma cell induction experiment in vitro. RESULTS The Tm9sf1-/- RA and SLE model mice produced fewer autoantibodies and showed reduced disease severity relative to wild-type (WT) mice. TM9SF1 levels in PBMCs of patients were higher than those in healthy controls, and were reduced in patients with low disease activity relative to those with active RA and SLE. Furthermore, TM9SF1 levels were positively linked with autoantibody titers and pro-inflammatory cytokine levels in both diseases. ROC analyses indicated TM9SF1 outperformed several important clinical indicators in predicting disease activity (area under the curve (AUC) were 0.858 and 0.876 for RA and SLE, respectively). In vitro experiments demonstrated that Tm9sf1 knockout blocked differentiation of B cells into antibody-producing plasma cells by activating mTOR and inhibiting autophagy, and mTOR inhibitors such as rapamycin could reverse this effect. CONCLUSIONS The primary finding was the identification of the molecular mechanism underlying autophagy regulation in B cells, in which Tm9sf1 knockout was found to modulate mTOR-dependent autophagy to block B cell differentiation into antibody-secreting plasma cells. It was also found that TM9SF1 expression level in PBMCs was an accurate indicator of disease activity in patients with RA and SLE, suggesting its clinical potential for monitoring disease activity in these patients.
Collapse
Affiliation(s)
- Juan Xiao
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
| | - Zhenwang Zhao
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
| | - Fengqiao Zhou
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
| | - Jinsong Xiong
- Gucheng People's Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441700, China
| | - Zean Yang
- Gucheng People's Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441700, China
| | - Baoxian Gong
- Gucheng People's Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441700, China
| | - Lei Xiang
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
| | - Mingming Liu
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
| | - Fengsheng Cao
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
| | - Hong Xiao
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China
| | - Huabo Chen
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China.
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China.
| | - Anbing Zhang
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China.
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China.
| | - Ke Wang
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China.
- Department of Rheumatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441021, China.
| |
Collapse
|
30
|
Ammons DT, Chow L, Goodrich L, Bass L, Larson B, Williams ZJ, Stoneback JW, Dow S, Pezzanite LM. Characterization of the single cell landscape in normal and osteoarthritic equine joints. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:88. [PMID: 39507442 PMCID: PMC11534742 DOI: 10.21037/atm-24-40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/30/2024] [Indexed: 11/08/2024]
Abstract
Background Osteoarthritis (OA) is a major source of pain and disability worldwide. Understanding of disease progression is evolving, but OA is increasingly thought to be a multifactorial disease in which the innate immune system plays a role in regulating and perpetuating low-grade inflammation. The aim of this study was to enhance our understanding of OA immunopathogenesis through characterization of the transcriptomic responses in OA joints, with the goal to facilitate the development of targeted therapies. Methods Single-cell RNA sequencing (scRNA-seq) was completed on cells isolated from the synovial fluid of three normal and three OA equine joints. In addition to synovial fluid, scRNA-seq was also performed on synovium from one normal joint and one OA joint. Results Characterization of 28,639 cells isolated from normal and OA-affected equine synovial fluid revealed the composition to be entirely immune cells (CD45+) with 8 major populations and 26 subpopulations identified. In synovial fluid, we found myeloid cells (macrophage and dendritic cells) to be overrepresented and T cells (CD4 and CD8) to be underrepresented in OA relative to normal joints. Through subcluster and differential abundance analysis of T cells we further identified a relative overrepresentation of IL23R+ gamma-delta (γδ) T cells in OA-affected joints (a cell type we report to be enriched in gene signatures associated with T helper 17 mediated immunity). Analysis of an additional 17,690 cells (11 distinct cell type clusters) obtained from synovium of one horse led to the identification of an OA-associated reduction in the relative abundance of synovial macrophages, which contrasts with the increased relative abundance of macrophages in synovial fluid. Completion of cell-cell interaction analysis implicated myeloid cells in disease progression, suggesting that the myeloid-myeloid interactions were increased in OA-affected joints. Conclusions Overall, this work provides key insights into the composition of equine synovial fluid and synovium in health and OA. The data generated in this study provides equine-specific cell type gene signatures which can be applied to future investigations. Furthermore, our analysis highlights the potential role of macrophages and IL23R+ γδ T cells in OA immunopathogenesis.
Collapse
Affiliation(s)
- Dylan T Ammons
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Lyndah Chow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Laurie Goodrich
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Luke Bass
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Blaine Larson
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Zoë J Williams
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jason W Stoneback
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Steven Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Lynn M Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
31
|
Bergström B, Selldén T, Bollmann M, Svensson MND, Ekwall AKH. Methotrexate promotes the release of granulocyte-macrophage colony-stimulating factor from rheumatoid arthritis fibroblast-like synoviocytes via autocrine interleukin-1 signaling. Arthritis Res Ther 2024; 26:178. [PMID: 39394168 PMCID: PMC11468154 DOI: 10.1186/s13075-024-03406-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Activated fibroblast-like synoviocytes (FLS) are drivers of synovitis and structural joint damage in rheumatoid arthritis (RA). Despite the use of disease-modifying drugs, only about 50% of RA patients reach remission in real-world settings. We used an unbiased approach to investigate the effects of standard-of-care methotrexate (MTX) and a Janus kinase inhibitor, tofacitinib (TOFA), on gene expression in RA-FLS, in order to identify untargeted disease mediators. METHODS Primary RA-FLS were activated by stimulation with interleukin-1β (IL-1β) or platelet-derived growth factor + IL-1β in the presence or absence of MTX or TOFA, with or without additional inhibitors. Co-cultures of synovial cells were performed in direct and indirect systems. Cells were collected for RNA sequencing or qPCR, and supernatants were analyzed for protein concentrations. RESULTS Six thousand three hundred fifty genes were differentially expressed, the majority being upregulated, in MTX-treated activated RA-FLS and 970 genes, the majority being downregulated, in TOFA-treated samples. Pathway analysis showed that MTX had largest effects on 'Molecular mechanisms of cancer' and TOFA on 'Interferon signaling'. Targeted analysis of disease-associated genes revealed that MTX increased the expression of cell cycle-regulating genes but also of pro-inflammatory mediators like IL-1α (IL1A) and granulocyte-macrophage colony-stimulating factor, GM-CSF (CSF2). The MTX-promoted expression of CSF2 in activated RA-FLS peaked at 48 h, could be mediated via either NF-κB or AP-1 transcription factors, and was abrogated by IL-1 inhibitors (IRAK4 inhibitor and anakinra). In a co-culture setting, MTX-treatment of activated RA-FLS induced IL1B expression in macrophages. CONCLUSIONS MTX treatment induces secretion of IL-1 from activated RA-FLS which by autocrine signaling augments their release of GM-CSF. This unexpected effect of MTX might contribute to the persistence of synovitis.
Collapse
Affiliation(s)
- Beatrice Bergström
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tilia Selldén
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Miriam Bollmann
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Mattias N D Svensson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin Hultgård Ekwall
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Rheumatology, Division 3, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
32
|
Cen Y, He D, Wang P, Qin Y, Huang M. Contribution of Musculoskeletal Ultrasound in the Diagnosis of Seronegative Rheumatoid Arthritis. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:1929-1936. [PMID: 39037203 DOI: 10.1002/jum.16527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVE This is a study to investigate the value of musculoskeletal ultrasound for the early diagnosis of seronegative rheumatoid arthritis (SNRA); and to study the relationship between anti-cyclic citrullinated peptide antibody (anti-CCP) and the occurrence of bone erosion in rheumatoid arthritis (RA) detected by ultrasound. METHODS A total of 101 patients with RA or osteoarthritis (OA) admitted to the First Affiliated Hospital of Anhui Medical University from July 2022 to December 2023 were selected and divided into the SNRA group, the SPRA group, and the OA group. The patients' metacarpophalangeal joints, proximal interphalangeal joints, distal interphalangeal joints, and wrist joints of both hands were ultrasonically examined separately, and the extensor tendon, flexor tendon, synovium, joint surface, joint cavity, and bone surface were observed. RESULTS The differences in the detection of joint effusion, bone erosion, and joint space narrowing were not statistically significant between SNRA group and OA group (P > .05), the differences in the detection of synovitis and tenosynovitis were statistically significant (P < .05). The mean levels of synovial hyperplasia grade and synovial blood flow grade between SNRA group and OA group were significantly different (P < .05). The differences in synovitis, tenosynovitis, joint effusion, and joint space narrowing were not statistically significant between SNRA and SPRA groups (P > .05), and the differences in bone erosion were statistically significant (P < .05). The mean levels of synovial hyperplasia grade and synovial blood flow grade between SNRA group and SPRA group were significantly different (P < .05). Logistic regression analysis showed that anti-CCP antibody was an influential factor for bone erosion in RA patients (P < .05). The ROC curve was plotted, and the optimal cut-off value of anti-CCP antibody was 356.5 U/mL, at which time the AUC was 0.716, the sensitivity of diagnosing bone erosion was 0.714, the specificity was 0.694, and the Yoden index was 0.408. CONCLUSION In summary, ultrasound is helpful for the early diagnosis of SNRA by evaluating the condition of joints, synovium, and tendon sheath, and when anti-CCP antibodies are positive, ultrasound is more likely to detect bone erosion. Ultrasound examination combined with anti-CCP antibody can further observe the joint lesions.
Collapse
Affiliation(s)
- Yang Cen
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Danqing He
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Peiyao Wang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yangyang Qin
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Meng Huang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Ultrasound, East District of First Affiliated Hospital of Anhui Medical University/Feidong People's Hospital, Hefei, China
| |
Collapse
|
33
|
Zack SR, Alzoubi O, Satoeya N, Singh KP, Deen S, Nijim W, Lewis MJ, Pitzalis C, Sweiss N, Ivashkiv LB, Shahrara S. Another Notch in the Belt of Rheumatoid Arthritis. Arthritis Rheumatol 2024; 76:1475-1487. [PMID: 38961731 PMCID: PMC11421962 DOI: 10.1002/art.42937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/28/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Notch ligands and receptors, including JAG1/2, DLL1/4, and Notch1/3, are enriched on macrophages (MΦs), fibroblast-like synoviocytes (FLS), and/or endothelial cells in rheumatoid arthritis (RA) compared with normal synovial tissues (ST). Power Doppler ultrasound-guided ST studies reveal that the Notch family is highly involved in early active RA, especially during neovascularization. In contrast, the Notch family is not implicated during the erosive stage, evidenced by their lack of correlation with radiographic damage in RA ST. Toll-like receptors and tumor necrosis factor (TNF) are the common inducers of Notch expression in RA MΦs, FLS, and endothelial cells. Among Notch ligands, JAG1 and/or DLL4 are most inducible by inflammatory responses in RA MΦs or endothelial cells and transactivate their receptors on RA FLS. TNF plays a central role on Notch ligands, as anti-TNF good responders display JAG1/2 and DLL1/4 transcriptional downregulation in RA ST myeloid cells. In in vitro studies, TNF increases Notch3 expression in MΦs, which is further amplified by RA FLS addition. Specific disease-modifying antirheumatic drugs reduced JAG1 and Notch3 expression in MΦ and RA FLS cocultures. Organoids containing FLS and endothelial cells have increased expression of JAG1 and Notch3. Nonetheless, Methotrexate, interleukin-6 receptor (IL-6R) antibodies, and B cell blockers are mostly ineffective at decreasing Notch family expression. NF-κB, MAPK, and AKT pathways are involved in Notch signaling, whereas JAK/STATs are not. Although Notch blockade has been effective in RA preclinical studies, its small molecule inhibitors have failed in phase I and II studies, suggesting that alternative strategies may be required to intercept their function.
Collapse
Affiliation(s)
- Stephanie R. Zack
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Osama Alzoubi
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Neha Satoeya
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Kunwar P. Singh
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Sania Deen
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Wes Nijim
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Myles J. Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, London, United Kingdom
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London, United Kingdom
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, London, United Kingdom
- Department of Biomedical Sciences, Humanitas University, and Humanitas Research Hospital, Milan, Italy
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| | - Lionel B. Ivashkiv
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York 10021, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, New York 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York 10065, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago; IL, USA
| |
Collapse
|
34
|
Navratilova HF, Whetton AD, Geifman N. Artificial intelligence driven definition of food preference endotypes in UK Biobank volunteers is associated with distinctive health outcomes and blood based metabolomic and proteomic profiles. J Transl Med 2024; 22:881. [PMID: 39354608 PMCID: PMC11443809 DOI: 10.1186/s12967-024-05663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Specific food preferences can determine an individual's dietary patterns and therefore, may be associated with certain health risks and benefits. METHODS Using food preference questionnaire (FPQ) data from a subset comprising over 180,000 UK Biobank participants, we employed Latent Profile Analysis (LPA) approach to identify the main patterns or profiles among participants. blood biochemistry across groups/profiles was compared using the non-parametric Kruskal-Wallis test. We applied the Limma algorithm for differential abundance analysis on 168 metabolites and 2923 proteins, and utilized the Database for Annotation, Visualization and Integrated Discovery (DAVID) to identify enriched biological processes and pathways. Relative risks (RR) were calculated for chronic diseases and mental conditions per group, adjusting for sociodemographic factors. RESULTS Based on their food preferences, three profiles were termed: the putative Health-conscious group (low preference for animal-based or sweet foods, and high preference for vegetables and fruits), the Omnivore group (high preference for all foods), and the putative Sweet-tooth group (high preference for sweet foods and sweetened beverages). The Health-conscious group exhibited lower risk of heart failure (RR = 0.86, 95%CI 0.79-0.93) and chronic kidney disease (RR = 0.69, 95%CI 0.65-0.74) compared to the two other groups. The Sweet-tooth group had greater risk of depression (RR = 1.27, 95%CI 1.21-1.34), diabetes (RR = 1.15, 95%CI 1.01-1.31), and stroke (RR = 1.22, 95%CI 1.15-1.31) compared to the other two groups. Cancer (overall) relative risk showed little difference across the Health-conscious, Omnivore, and Sweet-tooth groups with RR of 0.98 (95%CI 0.96-1.01), 1.00 (95%CI 0.98-1.03), and 1.01 (95%CI 0.98-1.04), respectively. The Health-conscious group was associated with lower levels of inflammatory biomarkers (e.g., C-reactive Protein) which are also known to be elevated in those with common metabolic diseases (e.g., cardiovascular disease). Other markers modulated in the Health-conscious group, ketone bodies, insulin-like growth factor-binding protein (IGFBP), and Growth Hormone 1 were more abundant, while leptin was less abundant. Further, the IGFBP pathway, which influences IGF1 activity, may be significantly enhanced by dietary choices. CONCLUSIONS These observations align with previous findings from studies focusing on weight loss interventions, which include a reduction in leptin levels. Overall, the Health-conscious group, with preference to healthier food options, has better health outcomes, compared to Sweet-tooth and Omnivore groups.
Collapse
Affiliation(s)
- Hana F Navratilova
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
- Veterinary Health Innovation Engine, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, UK
- Department of Community Nutrition, Faculty of Human Ecology, IPB University, Bogor, 16680, Indonesia
| | - Anthony D Whetton
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
- Veterinary Health Innovation Engine, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, UK
| | - Nophar Geifman
- Veterinary Health Innovation Engine, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, UK
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7YH, UK
| |
Collapse
|
35
|
Ruscitti P, Currado D, Rivellese F, Vomero M, Navarini L, Cipriani P, Pitzalis C, Giacomelli R. Diminished expression of the ubiquitin-proteasome system in early treatment-naïve patients with rheumatoid arthritis and concomitant type 2 diabetes may be linked to IL-1 pathway hyper-activity; results from PEAC cohort. Arthritis Res Ther 2024; 26:171. [PMID: 39342401 PMCID: PMC11437779 DOI: 10.1186/s13075-024-03392-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
OBJECTIVE Based on the recent evidence of IL-1 inhibition in patients with rheumatoid arthritis (RA) and concomitant type 2 diabetes (T2D), we evaluated the synovial tissue expression of IL-1 related genes in relationship to the ubiquitin-proteasome system and the effects of insulin on ubiquitinated proteins in fibroblast-like synoviocytes (FLSs). METHODS The synovial expression of IL-1 pathway genes was compared in early (< 1 year) treatment-naïve RA patients with T2D (RA/T2D n = 16) and age- and sex-matched RA patients without T2D (n = 16), enrolled in the Pathobiology of Early Arthritis Cohort (PEAC). The synovial expression of ubiquitin in macrophages and synovial lining fibroblasts was also assessed by Immunohistochemistry/immunofluorescence and correlated with synovial pathotypes. Finally, FLSs from RA patients (n = 5) were isolated and treated with human insulin (200 and 500 nM) and ubiquitinated proteins were assessed by western blot. RESULTS Synovial tissues of RA/T2D patients were characterised by a consistent reduced expression of ubiquitin-proteasome genes. More specifically, ubiquitin genes (UBB, UBC, and UBA52) and genes codifying proteasome subunits (PSMA2, PSMA6, PSMA7, PSMB1, PSMB3, PSMB4, PSMB6, PSMB8, PSMB9, PSMB10, PSMC1, PSMD9, PSME1, and PSME2) were significantly lower in RA/T2D patients. On the contrary, genes regulating fibroblast functions (FGF7, FGF10, FRS2, FGFR3, and SOS1), and genes linked to IL-1 pathway hyper-activity (APP, IRAK2, and OSMR) were upregulated in RA/T2D. Immunohistochemistry showed a significant reduction of the percentage of ubiquitin-positive cells in synovial tissues of RA/T2D patients. Ubiquitin-positive cells were also increased in patients with a lympho-myeloid pathotype compared to diffuse myeloid or pauci-immune-fibroid. Finally, in vitro experiments showed a reduction of ubiquitinated proteins in RA-FLSs treated with a high concentration of insulin (500 nM). CONCLUSIONS A different IL-1 pathway gene expression was observed in the synovial tissues of early treatment-naïve RA/T2D patients, linked to decreased expression of the ubiquitin-proteasome system. These findings may provide a mechanistic explanation of the observed clinical benefits of IL-1 inhibition in patients with RA and concomitant T2D.
Collapse
Affiliation(s)
- Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy.
| | - Damiano Currado
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust & Barts Biomedical Research Centre (BRC) National Institute for Health and Care Research (NIHR), London, UK
| | - Marta Vomero
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Luca Navarini
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Paola Cipriani
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Delta 6 Building, PO box 67100, L'Aquila, Italy
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Health NHS Trust & Barts Biomedical Research Centre (BRC) National Institute for Health and Care Research (NIHR), London, UK
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Roberto Giacomelli
- Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy
- Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| |
Collapse
|
36
|
Hanlon MM, Smith CM, Canavan M, Neto NGB, Song Q, Lewis MJ, O’Rourke AM, Tynan O, Barker BE, Gallagher P, Mullan R, Hurson C, Moran B, Monaghan MG, Pitzalis C, Fletcher JM, Nagpal S, Veale DJ, Fearon U. Loss of synovial tissue macrophage homeostasis precedes rheumatoid arthritis clinical onset. SCIENCE ADVANCES 2024; 10:eadj1252. [PMID: 39321281 PMCID: PMC11423874 DOI: 10.1126/sciadv.adj1252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/20/2024] [Indexed: 09/27/2024]
Abstract
This study performed an in-depth investigation into the myeloid cellular landscape in the synovium of patients with rheumatoid arthritis (RA), "individuals at risk" of RA, and healthy controls (HC). Flow cytometric analysis demonstrated the presence of a CD40-expressing CD206+CD163+ macrophage population dominating the inflamed RA synovium, associated with disease activity and treatment response. In-depth RNA sequencing and metabolic analysis demonstrated that this macrophage population is transcriptionally distinct, displaying unique inflammatory and tissue-resident gene signatures, has a stable bioenergetic profile, and regulates stromal cell responses. Single-cell RNA sequencing profiling of 67,908 RA and HC synovial tissue cells identified nine transcriptionally distinct macrophage clusters. IL-1B+CCL20+ and SPP1+MT2A+ are the principal macrophage clusters in RA synovium, displaying heightened CD40 gene expression, capable of shaping stromal cell responses, and are importantly enriched before disease onset. Combined, these findings identify the presence of an early pathogenic myeloid signature that shapes the RA joint microenvironment and represents a unique opportunity for early diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
- Megan M. Hanlon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Conor M. Smith
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Mary Canavan
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
- Translational Immunopathology, School of Biochemistry and Immunology and School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Nuno G. B. Neto
- Department of Mechanical and Manufacturing Engineering, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Qingxuan Song
- Immunology and Discovery Sciences, Janssen Research and Development, Spring House, PA, USA
| | - Myles J. Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC and Barts Health NHS Trust, London, UK
| | - Aoife M. O’Rourke
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
- Translational Immunopathology, School of Biochemistry and Immunology and School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Orla Tynan
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Brianne E. Barker
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Phil Gallagher
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Ronan Mullan
- Department of Rheumatology, Adelaide and Meath Hospital, Dublin, Ireland
| | - Conor Hurson
- Department of Orthopaedics, St. Vincent’s University Hospital, Dublin, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Michael G. Monaghan
- Department of Mechanical and Manufacturing Engineering, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC and Barts Health NHS Trust, London, UK
- Department of Biomedical Sciences, Humanitas University and Humanitas Research Hospital, Milan, Italy
| | - Jean M. Fletcher
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Sunil Nagpal
- Immunology and Discovery Sciences, Janssen Research and Development, Spring House, PA, USA
| | - Douglas J. Veale
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Chmiel J, Stasiak M, Skrzypkowska M, Samson L, Łuczkiewicz P, Trzonkowski P. Regulatory T lymphocytes as a treatment method for rheumatoid arthritis - Superiority of allogeneic to autologous cells. Heliyon 2024; 10:e36512. [PMID: 39319132 PMCID: PMC11419861 DOI: 10.1016/j.heliyon.2024.e36512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
Cellular therapies utilizing regulatory T cells (Tregs) have flourished in the autoimmunity space as a new pillar of medicine. These cells have shown a great promise in the treatment of such devastating conditions as type 1 diabetes mellitus (T1DM), systemic lupus erythematosus (SLE) and graft versus host disease (GVHD). Novel treatment protocols, which utilize Tregs-mediated suppressive mechanisms, are based on the two main strategies: administration of immunomodulatory factors affecting Tregs or adoptive cell transfer (ACT). ACT involves extraction, in vitro expansion and subsequent administration of Tregs that could be either of autologous or allogeneic origin. Rheumatoid arthritis (RA) is another autoimmune candidate where this treatment approach is being considered. RA remains an especially challenging adversary since it is one of the most frequent and debilitating conditions among all autoaggressive disorders. Noteworthy, Tregs circulating in RA patients' blood have been proven defective and unable to suppress inflammation and joint destruction. With this knowledge, adoptive transfer of compromised autologous Tregs in the fledgling clinical trials involving RA patients should be reconsidered. In this article we hypothesize that incorporation of healthy donor allogeneic Tregs may provide more lucid and beneficial results.
Collapse
Affiliation(s)
- Joanna Chmiel
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Mariusz Stasiak
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Maria Skrzypkowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Lucjan Samson
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | | | - Piotr Trzonkowski
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Poland
| |
Collapse
|
38
|
Bell RD, Brendel M, Konnaris MA, Xiang J, Otero M, Fontana MA, Bai Z, Krenitsky DM, Meednu N, Rangel-Moreno J, Scheel-Toellner D, Carr H, Nayar S, McMurray J, DiCarlo E, Anolik JH, Donlin LT, Orange DE, Kenney HM, Schwarz EM, Filer A, Ivashkiv LB, Wang F. Automated multi-scale computational pathotyping (AMSCP) of inflamed synovial tissue. Nat Commun 2024; 15:7503. [PMID: 39209814 PMCID: PMC11362542 DOI: 10.1038/s41467-024-51012-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Rheumatoid arthritis (RA) is a complex immune-mediated inflammatory disorder in which patients suffer from inflammatory-erosive arthritis. Recent advances on histopathology heterogeneity of RA synovial tissue revealed three distinct phenotypes based on cellular composition (pauci-immune, diffuse and lymphoid), suggesting that distinct etiologies warrant specific targeted therapy which motivates a need for cost effective phenotyping tools in preclinical and clinical settings. To this end, we developed an automated multi-scale computational pathotyping (AMSCP) pipeline for both human and mouse synovial tissue with two distinct components that can be leveraged together or independently: (1) segmentation of different tissue types to characterize tissue-level changes, and (2) cell type classification within each tissue compartment that assesses change across disease states. Here, we demonstrate the efficacy, efficiency, and robustness of the AMSCP pipeline as well as the ability to discover novel phenotypes. Taken together, we find AMSCP to be a valuable cost-effective method for both pre-clinical and clinical research.
Collapse
Affiliation(s)
- Richard D Bell
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Matthew Brendel
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Maxwell A Konnaris
- Huck Institute of the Life Sciences, Pennsylvania State University, State College, University Park, PA, USA
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY, USA
| | | | - Miguel Otero
- Weill Cornell Medical College, New York, NY, USA
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY, USA
| | - Mark A Fontana
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Zilong Bai
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Daria M Krenitsky
- Allergy, Immunology and Rheumatology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Nida Meednu
- Allergy, Immunology and Rheumatology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Javier Rangel-Moreno
- Allergy, Immunology and Rheumatology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Dagmar Scheel-Toellner
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Hayley Carr
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Saba Nayar
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Jack McMurray
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Edward DiCarlo
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Jennifer H Anolik
- Allergy, Immunology and Rheumatology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura T Donlin
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Dana E Orange
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA
- The Rockefeller University, New York, NY, USA
| | - H Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Andrew Filer
- Rheumatology Research Group, Institute for Inflammation and Ageing, University of Birmingham, NIHR Birmingham Biomedical Research Center and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Lionel B Ivashkiv
- Arthritis and Tissue Degeneration Program and Research Institute, Hospital for Special Surgery, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
39
|
Natalucci F, Triaille C, Van Mullem C, Sokolova T, Sapart E, de Bellefon LM, Nzeusseu A, Galant C, Lauwerys B, Durez P. Inclusion of fibrinoid necrosis increases the accuracy of synovial tissue assessment in predicting response to methotrexate: analysis of the UCLouvain Brussels ERA Cohort. Arthritis Res Ther 2024; 26:150. [PMID: 39160592 PMCID: PMC11331690 DOI: 10.1186/s13075-024-03384-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024] Open
Abstract
OBJECTIVE Rheumatoid Arthritis (RA) often exhibits suboptimal treatment response despite early diagnosis and treatment. This study aimed to analyze Early Rheumatoid Arthritis (ERA) synovial biopsies through histology and immunohistochemistry (IHC) to identify predictive factors for treatment response to Methotrexate (MTX). METHODS 140 ERA patients from the UCLouvain Arthritis Cohort underwent synovial biopsy and were monitored after initiating Disease-Modifying Antirheumatic Drug (DMARD) therapy. Histological features [Synovial Hyperplasia, Fibrinoid Necrosis (FN), Hypervascularization and Inflammatory Infiltrate] and IHC (CD3, CD20, CD138, CD68) were each semi-quantitatively assessed on a 0-3 scale with 7 levels. RESULTS A strong association was observed between synovial CD68 and Fibrinoid Necrosis scores [r = 0.44 (0.27 - 0.56); p < 0.0001]. CD68 correlated with C-Reactive Protein (CRP), DAS28, SDAI and CDAI. Fibrinoid Necrosis score correlated with CRP and DAS28. Patients were then categorized as CD68NecrosisHIGH (CD68 + Necrosis ≥ 3) and CD68NecrosisLOW (CD68 + Necrosis < 3). CD68NecrosisHIGH exhibited higher pre-treatment disease activity [5.48 (1.6) versus 4.8 (1.7); p = 0.03] and a greater fall in DAS28 [1.99 (2.06) versus 1.1 (2.27), p = 0.03], SDAI [21.45 (IQR 23.3) versus 11.65 (IQR 17.5); p = 0.003] and CDAI [16 [14.9] versus 10.5 (20.1), p = 0.04]. CD68NecrosisHIGH patients had a higher EULAR Moderate/Good Response rate. CD68Necrosis score was incorporated into a probability matrix model together with clinical features (SJC44 and DAS28) to predict achieving a Moderate/Good EULAR Response Criteria at 3 months with a good performance (AUC 0.724). CONCLUSION FN and CD68 + in ERA synovial biopsies identify patients with higher disease activity and predict a better treatment response at three months. A model including synovial CD68 and fibrinoid necrosis with baseline clinical features predicts EULAR response at 3 months.
Collapse
Affiliation(s)
- Francesco Natalucci
- Cliniques Universitaires Saint-Luc - Université catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique (IREC), Rheumatology, Brussels, Belgium
- Sapienza University of Roma, Rheumatology, Department of Clinical, Internistic, Anesthesiological and Cardiovascular Sciences, Roma, Italy
| | - Clément Triaille
- Pôle de pathologies rhumatismales systémiques et inflammatoires, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Cécile Van Mullem
- Cliniques Universitaires Saint-Luc - Université catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique (IREC), Rheumatology, Brussels, Belgium
| | - Tatiana Sokolova
- Pôle de pathologies rhumatismales systémiques et inflammatoires, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Emilie Sapart
- Cliniques Universitaires Saint-Luc - Université catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique (IREC), Rheumatology, Brussels, Belgium
| | - Laurent Meric de Bellefon
- Cliniques Universitaires Saint-Luc - Université catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique (IREC), Rheumatology, Brussels, Belgium
| | - Adrien Nzeusseu
- Cliniques Universitaires Saint-Luc - Université catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique (IREC), Rheumatology, Brussels, Belgium
| | - Christine Galant
- Cliniques Universitaires Saint-Luc - Université catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique (IREC), Rheumatology, Brussels, Belgium
| | - Bernard Lauwerys
- Pôle de pathologies rhumatismales systémiques et inflammatoires, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Patrick Durez
- Cliniques Universitaires Saint-Luc - Université catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique (IREC), Rheumatology, Brussels, Belgium.
- Pôle de pathologies rhumatismales systémiques et inflammatoires, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
40
|
Li MM, Huang Y, Sumathipala M, Liang MQ, Valdeolivas A, Ananthakrishnan AN, Liao K, Marbach D, Zitnik M. Contextual AI models for single-cell protein biology. Nat Methods 2024; 21:1546-1557. [PMID: 39039335 PMCID: PMC11310085 DOI: 10.1038/s41592-024-02341-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
Understanding protein function and developing molecular therapies require deciphering the cell types in which proteins act as well as the interactions between proteins. However, modeling protein interactions across biological contexts remains challenging for existing algorithms. Here we introduce PINNACLE, a geometric deep learning approach that generates context-aware protein representations. Leveraging a multiorgan single-cell atlas, PINNACLE learns on contextualized protein interaction networks to produce 394,760 protein representations from 156 cell type contexts across 24 tissues. PINNACLE's embedding space reflects cellular and tissue organization, enabling zero-shot retrieval of the tissue hierarchy. Pretrained protein representations can be adapted for downstream tasks: enhancing 3D structure-based representations for resolving immuno-oncological protein interactions, and investigating drugs' effects across cell types. PINNACLE outperforms state-of-the-art models in nominating therapeutic targets for rheumatoid arthritis and inflammatory bowel diseases and pinpoints cell type contexts with higher predictive capability than context-free models. PINNACLE's ability to adjust its outputs on the basis of the context in which it operates paves the way for large-scale context-specific predictions in biology.
Collapse
Affiliation(s)
- Michelle M Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yepeng Huang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Marissa Sumathipala
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Man Qing Liang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Alberto Valdeolivas
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Ashwin N Ananthakrishnan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Katherine Liao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Marbach
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Allston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Data Science Initiative, Cambridge, MA, USA.
| |
Collapse
|
41
|
Murillo-Saich JD, Coras R, Ramirez J, Quesada-Masachs E, Sala-Climent M, Eschelbach K, Mahony CB, Celis R, Armando A, Quehenberger O, Croft AP, Kavanaugh A, Chang E, Cañete JD, Singh A, Guma M. Synovial 5-Lipoxygenase-Derived Oxylipins Define a Lympho-Myeloid-Enriched Synovium. Arthritis Rheumatol 2024; 76:1230-1242. [PMID: 38508862 PMCID: PMC11288786 DOI: 10.1002/art.42848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 02/11/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
OBJECTIVE Oxylipins are bioactive lipids derived from polyunsaturated fatty acids (PUFAs) that modulate inflammation and may remain overexpressed in refractory synovitis. In plasma, they could also be biomarkers of synovial pathology. The aim of this study is to determine if synovial oxylipins in inflamed joints correlate with plasma oxylipins and with synovial histologic patterns. METHODS Patients with established rheumatoid or psoriatic arthritis with active disease despite treatment were recruited, and paired synovial tissue (ST) and plasma were collected. Oxylipins were determined by liquid chromatography with tandem mass spectrometry and were classified into groups according to their PUFA precursor and enzyme. The expression of CD20, CD68, CD3, and CD138 was obtained to describe synovial histology. Cell-specific expression of oxylipin-related genes was identified by examining available synovial single-cell RNA sequencing data. RESULTS We included a total of 32 ST and 26 paired-plasma samples. A total of 71 oxylipins were identified in ST, but only 24 were identified in plasma. Only levels of 9,10-dihydroxyoctadecenoic acid and tetranor-Prostaglandin FM had a significant positive correlation between plasma and ST. Several oxylipins and oxylipin-related genes were differentially expressed among synovial phenotypes. Specifically, several 5-lipoxygenase (LOX)-derived oxylipins were statistically elevated in the lympho-myeloid phenotype and associated with B cell expression in rheumatoid arthritis samples. CONCLUSION The lack of correlation between ST and plasma oxylipins suggests that ST lipid profiling better characterizes active pathways in treated joints. Synovial 5-LOX-derived oxylipins were highly expressed in lympho-myeloid-enriched synovium. Combination therapy with 5-LOX inhibitors to improve refractory inflammation may be needed in patients with this histologic group.
Collapse
Affiliation(s)
- Jessica D. Murillo-Saich
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Roxana Coras
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Julio Ramirez
- Arthritis Unit, Rheumatology Department, Hospital Clinic and IDIBAPS, Barcelona, Spain
| | | | - Marta Sala-Climent
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | | | - Christopher B Mahony
- Rheumatology Research Group, Institute of Inflammation and Ageing, Queen Elizabeth Hospital, University of Birmingham, Birmingham, UK
| | - Raquel Celis
- Arthritis Unit, Rheumatology Department, Hospital Clinic and IDIBAPS, Barcelona, Spain
| | - Aaron Armando
- VA San Diego Healthcare System, 3350 La Jolla Village Dr. San Diego, CA 92161, USA
| | - Oswald Quehenberger
- VA San Diego Healthcare System, 3350 La Jolla Village Dr. San Diego, CA 92161, USA
| | - Adam P Croft
- Rheumatology Research Group, Institute of Inflammation and Ageing, Queen Elizabeth Hospital, University of Birmingham, Birmingham, UK
| | - Arthur Kavanaugh
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Eric Chang
- Department of Radiology School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Juan D. Cañete
- Arthritis Unit, Rheumatology Department, Hospital Clinic and IDIBAPS, Barcelona, Spain
| | - Abha Singh
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Monica Guma
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
- VA San Diego Healthcare System, 3350 La Jolla Village Dr. San Diego, CA 92161, USA
| |
Collapse
|
42
|
Jeong S, Chokkalla AK, Davis CK, Jeong H, Chelluboina B, Arruri V, Kim B, Narman A, Bathula S, Arumugam TV, Bendlin BB, Vemuganti R. Circadian-Dependent Intermittent Fasting Influences Ischemic Tolerance and Dendritic Spine Remodeling. Stroke 2024; 55:2139-2150. [PMID: 38920050 PMCID: PMC11262964 DOI: 10.1161/strokeaha.124.046400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Preconditioning by intermittent fasting is linked to improved cognition and motor function, and enhanced recovery after stroke. Although the duration of fasting was shown to elicit different levels of neuroprotection after ischemic stroke, the impact of time of fasting with respect to the circadian cycles remains unexplored. METHODS Cohorts of mice were subjected to a daily 16-hour fast, either during the dark phase (active-phase intermittent fasting) or the light phase (inactive-phase intermittent fasting) or were fed ad libitum. Following a 6-week dietary regimen, mice were subjected to transient focal cerebral ischemia and underwent behavioral functional assessment. Brain samples were collected for RNA sequencing and histopathologic analyses. RESULTS Active-phase intermittent fasting cohort exhibited better poststroke motor and cognitive recovery as well as reduced infarction, in contrast to inactive-phase intermittent fasting cohort, when compared with ad libitum cohort. In addition, protection of dendritic spine density/morphology and increased expression of postsynaptic density protein-95 were observed in the active-phase intermittent fasting. CONCLUSIONS These findings indicate that the time of daily fasting is an important factor in inducing ischemic tolerance by intermittent fasting.
Collapse
Affiliation(s)
- Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Hyunmook Jeong
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University Hospital, Seoul, South Korea
| | - Bharath Chelluboina
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Bori Kim
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Ashlyn Narman
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | | | - Thiruma V Arumugam
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Barbara B Bendlin
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
- William S. Middleton Veterans Hospital, Madison, WI, USA
| |
Collapse
|
43
|
Favalli EG, Maioli G, Caporali R. Biologics or Janus Kinase Inhibitors in Rheumatoid Arthritis Patients Who are Insufficient Responders to Conventional Anti-Rheumatic Drugs. Drugs 2024; 84:877-894. [PMID: 38949688 PMCID: PMC11343917 DOI: 10.1007/s40265-024-02059-8] [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] [Accepted: 06/03/2024] [Indexed: 07/02/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic immune-mediated inflammatory disease which can induce progressive disability if not properly treated early. Over the last 20 years, the improvement of knowledge on the pathogenesis of the disease has made available several drugs targeting key elements of the pathogenetic process, which now represent the preferred treatment option after the failure of first-line therapy with conventional drugs such as methotrexate (MTX). To this category of targeted drugs belong anti-cytokine or cell-targeted biological agents and more recently also Janus kinase inhibitors (JAKis). In the absence to date of specific biomarkers to guide the therapeutic choice in the context of true precision medicine, the choice of the first targeted drug after MTX failure is guided by treatment cost (especially after the marketing of biosimilar products) and by the clinical characteristics of the patient (age, sex, comorbidities and compliance) and the disease (presence or absence of autoantibodies and systemic or extra-articular manifestations), which may influence the efficacy and safety profile of the available products. This viewpoint focuses on the decision-making process underlying the personalized approach to RA therapy and will analyse the evidence in the literature supporting the choice of individual products and in particular the differential choice between biological drugs and JAKis.
Collapse
Affiliation(s)
- Ennio Giulio Favalli
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122, Milan, Italy
- Department of Rheumatology and Medical Sciences, Gaetano Pini-CTO Hospital, P.zza Cardinal Ferrari 1, 20122, Milan, Italy
| | - Gabriella Maioli
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122, Milan, Italy.
- Department of Rheumatology and Medical Sciences, Gaetano Pini-CTO Hospital, P.zza Cardinal Ferrari 1, 20122, Milan, Italy.
| | - Roberto Caporali
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Via Festa del Perdono, 7, 20122, Milan, Italy
- Department of Rheumatology and Medical Sciences, Gaetano Pini-CTO Hospital, P.zza Cardinal Ferrari 1, 20122, Milan, Italy
| |
Collapse
|
44
|
Li K, Ling H, Huang W, Luo W, Gu C, Tao B, Xie Q, Qiu P. Single-cell RNA-sequencing analysis reveals α-syn induced astrocyte-neuron crosstalk-mediated neurotoxicity. Int Immunopharmacol 2024; 139:112676. [PMID: 39053230 DOI: 10.1016/j.intimp.2024.112676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Accumulation of alpha-synuclein (α-syn) is a key pathological hallmark of synucleinopathies and has been shown to negatively impact neuronal function and activity. α-syn is an important factor contributing to astrocyte overactivation, though the effect of astrocyte overactivation on neurons remains unclear. Single-cell RNA sequencing data of mouse brain frontal cortex and midbrain from Hua-Syn (A53T) and wild type mice were utilized from the GEO database. Enrichment analysis, protein-protein interaction networks, and cell-cell interaction networks all indicated enhanced communication between astrocytes and neurons, along with the involvement of TNF and inflammation-related signaling pathways. In vitro experiments were performed to further explore the mechanism of neurotoxicity in astrocyte-neuron crosstalk. Astrocytes were treated by α-syn, neuronal TNFR1 receptors were antagonized by R-7050, and the cells were co-cultured after 24 h treatment. ELISA results revealed that cytokines such as TNF-α and IL-6 were significantly upregulated in astrocytes following the endocytosis of α-syn. Immunofluorescence (IF) showed neuronal dendritic reduction, axon elongation and increased co-localisation of TNFR1 receptor expression. Western blot showed up-regulation of PKR, P-eIF2α and ATF4 protein expression. Conversely, after antagonizing neuronal TNFR1 receptors with the R-7050 chemical inhibitor, neuronal synaptic structure was significantly restored and the expression of PKR, P-eIF2α and ATF4 was down-regulated. In summary, TNF-α acts as a signaling molecule mediating the up-regulated astrocyte-neuron crosstalk, providing new insights into the pathogenesis of α-syn-related neurological disorders.
Collapse
Affiliation(s)
- Kuan Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Haosen Ling
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wei Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wenyu Luo
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Cihang Gu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Bowen Tao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Qiqian Xie
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Pingming Qiu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China; Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
45
|
Binvignat M, Miao BY, Wibrand C, Yang MM, Rychkov D, Flynn E, Nititham J, Tamaki W, Khan U, Carvidi A, Krueger M, Niemi E, Sun Y, Fragiadakis GK, Sellam J, Mariotti-Ferrandiz E, Klatzmann D, Gross AJ, Ye CJ, Butte AJ, Criswell LA, Nakamura MC, Sirota M. Single-cell RNA-Seq analysis reveals cell subsets and gene signatures associated with rheumatoid arthritis disease activity. JCI Insight 2024; 9:e178499. [PMID: 38954480 PMCID: PMC11343607 DOI: 10.1172/jci.insight.178499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024] Open
Abstract
Rheumatoid arthritis (RA) management leans toward achieving remission or low disease activity. In this study, we conducted single-cell RNA sequencing (scRNA-Seq) of peripheral blood mononuclear cells (PBMCs) from 36 individuals (18 patients with RA and 18 matched controls, accounting for age, sex, race, and ethnicity), to identify disease-relevant cell subsets and cell type-specific signatures associated with disease activity. Our analysis revealed 18 distinct PBMC subsets, including an IFN-induced transmembrane 3-overexpressing (IFITM3-overexpressing) IFN-activated monocyte subset. We observed an increase in CD4+ T effector memory cells in patients with moderate-high disease activity (DAS28-CRP ≥ 3.2) and a decrease in nonclassical monocytes in patients with low disease activity or remission (DAS28-CRP < 3.2). Pseudobulk analysis by cell type identified 168 differentially expressed genes between RA and matched controls, with a downregulation of proinflammatory genes in the γδ T cell subset, alteration of genes associated with RA predisposition in the IFN-activated subset, and nonclassical monocytes. Additionally, we identified a gene signature associated with moderate-high disease activity, characterized by upregulation of proinflammatory genes such as TNF, JUN, EGR1, IFIT2, MAFB, and G0S2 and downregulation of genes including HLA-DQB1, HLA-DRB5, and TNFSF13B. Notably, cell-cell communication analysis revealed an upregulation of signaling pathways, including VISTA, in both moderate-high and remission-low disease activity contexts. Our findings provide valuable insights into the systemic cellular and molecular mechanisms underlying RA disease activity.
Collapse
Affiliation(s)
- Marie Binvignat
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
- Immunology Immunopathology Immunotherapy, Pitie Salpetriere Hospital UMRS 959, Sorbonne University, Paris, France
- Department of Rheumatology, Research Center Saint Antoine, UMRS 938, Sorbonne University, AP-HP, Saint-Antoine Hospital, Inserm UMRS 938, Paris, France
| | - Brenda Y. Miao
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
| | - Camilla Wibrand
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
- Aarhus University, Aarhus, Denmark
| | - Monica M. Yang
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
| | - Dmitry Rychkov
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
| | - Emily Flynn
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
- CoLabs, UCSF, San Francisco, California, USA
| | - Joanne Nititham
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
| | - Whitney Tamaki
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
| | - Umair Khan
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
| | - Alexander Carvidi
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
| | - Melissa Krueger
- Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Erene Niemi
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
| | - Yang Sun
- Department of Human Genetics and
| | - Gabriela K. Fragiadakis
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
- CoLabs, UCSF, San Francisco, California, USA
| | - Jérémie Sellam
- Department of Rheumatology, Research Center Saint Antoine, UMRS 938, Sorbonne University, AP-HP, Saint-Antoine Hospital, Inserm UMRS 938, Paris, France
| | - Encarnita Mariotti-Ferrandiz
- Immunology Immunopathology Immunotherapy, Pitie Salpetriere Hospital UMRS 959, Sorbonne University, Paris, France
| | - David Klatzmann
- Immunology Immunopathology Immunotherapy, Pitie Salpetriere Hospital UMRS 959, Sorbonne University, Paris, France
| | - Andrew J. Gross
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
| | | | - Atul J. Butte
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Lindsey A. Criswell
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Mary C. Nakamura
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, and
- San Francisco VA Health Care System, San Francisco, California, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, California, USA
| |
Collapse
|
46
|
Dreo B, Muralikrishnan AS, Husic R, Lackner A, Brügmann T, Haudum P, Bosch P, Thiel J, Fessler J, Stradner M. JAK/STAT signaling in rheumatoid arthritis leukocytes is uncoupled from serum cytokines in a subset of patients. Clin Immunol 2024; 264:110238. [PMID: 38729230 DOI: 10.1016/j.clim.2024.110238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
OBJECTIVE Rheumatoid Arthritis (RA) is a systemic autoimmune disease involving pro-inflammatory cytokines that can be therapeutically targeted by antibodies or kinase inhibitors. Nevertheless, these drugs fail in a subset of patients independent of the abundance of the targeted cytokines. We aim to explore the cellular basis of this phenomenon by analyzing the relation of cytokine abundance and activation of downstream signaling pathways in RA. METHODS The study included 62 RA patients and 9 healthy controls (HC). Phosphorylation of STAT 1-6 in various immune cell subsets was determined ex vivo using a novel robust flow cytometry-based protocol. Serum concentrations of IL-6, IL-10, IL-12p70, IL-17 A, interferon gamma, and TNFα in the same samples were measured using highly sensitive single molecule array (SIMOA). RESULTS We found an increase in circulating cytokines in RA patients, while STAT activity was lower in RA patients compared to HC. Based on STAT activity we determined three endotypes in active RA patients (cDAI>10, n = 28): 1) those with active STAT5a/b signaling in T cells (n = 7/28), 2) those with a low STAT activity in all assessed cell types (n = 14/28), and 3) those with active STAT1 and STAT3 signaling mainly in myeloid cells (n = 7/28). Integrating intracellular STAT activation and cytokine analysis revealed diminished JAK/STAT signaling in a subset of patients (n = 8/20) despite elevated serum cytokine concentrations. CONCLUSION Diminished JAK/STAT signaling in active RA may partly explain unresponsiveness to therapy targeting cytokine signaling. Analysis of JAK/STAT phosphorylation may identify patients at risk for non-response to these therapies.
Collapse
Affiliation(s)
- Barbara Dreo
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | | | - Rusmir Husic
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Angelika Lackner
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Theresa Brügmann
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Patrizia Haudum
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Philipp Bosch
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Jens Thiel
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Johannes Fessler
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Austria.
| | - Martin Stradner
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| |
Collapse
|
47
|
Chang MJ, Feng QF, Hao JW, Zhang YJ, Zhao R, Li N, Zhao YH, Han ZY, He PF, Wang CH. Deciphering the molecular landscape of rheumatoid arthritis offers new insights into the stratified treatment for the condition. Front Immunol 2024; 15:1391848. [PMID: 38983856 PMCID: PMC11232074 DOI: 10.3389/fimmu.2024.1391848] [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: 02/26/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
Background For Rheumatoid Arthritis (RA), a long-term chronic illness, it is essential to identify and describe patient subtypes with comparable goal status and molecular biomarkers. This study aims to develop and validate a new subtyping scheme that integrates genome-scale transcriptomic profiles of RA peripheral blood genes, providing a fresh perspective for stratified treatments. Methods We utilized independent microarray datasets of RA peripheral blood mononuclear cells (PBMCs). Up-regulated differentially expressed genes (DEGs) were subjected to functional enrichment analysis. Unsupervised cluster analysis was then employed to identify RA peripheral blood gene expression-driven subtypes. We defined three distinct clustering subtypes based on the identified 404 up-regulated DEGs. Results Subtype A, named NE-driving, was enriched in pathways related to neutrophil activation and responses to bacteria. Subtype B, termed interferon-driving (IFN-driving), exhibited abundant B cells and showed increased expression of transcripts involved in IFN signaling and defense responses to viruses. In Subtype C, an enrichment of CD8+ T-cells was found, ultimately defining it as CD8+ T-cells-driving. The RA subtyping scheme was validated using the XGBoost machine learning algorithm. We also evaluated the therapeutic outcomes of biological disease-modifying anti-rheumatic drugs. Conclusions The findings provide valuable insights for deep stratification, enabling the design of molecular diagnosis and serving as a reference for stratified therapy in RA patients in the future.
Collapse
Affiliation(s)
- Min-Jing Chang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Qi-Fan Feng
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, China
| | - Jia-Wei Hao
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Ya-Jing Zhang
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Rong Zhao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, China
| | - Nan Li
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Yu-Hui Zhao
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Zi-Yi Han
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Pei-Feng He
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Cai-Hong Wang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, China
| |
Collapse
|
48
|
Erensoy G, Råberg L, von Mentzer U, Menges LD, Bardhi E, Hultgård Ekwall AK, Stubelius A. Dynamic Release from Acetalated Dextran Nanoparticles for Precision Therapy of Inflammation. ACS APPLIED BIO MATERIALS 2024; 7:3810-3820. [PMID: 38795048 PMCID: PMC11191005 DOI: 10.1021/acsabm.4c00182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 05/27/2024]
Abstract
Polymer-based nanoparticles (NPs) that react to altered physiological characteristics have the potential to enhance the delivery of therapeutics to a specific area. These materials can utilize biochemical triggers, such as low pH, which is prone to happen locally in an inflammatory microenvironment due to increased cellular activity. This reduced pH is neutralized when inflammation subsides. For precise delivery of therapeutics to match this dynamic reaction, drug delivery systems (DDS) need to not only release the drug (ON) but also stop the release (OFF) autonomously. In this study, we use a systematic approach to optimize the composition of acetalated dextran (AcDex) NPs to start (ON) and stop (OFF) releasing model cargo, depending on local pH changes. By mixing ratios of AcDex polymers (mixed NPs), we achieved a highly sensitive material that was able to rapidly release cargo when going from pH 7.4 to pH 6.0. At the same time, the mix also offered a stable composition that enabled a rapid ON/OFF/ON/OFF switching within this narrow pH range in only 90 min. These mixed NPs were also sensitive to biological pH changes, with increased release in the presence of inflammatory cells compared to healthy cells. Such precise and controllable characteristics of a DDS position mixed NPs as a potential treatment platform to inhibit disease flare-ups, reducing both systemic and local side effects to offer a superior treatment option for inflammation compared to conventional systems.
Collapse
Affiliation(s)
- Gizem Erensoy
- Division
of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Loise Råberg
- Division
of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Ula von Mentzer
- Division
of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Luca Dirk Menges
- Division
of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Endri Bardhi
- Division
of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Anna-Karin Hultgård Ekwall
- The
Rheumatology Clinic, Sahlgrenska University
Hospital, Gothenburg 413 45, Sweden
- Department
of Rheumatology and Inflammation Research, Institute of Medicine,
Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 46, Sweden
| | - Alexandra Stubelius
- Division
of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
49
|
Barbuti PA, Guardia-Laguarta C, Yun T, Chatila ZK, Flowers X, Santos BFR, Larsen SB, Hattori N, Bradshaw E, Dettmer U, Fanning S, Vilas M, Reddy H, Teich AF, Krüger R, Area-Gomez E, Przedborski S. The Role of Alpha-Synuclein in Synucleinopathy: Impact on Lipid Regulation at Mitochondria-ER Membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599406. [PMID: 38948777 PMCID: PMC11212931 DOI: 10.1101/2024.06.17.599406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The protein alpha-synuclein (αSyn) plays a critical role in the pathogenesis of synucleinopathy, which includes Parkinson's disease and multiple system atrophy, and mounting evidence suggests that lipid dyshomeostasis is a critical phenotype in these neurodegenerative conditions. Previously, we identified that αSyn localizes to mitochondria-associated endoplasmic reticulum membranes (MAMs), temporary functional domains containing proteins that regulate lipid metabolism, including the de novo synthesis of phosphatidylserine. In the present study, we have analyzed the lipid composition of postmortem human samples, focusing on the substantia nigra pars compacta of Parkinson's disease and controls, as well as three less affected brain regions of Parkinson's donors. To further assess synucleinopathy-related lipidome alterations, similar analyses were performed on the striatum of multiple system atrophy cases. Our data show region-and disease-specific changes in the levels of lipid species. Specifically, our data revealed alterations in the levels of specific phosphatidylserine species in brain areas most affected in Parkinson's disease. Some of these alterations, albeit to a lesser degree, are also observed multiples system atrophy. Using induced pluripotent stem cell-derived neurons, we show that αSyn contributes to regulating phosphatidylserine metabolism at MAM domains, and that αSyn dosage parallels the perturbation in phosphatidylserine levels. Our results support the notion that αSyn pathophysiology is linked to the dysregulation of lipid homeostasis, which may contribute to the vulnerability of specific brain regions in synucleinopathy. These findings have significant therapeutic implications.
Collapse
Affiliation(s)
- Peter A. Barbuti
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445, Luxembourg
| | - Cristina Guardia-Laguarta
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Taekyung Yun
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, 28040, Spain
| | - Zena K. Chatila
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xena Flowers
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY 10032, USA
| | - Bruno FR. Santos
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445, Luxembourg
- Disease Modelling and Screening Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg RRID:SCR_025237
| | - Simone B. Larsen
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Elizabeth Bradshaw
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY 10032, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Manon Vilas
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY 10032, USA
| | - Hasini Reddy
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andrew F. Teich
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445, Luxembourg
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, 28040, Spain
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - Serge Przedborski
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neuroscience, Columbia University, New York, NY 10032, USA
| |
Collapse
|
50
|
Çubuk C, Lau R, Cutillas P, Rajeeve V, John CR, Surace AEA, Hands R, Fossati-Jimack L, Lewis MJ, Pitzalis C. Phosphoproteomic profiling of early rheumatoid arthritis synovium reveals active signalling pathways and differentiates inflammatory pathotypes. Arthritis Res Ther 2024; 26:120. [PMID: 38867295 PMCID: PMC11167927 DOI: 10.1186/s13075-024-03351-4] [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: 01/10/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Kinases are intracellular signalling mediators and key to sustaining the inflammatory process in rheumatoid arthritis (RA). Oral inhibitors of Janus Kinase family (JAKs) are widely used in RA, while inhibitors of other kinase families e.g. phosphoinositide 3-kinase (PI3K) are under development. Most current biomarker platforms quantify mRNA/protein levels, but give no direct information on whether proteins are active/inactive. Phosphoproteome analysis has the potential to measure specific enzyme activation status at tissue level. METHODS We validated the feasibility of phosphoproteome and total proteome analysis on 8 pre-treatment synovial biopsies from treatment-naive RA patients using label-free mass spectrometry, to identify active cell signalling pathways in synovial tissue which might explain failure to respond to RA therapeutics. RESULTS Differential expression analysis and functional enrichment revealed clear separation of phosphoproteome and proteome profiles between lymphoid and myeloid RA pathotypes. Abundance of specific phosphosites was associated with the degree of inflammatory state. The lymphoid pathotype was enriched with lymphoproliferative signalling phosphosites, including Mammalian Target Of Rapamycin (MTOR) signalling, whereas the myeloid pathotype was associated with Mitogen-Activated Protein Kinase (MAPK) and CDK mediated signalling. This analysis also highlighted novel kinases not previously linked to RA, such as Protein Kinase, DNA-Activated, Catalytic Subunit (PRKDC) in the myeloid pathotype. Several phosphosites correlated with clinical features, such as Disease-Activity-Score (DAS)-28, suggesting that phosphosite analysis has potential for identifying novel biomarkers at tissue-level of disease severity and prognosis. CONCLUSIONS Specific phosphoproteome/proteome signatures delineate RA pathotypes and may have clinical utility for stratifying patients for personalised medicine in RA.
Collapse
Affiliation(s)
- Cankut Çubuk
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Rachel Lau
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Pedro Cutillas
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Vinothini Rajeeve
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Christopher R John
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Anna E A Surace
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Rebecca Hands
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Liliane Fossati-Jimack
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK.
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust, Charterhouse Square, London, EC1M 6BQ, UK.
- IRCCS Istituto Clinico Humanitas, Via Manzoni 56, Rozzao, Milan, Italy.
| |
Collapse
|