1
|
Briem O, Tahin B, Frank AM, Olsson L, Gerdtsson AS, Källberg E, Leandersson K. Altered immune signatures in breast cancer lymph nodes with metastases revealed by spatial proteome analyses. J Transl Med 2025; 23:422. [PMID: 40211433 PMCID: PMC11987258 DOI: 10.1186/s12967-025-06415-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/24/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Metastasis to lymph nodes is strongly associated with reduced survival in breast cancer patients. To increase the understanding on how lymph node metastasis impairs the local immune response in affected lymph nodes, we here studied spatial proteomic changes of critical lymph node immune populations in uninvolved lymph nodes (UnLN) and paired lymph nodes with metastases (LNM) from five breast cancer patients. METHODS The proteome was analyzed for cortical lymphocyte compartments, subcapsular sinus (SCS) and medullary sinus (MS) CD169+ macrophages, using the Digital Spatial Profiling (DSP) platform from NanoString. RESULTS Our results identified a stable proteome of SCS CD169+ macrophages in LNM, with the exception for downregulation of the anti-apoptotic protein Bcl-xL and FAPα, but a clear reduction in numbers of SCS CD169+ macrophages in LNM. In contrast, the proteome of MS CD169+ macrophages, B-cell compartments and interfollicular T-cells showed altered immune signatures in LNM, indicating that the decline in SCS CD169+ macrophages coincide with a malfunction in the local, anti-tumor immune responses. CONCLUSIONS The findings from our study support the notion that metastasis to lymph nodes in breast cancer patients modifies local immune responses. These changes may contribute to explain unsuccessful therapeutic responses, and thereby worsened prognosis, for breast cancer patients with LNM.
Collapse
Affiliation(s)
- Oscar Briem
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Jan Waldenströms gata 35, Malmö, SE-214 28, Sweden
| | - Balázs Tahin
- Division of Clinical Pathology, Department of Clinical Sciences, Lund University, Malmö, 214 28, Sweden
| | - Asger Meldgaard Frank
- Division of Immunotechnology, Faculty of Engineering, Lund University, Malmö, 211 00, Sweden
| | - Lina Olsson
- Division of Immunotechnology, Faculty of Engineering, Lund University, Malmö, 211 00, Sweden
| | | | - Eva Källberg
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Jan Waldenströms gata 35, Malmö, SE-214 28, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Jan Waldenströms gata 35, Malmö, SE-214 28, Sweden.
| |
Collapse
|
2
|
Mahyari E, Boggy GJ, McElfresh GW, Kaza M, Benjamin S, Varco-Merth B, Ojha S, Feltham S, Goodwin W, Nkoy C, Duell D, Selseth A, Bennett T, Barber-Axthelm A, Smedley JV, Labriola CS, Axthelm MK, Reeves RK, Okoye AA, Hansen SG, Picker LJ, Bimber BN. Enhanced interpretation of immune cell phenotype and function through a rhesus macaque single-cell atlas. CELL GENOMICS 2025:100849. [PMID: 40233746 DOI: 10.1016/j.xgen.2025.100849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
Single-cell RNA sequencing (scRNA-seq) allows cell classification using genome-wide transcriptional state; however, high-dimensional transcriptomic profiles, and the unsupervised analyses employed to interpret them, provide a systematically different view of biology than well-established functional/lineage definitions of immunocytes. Understanding these differences and limits is essential for accurate interpretation of these rich data. We present the Rhesus Immune Reference Atlas (RIRA), the first immune-focused macaque single-cell multi-tissue atlas. We contrasted transcriptional profiles against immune lineages, using surface protein and marker genes as ground truth. While the pattern of clustering can align with cell type, this is not always true. Especially within T and natural killer (NK) cells, many functionally distinct subsets lack defining markers, and strong shared expression programs, such as cytotoxicity, result in systematic intermingling by unsupervised clustering. We identified gene programs with high discriminatory/diagnostic value, including multi-gene signatures that model T/NK cell maturation. Directly measuring these diagnostic programs complements unsupervised analyses.
Collapse
Affiliation(s)
- Eisa Mahyari
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Gregory J Boggy
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - G W McElfresh
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Maanasa Kaza
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Sebastian Benjamin
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Benjamin Varco-Merth
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Sohita Ojha
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Shana Feltham
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - William Goodwin
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Candice Nkoy
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Derick Duell
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Andrea Selseth
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Tyler Bennett
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Jeremy V Smedley
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Caralyn S Labriola
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - R Keith Reeves
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, NC, USA; Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Afam A Okoye
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Scott G Hansen
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Louis J Picker
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Benjamin N Bimber
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA; Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
3
|
Venturini S, Crapis M, Zanus-Fortes A, Orso D, Cugini F, Fabro GD, Bramuzzo I, Callegari A, Pellis T, Sagnelli V, Marangone A, Pontoni E, Arcidiacono D, De Santi L, Ziraldo B, Valentini G, Santin V, Reffo I, Doretto P, Pratesi C, Pivetta E, Vattamattahil K, De Rosa R, Avolio M, Tedeschi R, Basaglia G, Bove T, Tascini C. Can nCD64 and mCD169 biomarkers improve the diagnosis of viral and bacterial respiratory syndromes in the emergency department? A prospective cohort pilot study. Infection 2025; 53:679-691. [PMID: 39821738 DOI: 10.1007/s15010-024-02468-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
PURPOSE Differentiating infectious from non-infectious respiratory syndromes is critical in emergency settings. This study aimed to assess whether nCD64 and mCD169 exhibit specific distributions in patients with respiratory infections (viral, bacterial, or co-infections) and to evaluate their diagnostic accuracy compared to non-infectious conditions. METHODS A prospective cohort study enrolled 443 consecutive emergency department patients with respiratory syndromes, categorized into four groups: no infection group (NOIG), bacterial infection group (BIG), viral infection group (VIG), and co-infection group (COING). Multinomial logistic regression was used to evaluate nCD64 and mCD169's association with diagnostic groups and estimate their predictive accuracy. RESULTS 290 patients were included in VIG, 53 in BIG, 46 in COING, and 54 in NOIG. nCD64 was associated with bacterial infections and co-infections (p = 2.73 × 10- 16 and p = 8.83 × 10- 11, respectively), but not viral infections. mCD169 was associated with viral infections and co-infections (p = < 2 × 10- 16 and p = 2.45 × 10- 13, respectively), but not bacterial infections. The sensitivity and specificity of nCD64 for detecting bacterial infections were 0.75 and 0.84 (AUC = 0.83), respectively, while for mCD169 they were 0.87 and 0.91 (AUC = 0.92), respectively, for diagnosing viral infections. A diagnostic algorithm incorporating fever, nasopharyngeal swabs for the main respiratory virus, C-reactive protein, procalcitonin, and mCD169 reached an accuracy of 0.79 (95% CI 0.72-0.85) in distinguishing among the different groups. CONCLUSIONS nCD64 and MCD169 seem valuable for distinguishing between bacterial and viral respiratory infections. Integrating these biomarkers into diagnostic algorithms could enhance diagnostic accuracy aiding patient management in emergency settings.
Collapse
Affiliation(s)
- Sergio Venturini
- Department of Infectious Diseases, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Massimo Crapis
- Department of Infectious Diseases, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Agnese Zanus-Fortes
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Infectious Diseases Clinic, ASUFC "Santa Maria della Misericordia" University Hospital of Udine, Udine, Italy
| | - Daniele Orso
- Department of Emergency, University Hospital of Udine, ASUFC "Santa Maria della Misericordia", Udine, Italy
| | - Francesco Cugini
- Department of Emergency Medicine, ASUFC Hospital of San Daniele, Udine, Italy
| | - Giovanni Del Fabro
- Department of Infectious Diseases, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Igor Bramuzzo
- Department of Infectious Diseases, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Astrid Callegari
- Department of Infectious Diseases, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Tommaso Pellis
- Department of Anesthesia and Intensive Care, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Vincenzo Sagnelli
- Department of Anesthesia and Intensive Care, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Anna Marangone
- Department of Anesthesia and Intensive Care, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Elisa Pontoni
- Department of Emergency Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Domenico Arcidiacono
- Department of Emergency Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Laura De Santi
- Department of Emergency Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Barbra Ziraldo
- Department of Emergency Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Giada Valentini
- Department of Emergency Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Veronica Santin
- Department of Emergency Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Ingrid Reffo
- Department of Anesthesia and Intensive Care, ASFO "Santa Maria dei Battuti" Hospital of San Vito al Tagliamento, Pordenone, Italy.
- Department of Anesthesia and Intensive Care, San Vito al Tagliamento (Pordenone), ASFO Santa Maria dei Battuti Hospital of San Vito al Tagliamento, via Savorgnano 24, Pordenone, 33078, Italy.
| | - Paolo Doretto
- Department of Laboratory Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Chiara Pratesi
- Department of Laboratory Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Eliana Pivetta
- Department of Laboratory Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Kathreena Vattamattahil
- Department of Laboratory Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Rita De Rosa
- Department of Laboratory Medicine, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Manuela Avolio
- Department of Microbiology, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Rosamaria Tedeschi
- Department of Microbiology, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Giancarlo Basaglia
- Department of Microbiology, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Tiziana Bove
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Emergency, University Hospital of Udine, ASUFC "Santa Maria della Misericordia", Udine, Italy
| | - Carlo Tascini
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Infectious Diseases Clinic, ASUFC "Santa Maria della Misericordia" University Hospital of Udine, Udine, Italy
| |
Collapse
|
4
|
Pratesi C, De Rosa R, Pivetta E, Vattamattathil K, Malipiero G, Fontana DE, Basaglia G, Doretto P. Validation of monocyte CD169 expression as a valuable rapid diagnostic marker of SARS-CoV-2 and other acute viral infections. Am J Clin Pathol 2025; 163:340-349. [PMID: 39305084 DOI: 10.1093/ajcp/aqae127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/27/2024] [Indexed: 03/11/2025] Open
Abstract
OBJECTIVES Acute infectious diseases are some of the most common reasons for receiving medical care, and analysis of the host immune response is an attractive approach for their diagnosis. The present study aimed to evaluate the potential usefulness of CD169 expression on peripheral monocytes (mCD169) as a marker of viral-associated host immune response. METHODS In a large mono-institutional cohort of 4,025 patients evaluated for SARS-CoV-2 (CoV2) and other viral infections, mCD169 analysis was performed by rapid flow cytometry assay. RESULTS Increased mCD169 values (median, 17.50; IQR, 8.40-25.72) were found in 1,631 patients with CoV2+ acute infection compared to 2,394 in CoV2- patients (median, 2.35; IQR, 2.0-3.25) (odds ratio [OR], 21.84; 95% CI ,17.53-27.21; P < .001). Among CoV2- patients, 1,484 (62.0%) were assessed for other viral infections, and viral etiology was laboratory confirmed in 428 patients (CoV2- Vir+), with RNA viruses most frequently detected (94.6%). Higher levels of mCD169 were also confirmed in CoV2- Vir+ compared to CoV2- Vir- patients (OR, 10.05; 95% CI, 7.35-13.74; P < .001). CONCLUSIONS mCD169 analysis by rapid flow cytometry assay may be a sensitive broad marker useful for the rapid triage of patients with suspected acute viral infections and could potentially be directly applied to eventual new emergent viral outbreaks.
Collapse
Affiliation(s)
- Chiara Pratesi
- Clinical Pathology Unit, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Rita De Rosa
- Clinical Pathology Unit, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Eliana Pivetta
- Clinical Pathology Unit, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Kathreena Vattamattathil
- Clinical Pathology Unit, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Giacomo Malipiero
- Clinical Pathology Unit, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Desré Ethel Fontana
- Clinical Pathology Unit, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Giancarlo Basaglia
- Department of Microbiology, Department of Microbiology, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| | - Paolo Doretto
- Clinical Pathology Unit, ASFO "Santa Maria degli Angeli" Hospital of Pordenone, Pordenone, Italy
| |
Collapse
|
5
|
Joshi JC, Joshi B, Zhang C, Banerjee S, Vellingiri V, Raghunathrao VAB, Anwar M, Rokade TP, Zhang L, Amin R, Song Y, Mehta D. RGS2 is an innate immune checkpoint for suppressing Gαq-mediated IFNγ generation and lung injury. iScience 2025; 28:111878. [PMID: 40041768 PMCID: PMC11876898 DOI: 10.1016/j.isci.2025.111878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/01/2024] [Accepted: 01/20/2025] [Indexed: 03/12/2025] Open
Abstract
Interferon gamma (IFNγ), a type II interferon, augments tissue inflammation following infections, leading to lethal acute lung injury (ALI), yet the mechanisms controlling IFNγ generation in the lungs remain elusive. Here, we identified regulator of G protein signaling 2 (RGS2) as a gatekeeper of the lung's IFNγ levels during infections. Deletion of RGS2 sustained an increase in IFNγ levels in macrophages, leading to unresolvable inflammatory lung injury. This response was not seen in RGS2 null chimeric mice receiving wild-type (WT) bone marrow or the RGS2 gene in alveolar macrophages (AMs) or IFNγ-blocking antibody. RGS2 functioned by suppressing Gαq-mediated IFNγ generation and AM inflammatory signaling. Thus, the inhibition of Gαq blocked IFNγ generation in AMs and rewired AM transcriptomes from an inflammatory to a reparative phenotype in RGS2 null mice, pointing to the RGS2-Gαq axis as a potential target for suppressing inflammatory injury.
Collapse
Affiliation(s)
- Jagdish Chandra Joshi
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
- Lake Erie College of Osteopathic Medicine, School of Pharmacy, Erie, PA, USA
| | - Bhagwati Joshi
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Somenath Banerjee
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Vigneshwaran Vellingiri
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Vijay Avin Balaji Raghunathrao
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Mumtaz Anwar
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Tejas Pravin Rokade
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Lianghui Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Vascular Medicine Institute, Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Ruhul Amin
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dolly Mehta
- Department of Pharmacology and Centre for Lung and Vascular Biology, University of Illinois, College of Medicine, Chicago, IL, USA
| |
Collapse
|
6
|
Zhu Y, Yao ZC, Li S, Ma J, Wei C, Yu D, Stelzel JL, Ni BYX, Miao Y, Van Batavia K, Lu X, Lin J, Dai Y, Kong J, Shen R, Goodier KD, Liu X, Cheng L, Vuong I, Howard GP, Livingston NK, Choy J, Schneck JP, Doloff JC, Reddy SK, Hickey JW, Mao HQ. mRNA lipid nanoparticle-incorporated nanofiber-hydrogel composite generates a local immunostimulatory niche for cancer immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.633179. [PMID: 39975373 PMCID: PMC11838205 DOI: 10.1101/2025.01.27.633179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Hydrogel materials have emerged as versatile platforms for various biomedical applications. Notably, the engineered nanofiber-hydrogel composite (NHC) has proven effective in mimicking the soft tissue extracellular matrix, facilitating substantial recruitment of host immune cells and the formation of a local immunostimulatory microenvironment. Leveraging this feature, here we report an mRNA lipid nanoparticle (LNP)-incorporated NHC microgel matrix, termed LiNx, by incorporating LNPs loaded with mRNA encoding tumour antigens. Harnessing the potent transfection efficiency of LNPs in antigen-presenting cells (APCs), LiNx demonstrates remarkable immune cell recruitment, antigen expression and presentation, and cellular interaction. These attributes collectively create an immunostimulating milieu and yield a potent immune response achievable with a single dose, comparable to the conventional three-dose LNP immunization regimen. Further investigations reveal that the LiNx not only generates heightened Th1 and Th2 responses but also elicits a distinctive Type 17 T helper cell-mediated response pivotal for bolstering antitumour efficacy. Our findings elucidate the mechanism underlying LiNx's role in potentiating antigen-specific immune responses, presenting a new strategy for cancer immunotherapy.
Collapse
|
7
|
Candela ME, Addison M, Aird R, Man TY, Cartwright JA, Ashmore-Harris C, Kilpatrick AM, Starkey Lewis PJ, Drape A, Barnett M, Mitchell D, McLean C, McGowan N, Turner M, Dear JW, Forbes SJ. Cryopreserved human alternatively activated macrophages promote resolution of acetaminophen-induced liver injury in mouse. NPJ Regen Med 2025; 10:5. [PMID: 39843512 PMCID: PMC11754469 DOI: 10.1038/s41536-025-00393-3] [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: 06/24/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
Acute liver failure is a rapidly progressing, life-threatening condition most commonly caused by an overdose of acetaminophen (paracetamol). The antidote, N-acetylcysteine (NAC), has limited efficacy when liver injury is established. If acute liver damage is severe, liver failure can rapidly develop with associated high mortality rates. We have previously demonstrated that alternatively, activated macrophages are a potential therapeutic option to reverse acute liver injury in pre-clinical models. In this paper, we present data using cryopreserved human alternatively activated macrophages (hAAMs)-which represent a potential, rapidly available treatment suitable for use in the acute setting. In a mouse model of APAP-induced injury, peripherally injected cryopreserved hAAMs reduced liver necrosis, modulated inflammatory responses, and enhanced liver regeneration. hAAMs were effective even when administered after the therapeutic window for NAC. This cell therapy approach represents a potential treatment for APAP overdose when NAC is ineffective because liver injury is established.
Collapse
Affiliation(s)
- Maria Elena Candela
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | - Melisande Addison
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Rhona Aird
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Tak-Yung Man
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Jennifer A Cartwright
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- The Royal (Dick) School of Veterinary Studies and the Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Candice Ashmore-Harris
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Philip J Starkey Lewis
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Anna Drape
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Mark Barnett
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Donna Mitchell
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Colin McLean
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Neil McGowan
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
| | - Marc Turner
- Scottish National Blood Transfusion Service (SNBTS), The Jack Copland Centre, Heriot-Watt Research Park, Edinburgh, UK
- Centre for Precision Cell Therapy for the Liver, Lothian Health Board, Queens Medical Research Institute, Edinburgh, UK
| | - James W Dear
- Centre for Precision Cell Therapy for the Liver, Lothian Health Board, Queens Medical Research Institute, Edinburgh, UK
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, The Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Centre for Precision Cell Therapy for the Liver, Lothian Health Board, Queens Medical Research Institute, Edinburgh, UK
| |
Collapse
|
8
|
Matter A, Budzik K, Mehta S, Hoyt K, Dambra R, Vigil A, Ashour J, Raymond E, Clark E, Wood C. Molecular Pathology Methods to Characterize Biodistribution and Pharmacodynamics of the Oncolytic Virus VSV-GP in a Nonclinical Tumor Model. Toxicol Pathol 2025; 53:65-82. [PMID: 40013517 DOI: 10.1177/01926233241303904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Replication-competent oncolytic virus (OV) therapies are a promising new modality for cancer treatment. However, they pose unique challenges for preclinical assessment, due in part to their tumor specificity and ability to self-replicate in vivo. Understanding biodistribution, immune cell responses, and potential effects of intratumoral replication on these outcomes are important aspects of the nonclinical profile for OVs. Herein, a single intravenous dose of vesicular stomatitis virus pseudotyped with the glycoprotein of lymphocytic choriomeningitis virus (VSV-GP), or a cargo-expressing variant (VSV-GP-[cargo]), was examined in both tumor-free and CT26.CL25.IFNAR-/- syngeneic tumor-bearing mouse models. Biodistribution and immune cell responses were characterized using different molecular pathology methods, including a strand-specific in situ hybridization method to differentiate administered viral genomes from replicated or transcribed viral anti-genome RNA. We identified distinct patterns of viral biodistribution and replication across tumor and nontumor sites but no major differences in biodistribution, off-tumor cell tropism, or immune cell responses between tumor-free and tumor-bearing mouse models. Our findings characterize key cellular changes following systemic exposure to VSV-GP, provide a better understanding of a nonclinical permissive tumor model for OV assessment, and demonstrate how current molecular pathology methods can provide a bridge between traditional biodistribution and pathology readouts.
Collapse
Affiliation(s)
- Andrea Matter
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Karol Budzik
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Saurin Mehta
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Kathleen Hoyt
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Richard Dambra
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
- Drexel University, Philadelphia, Pennsylvania, USA
| | - Adam Vigil
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Joseph Ashour
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Ernest Raymond
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Elizabeth Clark
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | - Charles Wood
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| |
Collapse
|
9
|
Gao X, Feng X, Hou T, Huang W, Ma Z, Zhang D. The roles of flavonoids in the treatment of inflammatory bowel disease and extraintestinal manifestations: A review. FOOD BIOSCI 2024; 62:105431. [DOI: 10.1016/j.fbio.2024.105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Ahmad S, Nasser W, Ahmad A. Epigenetic mechanisms of alveolar macrophage activation in chemical-induced acute lung injury. Front Immunol 2024; 15:1488913. [PMID: 39582870 PMCID: PMC11581858 DOI: 10.3389/fimmu.2024.1488913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/15/2024] [Indexed: 11/26/2024] Open
Abstract
Airways, alveoli and the pulmonary tissues are the most vulnerable to the external environment including occasional deliberate or accidental exposure to highly toxic chemical gases. However, there are many effective protective mechanisms that maintain the integrity of the pulmonary tissues and preserve lung function. Alveolar macrophages form the first line of defense against any pathogen or chemical/reactant that crosses the airway mucociliary barrier and reaches the alveolar region. Resident alveolar macrophages are activated or circulating monocytes infiltrate the airspace to contribute towards inflammatory or reparative responses. Studies on response of alveolar macrophages to noxious stimuli are rapidly emerging and alveolar macrophage are also being sought as therapeutic target. Here such studies have been reviewed and put together for a better understanding of the role pulmonary macrophages in general and alveolar macrophage in particular play in the pathogenesis of disease caused by chemical induced acute lung injury.
Collapse
Affiliation(s)
- Shama Ahmad
- Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | |
Collapse
|
11
|
Gryziak M, Kraj L, Stec R. The role of tumor-associated macrophages in hepatocellular carcinoma-from bench to bedside: A review. J Gastroenterol Hepatol 2024; 39:1489-1499. [PMID: 38651642 DOI: 10.1111/jgh.16564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024]
Abstract
Hepatocellular carcinoma is one of the most common cancers worldwide. Despite progress in treatment, recurrence after radical treatment is common, and the prognosis remains poor for patients with advanced disease. Therefore, there is a need to identify prognostic and predictive factors for the response to therapy or more intensive surveillance or treatment. Because the tumor microenvironment plays a crucial role in the development of cancer and metastasis, it is a crucial need to understand processes that are involved in carcinogenesis. Within the microenvironment, several immune cells with different roles are present. One of the most important of these is tumor-associated macrophages. These cells may exert either antitumor or protumor roles. Several studies have suggested that tumor-associated macrophages can be used as prognostic markers. Furthermore, they may be involved in resistance to immunotherapy or systemic treatment. As they play an important role in cancer development, tumor-associated macrophages are also a good target for therapy. In this review, we briefly summarize recent progress on knowledge regarding the basic molecular characteristics, impact on prognosis and potential clinical implications of tumor-associated macrophages in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Maciej Gryziak
- Department of Oncology, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Kraj
- Department of Oncology, Medical University of Warsaw, Warsaw, Poland
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology Polish Academy of Sciences, Jastrzebiec, Poland
| | - Rafał Stec
- Department of Oncology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
12
|
Li M, Yu W, Liu Z, Liu S. CD169 + Skin Macrophages Function as a Specialized Subpopulation in Promoting Psoriasis-like Skin Disease in Mice. Int J Mol Sci 2024; 25:5705. [PMID: 38891893 PMCID: PMC11171985 DOI: 10.3390/ijms25115705] [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: 04/21/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Skin macrophages are critical to maintain and restore skin homeostasis. They serve as major producers of cytokines and chemokines in the skin, participating in diverse biological processes such as wound healing and psoriasis. The heterogeneity and functional diversity of macrophage subpopulations endow them with multifaceted roles in psoriasis development. A distinct subpopulation of skin macrophages, characterized by high expression of CD169, has been reported to exist in both mouse and human skin. However, its role in psoriasis remains unknown. Here, we report that CD169+ macrophages exhibit increased abundance in imiquimod (IMQ) induced psoriasis-like skin lesions. Specific depletion of CD169+ macrophages in CD169-ditheria toxin receptor (CD169-DTR) mice inhibits IMQ-induced psoriasis, resulting in milder symptoms, diminished proinflammatory cytokine levels and reduced proportion of Th17 cells within the skin lesions. Furthermore, transcriptomic analysis uncovers enhanced activity in CD169+ macrophages when compared with CD169- macrophages, characterized by upregulated genes that are associated with cell activation and cell metabolism. Mechanistically, CD169+ macrophages isolated from IMQ-induced skin lesions produce more proinflammatory cytokines and exhibit enhanced ability to promote Th17 cell differentiation in vitro. Collectively, our findings highlight the crucial involvement of CD169+ macrophages in psoriasis development and offer novel insights into the heterogeneity of skin macrophages in the context of psoriasis.
Collapse
Affiliation(s)
| | | | - Zhiduo Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (M.L.); (W.Y.)
| | - Siming Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (M.L.); (W.Y.)
| |
Collapse
|
13
|
Xu L, Huang C, Zheng X, Gao H, Zhang S, Zhu M, Dai X, Wang G, Wang J, Chen H, Zhu H, Chen Z. Elevated CD169 expressing monocyte/macrophage promotes systemic inflammation and disease progression in cirrhosis. Clin Exp Med 2024; 24:45. [PMID: 38413535 PMCID: PMC10899294 DOI: 10.1007/s10238-024-01305-3] [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: 11/27/2023] [Accepted: 01/27/2024] [Indexed: 02/29/2024]
Abstract
Systemic inflammation is related to disease progression and prognosis in patients with advanced cirrhosis. However, the mechanisms underlying the initiation of inflammation are still not fully understood. The role of CD169+ monocyte/macrophage in cirrhotic systemic inflammation was undetected. Flow cytometry analysis was used to detect the percentage and phenotypes of CD169+ monocytes as well as their proinflammatory function in patient-derived cirrhotic tissue and blood. Transcriptome differences between CD169+ and CD169- monocytes were also compared. Additionally, a mouse model with specific depletion of CD169+ monocytes/macrophages was utilized to define their role in liver injury and fibrosis. We observed increased CD169 expression in monocytes from cirrhotic patients, which was correlated with inflammatory cytokine production and disease progression. CD169+ monocytes simultaneously highly expressed M1- and M2-like markers and presented immune-activated profiles. We also proved that CD169+ monocytes robustly prevented neutrophil apoptosis. Depletion of CD169+ monocytes/macrophages significantly inhibited inflammation and liver necrosis in acute liver injury, but the spontaneous fibrin resolution after repeated liver injury was impaired. Our results indicate that CD169 defines a subset of inflammation-associated monocyte that correlates with disease development in patients with cirrhosis. This provides a possible therapeutic target for alleviating inflammation and improving survival in cirrhosis.
Collapse
Affiliation(s)
- Lichen Xu
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Chunhong Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Xiaoping Zheng
- Department of Pathology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, People's Republic of China
| | - Hainv Gao
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, People's Republic of China
| | - Sainan Zhang
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, People's Republic of China
| | - Mengfei Zhu
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, People's Republic of China
| | - Xiahong Dai
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, People's Republic of China
| | - Gang Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People's Republic of China
| | - Jie Wang
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Haolu Chen
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Haihong Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.
| |
Collapse
|
14
|
Woottum M, Yan S, Sayettat S, Grinberg S, Cathelin D, Bekaddour N, Herbeuval JP, Benichou S. Macrophages: Key Cellular Players in HIV Infection and Pathogenesis. Viruses 2024; 16:288. [PMID: 38400063 PMCID: PMC10893316 DOI: 10.3390/v16020288] [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: 01/22/2024] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Although cells of the myeloid lineages, including tissue macrophages and conventional dendritic cells, were rapidly recognized, in addition to CD4+ T lymphocytes, as target cells of HIV-1, their specific roles in the pathophysiology of infection were initially largely neglected. However, numerous studies performed over the past decade, both in vitro in cell culture systems and in vivo in monkey and humanized mouse animal models, led to growing evidence that macrophages play important direct and indirect roles as HIV-1 target cells and in pathogenesis. It has been recently proposed that macrophages are likely involved in all stages of HIV-1 pathogenesis, including virus transmission and dissemination, but above all, in viral persistence through the establishment, together with latently infected CD4+ T cells, of virus reservoirs in many host tissues, the major obstacle to virus eradication in people living with HIV. Infected macrophages are indeed found, very often as multinucleated giant cells expressing viral antigens, in almost all lymphoid and non-lymphoid tissues of HIV-1-infected patients, where they can probably persist for long period of time. In addition, macrophages also likely participate, directly as HIV-1 targets or indirectly as key regulators of innate immunity and inflammation, in the chronic inflammation and associated clinical disorders observed in people living with HIV, even in patients receiving effective antiretroviral therapy. The main objective of this review is therefore to summarize the recent findings, and also to revisit older data, regarding the critical functions of tissue macrophages in the pathophysiology of HIV-1 infection, both as major HIV-1-infected target cells likely found in almost all tissues, as well as regulators of innate immunity and inflammation during the different stages of HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Marie Woottum
- Institut Cochin, Inserm U1016, CNRS UMR-8104, Université Paris Cité, 75014 Paris, France; (M.W.); (S.Y.); (S.S.)
| | - Sen Yan
- Institut Cochin, Inserm U1016, CNRS UMR-8104, Université Paris Cité, 75014 Paris, France; (M.W.); (S.Y.); (S.S.)
| | - Sophie Sayettat
- Institut Cochin, Inserm U1016, CNRS UMR-8104, Université Paris Cité, 75014 Paris, France; (M.W.); (S.Y.); (S.S.)
| | - Séverine Grinberg
- CNRS UMR-8601, Université Paris Cité, 75006 Paris, France; (S.G.); (D.C.); (N.B.); (J.-P.H.)
| | - Dominique Cathelin
- CNRS UMR-8601, Université Paris Cité, 75006 Paris, France; (S.G.); (D.C.); (N.B.); (J.-P.H.)
| | - Nassima Bekaddour
- CNRS UMR-8601, Université Paris Cité, 75006 Paris, France; (S.G.); (D.C.); (N.B.); (J.-P.H.)
| | - Jean-Philippe Herbeuval
- CNRS UMR-8601, Université Paris Cité, 75006 Paris, France; (S.G.); (D.C.); (N.B.); (J.-P.H.)
| | - Serge Benichou
- Institut Cochin, Inserm U1016, CNRS UMR-8104, Université Paris Cité, 75014 Paris, France; (M.W.); (S.Y.); (S.S.)
| |
Collapse
|
15
|
Shen W, Wang C, Jiang J, He Y, Liang Q, Hu K. Targeted delivery of herpes simplex virus glycoprotein D to CD169 + macrophages using ganglioside liposomes alleviates herpes simplex keratitis in mice. J Control Release 2024; 365:208-218. [PMID: 37981051 DOI: 10.1016/j.jconrel.2023.11.026] [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: 06/20/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Herpes simplex keratitis (HSK) is a common blinding corneal disease caused by herpes simplex virus type 1 (HSV-1) infection. Antiviral drugs and corticosteroids haven't shown adequate therapeutic efficacy. During the early stage of HSV-1 infection, macrophages serve as the first line of defense. In particular, CD169+ macrophages play an important role in phagocytosis and antigen presentation. Therefore, we constructed GM-gD-lip, a ganglioside GM1 liposome vaccine encapsulating HSV-1 glycoprotein D and targeting CD169+ macrophages. After subconjunctival injection of the vaccine, we evaluated the survival rate and ocular surface lesions of the HSK mice, as well as the virus levels in the tear fluid, corneas, and trigeminal ganglia. We discovered that GM-gD-lip reduced HSV-1 viral load and alleviated the clinical severity of HSK. The GM-gD-lip also increased the number of corneal infiltrating macrophages, especially CD169+ macrophages, and polarized them toward M1. Furthermore, the number of dendritic cells (DCs) and CD8+ T cells in the ocular draining lymph nodes was significantly increased. These findings demonstrated that GM-gD-lip polarized CD169+ macrophages toward M1 to eliminate the virus while cross-presenting antigens to CD8+ T cells via DCs to activate adaptive immunity, ultimately attenuating the severity of HSK. The use of GM-gD-lip as an immunotherapeutic method for the treatment of HSK has significant implications.
Collapse
Affiliation(s)
- Wenhao Shen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd,Nanjing, Jiangsu, China.
| | - Chenchen Wang
- The Eye Hospital of Wenzhou Medical University, Wenzhou Medical University, 618 Fengqi East Rd, Hangzhou, Zhejiang, China.
| | - Jiaxuan Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd,Nanjing, Jiangsu, China.
| | - Yun He
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd,Nanjing, Jiangsu, China.
| | - Qi Liang
- Department of Ophthalmology, Sir Run Run Shaw Hospital, Zhejiang University School of medicine, 3 Qingchun East Road, Hangzhou, Zhejiang, China.
| | - Kai Hu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Rd,Nanjing, Jiangsu, China.
| |
Collapse
|
16
|
Fujiwara Y, Yano H, Pan C, Shiota T, Komohara Y. Anticancer immune reaction and lymph node sinus macrophages: a review from human and animal studies. J Clin Exp Hematop 2024; 64:71-78. [PMID: 38925976 PMCID: PMC11303962 DOI: 10.3960/jslrt.24017] [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: 04/01/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 06/28/2024] Open
Abstract
Lymph nodes are secondary lymphoid organs localized throughout the body that typically appear as bean-like nodules. Numerous antigen-presenting cells, including dendritic cells and macrophages, that mediate host defense responses against pathogens, such as bacteria and viruses, reside within lymph nodes. To react to cancer cell-derived antigens in a variety of cancers, antigen-presenting cells induce cytotoxic T lymphocytes (CTLs). In relation to anticancer immune responses, macrophages in the lymph node sinus have been of particular interest because a number of studies involving both human specimens and animal models have reported that lymph node macrophages expressing CD169 play a key role in activating anticancer CTLs. Recent studies have indicated that dysfunction of lymph node macrophages potentially contributes to immune suppression in elderly patients and immunological "cold" tumors. Therefore, in anticancer therapy, the regulation of lymph node macrophages is a potentially promising approach.
Collapse
|
17
|
Xu HZ, Lin XY, Xu YX, Xue HB, Lin S, Xu TW. An emerging research: the role of hepatocellular carcinoma-derived exosomal circRNAs in the immune microenvironment. Front Immunol 2023; 14:1227150. [PMID: 37753074 PMCID: PMC10518420 DOI: 10.3389/fimmu.2023.1227150] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the most common primary malignancy of the liver, is one of the leading causes of cancer-related death and is associated with a poor prognosis. The tumor microenvironment (TME) of HCC comprises immune, immunosuppressive, and interstitial cells with hypoxic, angiogenic, metabolic reprogramming, inflammatory, and immunosuppressive features. Exosomes are nanoscale extracellular vesicles that secrete biologically active signaling molecules such as deoxyribonucleic acid (DNA), messenger ribonucleic acid (mRNA), microribonucleic acid (miRNA), proteins, and lipids. These signaling molecules act as messengers in the tumor microenvironment, especially the tumor immunosuppressive microenvironment. Exosomal circRNAs reshape the tumor microenvironment by prompting hypoxic stress response, stimulating angiogenesis, contributing to metabolic reprogramming, facilitating inflammatory changes in the HCC cells and inducing tumor immunosuppression. The exosomes secreted by HCC cells carry circRNA into immune cells, which intervene in the activation of immune cells and promote the overexpression of immune checkpoints to regulate immune response, leading tumor cells to acquire immunosuppressive properties. Furthermore, immunosuppression is the final result of a combination of TME-related factors, including hypoxia, angiogenesis, metabolic reprogramming, and inflammation changes. In conclusion, exosomal circRNA accelerates the tumor progression by adjusting the phenotype of the tumor microenvironment and ultimately forming an immunosuppressive microenvironment. HCC-derived exosomal circRNA can affect HCC cell proliferation, invasion, metastasis, and induction of chemoresistance. Therefore, this review aimed to summarize the composition and function of these exosomes, the role that HCC-derived exosomal circRNAs play in microenvironment formation, and the interactions between exosomes and immune cells. This review outlines the role of exosomal circRNAs in the malignant phenotype of HCC and provides a preliminary exploration of the clinical utility of exosomal circRNAs.
Collapse
Affiliation(s)
- Huang-Zhen Xu
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xin-Yi Lin
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yun-Xian Xu
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hui-Bin Xue
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Tian-Wen Xu
- Department of Digestive Tumor, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
18
|
Taylor X, Clark IM, Fitzgerald GJ, Oluoch H, Hole JT, DeMattos RB, Wang Y, Pan F. Amyloid-β (Aβ) immunotherapy induced microhemorrhages are associated with activated perivascular macrophages and peripheral monocyte recruitment in Alzheimer's disease mice. Mol Neurodegener 2023; 18:59. [PMID: 37649100 PMCID: PMC10469415 DOI: 10.1186/s13024-023-00649-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Amyloid-related imaging abnormalities (ARIA) have been identified as the most common and serious adverse events resulting from pathological changes in the cerebral vasculature during several recent anti-amyloid-β (Aβ) immunotherapy trials. However, the precise cellular and molecular mechanisms underlying how amyloid immunotherapy enhances cerebral amyloid angiopathy (CAA)-mediated alterations in vascular permeability and microhemorrhages are not currently understood. Interestingly, brain perivascular macrophages have been implicated in regulating CAA deposition and cerebrovascular function however, further investigations are required to understand how perivascular macrophages play a role in enhancing CAA-related vascular permeability and microhemorrhages associated with amyloid immunotherapy. METHODS In this study, we examined immune responses induced by amyloid-targeting antibodies and CAA-induced microhemorrhages using histology and gene expression analyses in Alzheimer's disease (AD) mouse models and primary culture systems. RESULTS In the present study, we demonstrate that anti-Aβ (3D6) immunotherapy leads to the formation of an antibody immune complex with vascular amyloid deposits and induces the activation of CD169+ perivascular macrophages. We show that macrophages activated by antibody mediated Fc receptor signaling have increased expression of inflammatory signaling and extracellular matrix remodeling genes such as Timp1 and MMP9 in vitro and confirm these key findings in vivo. Finally, we demonstrate enhanced vascular permeability of plasma proteins and recruitment of inflammatory monocytes around vascular amyloid deposits, which are associated with hemosiderin deposits from cerebral microhemorrhages, suggesting the multidimensional roles of activated perivascular macrophages in response to Aβ immunotherapy. CONCLUSIONS In summary, our study establishes a connection between Aβ antibodies engaged at CAA deposits, the activation of perivascular macrophages, and the upregulation of genes involved in vascular permeability. However, the implications of this phenomenon on the susceptibility to microhemorrhages remain to be fully elucidated. Further investigations are warranted to determine the precise role of CD169 + perivascular macrophages in enhancing CAA-mediated vascular permeability, extravasation of plasma proteins, and infiltration of immune cells associated with microhemorrhages.
Collapse
Affiliation(s)
- Xavier Taylor
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Isaiah M Clark
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Griffin J Fitzgerald
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Herold Oluoch
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Justin T Hole
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Ronald B DeMattos
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA.
| | - Yaming Wang
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Feng Pan
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| |
Collapse
|
19
|
Georgoulis V, Papoudou-Bai A, Makis A, Kanavaros P, Hatzimichael E. Unraveling the Immune Microenvironment in Classic Hodgkin Lymphoma: Prognostic and Therapeutic Implications. BIOLOGY 2023; 12:862. [PMID: 37372147 PMCID: PMC10294989 DOI: 10.3390/biology12060862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
Classic Hodgkin lymphoma (cHL) is a lymphoid neoplasm composed of rare neoplastic Hodgkin and Reed-Sternberg (HRS) cells surrounded by a reactive tumor microenvironment (TME) with suppressive properties against anti-tumor immunity. TME is mainly composed of T cells (CD4 helper, CD8 cytotoxic and regulatory) and tumor-associated macrophages (TAMs), but the impact of these cells on the natural course of the disease is not absolutely understood. TME contributes to the immune evasion of neoplastic HRS cells through the production of various cytokines and/or the aberrant expression of immune checkpoint molecules in ways that have not been fully understood yet. Herein, we present a comprehensive review of findings regarding the cellular components and the molecular features of the immune TME in cHL, its correlation with treatment response and prognosis, as well as the potential targeting of the TME with novel therapies. Among all cells, macrophages appear to be a most appealing target for immunomodulatory therapies, based on their functional plasticity and antitumor potency.
Collapse
Affiliation(s)
- Vasileios Georgoulis
- Department of Hematology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece;
| | - Alexandra Papoudou-Bai
- Department of Pathology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece;
| | - Alexandros Makis
- Department of Child Health, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece;
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 000 Ioannina, Greece;
| | - Eleftheria Hatzimichael
- Department of Hematology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece;
| |
Collapse
|
20
|
Nakamura M, Mochizuki C, Kuroda C, Shiohama Y, Nakamura J. Size effect of fluorescent thiol-organosilica particles on their distribution in the mouse spleen. Colloids Surf B Biointerfaces 2023; 228:113397. [PMID: 37348267 DOI: 10.1016/j.colsurfb.2023.113397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 05/22/2023] [Accepted: 06/07/2023] [Indexed: 06/24/2023]
Abstract
We investigated the distribution of intravenously administered thiol-organosilica particle (thiol-OS) in the spleen to evaluate their size effect in mice. A single administration of particles of thiol-OS containing rhodamine B (Rh) (90, 280, 340, 450, 630, 1110, 1670, and 3030 nm in diameter) was performed. After 24 h, we conducted a combination analysis using histological studies by fluorescent microscopy and quantitative inductively coupled plasma optical emission spectrometry (ICP-OES), which revealed no clear correlation between the particle size and spleen uptake of particle weight and number per tissue weight, and the injection dose. Moreover, Rh with 450 nm diameter (Rh450) showed the highest uptake, and Rh with 340 nm diameter (Rh340) showed the lowest uptake. Histologically, large fluorescent areas in the marginal zone (MZ) and red pulp (RP) of the spleen were observed for all particle sizes, but less in the follicle of white pulp. Using combination analysis using the particle weights of ICP-OES and the fluorescent area, we compared the distributions of each particle in each region. Rh450 had the largest accumulated weight in the MZ and RP. Particles larger than Rh450 showed negative correlations between their sizes and accumulated weight in the MZ and RP. Simultaneous dual administration of particles using Rhs and thiol-OS containing fluorescein (90 nm in diameter) showed the size-dependent difference in cellular distribution and intracellular localization. Immunohistochemical staining against macrophage markers, CD169, and F4/80 showed various colocalization patterns with macrophages that uptook particles, indicating differences in particle uptake in each macrophage may have novel significance.
Collapse
Affiliation(s)
- Michihiro Nakamura
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan; Core Clusters for Research Initiatives of Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan.
| | - Chihiro Mochizuki
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Chika Kuroda
- Yamaguchi University Faculty of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yasuo Shiohama
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Junna Nakamura
- Department of Organ Anatomy and Nanomedicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
21
|
Gusev E, Sarapultsev A. Atherosclerosis and Inflammation: Insights from the Theory of General Pathological Processes. Int J Mol Sci 2023; 24:ijms24097910. [PMID: 37175617 PMCID: PMC10178362 DOI: 10.3390/ijms24097910] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Recent advances have greatly improved our understanding of the molecular mechanisms behind atherosclerosis pathogenesis. However, there is still a need to systematize this data from a general pathology perspective, particularly with regard to atherogenesis patterns in the context of both canonical and non-classical inflammation types. In this review, we analyze various typical phenomena and outcomes of cellular pro-inflammatory stress in atherosclerosis, as well as the role of endothelial dysfunction in local and systemic manifestations of low-grade inflammation. We also present the features of immune mechanisms in the development of productive inflammation in stable and unstable plaques, along with their similarities and differences compared to canonical inflammation. There are numerous factors that act as inducers of the inflammatory process in atherosclerosis, including vascular endothelium aging, metabolic dysfunctions, autoimmune, and in some cases, infectious damage factors. Life-critical complications of atherosclerosis, such as cardiogenic shock and severe strokes, are associated with the development of acute systemic hyperinflammation. Additionally, critical atherosclerotic ischemia of the lower extremities induces paracoagulation and the development of chronic systemic inflammation. Conversely, sepsis, other critical conditions, and severe systemic chronic diseases contribute to atherogenesis. In summary, atherosclerosis can be characterized as an independent form of inflammation, sharing similarities but also having fundamental differences from low-grade inflammation and various variants of canonical inflammation (classic vasculitis).
Collapse
Affiliation(s)
- Evgenii Gusev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| |
Collapse
|
22
|
CD169 + Macrophages in Primary Breast Tumors Associate with Tertiary Lymphoid Structures, T regs and a Worse Prognosis for Patients with Advanced Breast Cancer. Cancers (Basel) 2023; 15:cancers15041262. [PMID: 36831605 PMCID: PMC9954705 DOI: 10.3390/cancers15041262] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
The presence of CD169+ macrophages in the draining lymph nodes of cancer patients is, for unknown reasons, associated with a beneficial prognosis. We here investigated the prognostic impact of tumor-infiltrating CD169+ macrophages in primary tumors (PTs) and their spatial relation to tumor-infiltrating B and T cells. Using two breast cancer patient cohorts, we show that CD169+ macrophages were spatially associated with the presence of B and T cell tertiary lymphoid-like structures (TLLSs) in both PTs and lymph node metastases (LNMs). While co-infiltration of CD169+/TLLS in PTs correlated with a worse prognosis, the opposite was found when present in LNMs. RNA sequencing of breast tumors further confirmed that SIGLEC1 (CD169) expression was associated with mature tertiary lymphoid structure (TLS), and Treg and Breg signatures. We propose that the negative prognostic value related to CD169+ macrophages in PTs is a consequence of an immunosuppressive tumor environment rich in TLSs, Tregs and Bregs.
Collapse
|
23
|
Preglej T, Brinkmann M, Steiner G, Aletaha D, Göschl L, Bonelli M. Advanced immunophenotyping: A powerful tool for immune profiling, drug screening, and a personalized treatment approach. Front Immunol 2023; 14:1096096. [PMID: 37033944 PMCID: PMC10080106 DOI: 10.3389/fimmu.2023.1096096] [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: 11/11/2022] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Various autoimmune diseases are characterized by distinct cell subset distributions and activation profiles of peripheral blood mononuclear cells (PBMCs). PBMCs can therefore serve as an ideal biomarker material, which is easily accessible and allows for screening of multiple cell types. A detailed understanding of the immune landscape is critical for the diagnosis of patients with autoimmune diseases, as well as for a personalized treatment approach. In our study, we investigate the potential of multi-parameter spectral flow cytometry for the identification of patients suffering from autoimmune diseases and its power as an evaluation tool for in vitro drug screening approaches (advanced immunophenotyping). We designed a combination of two 22-color immunophenotyping panels for profiling cell subset distribution and cell activation. Downstream bioinformatics analyses included percentages of individual cell populations and median fluorescent intensity of defined markers which were then visualized as heatmaps and in dimensionality reduction approaches. In vitro testing of epigenetic immunomodulatory drugs revealed an altered activation status upon treatment, which supports the use of spectral flow cytometry as a high-throughput drug screening tool. Advanced immunophenotyping might support the exploration of novel therapeutic drugs and contribute to future personalized treatment approaches in autoimmune diseases and beyond.
Collapse
Affiliation(s)
| | | | | | | | - Lisa Göschl
- *Correspondence: Lisa Göschl, ; Michael Bonelli,
| | | |
Collapse
|
24
|
Suthen S, Lim CJ, Nguyen PHD, Dutertre CA, Lai HLH, Wasser M, Chua C, Lim TKH, Leow WQ, Loh TJ, Wan WK, Pang YH, Soon G, Cheow PC, Kam JH, Iyer S, Kow A, Tam WL, Shuen TWH, Toh HC, Dan YY, Bonney GK, Chan CY, Chung A, Goh BKP, Zhai W, Ginhoux F, Chow PKH, Albani S, Chew V. Hypoxia-driven immunosuppression by Treg and type-2 conventional dendritic cells in HCC. Hepatology 2022; 76:1329-1344. [PMID: 35184329 DOI: 10.1002/hep.32419] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/25/2022] [Accepted: 02/07/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND AIMS Hypoxia is one of the central players in shaping the immune context of the tumor microenvironment (TME). However, the complex interplay between immune cell infiltrates within the hypoxic TME of HCC remains to be elucidated. APPROACH AND RESULTS We analyzed the immune landscapes of hypoxia-low and hypoxia-high tumor regions using cytometry by time of light, immunohistochemistry, and transcriptomic analyses. The mechanisms of immunosuppression in immune subsets of interest were further explored using in vitro hypoxia assays. Regulatory T cells (Tregs) and a number of immunosuppressive myeloid subsets, including M2 macrophages and human leukocyte antigen-DR isotype (HLA-DRlo ) type 2 conventional dendritic cell (cDC2), were found to be significantly enriched in hypoxia-high tumor regions. On the other hand, the abundance of active granzyme Bhi PD-1lo CD8+ T cells in hypoxia-low tumor regions implied a relatively active immune landscape compared with hypoxia-high regions. The up-regulation of cancer-associated genes in the tumor tissues and immunosuppressive genes in the tumor-infiltrating leukocytes supported a highly pro-tumorigenic network in hypoxic HCC. Chemokine genes such as CCL20 (C-C motif chemokine ligand 20) and CXCL5 (C-X-C motif chemokine ligand 5) were associated with recruitment of both Tregs and HLA-DRlo cDC2 to hypoxia-high microenvironments. The interaction between Tregs and cDC2 under a hypoxic TME resulted in a loss of antigen-presenting HLA-DR on cDC2. CONCLUSIONS We uncovered the unique immunosuppressive landscapes and identified key immune subsets enriched in hypoxic HCC. In particular, we identified a potential Treg-mediated immunosuppression through interaction with a cDC2 subset in HCC that could be exploited for immunotherapies.
Collapse
Affiliation(s)
- Sheena Suthen
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Chun Jye Lim
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Phuong H D Nguyen
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France.,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Hannah L H Lai
- Agency for Science, Technology and Research, Genome Institute of Singapore, Singapore
| | - Martin Wasser
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Camillus Chua
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Tony K H Lim
- Duke-NUS Medical School, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wei Qiang Leow
- Duke-NUS Medical School, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Tracy Jiezhen Loh
- Duke-NUS Medical School, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wei Keat Wan
- Duke-NUS Medical School, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Yin Huei Pang
- Department of Pathology, National University Hospital, Singapore
| | - Gwyneth Soon
- Department of Pathology, National University Hospital, Singapore
| | - Peng Chung Cheow
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Juinn Huar Kam
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Shridhar Iyer
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Alfred Kow
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Wai Leong Tam
- Agency for Science, Technology and Research, Genome Institute of Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Timothy W H Shuen
- Division of Medical Oncology, National Cancer Center Singapore, Singapore
| | - Han Chong Toh
- Duke-NUS Medical School, Singapore.,Division of Medical Oncology, National Cancer Center Singapore, Singapore
| | - Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Glenn K Bonney
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Chung Yip Chan
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Alexander Chung
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Brian K P Goh
- Duke-NUS Medical School, Singapore.,Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore
| | - Weiwei Zhai
- Gustave Roussy Cancer Campus, Villejuif, France.,Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France.,Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Pierce K H Chow
- Division of Surgery and Surgical Oncology, Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Center Singapore, Singapore.,Academic Clinical Programme for Surgery, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Salvatore Albani
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| | - Valerie Chew
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, Singapore
| |
Collapse
|
25
|
Skeletal muscle provides the immunological micro-milieu for specific plasma cells in anti-synthetase syndrome-associated myositis. Acta Neuropathol 2022; 144:353-372. [PMID: 35612662 PMCID: PMC9288384 DOI: 10.1007/s00401-022-02438-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/08/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
Abstract
Anti-synthetase syndrome (ASyS)-associated myositis is a major subgroup of the idiopathic inflammatory myopathies (IIM) and is characterized by disease chronicity with musculoskeletal, dermatological and pulmonary manifestations. One of eight autoantibodies against the aminoacyl-transferase RNA synthetases (ARS) is detectable in the serum of affected patients. However, disease-specific therapeutic approaches have not yet been established.To obtain a deeper understanding of the underlying pathogenesis and to identify putative therapeutic targets, we comparatively investigated the most common forms of ASyS associated with anti-PL-7, anti-PL-12 and anti-Jo-1. Our cohort consisted of 80 ASyS patients as well as healthy controls (n = 40), diseased controls (n = 40) and non-diseased controls (n = 20). We detected a reduced extent of necrosis and regeneration in muscle biopsies from PL-12+ patients compared to Jo-1+ patients, while PL-7+ patients had higher capillary dropout in biopsies of skeletal muscle. Aside from these subtle alterations, no significant differences between ASyS subgroups were observed. Interestingly, a tissue-specific subpopulation of CD138+ plasma cells and CXCL12+/CXCL13+CD20+ B cells common to ASyS myositis were identified. These cells were localized in the endomysium associated with alkaline phosphatase+ activated mesenchymal fibroblasts and CD68+MHC-II+CD169+ macrophages. An MHC-I+ and MHC-II+ MxA negative type II interferon-driven milieu of myofiber activation, topographically restricted to the perifascicular area and the adjacent perimysium, as well as perimysial clusters of T follicular helper cells defined an extra-medullary immunological niche for plasma cells and activated B cells. Consistent with this, proteomic analyses of muscle tissues from ASyS patients demonstrated alterations in antigen processing and presentation. In-depth immunological analyses of peripheral blood supported a B-cell/plasma-cell-driven pathology with a shift towards immature B cells, an increase of B-cell-related cytokines and chemokines, and activation of the complement system. We hypothesize that a B-cell-driven pathology with the presence and persistence of a specific subtype of plasma cells in the skeletal muscle is crucially involved in the self-perpetuating chronicity of ASyS myositis. This work provides the conceptual framework for the application of plasma-cell-targeting therapies in ASyS myositis.
Collapse
|
26
|
Zhou Y, Wang S, Liang X, Heger Z, Xu M, Lu Q, Yu M, Adam V, Li N. Turning Hot into Cold: Immune Microenvironment Reshaping for Atherosclerosis Attenuation Based on pH-Responsive shSiglec-1 Delivery System. ACS NANO 2022; 16:10517-10533. [PMID: 35762565 DOI: 10.1021/acsnano.2c01778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Current atherosclerosis treatment is based on a combination of cholesterol-lowering medication and low-fat diets; however, the clinical effect is unsatisfactory. It has been shown that the level of immune cell infiltration and pro-inflammatory factors in the atherosclerotic immune microenvironment (AIM) play important roles in the development and progression of atherosclerosis. Therefore, we hypothesized that reshaping "hot AIM" into "cold AIM" could attenuate atherosclerosis. For this purpose, we designed a pH-responsive and charge-reversible nanosystem, referred to as Au-PEI/shSiglec-1/PEI-acetylsalicylic acid (ASPA NPs) to effectively deliver shSiglec-1, which blocked the interactions between macrophages with CD8+ T/NKT cells, thus inhibiting immune cell infiltration. Further, we demonstrated that acetylsalicylic acid (ASA), detached from the pH-responsive PEI-ASA polymer, and inhibited lipid accumulation in macrophage, thereby decreasing the lipid antigen presentation. Additionally, reduced macrophage-produced inflammatory factors by ASA and low CD8+ T/NKT cell infiltration levels synergistically inhibit Th17 cell differentiation, thus further dramatically attenuating inflammation in AIM by decreasing the IL-17A production. Eventually, ASPA NPs efficiently reshaped AIM by inhibiting immune cell infiltration, lipid antigen presentation, and pro-inflammation, which provided a feasible therapeutic strategy for atherosclerosis immunotherapy.
Collapse
Affiliation(s)
- Yue Zhou
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Siyu Wang
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Xiaoyang Liang
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-61200 Brno, Czech Republic
| | - Min Xu
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Qiang Lu
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Meng Yu
- School of Pharmaceutical Science Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-61200 Brno, Czech Republic
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
27
|
Takenouchi T, Masujin K, Miyazaki A, Suzuki S, Takagi M, Kokuho T, Uenishi H. Isolation and immortalization of macrophages derived from fetal porcine small intestine and their susceptibility to porcine viral pathogen infections. Front Vet Sci 2022; 9:919077. [PMID: 35923820 PMCID: PMC9339801 DOI: 10.3389/fvets.2022.919077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Macrophages are a heterogeneous population of cells that are present in all vertebrate tissues. They play a key role in the innate immune system, and thus, in vitro cultures of macrophages provide a valuable model for exploring their tissue-specific functions and interactions with pathogens. Porcine macrophage cultures are often used for the identification and characterization of porcine viral pathogens. Recently, we have developed a simple and efficient method for isolating primary macrophages from the kidneys and livers of swine. Here, we applied this protocol to fetal porcine intestinal tissues and demonstrated that porcine intestinal macrophages (PIM) can be isolated from mixed primary cultures of porcine small intestine-derived cells. Since the proliferative capacity of primary PIM is limited, we attempted to immortalize them by transferring the SV40 large T antigen and porcine telomerase reverse transcriptase genes using lentiviral vectors. Consequently, immortalized PIM (IPIM) were successfully generated and confirmed to retain various features of primary PIM. We further revealed that IPIM are susceptible to infection by the African swine fever virus and the porcine reproductive and respiratory syndrome virus and support their replication. These findings suggest that the IPIM cell line is a useful tool for developing in vitro models that mimic the intestinal mucosal microenvironments of swine, and for studying the interactions between porcine pathogens and host immune cells.
Collapse
Affiliation(s)
- Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
- *Correspondence: Takato Takenouchi
| | - Kentaro Masujin
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Kodaira, Japan
- Kentaro Masujin
| | - Ayako Miyazaki
- Division of Infectious Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Japan
- Ayako Miyazaki
| | - Shunichi Suzuki
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Michihiro Takagi
- Division of Infectious Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Takehiro Kokuho
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Kodaira, Japan
| | - Hirohide Uenishi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| |
Collapse
|
28
|
Al-kuraishy HM, Al-Gareeb AI, Al-Hamash SM, Cavalu S, El-Bouseary MM, Sonbol FI, Batiha GES. Changes in the Blood Viscosity in Patients With SARS-CoV-2 Infection. Front Med (Lausanne) 2022; 9:876017. [PMID: 35783600 PMCID: PMC9247235 DOI: 10.3389/fmed.2022.876017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/31/2022] [Indexed: 12/18/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a novel virus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2-induced hyperinflammation together with alteration of plasma proteins, erythrocyte deformability, and platelet activation, may affect blood viscosity. Thus, this review aimed to study the link between SARS-CoV-2 infection and alteration of blood viscosity in COVID-19 patients. In order to review findings related to hyperviscosity in COVID-19, we suggested a protocol for narrative review of related published COVID-19 articles. Hyperviscosity syndrome is developed in different hematological disorders including multiple myeloma, sickle cell anemia, Waldenstorm macroglobulinemia, polycythemia, and leukemia. In COVID-19, SARS-CoV-2 may affect erythrocyte morphology via binding of membrane cluster of differentiation 147 (CD147) receptors, and B and 3 proteins on the erythrocyte membrane. Variations in erythrocyte fragility and deformability with endothelial dysfunction and oxidative stress in SARS-CoV-2 infection may cause hyperviscosity syndrome in COVID-19. Of interest, hyperviscosity syndrome in COVID-19 may cause poor tissue perfusion, peripheral vascular resistance, and thrombosis. Most of the COVID-19 patients with a blood viscosity more than 3.5 cp may develop coagulation disorders. Of interest, hyperviscosity syndrome is more commonly developed in vaccine recipients who had formerly received the COVID-19 vaccine due to higher underlying immunoglobulin concentrations, and only infrequently in those who have not received the COVID-19 vaccine. Taken together, these observations are untimely too early to give a final connotation between COVID-19 vaccination and the risk for development of hyperviscosity syndrome, consequently prospective and retrospective studies are necessary in this regard.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | | | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
- *Correspondence: Maisra M. El-Bouseary,
| | - Fatma I. Sonbol
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
29
|
Flaherty S, Strauch P, Maktabi M, Pybus BS, Reichard G, Walker LA, Rochford R. Mechanisms of 8-aminoquinoline induced haemolytic toxicity in a G6PDd humanized mouse model. J Cell Mol Med 2022; 26:3675-3686. [PMID: 35665597 PMCID: PMC9258708 DOI: 10.1111/jcmm.17362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022] Open
Abstract
Primaquine (PQ) and Tafenoquine (TQ) are clinically important 8‐aminoquinolines (8‐AQ) used for radical cure treatment of P. vivax infection, known to target hepatic hypnozoites. 8‐AQs can trigger haemolytic anaemia in individuals with glucose‐6‐phosphate dehydrogenase deficiency (G6PDd), yet the mechanisms of haemolytic toxicity remain unknown. To address this issue, we used a humanized mouse model known to predict haemolytic toxicity responses in G6PDd human red blood cells (huRBCs). To evaluate the markers of eryptosis, huRBCs were isolated from mice 24–48 h post‐treatment and analysed for effects on phosphatidylserine (PS), intracellular reactive oxygen species (ROS) and autofluorescence. Urinalysis was performed to evaluate the occurrence of intravascular and extravascular haemolysis. Spleen and liver tissue harvested at 24 h and 5–7 days post‐treatment were stained for the presence of CD169+ macrophages, F4/80+ macrophages, Ter119+ mouse RBCs, glycophorin A+ huRBCs and murine reticulocytes (muRetics). G6PDd‐huRBCs from PQ/TQ treated mice showed increased markers for eryptosis as early as 24 h post‐treatment. This coincided with an early rise in levels of muRetics. Urinalysis revealed concurrent intravascular and extravascular haemolysis in response to PQ/TQ. Splenic CD169+ macrophages, present in all groups at day 1 post‐dosing were eliminated by days 5–7 in PQ/TQ treated mice only, while liver F4/80 macrophages and iron deposits increased. Collectively, our data suggest 8‐AQ treated G6PDd‐huRBCs have early physiological responses to treatment, including increased markers for eryptosis indicative of oxidative stress, resulting in extramedullary haematopoiesis and loss of splenic CD169+ macrophages, prompting the liver to act as the primary site of clearance.
Collapse
Affiliation(s)
- Siobhan Flaherty
- Department of Immunology and Microbiology, The University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Pamela Strauch
- Department of Immunology and Microbiology, The University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Mahdi Maktabi
- Department of Immunology and Microbiology, The University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Brandon S Pybus
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Gregory Reichard
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Larry A Walker
- National Center for Natural Products Research and Department of Biomolecular Sciences, School of Pharmacy, The University of Mississippi, University, Mississippi, USA
| | - Rosemary Rochford
- Department of Immunology and Microbiology, The University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
30
|
Al-Kuraishy HM, Al-Gareeb AI, El-Bouseary MM, Sonbol FI, Batiha GES. Hyperviscosity syndrome in COVID-19 and related vaccines: exploring of uncertainties. Clin Exp Med 2022:10.1007/s10238-022-00836-x. [PMID: 35608715 PMCID: PMC9128329 DOI: 10.1007/s10238-022-00836-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/26/2022] [Indexed: 12/31/2022]
Abstract
Hyperviscosity syndrome (HVS) recently emerged as a complication of coronavirus disease 2019 (COVID-19) and COVID-19 vaccines. Therefore, the objectives of this critical review are to establish the association between COVID-19 and COVID-19 vaccines with the development of HVS. HVS may develop in various viral infections due to impairment of humoral and cellular immunity with elevation of immunoglobulins. COVID-19 can increase blood viscosity (BV) through modulation of fibrinogen, albumin, lipoproteins, and red blood cell (RBC) indices. HVS can cause cardiovascular and neurological complications in COVID-19 like myocardial infarction (MI) and stroke. HVS with or without abnormal RBCs function in COVID-19 participates in the reduction of tissue oxygenation with the development of cardio-metabolic complications and long COVID-19. Besides, HVS may develop in vaccine recipients with previous COVID-19 due to higher underlying Ig concentrations and rarely without previous COVID-19. Similarly, patients with metabolic syndrome are at the highest risk for propagation of HVS after COVID-19 vaccination. In conclusion, COVID-19 and related vaccines are linked with the development of HVS, mainly in patients with previous COVID-19 and underlying metabolic derangements. The possible mechanism of HVS in COVID-19 and related vaccines is increasing levels of fibrinogen and immunoglobulins. However, dehydration, oxidative stress, and inflammatory reactions are regarded as additional contributing factors in the pathogenesis of HVS in COVID-19. However, this critical review cannot determine the final causal relationship between COVID-19 and related vaccines and the development of HVS. Prospective and retrospective studies are warranted in this field.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Maisra M El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Fatma I Sonbol
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
31
|
Zhang J, Liu Z, Cao P, Wang H, Liu H, Hua L, Xue H, Fu R. Tumor-associated macrophages regulate the function of cytotoxic T lymphocyte through PD-1/PD-L1 pathway in multiple myeloma. Cancer Med 2022; 11:4838-4848. [PMID: 35593325 PMCID: PMC9761071 DOI: 10.1002/cam4.4814] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are originated from circulating mononuclear cells in peripheral blood. They result from the recruitment of tumor cells and are a vital constituent of the tumor microenvironment. TAMs may be involved in the immunological escape of vicious clonal plasma cells (PC) in the bone marrow (BM) of sufferers with myeloma. METHODS From March 2020 to January 2021, 28 healthy controls (HC) and 86 multiple myeloma (MM) (53 newly diagnosed MM [NDMM] and 33 remissions) patients were enrolled as objects of the study. The expression of TAMs in the BM, CSF1 on CD138 + cells, and CSF1R on macrophages were detected by the method of flow cytometry, and the expression of PD-1 on CD8 + T cells and PD-L1 on TAMs were also done. Bone marrow mononuclear cells (BMMNCs) were extracted and cultured into TAMs, CD8 + T cells were sorted by magnetic beads and cultured, a coculture system was established and different inhibitors were added. The expression of the perforin and granzyme B was detected by flow cytometry. RESULTS The percentage of TAMs in NDMM group (61.49 ± 2.176%) increased when compared with remission (23.08 ± 1.699%, p < 0.001) and HC group (17.95 ± 1.865%, p < 0.001), and TAMs decreased after adding CSF1R inhibitor. Moreover, the expression of CSF1 on CD138 + cells increased significantly in NDMM group (17.090 ± 0.9156%) than remission (8.214 ± 0.5911% p < 0.001), and HC group (5.257 ± 0.6231%, p < 0.001), and CSF1R on macrophages increased significantly in NDMM group (58.78 ± 2.286%) than remission (20.74 ± 1.376%, p < 0.001) and HC group (17.42 ± 1.081%, p < 0.001). The expression of PD-1 on CD8 + T cells in NDMM group (32.64 ± 2.982%) increased than remission (20.35 ± 2.335% p < 0.01) and HC group (17.53 ± 1.349%, p < 0.001), and PD-L1 on TAMs also increased in NDMM group (50.92 ± 2.554%) than remission (20.02 ± 1.893%, p < 0.001) and HC group (13.08 ± 1.289%, p < 0.001). When CD8 + T cells were cocultured with TAMs, the perforin and granzyme B levels decreased significantly. However, the perforin and granzyme B levels were partly restored after adding CSF1R inhibitor and anti-PD-L1 antibody. CONCLUSION Our study shows that TAMs were increased in MM patients which can inhibit the function of cytotoxic T lymphocyte (CTL) through the PD-1/ PD-L1 signaling pathway and participate in the occurrence of immune escape of myeloma cells.
Collapse
Affiliation(s)
- Jiangbo Zhang
- Department of HematologyTianjin Medical University General HospitalTianjinPeople's Republic of China,Department of HematologyHebei University Affiliated HospitalBaodingPeople's Republic of China
| | - Zhaoyun Liu
- Department of HematologyTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Panpan Cao
- Department of HematologyTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Hao Wang
- Department of HematologyTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Hui Liu
- Department of HematologyTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Luoming Hua
- Department of HematologyHebei University Affiliated HospitalBaodingPeople's Republic of China
| | - Hua Xue
- Department of HematologyHebei University Affiliated HospitalBaodingPeople's Republic of China
| | - Rong Fu
- Department of HematologyTianjin Medical University General HospitalTianjinPeople's Republic of China
| |
Collapse
|
32
|
Jiang KY, Qi LL, Kang FB, Wang L. The intriguing roles of Siglec family members in the tumor microenvironment. Biomark Res 2022; 10:22. [PMID: 35418152 PMCID: PMC9008986 DOI: 10.1186/s40364-022-00369-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Sialic acid-binding receptors are expressed on the surfaces of a variety of immune cells and have complex and diverse immunoregulatory functions in health and diseases. Recent studies have shown that Siglecs could play diverse immune and nonimmune regulatory roles in the tumor microenvironment (TME) and participate in tumor progression through various mechanisms, such as regulating tumor growth and metastasis, mediating the inflammatory response, and promoting tumor immune escape, thereby affecting the prognoses and outcomes of patients. However, depending on the cell type in which they are expressed, each Siglec member binds to corresponding ligands in the microenvironment milieu to drive diverse cell physiological and pathological processes in tumors. Therefore, we herein summarize the expression spectra and functions of the Siglec family in human diseases, particularly cancer, and highlight the possibility of therapeutic interventions targeting the TME in the future.
Collapse
Affiliation(s)
- Kui-Ying Jiang
- Department of Orthopedic Oncology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Li-Li Qi
- Experimental Center for Teaching of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Fu-Biao Kang
- The Liver Disease Center of PLA, the 980Th Hospital of PLA Joint Logistics Support Force, Shijiazhuang, Hebei, People's Republic of China.
| | - Ling Wang
- Department of Orthopedic Oncology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
33
|
Minutolo A, Petrone V, Fanelli M, Iannetta M, Giudice M, Ait Belkacem I, Zordan M, Vitale P, Rasi G, Sinibaldi-Vallebona P, Sarmati L, Andreoni M, Malergue F, Balestrieri E, Grelli S, Matteucci C. High CD169 Monocyte/Lymphocyte Ratio Reflects Immunophenotype Disruption and Oxygen Need in COVID-19 Patients. Pathogens 2021; 10:1639. [PMID: 34959594 PMCID: PMC8715749 DOI: 10.3390/pathogens10121639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/28/2021] [Accepted: 12/15/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Sialoadhesin (CD169) has been found to be overexpressed in the blood of COVID-19 patients and identified as a biomarker in early disease. We analyzed CD169 in the blood cells of COVID-19 patients to assess its role as a predictive marker of disease progression and clinical outcomes. METHODS The ratio of the median fluorescence intensity of CD169 between monocytes and lymphocytes (CD169 RMFI) was analyzed by flow cytometry in blood samples of COVID-19 patients (COV) and healthy donors (HDs) and correlated with immunophenotyping, inflammatory markers, cytokine mRNA expression, pulmonary involvement, and disease progression. RESULTS CD169 RMFI was high in COV but not in HDs, and it correlated with CD8 T-cell senescence and exhaustion markers, as well as with B-cell maturation and differentiation in COV. CD169 RMFI correlated with blood cytokine mRNA levels, inflammatory markers, and pneumonia severity in patients who were untreated at sampling, and was associated with the respiratory outcome throughout hospitalization. Finally, we also report the first evidence of the specific ability of the spike protein of SARS-CoV-2 to trigger CD169 RMFI in a dose-dependent manner in parallel with IL-6 and IL-10 gene transcription in HD PBMCs stimulated in vitro. CONCLUSION CD169 is induced by the spike protein and should be considered as an early biomarker for evaluating immune dysfunction and respiratory outcomes in COVID-19 patients.
Collapse
Affiliation(s)
- Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
| | - Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
| | - Marco Iannetta
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.I.); (M.Z.); (L.S.); (M.A.)
- Infectious Diseases Clinic, Policlinic of Tor Vergata, 00133 Rome, Italy;
| | - Martina Giudice
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
| | - Ines Ait Belkacem
- CNRS, INSERM, CIML, Centre d’Immunologie de Marseille-Luminy, Aix Marseille Université, 13009 Marseille, France;
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, 13009 Marseille, France;
| | - Marta Zordan
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.I.); (M.Z.); (L.S.); (M.A.)
- Infectious Diseases Clinic, Policlinic of Tor Vergata, 00133 Rome, Italy;
| | - Pietro Vitale
- Infectious Diseases Clinic, Policlinic of Tor Vergata, 00133 Rome, Italy;
| | - Guido Rasi
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Loredana Sarmati
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.I.); (M.Z.); (L.S.); (M.A.)
- Infectious Diseases Clinic, Policlinic of Tor Vergata, 00133 Rome, Italy;
| | - Massimo Andreoni
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.I.); (M.Z.); (L.S.); (M.A.)
- Infectious Diseases Clinic, Policlinic of Tor Vergata, 00133 Rome, Italy;
| | - Fabrice Malergue
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, 13009 Marseille, France;
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
- Virology Unit, Policlinic of Tor Vergata, 00133 Rome, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.M.); (V.P.); (M.F.); (M.G.); (G.R.); (P.S.-V.); (E.B.); (S.G.)
| |
Collapse
|
34
|
Anti-Cancer Immune Reaction and Lymph Node Macrophage; A Review from Human and Animal Studies. IMMUNO 2021. [DOI: 10.3390/immuno1030014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Lymph nodes are secondary lymphoid organs that appear as bean-like nodules usually <1 cm in size, and they are localized throughout the body. Many antigen-presenting cells such as dendritic cells and macrophages reside in lymph nodes, where they mediate host defense responses against pathogens such as viruses and bacteria. In cancers, antigen-presenting cells induce cytotoxic T lymphocytes (CTLs) to react to cancer cell-derived antigens. Macrophages located in the lymph node sinus are of particular interest in relation to anti-cancer immune responses because many studies using both human specimens and animal models have suggested that lymph node macrophages expressing CD169 play a key role in activating anti-cancer CTLs. The regulation of lymph node macrophages therefore represents a potentially promising novel approach in anti-cancer therapy.
Collapse
|
35
|
Kumamoto K, Tasaki T, Ohnishi K, Shibata M, Shimajiri S, Harada M, Komohara Y, Nakayama T. CD169 Expression on Lymph Node Macrophages Predicts in Patients With Gastric Cancer. Front Oncol 2021; 11:636751. [PMID: 33816277 PMCID: PMC8017296 DOI: 10.3389/fonc.2021.636751] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/01/2021] [Indexed: 12/31/2022] Open
Abstract
The induction of an anti-cancer immune responses is potentially associated with the efficacy of anti-cancer therapy. Recent studies have indicated that sinus macrophages in regional lymph nodes are involved in anti-cancer immune responses in the cancer microenvironment. In the present study, we investigated the correlation between lymphocyte infiltration in cancer tissues and macrophage activation in regional lymph nodes. We retrospectively identified 294 patients with gastric cancer who underwent surgery from 2008 to 2012. Using immunohistochemistry, we evaluated CD169-expression on CD68-positive macrophages, and the density of CD8-postive lymphocytes in tumor microenvironment. We statistically examined the correlation between CD169 and CD8 expression, and performed Cox regression analysis of potential prognostic factors, including CD169 and CD8 expression, for cancer-specific survival (CSS) in patients with total and advanced gastric cancer. CD169 overexpression in lymph node sinus macrophages (LySMs) was positively correlated to the density of CD8-positive lymphocytes in primary cancer tissues (R = 0.367, p < 0.001). A high density of CD8-positive T lymphocytes in the primary site and a high level of CD169 expression in LySMs were independently associated with greater CSS in patients with total and advanced gastric cancer (p < 0.05 for all). The expression on CD169 in LySMs is a predictor of a favorable clinical course in patients with gastric cancer, and might be useful for evaluating anti-cancer immune responses.
Collapse
Affiliation(s)
- Keiichiro Kumamoto
- Department of Pathology, University of Environmental and Occupational Health, Fukuoka, Japan.,Third Department of Internal Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Takashi Tasaki
- Department of Pathology, University of Environmental and Occupational Health, Fukuoka, Japan.,Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Koji Ohnishi
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Michihiko Shibata
- Third Department of Internal Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Shohei Shimajiri
- Department of Pathology, University of Environmental and Occupational Health, Fukuoka, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| | - Toshiyuki Nakayama
- Department of Pathology, University of Environmental and Occupational Health, Fukuoka, Japan
| |
Collapse
|